Vaccine against RSV

Information

  • Patent Grant
  • 11801297
  • Patent Number
    11,801,297
  • Date Filed
    Friday, May 20, 2022
    a year ago
  • Date Issued
    Tuesday, October 31, 2023
    6 months ago
Abstract
The present invention relates to novel nucleic acid molecules encoding a pre-fusion RSV F protein or immunologically active part thereof, wherein the pre-fusion RSV F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2. The invention further relates to the use of the nucleic acid molecules, or vectors comprising said nucleic acid molecules, as a vaccine against respiratory syncytial virus (RSV).
Description
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY

This application contains a sequence listing, which is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file name “Sequence_Listing_004852-111US3”, creation date of May 12, 2022, and having a size of about 16 KB. The sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.


FIELD OF THE INVENTION

The invention relates to the field of medicine. More in particular, the invention relates to vaccines against RSV.


BACKGROUND OF THE INVENTION

Respiratory syncytial virus (RSV) is a highly contagious childhood pathogen of the respiratory tract which is believed to be responsible for 200,000 childhood deaths annually. In children younger than 2 years, RSV accounts for approximately 50% of the hospitalizations due to respiratory infections, with a peak of hospitalization occurring at 2-4 months of age. It has been reported that almost all children will have experienced infection with RSV by the age of two, and repeated infection during life is attributed to low natural immunity. In the elderly, the RSV disease burden is similar to those caused by non-pandemic influenza A infections. A vaccine against RSV is currently not available, but is desired due to the high disease burden.


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.


Unlike the RSV G protein, the F protein is conserved between RSV strains; which makes it an attractive vaccine candidate able to elicit broadly neutralizing antibodies. The F protein is a transmembrane protein and it is incorporated in the virion membrane from cellular membrane during virus budding. The RSV F protein facilitates infection by fusing the viral and host-cell membranes. In the process of fusion, the F protein refolds irreversibly from a labile pre-fusion conformation to a stable post-fusion conformation. The protein precursor, F0, requires cleavage during intracellular maturation by a furin-like protease. There are two furin sites, cleavage of which results in removal of a p27 peptide and formation of two domains: an N-terminal F2 domain and a C-terminal F1 domain (FIG. 1). The F2 and F1 domains are connected by two cystine bridges. Antibodies against the fusion protein can prevent virus uptake in the cell and thus have a neutralizing effect. Besides being a target for neutralizing antibodies, RSV F contains cytotoxic T cell epitopes (Pemberton et al, 1987, J. Gen. Virol. 68: 2177-2182).


Despite 50 years of research, there is still no licensed vaccine against RSV. One major obstacle to the vaccine development is the legacy of vaccine-enhanced disease in a clinical trial in the 1960s with a formalin-inactivated (FI) RSV vaccine. FI-RSV vaccinated children were not protected against natural infection and infected children experienced more severe illness than non-vaccinated children, including two deaths. This phenomenon is referred to as ‘enhanced disease’.


Since the trial with the FI-RSV vaccine, various approaches to generate an RSV vaccine have been pursued. Attempts include classical live attenuated cold passaged or temperature sensitive mutant strains of RSV, (chimeric) protein subunit vaccines, peptide vaccines and RSV proteins expressed from recombinant viral vectors, including adenoviral vectors. Although some of these vaccines showed promising pre-clinical data, no vaccine has been licensed for human use due to safety concerns or lack of efficacy.


Therefore, a need remains for efficient vaccines and methods of vaccinating against RSV, in particular vaccines that do not lead to enhanced disease. The present invention aims at providing such vaccines and methods for vaccinating against RSV in a safe and efficacious manner.


SUMMARY OF THE INVENTION

The present invention provides novel nucleic acid molecules encoding stable RSV pre-fusion F proteins, wherein the RSV pre-fusion F proteins comprise the amino acid sequence of SEQ ID NO: 1 or 2.


In certain embodiments, the nucleic acid molecules encoding the RSV pre-fusion F proteins are codon optimized for expression in human cells.


In certain embodiments, the nucleic acid molecules encoding the RSV pre-fusion F proteins comprise the nucleic acid sequence of SEQ ID NO: 3 or 4.


The invention further provides vectors comprising the nucleic acid molecules encoding RSV pre-fusion F proteins, wherein the RSV F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2.


In certain embodiments, the vector is a human recombinant adenovirus.


In certain embodiments, the recombinant adenovirus is of serotype 26 or 35.


In certain embodiments, the recombinant human adenovirus has a deletion in the E1 region, a deletion in the E3 region, or a deletion in both the E1 and the E3 region of the adenoviral genome.


In certain embodiments, the recombinant adenovirus has a genome comprising at its 5′ terminal ends the sequence CTATCTAT.


The invention also provides compositions, e.g. vaccines against respiratory syncytial virus (RSV), comprising a nucleic acid molecule or a vector according to the invention.


The invention further provides a method for vaccinating a subject against RSV, the method comprising administering to the subject a composition according to the invention.


In certain embodiments, the method of vaccinating a subject against RSV further comprises administering RSV F protein (preferably formulated as a pharmaceutical composition, thus a protein vaccine) to the subject.


The invention also provides a method for reducing infection and/or replication of RSV in, e.g. the nasal tract and lungs of, a subject, comprising administering to the subject a composition comprising a nucleic acid or vector according to the invention. This will reduce adverse effects resulting from RSV infection in a subject, and thus contribute to protection of the subject against such adverse effects upon administration of the composition. In certain embodiments, adverse effects of RSV infection may be essentially prevented, i.e. reduced to such low levels that they are not clinically relevant.


The invention also provides an isolated host cell comprising a nucleic acid molecule encoding a RSV pre-fusion F protein, wherein the RSV pre-fusion F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2. In certain embodiments, the nucleic acid molecule encoding the RSV pre-fusion F protein comprises the nucleic acid sequence of SEQ ID NO: 3 or 4.


The invention further provides a method for making a vaccine against respiratory syncytial virus (RSV), comprising providing a recombinant human adenovirus that comprises nucleic acid encoding a RSV pre-fusion F protein or fragment thereof, propagating said recombinant adenovirus in a culture of host cells, isolating and purifying the recombinant adenovirus, and formulating the recombinant adenovirus in a pharmaceutically acceptable composition, wherein the RSV pre-fusion F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2. In certain embodiments, the recombinant adenovirus is of serotype 26 or 35.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1: Schematic representation of the RSV F protein. A protein precursor includes F2 and F1 domains and p27 peptide that is removed from the mature proteins by cleavage with furin-like proteases. The cleavage sites are indicated by arrows. The numbers on top of the boxes indicate amino acid positions in the full length protein excluding signal peptide. In the F1, structural elements are shown: fusion peptide (FP), refolding region 1 (RR1) including heptad repeat A (HRA) and refolding region 2 (RR2) including heptad repeat B (HRB).



FIG. 2: Relative surface expression of F protein variants. Full length variants of F protein were expressed in HEK293T cells. The cell were stained with anti-RSV F antibody (CR9503) and analyzed by Flow Cytometry (FACS). The Mean Fluorescent Intensity (MFI) values were calculated and normalized to MFI of control F wild type (Fwt)-transfected cell sample. The MFI of Fwt was set to 1. Bars represent mean values, error bars represent range of values.



FIG. 3: Fraction of pre-fusion F protein on cell surface. Full length variants of F protein were expressed in HEK293T cells. The cell were stained with anti-RSV F antibody CR9503 and anti-pre-fusion RSV F antibody CR9501 and analyzed by Flow Cytometry. The Mean Fluorescent Intensity (MFI) values were calculated and MFI measured by CR9501 was normalized to MFI measured by CR9503. The normalized MFI values indicate fraction of the pre-F on the cell surface. Bars represent mean values, error bars represent range of values.



FIG. 4: Relative surface expression of F protein variants. Soluble versions with the transmembrane region and cytoplasmic region deleted (Fsl) and full length variants of F protein were expressed in HEK293T cells. The expression level of the soluble protein was measured in the culture supernatant by octet and the full length variants were tested by cell staining using anti-RSV F antibody (CR9503) and analyzed by Flow Cytometry. The Mean Fluorescent Intensity (MFI) values were calculated and normalized to MFI of control F wild type (Fwt)-transfected cell sample. The MFI of Fwt was set to 1. Bars represent mean values, error bars represent range of values.



FIGS. 5A and 5B: Temperature stability of the F protein variants. Full length variants of F protein were expressed in HEK293T cells. After heat-shock, the cell were stained with anti-RSV F antibody (CR9501—solid lines and CR9503—dashed lines) and analyzed by Flow Cytometry. The percentage of cells positive for the staining was determined. Symbols represent mean values, error bars represent range of values. (FIG. 5A) The percentage of cells positive for the staining was determined. (FIG. 5B) The Mean Fluorescent Intensity (MFI) values were calculated and normalized to MFI of 37 C cell sample. The MFI of 37° C. sample was set to 1. Dotted lines correspond to background staining at 60° C.



FIG. 6: Stability of the F proteins. PreF is more stable than FA2 protein in prefusion conformation on the cell surface in the heat-stress assay. A549 cells were infected with Ad26 and Ad35 comprising the insert FA2 (wt RSV F, grey bars) or prefusion F stabilized insert (preF2.2, black bars) at the indicated MOI. The cells were temperature treated at the indicated temperature for 15 minutes before staining. Top: percentage of cells presenting prefusion F on their surface (detected by CR9501 antibody); bottom: percentage of cells presenting any form of the F protein, prefusion and post fusion (detected by CR9503 antibody). The values were normalized to 37° C. samples. All bars represent a single measurement.



FIG. 7: Ad26.RSV.preF2.1 and F2.2 elicit a cellular immune response after single administration in mice. Horizontal bars depict the geometric mean of the response within a group. The background level is calculated as the 95% percentile of the spot forming units (SFU) observed in non-stimulated splenocytes, and is indicated with a dotted line.



FIGS. 8A, 8B, and 8C: Ad26.RSV.preF2.1 and F2.2 induce increased virus neutralizing antibodies when compared to Ad26.RSV.FA2, after single immunization in mice. Balb/c mice (n=4 per group) were immunized with the indicated dose of 108 to 1010 viral particles (vp) Ad26.RSV.FA2 or Ad26.RSV.preF2.1 or Ad26.RSV.preF2.2, or with formulation buffer, and humoral immune responses were assayed in the serum isolated 8 weeks after immunization. (FIG. 8A) Virus neutralizing antibodies were determined against RSV A Long using a micro-neutralization assay with an ELISA based read out. Titers are given as the log 2 value of the IC50. (FIG. 8B) Pre-fusion or post-fusion F antibody titers were determined by ELISA, and the ratio between pre- and post-fusion F antibodies for all samples that showed pre- and post-fusion F titers above lower limit of quantification (LLoQ) was calculated. (FIG. 8C) Subclass ELISA was performed using post-fusion RSV F A2 as coating reagent, and the IgG2a/IgG1 ratio (log 10) is plotted. Ratio's observed for Th1 (serum derived from animals immunized with RSV F expression Adenoviral vectors) and Th2 (serum derived from FI-RSV immunized animals) references samples are indicated with dashed lines. The LLoQ is indicated with a dotted lines (panels A), and horizontal bars represent the mean responses per group.



FIG. 9: Ad26.RSV.preF2.2 elicits antibody responses that neutralize a wide range of RSV isolates. Sera from Balb/c mice that were immunized with 1010 viral particles (vp) Ad26.RSV.FA2 (n=3) or Ad26.RSV.preF (n=4) or formulation buffer (n=2) were used in virus neutralization assays (micro-neutralization assay with an ELISA based read out) with the RSV A (upper panels) and B strains (lower panels) indicated. Titers are given as the log 2 value of the IC50, and horizontal bars represent the mean response per group. LLoQ is indicated with a dashed line.



FIGS. 10A, 10B, 10C, 10D, 10E, and 10F: Single immunization with Ad26.RSV.preF2.2 or Ad35.RSV.preF2.2 at low doses protects cotton rats against challenge with the homologous RSV A2. Cotton rats (Sigmodon hispidus) (n=7 to 9 per group) were immunized with the indicated doses (in vp/animal) of Ad26.RSV.preF2.2, Ad26.RSV.FA2 (left panels), Ad35.RSV.preF2.2 or Ad35.RSV.FA2 (right panels) by single intramuscular administration. Control immunizations were performed with formulation buffer, FI-RSV, or intranasal application of a low dose of RSV A2. At seven weeks post-immunization animals were challenged intranasally with 105 pfu RSV A2. The lung (FIGS. 10A and 10B) and nose viral titers (FIGS. 10C and 10D) were determined by plaque assay 5 days after challenge. (FIGS. 10E and 10F) Sera taken just before challenge were used to perform a virus neutralizing assay with the RSV A Long strain (micro-neutralization assay with an ELISA based read out). The dotted line represents the lower level of quantification (LLoQ). Horizontal bars represent the mean titer per group.



FIG. 11: Ad26.RSV.preF2.2 or Ad35.RSV.preF2.2 immunization of cotton rats does not result in increased alveolitis scores after RSV A2 challenge. Cotton rats (Sigmodon hispidus) (n=7 to 9 per group) were immunized with the indicated doses (in vp/animal) of Ad26.RSV.preF2.2, Ad26.RSV.FA2 (upper panel), Ad35.RSV.preF2.2, or Ad35.RSV.FA2 (lower panel) by single intramuscular administration. Control immunizations were performed with formulation buffer, FI-RSV, or intranasal application of a low dose of RSV A2. At seven weeks post-immunization animals were challenged intranasally with 105 pfu RSV A2. Alveolitis was scored by histopathological examination of one lung lobe 5 days after challenge on a non-linear scale from 0 to 4. The horizontal dotted line marks the maximal score of the control animals that were pre-exposed to RSV-A2 before challenge to mimic a natural exposure to RSV that does not lead to ERD.





DETAILED DESCRIPTION OF THE INVENTION

RSV vaccines comprising human adenovirus comprising nucleic acid encoding RSV F protein have previously been described in WO2013/139911 and WO2013/139916. It was shown therein that recombinant adenoviruses of serotype 26 or 35 that comprise a nucleic acid encoding an RSV F protein are very effective vaccines against RSV in a well established cotton rat model and have improved efficacy as compared to data described earlier for Ad5 encoding RSV F protein. It was demonstrated that a single administration, even administered intramuscularly, of Ad26 or Ad35 encoding RSV F is sufficient to provide complete protection against challenge RSV replication.


Because of the instability of the RSV F protein, however, the RSV F protein has the propensity to prematurely refold into its more stable post-fusion conformation. This phenomenon is an intrinsic feature of the protein both in solution and on the surface of the virions. In human sera most RSV neutralizing antibodies are, however, directed against the RSV F in the pre-fusion conformation. In the research that led to the present invention, effort was therefore undertaken to identify modifications that stabilize RSV F protein in its pre-fusion conformation in the full length protein.


Several possible mutations stabilizing RSV F protein in the pre-fusion conformation have previously been described in WO2014/174018 and WO2014/202570. The nucleic acid molecules according to the present invention encode RSV F proteins comprising a unique and specific subset of mutations. According to the invention it has been shown that this unique combination of mutations of the present invention results in increased RSV F protein expression levels and stability of the RSV F protein in the pre-fusion conformation. In addition, it has been shown that the nucleic acid molecules according to the invention encode RSV F protein which is stabilized in the pre-fusion conformation, and which induces higher titers of neutralizing antibody as compared to wild type RSV F protein (as disclosed in WO2013/139911 and WO2013/139916).


In a first aspect, the present invention thus provides novel nucleic acid molecules encoding a pre-fusion respiratory syncytial virus F protein (RSV pre-fusion F protein, or RSV pre-F protein), wherein the RSV pre-F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2.


In certain embodiments, the nucleic acid molecules encode the RSV pre-F proteins of SEQ ID NO: 1 or SEQ ID NO: 2.


As used herein, the terms nucleic acid, nucleic acid molecule, nucleic acid or nucleotide sequence, and polynucleotide are used interchangeably and all refer to the linear biopolymers (chains) made from nucleotides, including DNA and RNA.


It is understood by a skilled person that numerous different nucleic acid molecules can encode the same polypeptide as a result of the degeneracy of the genetic code. It is also understood that skilled persons may, using routine techniques, make nucleotide substitutions that do not affect the polypeptide sequence encoded by the polynucleotides described there to reflect the codon usage of any particular host organism in which the polypeptides are to be expressed. Therefore, unless otherwise specified, a “nucleic acid molecule encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns. Sequences herein are provided from 5′ to 3′ direction, as custom in the art.


In certain embodiments, the nucleic acid molecule encodes a fragment of pre-fusion RSV F comprising the amino acid sequence of SEQ ID NO: 1 or 2.


In certain embodiments, the nucleic acid molecules encoding the RSV pre-fusion F protein, or fragment thereof, are codon optimized for expression in mammalian cells, such as human cells. Methods of codon-optimization are known and have been described previously (e.g. WO 96/09378).


In certain embodiments, the nucleic acid molecule encoding the RSV pre-fusion F protein comprises the nucleic acid sequence of SEQ ID NO: 3. In certain embodiments, the nucleic acid encoding the RSV F protein comprises the nucleic acid sequence of SEQ ID NO: 4.


In certain embodiments, the nucleic acid encoding the RSV F protein consists of the nucleic acid sequence of SEQ ID NO: 3 or 4.


The term “fragment” as used herein refers to a peptide that has an amino-terminal and/or carboxy-terminal and/or internal deletion, but where the remaining amino acid sequence is identical to the corresponding positions in the sequence of the full length RSV F protein, for example, the RSV F protein of SEQ ID NO. 1 or 2. It will be appreciated that for inducing an immune response and in general for vaccination purposes, a protein needs not to be full length nor have all its wild type functions, and fragments of the protein are equally useful. The person skilled in the art will also appreciate that changes can be made to a protein, e.g. by amino acid substitutions, deletions, additions, etc, e.g. using routine molecular biology procedures. Generally, conservative amino acid substitutions may be applied without loss of function or immunogenicity of a polypeptide. This can easily be checked according to routine procedures well known to the skilled person.


The present invention also relates to vectors comprising a nucleic acid molecule encoding a pre-fusion RSV F protein comprising the amino acid sequence of SEQ ID NO: 1 or 2, or a fragment thereof.


According to the invention, the vector may be any vector that can be conveniently subjected to recombinant DNA procedures and can bring about expression of the nucleic acid molecule of the invention. The choice of the vector will typically depend on the compatibility of the vector with the host cell into which the vector is to be introduced.


In certain embodiments, the vector is a human recombinant adenovirus, also referred to as recombinant adenoviral vectors. The preparation of recombinant adenoviral vectors is well known in the art. The term ‘recombinant’ for an adenovirus, as used herein implicates that it has been modified by the hand of man, e.g. it has altered terminal ends actively cloned therein and/or it comprises a heterologous gene, i.e. it is not a naturally occurring wild type adenovirus.


In certain embodiments, an adenoviral vector according to the invention is deficient in at least one essential gene function of the E1 region, e.g. the Ela region and/or the E1b region, of the adenoviral genome that is required for viral replication. In certain embodiments, an adenoviral vector according to the invention is deficient in at least part of the non-essential E3 region. In certain embodiments, the vector is deficient in at least one essential gene function of the E1 region and at least part of the non-essential E3 region. The adenoviral vector can be “multiply deficient,” meaning that the adenoviral vector is deficient in one or more essential gene functions in each of two or more regions of the adenoviral genome. For example, the aforementioned E1-deficient or E1-, E3-deficient adenoviral vectors can be further deficient in at least one essential gene of the E4 region and/or at least one essential gene of the E2 region (e.g., the E2A region and/or E2B region).


Adenoviral vectors, methods for construction thereof and methods for propagating thereof, are well known in the art and are described in, for example, U.S. Pat. Nos. 5,559,099, 5,837,511, 5,846,782, 5,851,806, 5,994,106, 5,994,128, 5,965,541, 5,981,225, 6,040,174, 6,020,191, and 6,113,913, and Thomas Shenk, “Adenoviridae and their Replication”, M. S. Horwitz, “Adenoviruses”, Chapters 67 and 68, respectively, in Virology, B. N. Fields et al., eds., 3d ed., Raven Press, Ltd., New York (1996), and other references mentioned herein. Typically, construction of adenoviral vectors involves the use of standard molecular biological techniques, such as those described in, for example, Sambrook et al., Molecular Cloning, a Laboratory Manual, 2d ed., Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1989), Watson et al., Recombinant DNA, 2d ed., Scientific American Books (1992), and Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, NY (1995), and other references mentioned herein.


In certain embodiments, the adenovirus is a human adenovirus of the serotype 26 or 35. The vaccines according to the invention based on these serotypes appear more potent than the ones described in the prior art that were based on Ad5, since those failed to provide complete protection against RSV challenge replication after a single intramuscular administration (Kim et al, 2010, Vaccine 28: 3801-3808; Kohlmann et al, 2009, J Virol 83: 12601-12610; Krause et al, 2011, Virology Journal 8:375). The serotype of the invention further generally has a low seroprevalence and/or low pre-existing neutralizing antibody titers in the human population. Recombinant adenoviral vectors of these serotypes with different transgenes are evaluated in clinical trials, and thus far shows to have an excellent safety profile. Preparation of rAd26 vectors is described, for example, in WO 2007/104792 and in Abbink et al., (2007) Virol 81(9): 4654-63. Exemplary genome sequences of Ad26 are found in GenBank Accession EF 153474 and in SEQ ID NO:1 of WO 2007/104792. Preparation of rAd35 vectors is described, for example, in U.S. Pat. No. 7,270,811, in WO 00/70071, and in Vogels et al., (2003) J Virol 77(15): 8263-71. Exemplary genome sequences of Ad35 are found in GenBank Accession AC_000019 and in FIG. 6 of WO 00/70071.


A recombinant adenovirus according to the invention may be replication-competent or replication-deficient. In certain embodiments, the adenovirus is replication deficient, e.g. because it contains a deletion in the E1 region of the genome. As known to the skilled person, in case of deletions of essential regions from the adenovirus genome, the functions encoded by these regions have to be provided in trans, preferably by the producer cell, i.e. when parts or whole of E1, E2 and/or E4 regions are deleted from the adenovirus, these have to be present in the producer cell, for instance integrated in the genome thereof, or in the form of so-called helper adenovirus or helper plasmids. The adenovirus may also have a deletion in the E3 region, which is dispensable for replication, and hence such a deletion does not have to be complemented.


A producer cell (sometimes also referred to in the art and herein as ‘packaging cell’ or ‘complementing cell’ or ‘host cell’) that can be used can be any producer cell wherein a desired adenovirus can be propagated. For example, the propagation of recombinant adenovirus vectors is done in producer cells that complement deficiencies in the adenovirus. Such producer cells preferably have in their genome at least an adenovirus E1 sequence, and thereby are capable of complementing recombinant adenoviruses with a deletion in the E1 region. Any E1-complementing producer cell can be used, such as human retina cells immortalized by E1, e.g. 911 or PER.C6 cells (see U.S. Pat. No. 5,994,128), E1-transformed amniocytes (See EP patent 1230354), E1-transformed A549 cells (see e.g. WO 98/39411, U.S. Pat. No. 5,891,690), GH329:HeLa (Gao et al, 2000, Human Gene Therapy 11: 213-219), 293, and the like. In certain embodiments, the producer cells are for instance HEK293 cells, or PER.C6 cells, or 911 cells, or IT293SF cells, and the like.


For non-subgroup C E1-deficient adenoviruses such as Ad35 (subgroup B) or Ad26 (subgroup D), it is preferred to exchange the E4-orf6 coding sequence of these non-subgroup C adenoviruses with the E4-orf6 of an adenovirus of subgroup C such as Ad5. This allows propagation of such adenoviruses in well known complementing cell lines that express the E1 genes of Ad5, such as for example 293 cells or PER.C6 cells (see, e.g. Havenga et al, 2006, J. Gen. Virol. 87: 2135-2143; WO 03/104467, incorporated in its entirety by reference herein). In certain embodiments, an adenovirus that can be used is a human adenovirus of serotype 35, with a deletion in the E1 region into which the nucleic acid encoding RSV F protein antigen has been cloned, and with an E4 orf6 region of Ad5. In certain embodiments, the adenovirus in the vaccine composition of the invention is a human adenovirus of serotype 26, with a deletion in the E1 region into which the nucleic acid encoding RSV F protein antigen has been cloned, and with an E4 orf6 region of Ad5.


In alternative embodiments, there is no need to place a heterologous E4orf6 region (e.g. of Ad5) in the adenoviral vector, but instead the E1-deficient non-subgroup C vector is propagated in a cell line that expresses both E1 and a compatible E4orf6, e.g. the 293-ORF6 cell line that expresses both E1 and E4orf6 from Ad5 (see e.g. Brough et al, 1996, J Virol 70: 6497-501 describing the generation of the 293-ORF6 cells; Abrahamsen et al, 1997, J Virol 71: 8946-51 and Nan et al, 2003, Gene Therapy 10: 326-36 each describing generation of E1 deleted non-subgroup C adenoviral vectors using such a cell line).


Alternatively, a complementing cell that expresses E1 from the serotype that is to be propagated can be used (see e.g. WO 00/70071, WO 02/40665).


For subgroup B adenoviruses, such as Ad35, having a deletion in the E1 region, it is preferred to retain the 3′ end of the E1B 55K open reading frame in the adenovirus, for instance the 166 bp directly upstream of the pIX open reading frame or a fragment comprising this such as a 243 bp fragment directly upstream of the pIX start codon (marked at the 5′ end by a Bsu36I restriction site in the Ad35 genome), since this increases the stability of the adenovirus because the promoter of the pIX gene is partly residing in this area (see, e.g. Havenga et al, 2006, J. Gen. Virol. 87: 2135-2143; WO 2004/001032, incorporated by reference herein).


“Heterologous nucleic acid” (also referred to herein as ‘transgene’) in adenoviruses of the invention is nucleic acid that is not naturally present in the adenovirus. It is introduced into the adenovirus for instance by standard molecular biology techniques. In the present invention, the heterologous nucleic acid encodes the RSV pre-F protein (or fragment thereof) comprising the amino acid sequence of SEQ ID NO: 1 or 2. It can for instance be cloned into a deleted E1 or E3 region of an adenoviral vector. 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. A non-limiting example of a suitable promoter for obtaining expression in eukaryotic cells is a CMV-promoter (U.S. Pat. No. 5,385,839), e.g. the CMV immediate early promoter, for instance comprising nt. −735 to +95 from the CMV immediate early gene enhancer/promoter. A polyadenylation signal, for example the bovine growth hormone polyA signal (U.S. Pat. No. 5,122,458), may be present behind the transgene(s).


In certain embodiments, the recombinant adenovectors of the invention comprise as the 5′ terminal nucleotides the nucleotide sequence: CTATCTAT. These embodiments are advantageous because such vectors display improved replication in production processes, resulting in batches of adenovirus with improved homogeneity, as compared to vectors having the original 5′ terminal sequences (generally CATCATCA) (see also patent application nos. PCT/EP2013/054846 and U.S. Ser. No. 13/794,318, entitled ‘Batches of recombinant adenovirus with altered terminal ends’ filed on 12 Mar. 2012 in the name of Crucell Holland B.V.), incorporated in its entirety by reference herein. The invention thus also provides batches of recombinant adenovirus encoding RSV F protein or a part thereof, wherein the RSV F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2, and wherein essentially all (e.g. at least 90%) of the adenoviruses in the batch comprise a genome with terminal nucleotide sequence CTATCTAT.


In certain embodiments, the nucleic acid molecule may encode a fragment of the pre-fusion F protein of RSV. The fragment may result from either or both of amino-terminal and carboxy-terminal deletions. The extent of deletion may be determined by a person skilled in the art to, for example, achieve better yield of the recombinant adenovirus. The fragment will 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, all routine to the skilled person.


The invention furthermore provides compositions comprising a nucleic acid molecule encoding a pre-fusion RSV F comprising the amino acid sequence of SEQ ID NO: 1 or 2.


Also, the invention provides compositions comprising a vector as described herein.


In certain embodiments, the compositions comprising a nucleic acid molecule and/or a vector are for use in reducing infection and/or replication of RSV in a subject. In certain preferred embodiments, the compositions are for use as a vaccine against RSV. The term “vaccine” refers to an agent or composition containing an active component effective to induce a therapeutic degree of immunity in a subject against a certain pathogen or disease. In the present invention, the vaccine comprises an effective amount of a nucleic acid molecule that encodes an RSV pre-fusion F protein comprising SEQ ID NO: 1 or 2, or an antigenic fragment thereof, which results in an immune response against the F protein of RSV. This provides a method of preventing serious lower respiratory tract disease leading to hospitalization and the decrease the frequency of complications such as pneumonia and bronchiolitis due to RSV infection and replication in a subject. Thus, the invention also provides a method for preventing or reducing serious lower respiratory tract disease, preventing or reducing (e.g. shortening) hospitalization, and/or reducing the frequency and/or severity of pneumonia or bronchiolitis caused by RSV in a subject, comprising administering to the subject a composition comprising a nucleic acid molecule encoding a RSV pre-F protein or fragment thereof, wherein the RSV F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2. 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 ingredients. 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.


In certain embodiments the compositions comprising the nucleic acid molecule or vector further comprise, or are administered together with, one or more adjuvants. Adjuvants are known in the art to further increase the immune response to an applied antigenic determinant, and pharmaceutical compositions comprising adenovirus and suitable adjuvants are for instance disclosed in WO 2007/110409, incorporated by reference herein. 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 prefusion 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 emulsions, such as MF59 (see e.g. WO 90/14837); saponin formulations, such as for example QS21 and Immunostimulating Complexes (ISCOMS) (see e.g. U.S. Pat. No. 5,057,540; WO 90/03184, WO 96/11711, WO 2004/004762, WO 2005/002620); bacterial or microbial derivatives, 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, cholera toxin CT, and the like. It is also possible to use vector-encoded adjuvant, e.g. by using heterologous nucleic acid that encodes a fusion of the oligomerization domain of C4-binding protein (C4 bp) to the antigen of interest (e.g. Solabomi et al, 2008, Infect Immun 76: 3817-23). 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.


In other embodiments, the compositions do not comprise adjuvants.


It is also possible according to the invention to administer further active components, in combination with the compositions, e.g. vaccines, according to the invention. Such further active components may comprise e.g. other RSV antigens or vectors comprising nucleic acid encoding these. Such vectors again may be non-adenoviral or adenoviral, of which the latter can be of any serotype. An example of other RSV antigens includes RSV F or G protein or immunologically active parts thereof. For instance, intranasally applied recombinant replication-deficient Ad5 based adenovector rAd/3×G, expressing the soluble core domain of G glycoprotein (amino acids 130 to 230) was protective in a murine model (Yu et al, 2008, J Virol 82: 2350-2357), and although it was not protective when applied intramuscularly, it is clear from these data that RSV G is a suitable antigen for inducing protective responses. Further active components may also comprise non-RSV antigens, e.g. from other pathogens such as viruses, bacteria, parasites, and the like. 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. In certain embodiments, further non-adenoviral antigens (besides RSV.F), may be encoded in the vectors of the invention. In certain embodiments, it may thus be desired to express more than one protein from a single adenovirus, and in such cases more coding sequences for instance may be linked to form a single transcript from a single expression cassette or may be present in two separate expression cassettes cloned in different parts of the adenoviral genome.


The compositions of the invention, e.g. the adenovirus compositions, may be administered to a subject, e.g. a human subject. The total dose of the adenovirus provided to a subject during one administration can be varied as is known to the skilled practitioner, and is generally between 1×107 viral particles (vp) and 1×1012 vp, preferably between 1×108 vp and 1×1011 vp, for instance between 3×108 and 5×1010 vp, for instance between 109 and 3×1010 vp.


Administration of the compositions can be performed using standard routes of administration. Non-limiting embodiments include parenteral administration, such as by injection e.g. intradermal, intramuscular, etc, or subcutaneous, transcutaneous, or mucosal administration, e.g. intranasal, oral, and the like. Intranasal administration has generally been seen as a preferred route for vaccines against RSV. The most important advantage of the live intrasal strategy is the direct stimulation of local respiratory tract immunity and the lack of associated disease enhancement. The only vaccines under clinical evaluation for pediatric use at the present time are live intranasal vaccine (Collins and Murphy. Vaccines against human respiratory syncytial virus). In: Perspectives in Medical Virology 14: Respiratory Syncytial Virus (Ed. Cane, P.), Elsevier, Amsterdam, the Netherlands, pp 233-277). Intranasal administration is a suitable preferred route according to the present invention as well. The advantage of intramuscular administration is that it is simple and well-established, and does not carry the safety concerns for intranasal application in infants younger than 6 months. In one embodiment a composition is administered by intramuscular injection, e.g. into the deltoid muscle of the arm, or vastus lateralis muscle of the thigh. 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 subject can be of any age, e.g. from about 1 month to 100 years old, e.g. from about 2 months to about 80 years old, e.g. from about 1 month to about 3 years old, from about 3 years to about 50 years old, from about 50 years to about 75 years old, etc.


It is also possible to provide one or more booster administrations of one or compositions, e.g. the vaccines, of the invention. If a boosting vaccination is performed, typically, such a boosting vaccination will be administered to the same subject at a moment 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 alternative boosting regimens, it is also possible to administer different vectors, e.g. one or more adenoviruses of different serotype, or other vectors such as MVA, or DNA, or protein, to the subject after the priming vaccination. It is for instance possible to administer to the subject a recombinant adenoviral vector comprising a nucleic acid sequence encoding the pre-fusion RSV F protein as a prime, and to boost with a composition comprising a RSV F protein. In certain embodiments, the RSV F protein also is stabilized in the pre-fusion conformation.


In certain embodiments, the administration comprises a priming and at least one booster administration. In certain embodiments, the composition is administered as a priming composition and/or as a boosting composition in a prime-boost vaccination regimen. In certain embodiments thereof, the priming administration is with a rAd35 comprising nucleic acid encoding RSV pre-F protein or a fragment thereof (rAd35-RSV.pre-F′), wherein the RSV F pre-F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2, and the booster administration is with a rAd26 comprising nucleic acid encoding RSV F protein (rAd26-RSV.pre-F′), wherein the RSV pre-F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2. In other embodiments thereof, the priming administration is with rAd26-RSV.pre-F and the booster administration is with rAd35-RSV.pre-F. In other embodiments, both the priming and booster administration are with rAd26.RSV.pre-F. In certain embodiments, the priming administration is with rAd26-RSV.pre-F or rAd35-RSV.pre-F and the booster administration is with RSV F protein. In all these embodiments, it is possible to provide further booster administrations with the same or other vectors or protein. Embodiments where boosting with RSV F protein may be particularly beneficial include e.g. in elder subjects in risk groups (e.g. having COPD or asthma) of 50 years or older, or e.g. in healthy subjects of 60 years or older or 65 years or older.


In certain embodiments, the administration comprises a single administration of a composition according to the invention, without further (booster) administrations. Such embodiments are advantageous in view of the reduced complexity and costs of a single administration regimen as compared to a prime-boost regimen. Complete protection is already observed after single administration of the recombinant adenoviral vectors of the invention without booster administrations in the cotton rat model in the examples herein.


In a further aspect, the invention provides methods for making a vaccine against respiratory syncytial virus (RSV), comprising providing a recombinant human adenovirus that comprises nucleic acid encoding a RSV F protein or fragment thereof, propagating said recombinant adenovirus in a culture of host cells, isolating and purifying the recombinant adenovirus, and bringing the recombinant adenovirus in a pharmaceutically acceptable composition, wherein the RSV F protein comprises the amino acid sequence of SEQ ID NO: 1 or 2.


Recombinant adenovirus can be prepared and propagated in host cells, according to well known methods, which entail cell culture of the host cells that are infected with the adenovirus. The cell culture can be any type of cell culture, including adherent 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 (see e.g. WO 2010/060719, and WO 2011/098592, both incorporated by reference herein, which describe suitable methods for obtaining and purifying large amounts of recombinant adenoviruses).


Producer cells are cultured to increase cell and virus numbers and/or virus titers. Culturing a cell is done to enable it to metabolize, and/or grow and/or divide and/or produce virus of interest according to the invention. This can be accomplished by methods as such well known to persons skilled in the art, and includes but is not limited to providing nutrients for the cell, for instance in the appropriate culture media. Suitable culture media are well known to the skilled person and can generally be obtained from commercial sources in large quantities, 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, Academic Press, Kruse and Paterson, editors (1973), and R. I. Freshney, Culture of animal cells: A manual of basic technique, fourth edition (Wiley-Liss Inc., 2000, ISBN 0-471-34889-9).


Typically, the adenovirus will be exposed to the appropriate producer cell in a culture, permitting uptake of the virus. Usually, the optimal agitation is between about 50 and 300 rpm, typically about 100-200, e.g. about 150, typical DO is 20-60%, e.g. 40%, the optimal pH is between 6.7 and 7.7, the optimal temperature between 30 and 39° C., e.g. 34-37° C., and the optimal MOI between 5 and 1000, e.g. about 50-300. Typically, adenovirus infects producer cells spontaneously, and bringing the producer cells into contact with rAd particles is sufficient for infection of the cells. Generally, an adenovirus seed stock is added to the culture to initiate infection, and subsequently the adenovirus propagates in the producer cells. This is all routine for the person skilled in the art.


After infection of an adenovirus, the virus replicates inside the cell and is thereby amplified, a process referred to herein as propagation of adenovirus. Adenovirus infection results finally in the lysis of the cells being infected. The lytic characteristics of adenovirus therefore permits two different modes of virus production. The first mode is harvesting virus prior to cell lysis, employing external factors to lyse the cells. The second mode is harvesting virus supernatant after (almost) complete cell lysis by the produced virus (see e.g. U.S. Pat. No. 6,485,958, describing the harvesting of adenovirus without lysis of the host cells by an external factor). It is preferred to employ external factors to actively lyse the cells for harvesting the adenovirus.


Methods that can be used for active cell lysis are known to the person skilled in the art, and have for instance been discussed in WO 98/22588, p. 28-35. Useful methods in this respect are for example, freeze-thaw, solid shear, hypertonic and/or hypotonic lysis, liquid shear, sonication, high pressure extrusion, detergent lysis, combinations of the above, and the like. In one embodiment of the invention, the cells are lysed using at least one detergent. Use of a detergent for lysis has the advantage that it is an easy method, and that it is easily scalable.


Detergents that can be used, and the way they are employed, are generally known to the person skilled in the art. Several examples are for instance discussed in WO 98/22588, p. 29-33. Detergents can include anionic, cationic, zwitterionic, and nonionic detergents. The concentration of the detergent may be varied, for instance within the range of about 0.1%-5% (w/w). In one embodiment, the detergent used is Triton X-100.


Nuclease may be employed to remove contaminating, i.e. mostly from the producer cell, nucleic acids. Exemplary nucleases suitable for use in the present invention include Benzonase®, Pulmozyme®, or any other DNase and/or RNase commonly used within the art. In preferred embodiments, the nuclease is Benzonase®, which rapidly hydrolyzes nucleic acids by hydrolyzing internal phosphodiester bonds between specific nucleotides, thereby reducing the viscosity of the cell lysate. Benzonase® can be commercially obtained from Merck KGaA (code W214950). The concentration in which the nuclease is employed is preferably within the range of 1-100 units/ml. Alternatively, or in addition to nuclease treatment, it is also possible to selectively precipitate host cell DNA away from adenovirus preparations during adenovirus purification, using selective precipitating agents such as domiphen bromide (see e.g. U.S. Pat. No. 7,326,555; Goerke et al., 2005, Biotechnology and bioengineering, Vol. 91: 12-21; WO 2011/045378; WO 2011/045381).


Methods for harvesting adenovirus from cultures of producer cells have been extensively described in WO 2005/080556.


In certain embodiments, the harvested adenovirus is further purified. Purification of the adenovirus can be performed in several steps comprising clarification, ultrafiltration, diafiltration or separation with chromatography as described in for instance WO 05/080556, incorporated by reference herein. Clarification may be done by a filtration step, removing cell debris and other impurities from the cell lysate. Ultrafiltration is used to concentrate the virus solution. Diafiltration, or buffer exchange, using ultrafilters is a way for removal and exchange of salts, sugars and the like. The person skilled in the art knows how to find the optimal conditions for each purification step. Also WO 98/22588, incorporated in its entirety by reference herein, describes methods for the production and purification of adenoviral vectors. The methods comprise growing host cells, infecting the host cells with adenovirus, harvesting and lysing the host cells, concentrating the crude lysate, exchanging the buffer of the crude lysate, treating the lysate with nuclease, and further purifying the virus using chromatography.


Preferably, purification employs at least one chromatography step, as for instance discussed in WO 98/22588, p. 61-70. Many processes have been described for the further purification of adenoviruses, wherein chromatography steps are included in the process. The person skilled in the art will be aware of these processes, and can vary the exact way of employing chromatographic steps to optimize the process. It is for instance possible to purify adenoviruses by anion exchange chromatography steps, see for instance WO 2005/080556 and Konz et al, 2005, Hum Gene Ther 16: 1346-1353. Many other adenovirus purification methods have been described and are within the reach of the skilled person. Further methods for producing and purifying adenoviruses are disclosed in for example (WO 00/32754; WO 04/020971; U.S. Pat. Nos. 5,837,520; 6,261,823; WO 2006/108707; Konz et al, 2008, Methods Mol Biol 434: 13-23; Altaras et al, 2005, Adv Biochem Eng Biotechnol 99: 193-260), all incorporated by reference herein.


For administering to humans, the invention may employ pharmaceutical compositions comprising the rAd and a pharmaceutically acceptable carrier or excipient. In the present 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 edition, 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 edition, A. Kibbe, Ed., Pharmaceutical Press [2000]). The purified rAd preferably is formulated and administered as a sterile solution although it is also possible to utilize lyophilized preparations. Sterile solutions are prepared by sterile 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 rAd typically is in a solution having a suitable pharmaceutically acceptable buffer, and the solution of rAd may also contain a salt. Optionally stabilizing agent may be present, such as albumin. In certain embodiments, detergent is added. In certain embodiments, rAd may be formulated into an injectable preparation. These formulations contain effective amounts of rAd, are either sterile liquid solutions, liquid suspensions or lyophilized versions and optionally contain stabilizers or excipients. An adenovirus vaccine can also be aerosolized for intranasal administration (see e.g. WO 2009/117134).


For instance, adenovirus may be stored in the buffer that is also used for the Adenovirus World Standard (Hoganson et al, Development of a stable adenoviral vector formulation, Bioprocessing March 2002, p. 43-48): 20 mM Tris pH 8, 25 mM NaCl, 2.5% glycerol. Another useful formulation buffer suitable for administration to humans is 20 mM Tris, 2 mM MgCl2, 25 mM NaCl, sucrose 10% w/v, polysorbate-80 0.02% w/v. Obviously, many other buffers can be used, and several examples of suitable formulations for the storage and for pharmaceutical administration of purified (adeno)virus preparations can for instance be found in European patent no. 0853660, U.S. Pat. No. 6,225,289 and in international patent applications WO 99/41416, WO 99/12568, WO 00/29024, WO 01/66137, WO 03/049763, WO 03/078592, WO 03/061708.


The invention is further explained in the following examples. The examples do not limit the invention in any way. They merely serve to clarify the invention.


EXAMPLES
Example 1. Stabilizing the RSV F Protein in its Pre Fusion Conformation

Plasmids encoding basic RSV F sequences were synthesized and the amino acid substitutions were introduced in the protein by site-directed mutagenesis. The protein variants were transiently expressed in HEK293 cells. The relative protein expression on the cell surface was assessed by Flow Cytometry. The stability of the F proteins in pre-fusion conformation was evaluated in a heat-stability assay.


The protein sequence used for RSV A2 F protein variants was retrieved from the GenBank, accession number ACO83301.1. The amino acid substitutions were introduced in the sequence by site-directed mutagenesis (QuikChange II XL Site-Directed Mutagenesis Kit, Agilent technologies). The mutagenesis primers were designed using on-line tool PrimerX. HEK293T cells (CRL-11268) were purchased from American Tissue Culture Collection and cultured under standard cell culture conditions (37° C., 10% CO2).


Fully human IgG1 anti-RSV F protein antibodies CR9501 and CR9503 were constructed by cloning the heavy (VH) and light (VL) chain variable regions into a single IgG1 expression vector. PER.C6® cells (Crucell) were transfected with the IgG1 expression constructs and the expressed antibodies were purified from culture supernatants using POROS Mabcapture A chromatography (Applied Biosystems) and then buffer exchanged to 50 mM NaAc, 50 mM NaCl, pH 5.5. Antibody concentration was measured by optical absorption at 280 nm. Antibody quality was also confirmed by size-exclusion chromatography (SEC), SDS-PAGE and isoelectric focusing. The antibody CR9501 comprises VH and VL regions of 58C5 (as described in WO2011/020079) which binds specifically to RSV F protein in its pre-fusion conformation and not to the post-fusion conformation. CR9503 comprises VH and VL regions of motavizumab, which recognizes both the pre-fusion and post-fusion conformation of RSV F.


Protein Expression and Temperature Treatment:


The plasmids were transiently transfected into adherent HEK293T cells using 293fectine (Cat #12347-019) transfection reagents (Life Technologies) according to suppliers recommendations. 48 hours post transfection the cells were harvested by detaching with EDTA-containing FACS buffer (no trypsin, see next section) and cell suspension was heat-treated for 10 minutes either in a water bath or in PCR block for the temperature stability experiments. After the heat-treatment, the cells were prepared for the Flow Cytometry analysis.


For analysis of adeno expressed F proteins, A549 cells were infected with Ad26 virus at a MOI of 10 000 or 5000 and Ad35 viruses at a MOI of 5000, 2500 or 1.250. After 48h, the cells were detached and heat treated for 15 minutes at 37° C., 50° C. and 56° C. Upon heat treatment cells were stained using CR9501-Alexa647 or CR9503-Alexa647 and Propidium Iodide (PI). After staining, the cells were fixed and analyzed using the BD FACS CANTO II cell analyzer.


Flow Cytometry Analysis:


For each staining, the following controls were included: 1) negative control sample. i.e. cells that were not subjected to any treatment and not stained with any antibody labeled with a fluorophore; 2) positive control samples, i.e. cells that are stained with only one fluorophore (one of each that are used for the experiment).


The cells were resuspended in the Flow Cytometry (FC Buffer, 5 mM EDTA, 1% FBS in PBS) and distributed in volumes of 50 μl of the cell suspension per well in a 96-well plate with a lid (U- or V-bottom plates). Two-step or one-step protocols were used for staining.


In case of the two-step protocol 50 μl of the first Abs (or buffer for negative controls) was added to the wells and incubated at RT for 30 min. Biotinylated CR9501 and CR9503 were used at 2 μg/ml (final concentration in a well 1 μg/ml). After incubation, the cells were washed 2 times with the FC buffer. Afterwards 50 μl of Streptavidin-APC (Molecular Probes cat #SA1005, 0.1 mg/ml is used at 1:100) or buffer for negative controls was added to the wells and incubated at RT for 30 min. The cells were washed again 2 times with the FC buffer. After the last wash, the cells were resuspended in 100 μl of FC buffer+/−live-dead stain (PI from Invitrogen, cat #P1304MP, used at 2 μg/ml) and incubated at RT for 15 minutes. The cells were centrifuged at 200 g (1000 rpm) for 5 min., the buffer with PI was removed and the cells were resuspended in 150 μl of the FC buffer.


In case of a one-step protocol, CR9501 and CR9503 antibodies were labeled with fluorescent probe Alexa647 (Molecular Probes, cat #A-20186) according to manufacturer's instructions. Cells were stained according to the protocol above excluding the Streptavidin-APC step.


From the live cell population, the percentage of cells positive for CR9501/CR9503 antibody binding was determined. The cells positive for CR9503 binding express RSV F protein on their surface. The cells positive for CR9501 binding express pre-fusion RSV F on their surface.


The intensity of the antibody staining (Median fluorescence intensity—MFI) is proportional to the amount of F protein on the cell surface. MFI was calculated from the live cell population expressing F protein.


Results:


Surface Cell Expression of the Full Length F Protein Variants:


A subset of mutations that was previously identified to increase expression or stability of the RSV F protein ectodomain in pre-fusion conformation was introduced in the wild type full length RSV A2 F sequence (accession number Genbank ACO83301). The mutations were introduced alone or in multiple combinations, and the effect on protein expression and stability was assessed.


The expression level of the protein was measured as mean fluorescence intensity (MFI) by Flow Cytometry after staining with the CR9503 antibody that is recognizing both pre-fusion and post-fusion F protein. The combination of the two amino acid substitutions that were previously described for stabilization of the soluble RSV pre-F protein (i.e. N67I and S215P) also increased the expression level of the full length RSV F protein by 2.3-fold, relative to wild type full length RSV F (FIG. 2).


A prominent increase in expression was observed for variants with 3 amino acid substitutions combined. Interestingly, combination of more than three mutations in one variant did not further increase protein expression. This may be due to limited capacity of the cellular membrane to accommodate multiple copies of F protein.


The amount of the pre-fusion F on the surface of the cell was assessed by staining with pre-fusion specific antibody CR9501 (FIG. 3). Transfection of the cells with all F variants resulted in a more or less similar amount of pre-fusion F protein on the cell surface. Presence of the transmembrane domain stabilizes the full length protein to certain extent and therefore differences in the pre-fusion stability are not as apparent under ambient conditions between the full length F proteins. Therefore the heat-stability assay was developed to better discriminate stability of full length variants, as described below.


The A2 strain that was used as a parental sequence for the previously described F protein variants (WO2014/174018 and WO2014/202570) is a cell line adapted laboratory strain which has accumulated two unique and rare mutations (i.e. of Lysine 66 and Isoleucine 76). In the present invention, these two residues were mutated to match the natural clinical isolates (K66E, I76V). The K66E and I76V mutations were included in selected protein designs. In comparison to variants with Lys66 and Ile76, variants with glutamate at 66 (K66E) have a tendency to express slightly higher. Addition of valine at residue 76 (a double substitution of K66E and I76V) does not influence expression level when compared to variants with K66E substitution alone (FIG. 4).


Stability of the Full Length F Protein Variants on the Cell Surface:


In ambient conditions on a short time scale, no significant difference in stability of pre-fusion conformation was observed between full length F variants with the different combinations of stabilizing mutations. An elevated temperature is known to serve as an efficient in vitro trigger for refolding of RSV F protein from pre-fusion to post-fusion conformation. Therefore, a heat-shock assay was established and used to assess stability of the membrane-bound full length proteins. Shortly, the HEK293T cells were transfected with the F protein constructs and used for the assay 48 hours after transfection. The cells were detached from cell culture dishes and the cell suspension was heat-treated at increasing temperatures for 10 minutes. After the heat-treatment, cells were stained with the anti-RSV F antibodies and analyzed by Flow Cytometry. The Flow Cytometry data was analyzed in two different ways. The percentage of the cells, positive for staining with the anti-F antibodies was analyzed, and also mean fluorescence intensity (MFI) of the positive cells was calculated (FIGS. 5A and 5B).


Both staining with CR9501 (antibody recognizing only pre-fusion F protein) and CR9503 (antibody recognizing both pre- and post-fusion F protein) were used in the Flow Cytometry assays. CR9503 antibody served as a positive control. In case when F protein loses pre-fusion conformation but still is on the surface of the cell, the protein is still detected with the CR9503 antibody. Loss of staining with both antibodies indicates that protein is not available on the cell surface for antibody binding, e.g. due to aggregation.


Full length proteins with three of more amino acid substitutions were tested in the assay and compared to the wild type RSV F. The expression of these variants was the highest and therefore these variants were preferred candidates. All of the proteins contained the N67I and S215P substitutions, and one or two extra stabilizing mutations were added.


The unmodified wild type protein had a rather stable staining with CR9503 antibody. The MFI of the CR9503 staining was elevated at higher temperatures however the spread of values was also very high. This indicated that no protein aggregation occurred after the heat-shock. Half of the pre-fusion conformation was lost after incubation of cells at approximately 55° C., after incubation of at 60° C. all pre-fusion conformation was lost as was demonstrated by decreased CR9501 binding to the wild type F samples after heat-shock at increasing temperatures.


All tested pre-fusion F protein variants were more stable than the wild type RSV F with majority of the CR9501 staining retaining also after treatment at higher temperatures (FIGS. 5A, 5B, and 6). Proteins with K498R amino acid substitution were less stable than the others. Addition of the K66E mutation further stabilized the proteins as also variants with K498R amino acid substitution became as stable as others and no loss of the pre-fusion conformation was observed at 60° C. Only selected combinations of the stabilizing mutations were tested with K66E and I76V combined. All four tested proteins were stable when percentage of positive cells was analyzed, however when MFI was analyzed variant with K498R showed clear decrease in CR9501 binding after treatment with 60° C., indicating that this variant is less stable when evaluated in the temperature stress assay.


In conclusion, a combination of three stabilizing mutations (including N67I and S215P) was considered sufficient for high expression level and stability. The S46G or D486N mutations was selected as a third stabilizing mutation because of their position in the protein structure. K66E and I76V were included in the as they did not have negative effect on the protein expression and stability but made the sequence closer to naturally occurring ones.


Thus, the pre-fusion RSV F protein with the mutations K66E, N67I, I76V, S215P and D486N (F2.2) (SEQ ID NO: 2) and the pre-fusion RSV F protein with the mutations K66E, N67I, I76V, S215P and S46G (F2.1) (SEQ ID NO: 1) were selected for the construction adenoviral vectors. These proteins were shown to be stable in the pre-fusion conformation in the temperature stability assay up to 60° C., and to be expressed in high levels.


Example 2. Preparation of Adenoviral Vectors

Cloning RSV F Gene into E1 Region of Ad35 and Ad26:


The nucleic acid sequences, coding for the pre-fusion F proteins of the invention were gene optimized for human expression and synthesized, by Geneart. A Kozak sequence (5′ GCCACC 3′ (SEQ ID NO: 5)) was included directly in front of the ATG start codon, and two stop codons (5′ TGA TAA 3′ (SEQ ID NO: 6)) were added at the end of the RSV.pre-F coding sequence. The RSV.pre-F genes were inserted in the pAdApt35BSU plasmid and in the pAdApt26 plasmid via HindIII and XbaI sites.


Cell Culture:


PER.C6 cells (Fallaux et al., 1998, Hum Gene Ther 9: 1909-1917) were maintained in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum (FBS), supplemented with 10 mM MgCl2.


Adenovirus Generation, Infections and Passaging:


All adenoviruses were generated in PER.C6® cells by single homologous recombination and produced as previously described (for rAd35: Havenga et al., 2006, J Gen. Virol. 87: 2135-2143; for rAd26: Abbink et al., 2007, J. Virol. 81: 4654-4663). Briefly, PER.C6 cells were transfected with Ad vector plasmids, using Lipofectamine according to the instructions provided by the manufacturer (Life Technologies). For rescue of e.g. Ad35 vectors carrying the RSV.pre-F transgenes expression cassette, the pAdApt35BSU.RSV.pre-F plasmid and pWE/Ad35.pIX-rITR.dE3.5orf6 cosmid were used, whereas for Ad26 vectors carrying the RSV.pre-F transgene expression cassette, the pAdApt26.RSV.pre-F plasmid and pWE.Ad26.dE3.5orf6.cosmid were used. Cells were harvested one day after full CPE, freeze-thawed, centrifuged for 5 min at 3,000 rpm, and stored at −20° C. Next the viruses were plaque purified and amplified in PER.C6 cultured on a single well of a multiwell 24 tissue culture plate. Further amplification was carried out in PER.C6 cultured using a T25 tissue culture flask and a T175 tissue culture flask. Of the T175 crude lysate, 3 to 5 ml was used to inoculate 20×T175 triple-layer tissue culture flasks containing 70% confluent layers of PER.C6 cells. The virus was purified using a two-step CsCl purification method. Finally, the virus was stored in aliquots at −85° C.


Example 3. Induction of Immunity Against RSV F Using Recombinant Adenovirus Serotypes 26 and 35 Expressing Pre Fusion RSV F In Vivo

The immunogenicity of Ad26.RSV.preF2.1 and Ad26.RSV.preF.2.2 was evaluated in mice, comparing cellular and humoral immune responses to responses induced by identical doses of Ad26.RSV.FA2 (i.e. expressing the wild type RSV F protein). Balb/c mice (n=4 per group) were immunized with the indicated dose of 108 to 1010 viral particles (vp) Ad26.RSV.FA2 or Ad26.RSV.preF2.1 or Ad26.RSV.preF2.2, or with formulation buffer. At 8 weeks after prime, the number of RSV F A2 specific IFNγ spot forming units (SFU) per 106 splenocytes was determined using ELISpot. It was shown that Ad26.RSV.preF2.1 and Ad26.RSV.preF.2.2 induced increased humoral immune responses in mice when compared to Ad26.RSV.FA2, with broad neutralizing capacity and maintained cellular responses. A single intramuscular immunization with Ad26.RSV.preF2.1 and Ad26.RSV.preF.2.2 elicited a cellular response (FIG. 7) which was characterized by induction of CD8+ T cells positive for IFNγ, IL2 and/or TNFα (data not shown).


The quantity and quality of the cellular responses were comparable between Ad26.RSV.preF2.1, Ad26.RSV.preF.2.2 and Ad26.RSV.FA2. In contrast, Ad26.RSV.preF2.1 and Ad26.RSV.preF.2.2 induced significantly higher RSV neutralizing antibody titers than Ad26.RSV.FA2. Closer analysis of the antibody responses demonstrated that Ad26.RSV.preF2.1 and Ad26.RSV.preF.2.2 induced higher levels of antibodies against pre-fusion F, while post-fusion F titers remained comparable to Ad26.RSV.FA2, resulting in significantly increased preF/postF antibody ratios. In addition, the IgG2a/IgG1 ratio of the antibody response remained unaltered, demonstrating a similar Th1 skewing of the humoral response as previously demonstrated for Ad26.RSV.FA2 (FIGS. 8A, 8B, and 8C).


For Ad26.RSV.preF2.2 it was furthermore demonstrated that the antibodies elicited were capable of neutralizing various RSV A and B strains, laboratory strains as well as clinical isolates, similar as observed for Ad26.RSV.FA2 (FIG. 9).


Subsequently, the efficacy and immunogenicity of Ad26.RSV.preF2.2 and Ad35.RSV.preF2.2 vector constructs was evaluated in the cotton rat model. These animals are permissive to replication of human RSV, with peak RSV titers in the lungs at days 4 and 5. Control groups in the experiments included groups intranasally infected with a low dose RSV A2, thereby mimicking natural exposure, as well as groups immunized with FI-RSV, using the original lot 100 that induced enhanced respiratory disease (ERD) in clinical studies in the dilution that was shown to induce ERD in cotton rats.


Single intramuscular immunization of animals with Ad26.RSV.preF2.2 in doses ranging from 105 to 108 vp/animal, or Ad35.RSV.preF2.2 in doses ranging from 106 to 109 vp/animal resulted in complete protection of the lungs from infection with the vaccine homologous RSV A2 strain, except for 3 animals immunized with 105 vp Ad26.RSV.preF2.2 (FIGS. 10A and 10B). Dose dependent protection of RSV replication in the nose was observed for both vectors. This ranged from full protection at 108 vp/animal, to partial protection at 105 vp for Ad26.RSV.preF2.2, whereas for Ad35.RSV.preF2.2, noses of animals immunized with 109 vp were fully protected from RSV A2, and 106 vp resulted in partial protection (FIGS. 10C and 10D) Noses of animals immunized with Ad26.RSV.preF2.2 and Ad35.RSV.preF2.2 were better protected from RSV A2 infection than when immunized with their respective wild type F counterparts Ad26.RSV.FA2 and Ad35.RSV.FA2, when analyzed across dose (p=0.0003, and p=0.0001). Protection from RSV infection was accompanied by dose-dependent induction of virus neutralization titers against RSV A Long, already elicited by the lowest doses of Ad26.RSV.preF2.2 or Ad35.RSV.preF2.2 applied (FIGS. 10E and 10F). Across dose statistical comparisons of VNA A Long titers revealed that Ad26.RSV.preF2.2 is more immunogenic than Ad26.RSV.FA2 (p=0.0414), whereas elicitation of VNA titers was not significantly different between Ad35.RSV.preF2.2 and Ad35.RSV.FA2.


It was further demonstrated that Ad26.RSV.preF and Ad35.RSV.preF do not induce histopathological signs of Enhanced Respiratory Disease (ERD) after RSV A2 challenge, at any of the concentrations tested. The cotton rat is the most used and best studied model to monitor ERD. In this animal model, vaccination with FI-RSV consistently induces ERD after RSV challenge, which is visible by histopathological analysis of sections of the infected lungs for parameters as alveolitits, consisting primarily of neutrophil infiltrates, and peribronchiolitis, consisting primarily of lymphocyte infiltrates. In cotton rats, FI-RSV-induced scores for these parameters can be observed from day 1 after RSV infection, and peak at 4 to 5 days after RSV challenge.


ERD was analyzed 5 days after challenge with RSV A2 by scoring 4 parameters of pulmonary inflammatory changes (peribronchiolitis, perivasculitis, interstitial pneumonia, alveolitis). Immunization with FI-RSV resulted in enhanced scores for most histopathological markers, which was especially apparent for alveolitis (FIG. 11), the marker that was previously shown to be the most discriminating marker for ERD. No increases in alveolitis or any other ERD histopathological marker was observed in animals immunized by either Ad26.RSV.preF2.2 or Ad35.RSV.preF2.2 in a prime-only regimen after RSV challenge, even at low vaccine doses that may induce low affinity and/or low levels of antibodies (FIG. 11). This is confirming our previous results with Ad26.RSV.FA2 and Ad35.RSV.FA2 vectors.


According to the invention, it has thus been shown that Ad26.RSV.preF and Ad35.RSV.preF are potent adenoviral vectors expressing RSV F A2 which is stabilized in the pre-fusion conformation. These vectors induce strong humoral and cellular immune responses. The immune response elicited is protective against RSV A2 challenge and provides a wide range of virus neutralization in vitro against clinical and laboratory isolates of RSV. No ERD induction was observed in cotton rats after RSV exposure of vaccinated animals and therefore confirms the data generated with Ad26 and Ad35 encoding for the wild type RSV F A2 antigen. Neither mice nor cotton rats showed overt signs of reactogenicity after injection of either Ad26.RSV.preF or Ad35.RSV.preF.









TABLE 1





Amino acid sequences of the RSV pre-fusion F


proteins encoded by the nucleic acid molecules of


the invention (mutations are underlined)















SEQ ID NO: 1: RSV preF2.1 amino acid sequence:


MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLGALRT





GWYTSVITIELSNIKEIKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST





PATNNRARRELPRFMNYTLNNAKKTNVTLSKKRKRRFLGFLLGVGSAIAS





GVAVSKVLHLEGEVNKIKSALLSTNKAVVSLSNGVSVLTSKVLDLKNYID





KQLLPIVNKQSCSIPNIETVIEFQQKNNRLLEITREFSVNAGVTTPVSTY





MLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSIIKEEVLAYV





VQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGSVS





FFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNVDIFNPKYDCKIMTSKT





DVSSSVITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTV





SVGNTLYYVNKQEGKSLYVKGEPIINFYDPLVFPSDEFDASISQVNEKIN





QSLAFIRKSDELLHNVNAVKSTTNIMITTIIIVIIVILLSLIAVGLLLYC





KARSTPVTLSKDQLSGINNIAFSN





SEQ ID NO: 2: RSV preF2.2 amino acid sequence:


MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSALRT





GWYTSVITIELSNIKEIKCNGTDAKVKLIKQELDKYKNAVTELQLLMQST





PATNNRARRELPRFMNYTLNNAKKTNVTLSKKRKRRFLGFLLGVGSAIAS





GVAVSKVLHLEGEVNKIKSALLSTNKAVVSLSNGVSVLTSKVLDLKNYID





KQLLPIVNKQSCSIPNIETVIEFQQKNNRLLEITREFSVNAGVTTPVSTY





MLTNSELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSIIKEEVLAYV





VQLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGSVS





FFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNVDIFNPKYDCKIMTSKT





DVSSSVITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYVSNKGVDTV





SVGNTLYYVNKQEGKSLYVKGEPIINFYDPLVFPSNEFDASISQVNEKIN





QSLAFIRKSDELLHNVNAVKSTTNIMITTIIIVIIVILLSLIAVGLLLYC





KARSTPVTLSKDQLSGINNIAFSN
















TABLE 2





Nucleotide sequence of preferred nucleic acid


molcules of the invention















SEQ ID NO: 3: codon optimized nucleic acid


encoding the RSV F pre-F2.1 pre-fusion protein


PreF2.1


ATGGAGCTGCTGATCCTGAAGGCCAACGCCATCACCACCATCCTGACCGC





CGTGACCTTCTGCTTCGCCAGCGGCCAGAACATCACCGAGGAGTTCTACC





AGAGCACCTGCAGCGCCGTGAGCAAGGGCTACCTGGGCGCCCTGAGAACC





GGCTGGTACACCAGCGTGATCACCATCGAGCTGAGCAACATCAAGGAGAT





CAAGTGCAACGGCACCGACGCCAAGGTGAAGCTGATCAAGCAGGAGCTGG





ACAAGTACAAGAACGCCGTGACCGAGCTGCAGCTGCTGATGCAGAGCACC





CCCGCCACCAACAACAGAGCCAGAAGAGAGCTGCCCAGATTCATGAACTA





CACCCTGAACAACGCCAAGAAGACCAACGTGACCCTGAGCAAGAAGAGAA





AGAGAAGATTCCTGGGCTTCCTGCTGGGCGTGGGCAGCGCCATCGCCAGC





GGCGTGGCCGTGAGCAAGGTGCTGCACCTGGAGGGCGAGGTGAACAAGAT





CAAGAGCGCCCTGCTGAGCACCAACAAGGCCGTGGTGAGCCTGAGCAACG





GCGTGAGCGTGCTGACCAGCAAGGTGCTGGACCTGAAGAACTACATCGAC





AAGCAGCTGCTGCCCATCGTGAACAAGCAGAGCTGCAGCATCCCCAACAT





CGAGACCGTGATCGAGTTCCAGCAGAAGAACAACAGACTGCTGGAGATCA





CCAGAGAGTTCAGCGTGAACGCCGGCGTGACCACCCCCGTGAGCACCTAC





ATGCTGACCAACAGCGAGCTGCTGAGCCTGATCAACGACATGCCCATCAC





CAACGACCAGAAGAAGCTGATGAGCAACAACGTGCAGATCGTGAGACAGC





AGAGCTACAGCATCATGAGCATCATCAAGGAGGAGGTGCTGGCCTACGTG





GTGCAGCTGCCCCTGTACGGCGTGATCGACACCCCCTGCTGGAAGCTGCA





CACCAGCCCCCTGTGCACCACCAACACCAAGGAGGGCAGCAACATCTGCC





TGACCAGAACCGACAGAGGCTGGTACTGCGACAACGCCGGCAGCGTGAGC





TTCTTCCCCCAGGCCGAGACCTGCAAGGTGCAGAGCAACAGAGTGTTCTG





CGACACCATGAACAGCCTGACCCTGCCCAGCGAGGTGAACCTGTGCAACG





TGGACATCTTCAACCCCAAGTACGACTGCAAGATCATGACCAGCAAGACC





GACGTGAGCAGCAGCGTGATCACCAGCCTGGGCGCCATCGTGAGCTGCTA





CGGCAAGACCAAGTGCACCGCCAGCAACAAGAACAGAGGCATCATCAAGA





CCTTCAGCAACGGCTGCGACTACGTGAGCAACAAGGGCGTGGACACCGTG





AGCGTGGGCAACACCCTGTACTACGTGAACAAGCAGGAGGGCAAGAGCCT





GTACGTGAAGGGCGAGCCCATCATCAACTTCTACGACCCCCTGGTGTTCC





CCAGCGACGAGTTCGACGCCAGCATCAGCCAGGTGAACGAGAAGATCAAC





CAGAGCCTGGCCTTCATCAGAAAGAGCGACGAGCTGCTGCACAACGTGAA





CGCCGTGAAGAGCACCACCAACATCATGATCACCACCATCATCATCGTGA





TCATCGTGATCCTGCTGAGCCTGATCGCCGTGGGCCTGCTGCTGTACTGC





AAGGCCAGAAGCACCCCCGTGACCCTGAGCAAGGACCAGCTGAGCGGCAT





CAACAACATCGCCTTCAGCAACTGA





SEQ ID NO: 4: codon optimized nucleic acid


encoding the RSV F pre-F2.2 pre-fusion protein


PreF2.2


ATGGAGCTGCTGATCCTGAAGGCCAACGCCATCACCACCATCCTGACCGC





CGTGACCTTCTGCTTCGCCAGCGGCCAGAACATCACCGAGGAGTTCTACC





AGAGCACCTGCAGCGCCGTGAGCAAGGGCTACCTGAGCGCCCTGAGAACC





GGCTGGTACACCAGCGTGATCACCATCGAGCTGAGCAACATCAAGGAGAT





CAAGTGCAACGGCACCGACGCCAAGGTGAAGCTGATCAAGCAGGAGCTGG





ACAAGTACAAGAACGCCGTGACCGAGCTGCAGCTGCTGATGCAGAGCACC





CCCGCCACCAACAACAGAGCCAGAAGAGAGCTGCCCAGATTCATGAACTA





CACCCTGAACAACGCCAAGAAGACCAACGTGACCCTGAGCAAGAAGAGAA





AGAGAAGATTCCTGGGCTTCCTGCTGGGCGTGGGCAGCGCCATCGCCAGC





GGCGTGGCCGTGAGCAAGGTGCTGCACCTGGAGGGCGAGGTGAACAAGAT





CAAGAGCGCCCTGCTGAGCACCAACAAGGCCGTGGTGAGCCTGAGCAACG





GCGTGAGCGTGCTGACCAGCAAGGTGCTGGACCTGAAGAACTACATCGAC





AAGCAGCTGCTGCCCATCGTGAACAAGCAGAGCTGCAGCATCCCCAACAT





CGAGACCGTGATCGAGTTCCAGCAGAAGAACAACAGACTGCTGGAGATCA





CCAGAGAGTTCAGCGTGAACGCCGGCGTGACCACCCCCGTGAGCACCTAC





ATGCTGACCAACAGCGAGCTGCTGAGCCTGATCAACGACATGCCCATCAC





CAACGACCAGAAGAAGCTGATGAGCAACAACGTGCAGATCGTGAGACAGC





AGAGCTACAGCATCATGAGCATCATCAAGGAGGAGGTGCTGGCCTACGTG





GTGCAGCTGCCCCTGTACGGCGTGATCGACACCCCCTGCTGGAAGCTGCA





CACCAGCCCCCTGTGCACCACCAACACCAAGGAGGGCAGCAACATCTGCC





TGACCAGAACCGACAGAGGCTGGTACTGCGACAACGCCGGCAGCGTGAGC





TTCTTCCCCCAGGCCGAGACCTGCAAGGTGCAGAGCAACAGAGTGTTCTG





CGACACCATGAACAGCCTGACCCTGCCCAGCGAGGTGAACCTGTGCAACG





TGGACATCTTCAACCCCAAGTACGACTGCAAGATCATGACCAGCAAGACC





GACGTGAGCAGCAGCGTGATCACCAGCCTGGGCGCCATCGTGAGCTGCTA





CGGCAAGACCAAGTGCACCGCCAGCAACAAGAACAGAGGCATCATCAAGA





CCTTCAGCAACGGCTGCGACTACGTGAGCAACAAGGGCGTGGACACCGTG





AGCGTGGGCAACACCCTGTACTACGTGAACAAGCAGGAGGGCAAGAGCCT





GTACGTGAAGGGCGAGCCCATCATCAACTTCTACGACCCCCTGGTGTTCC





CCAGCAACGAGTTCGACGCCAGCATCAGCCAGGTGAACGAGAAGATCAAC





CAGAGCCTGGCCTTCATCAGAAAGAGCGACGAGCTGCTGCACAACGTGAA





CGCCGTGAAGAGCACCACCAACATCATGATCACCACCATCATCATCGTGA





TCATCGTGATCCTGCTGAGCCTGATCGCCGTGGGCCTGCTGCTGTACTGC





AAGGCCAGAAGCACCCCCGTGACCCTGAGCAAGGACCAGCTGAGCGGCAT





CAACAACATCGCCTTCAGCAACTGA








Claims
  • 1. A vaccine comprising a polynucleotide sequence that encodes a respiratory syncytial virus (RSV) pre-fusion F protein having the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
Priority Claims (1)
Number Date Country Kind
16163807 Apr 2016 EP regional
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/946,485 filed Jun. 24, 2020, which is a continuation of U.S. patent application Ser. No. 16/091,257, filed Oct. 4, 2018, which is a Section 371 of International Application No. PCT/EP2017/057957, filed Apr. 4, 2017, which was published in the English language on Oct. 12, 2017, under International Publication No. WO 2017/174564 A1, which claims priority under 35 U.S.C. § 119(b) to European Application No. 16163807.7, filed Apr. 5, 2016, the disclosures of which are incorporated herein by reference in their entirety.

US Referenced Citations (63)
Number Name Date Kind
4235877 Fullerton Nov 1980 A
4372945 Likhite Feb 1983 A
4474757 Amon et al. Oct 1984 A
5057540 Kensil et al. Oct 1991 A
5122458 Post et al. Jun 1992 A
5385839 Stinski Jan 1995 A
5559099 Wickham et al. Sep 1996 A
5837511 Falck-Pedersen et al. Nov 1998 A
5837520 Shabram et al. Nov 1998 A
5846782 Wickham et al. Dec 1998 A
5851806 Kovesdi Dec 1998 A
5891690 Massie Apr 1999 A
5965541 Wickham et al. Oct 1999 A
5981225 Kochanek et al. Nov 1999 A
5994106 Kovesdi Nov 1999 A
5994128 Fallaux et al. Nov 1999 A
6020191 Scaria et al. Feb 2000 A
6040174 Imler et al. Mar 2000 A
6083716 Wilson et al. Jul 2000 A
6113913 Brough et al. Sep 2000 A
6225289 Kovesdi May 2001 B1
6261823 Tang Jul 2001 B1
6485958 Blanche Nov 2002 B2
7270811 Bout et al. Sep 2007 B2
7326555 Konz, Jr. et al. Feb 2008 B2
8772256 Graham Jul 2014 B2
8932607 Custers Jan 2015 B2
10294279 Langedijk May 2019 B2
10456462 Langedijk Oct 2019 B2
10457708 Langedijk Oct 2019 B2
10729757 Langedijk Aug 2020 B2
10899800 Langedijk Jan 2021 B2
10953087 Langedijk Mar 2021 B2
11034731 Langedijk Jun 2021 B2
11155583 Krarup Oct 2021 B2
11229692 Godeaux Jan 2022 B2
11229694 Langedijk Jan 2022 B2
11229695 Widjojoatmodjo Jan 2022 B2
11338031 Langedijk May 2022 B2
20110305727 Swanson et al. Dec 2011 A1
20120164176 Swanson et al. Jun 2012 A1
20120315270 McLellan et al. Dec 2012 A1
20130177573 Williamson et al. Jul 2013 A1
20140073032 Custers et al. Mar 2014 A1
20140248314 Swanson et al. Sep 2014 A1
20140271699 Kwong Sep 2014 A1
20160102123 Langedijk Apr 2016 A1
20160145321 Wadia May 2016 A1
20160145322 Wadia May 2016 A1
20160176932 Langedijk et al. Jun 2016 A1
20200061181 Godeaux Feb 2020 A1
20200197509 Widjojoatmodjo et al. Jun 2020 A1
20200360506 Langedijk Nov 2020 A1
20210101940 Langedijk Apr 2021 A1
20210205440 Langedijk Jul 2021 A1
20210284698 Langedijk Sep 2021 A1
20220017574 Langedijk Jan 2022 A1
20220089652 Krarup Mar 2022 A1
20220125910 Godeaux Apr 2022 A1
20220125912 Langedijk Apr 2022 A1
20220133878 Widjojoatmodjo May 2022 A1
20220193219 Callendret Jun 2022 A1
20220204567 Brandenburg Jun 2022 A1
Foreign Referenced Citations (65)
Number Date Country
0853660 Jul 1998 EP
1230354 Aug 2002 EP
2015512380 Apr 2015 JP
2015171378 Oct 2015 JP
20140138765 Dec 2014 KR
9003184 Apr 1990 WO
9014837 Dec 1990 WO
9609378 Mar 1996 WO
9611711 Apr 1996 WO
9822588 May 1998 WO
9839411 Sep 1998 WO
9912568 Mar 1999 WO
9941416 Aug 1999 WO
200029024 May 2000 WO
200032754 Jun 2000 WO
200070071 Nov 2000 WO
200166137 Sep 2001 WO
2001085984 Nov 2001 WO
200240665 May 2002 WO
03040178 May 2003 WO
2003049763 Jun 2003 WO
2003061708 Jul 2003 WO
2003078592 Sep 2003 WO
2003104467 Dec 2003 WO
2004001032 Dec 2003 WO
2004004762 Jan 2004 WO
2004020971 Mar 2004 WO
2005002620 Jan 2005 WO
2005071093 Aug 2005 WO
2005080556 Sep 2005 WO
2006108707 Oct 2006 WO
2007104792 Sep 2007 WO
2007110409 Oct 2007 WO
200911713 Jan 2009 WO
2009079796 Jul 2009 WO
2009106580 Sep 2009 WO
2010060719 Jun 2010 WO
2010060719 Jun 2010 WO
2010086189 Aug 2010 WO
2010149743 Dec 2010 WO
2010149745 Dec 2010 WO
2011008974 Jan 2011 WO
2011020079 Feb 2011 WO
2011045378 Apr 2011 WO
2011045381 Apr 2011 WO
2011050168 Apr 2011 WO
2011098592 Aug 2011 WO
2012006596 Jan 2012 WO
2012158613 Nov 2012 WO
2013139911 Sep 2013 WO
2013139916 Sep 2013 WO
2013135615 Sep 2013 WO
2014005643 Jan 2014 WO
2014077096 May 2014 WO
2014160463 Oct 2014 WO
2014174018 Oct 2014 WO
2014202570 Dec 2014 WO
WO-2014202570 Dec 2014 WO
2015013551 Jan 2015 WO
2015040002 Mar 2015 WO
2015189425 Dec 2015 WO
2017174564 Oct 2017 WO
2017174568 Oct 2017 WO
2021198413 Oct 2021 WO
2022002894 Jan 2022 WO
Non-Patent Literature Citations (69)
Entry
Gilman et al., “Rapid profiling of RSV antibody repertories from the memory B cells of naturally infected adult donors”, Science Immunology, 11 pages, Dec. 2016.
Int'l Search Report and Written Opinion dated Dec. 17, 2018 in Int'l Application No. PCT/EP2018/074710, 14 pages.
International Search Report and Written Opinion issued in PCT/EP2017/062875, dated Aug. 14, 2017, 10 pages.
Janssen Vaccines & Prevention B.V.: A Study to Evaluate the Safety, Tolerability and Immunogenicity of Two Vaccinations of Ad26.RSV. preF One Year Apart in Adults Aged 60 Years and Older in Stable Health, Oct. 2016, retrieved from the Internet: http://clinicaltrials.gov/ct2/show/record/NCT02926430 (retrieved on Nov. 30, 2018) 10 pages.
Openshaw et al., “Protective and Harmful Immunity to RSV Infection”, Annu Rev. Immunol, vol. 35, pp. 501-532, 2017.
U.S. Appl. No. 17/594,394, filed Oct. 14, 2021. Inventor, Benoit Christophe Stephan Callendret.
U.S. Appl. No. 17/595,255, filed Nov. 12, 2021. Inventor, Benoit Christophe Stephan Callendret.
U.S. Appl. No. 17/664,290, filed May 20, 2022, Inventors: Langedijk, Johannes; Verhagen, Janneke M.
Hotard, et al., “Identification of Residues in the Human Respiratory Syncytial Virus Fusion Protein That Modulate Fusion Activity and Pathogenesis,” Journal of Virology, vol. 89, No. 1, pp. 512-522, Jan. 2015.
Cohen et al., “Chimpanzee adenovirus CV-68 adapted as a gene delivery vector interacts with the coxsackievirus and adenovirus receptor”, Journal of General Virology, 83, pp. 151-155, 2002.
Farina et al., “Replication-Defective Vector Based on a Chimpanzee Adenovirus”, Journal of Virology, vol. 75, No. 23, pp. 11603-11613, Dec. 2001.
Kobinger et al., “Chimpanzee adenovirus vaccine protects against Zaire Ebola Virus”, Science Direct, Virology, 346, pp. 394-401, 2006.
Lasaro et al., “New Insights on Adenovirus as Vaccine Vectors”, Molecular Therapy, vol. 17, No. 8, pp. 1333-1339, Aug. 2009.
Tatsis et al., “A CD46-binding Chimpanzee Adenovirus Vector as a Vaccine Carrier”, American Society of Gene Therapy, vol. 15, No. 3, pp. 608-617, Mar. 2007.
Widjojatomodjo et al., “Recombianant Low-seroprevalent adenoviral vectors Ad26 and Ad35 expressing the respiratory syncytial virus (RSV) fusion protein induce protective immunity against RSV infection in cotton rats”, Vaccine, 33, pp. 5406-5414, 2015.
Green et al., “Safety and Immunogenicity of novel respiratory syncytial virus (RSV) vaccines based on the RSV viral proteins F, N and M2-1 encoded by simian adenovirus (PanAd3-RSV) and MVA (MVA-RSV): protocol for an open-label, dose-escalation, single-centre, phase 1 clinical trial in healthy adults”, BMJ Open, 13 pages, Oct. 2015.
Grunwald et al., “Novel Vaccine Regimen Elicits Strong Airway Immune Responses and Control of Respiratory Syncytial Virus in Nonhuman Primates”, Journal of Virology, vol. 88, No. 8, pp. 3997-4007, Apr. 2014.
Comparison to Sequence 16, U.S. Appl. No. 12/517,194; U.S. Pat. No. 8,772,256 (Year: 2014).
McLellan et al., “Structural Basis of Respiratory Syncytial Virus Neutralization by Motavizumab,” Nature Structural & Molecular Biology, vol. 17, pp. 248-250 (2010).
McLellan et al., “Structure of RSV Fusion Glycoprotein Trimer Bound to a Prefusion-Specific Neutralizing Antibody,” Science, vol. 340, pp. 1113-1117 (2013).
McLellan et al., “Structure-Based Design of a Fusion Glycoprotein Vaccine for Respiratory Syncytial Virus,” Science, vol. 342, pp. 592-598 (2013).
Swanson et al., “Structural Basis for Immunization with Postfusion Respiratory Syncytial Virus Fusion F Glycoprotein (RSV F) to Elicit High Neutralizing Antibody Titers,” PNAS, vol. 108, pp. 9619-9624 (2011).
Letarov et al., “The Carboxy-Terminal Domain Initiates Trimerization of Bacteriophage T4 Fibritin,” Biochemistry (Moscow), vol. 64, No. 7, pp. 817-823 (1999).
Guthe et al., “Very Fast Folding and Association of a Trimerization Domain from Bacteriophage T4 Fibritin,” J. Mol. Biol., vol. 337, pp. 905-915 (2004).
“Database UniProt Accession W8CJC7,” http://ibis.internal.epo.org/exam/dbfetch.jsp?id=UNIPROT:W8CJC7, Download date: Aug. 12, 2015, 1 page.
Widjaja et al., “Recombinant soluble Respiratory Syncytial Virus F Protein That Lacks Heptad Repeat B, Contains a GCN4 Trimerization Motif and Is Not Cleaved Displays Prefusion-Like Characteristics,” Plos One, 20 pages, Jun. 24, 2015.
Written Opinion dated Oct. 10, 2016 in Int'l Application No. PCT/EP2016/066104.
Int'l Search Report dated Oct. 10, 2016 in Int'l Application No. PCT/EP2016/066104.
Written Opinion dated Oct. 12, 2016 in Int'l Application No. PCT/EP2016/066098.
Int'l Search Report dated Oct. 12, 2016 in Int'l Application No. PCT/EP2016/066098.
Suzuki et al., “An Isoleucine Zipper Peptide Forms a Native-like Triple Stranded Coiled Coil in Solution,” Protein Engineering, vol. 11, No. 11, pp. 1051-1055 (1998).
Dames et al., “NMR Structure of a Parallel Homotrimeric Coiled Coil,” Nature Structural Biology, vol. 5, No. 8, pp. 687-691 (Aug. 1998).
Calder et al., “Electron Microscopy of the Human Respiratory Syncytial Virus Fusion Protein and Complexes That It Forms With Monoclonal Antibodies,” Virology, vol. 271, pp. 122-131 (2000).
Harbury et al., “A Switch Between Two-, Three-, and Four-Stranded Coiled Coils in GON4 Leucine Zipper Mutants,” Science, vol. 262, pp. 1401-1407.
O'Shea et al., “Evidence That the Leucine Zipper Is a Coiled Coil,” Science, vol. 243, pp. 538-542 (Jan. 27, 1989).
Database EMBL, Aug. 28, 1995, Human respiratory syncytial virus, strain RSB89-1734, fusion protein (F) mRNA, complete CDS, XP002729919.
Int'l Search Report and Written Opinion dated Oct. 9, 2014 in Int'l Application No. PCT/EP2014/062655.
Int'l Search Report and Written Opinion dated Aug. 12, 2014 in Int'l Application No. PCT/EP2014/058353.
Magro et al., “Neutralizing Antibodies Against the Preactive Form of Respiratory Syncytial Virus Protein Offer Unique Possibilities for Clinical Intervention,” PNAS, vol. 109, No. 8, pp. 3089-3094 (Feb. 21, 2012).
Yin et al., “Structure of the Parainfluenza Virus 5 F Protein in its Metastable, Prefusion Conformation,” Nature, vol. 439, pp. 38-44 (Jan. 5, 2006).
Ngwuta et al, “Prefusion F-Specific Antibodies Determine the Magnitude of RSV Neutralizing Activity in Human Sera,” Science Translational Medicine, vol. 7, No. 309, pp. 1-9 (2015).
Int'l Search Report and Written Opinion dated Jun. 12, 2017 in Int'l Application No. PCT/EP2017/057962.
Abbink et al, “Comparative Seroprevalence and Immunogenicity of Six Rare Serotype Recombinant Adenovirus Vaccine Vectors from Subgroups B and D,” Journal of Virology, vol. 81, No. 9, pp. 4654-4663 (May 2007).
Abrahamsen et al, “Construction of an Adenovirus Type 7a E1A-Vector,” Journal of Virology, vol. 71, No. 11, pp. 8946-8951 (Nov. 1997).
Altaras et al, “Production and Formulation of Adenovirus Vectors,” Advances in Biochemical Engineering / Biotechnology, vol. 99, pp. 193-260 (2005).
Brough et al, “A Gene Transfer Vector-Cell Line System for Complete Functional Complementation of Adenovirus Early Regions E1 and E4,” Journal of Virology, vol. 70, No. 9, pp. 6497-6501 (Sep. 1996).
Fallaux et al, “New Helper Cells and Matched Early Region 1-Deleted Adenovirus Vectors Prevent Generation of Replication-Competent Adenoviruses,” Human Gene Therapy, vol. 9, pp. 1909-1917 (Sep. 1998).
Gao et al, “A Cell Line for High-Yield Production of E1-Deleted Adenovirus Vectors without the Emergence of Replication-Competent Virus,” Human Gene Therapy, vol. 11, pp. 213-219 (Jan. 2000).
Goerke et al, “Development of a Novel Adenovirus Purification Process Utilizing Selective Precipitation of Cellular DNA,” Biotechnology and Bioengineering, vol. 91, pp. 12-21 (2005).
Havenga et al, “Novel replication-incompetent adenoviral B-group vectors: high vector stability and yield in PER.C6 cells,” Journal of General Virology, vol. 87, pp. 2135-2143 (2006).
Hoganson et al, “Development of a Stable Adenoviral Vector Formulation,” BioProcessing Journal, vol. 1, No. 1, pp. 43-48 (Mar. 2002).
Kim et al, “Single mucosal immunization of recombinant adenovirus-based vaccine expressing F1 protein fragment induces protective mucosal immunity against respiratory syncytial virus infection,” Vaccine, vol. 28, pp. 3801-3808 (2010).
Kohlmann et al, “Protective Efficacy and Immunology of an Adenoviral Vector Vaccine Encoding the Codon-Optimized F Protein of Respiratory Syncytial Virus,” Journal of Virology, vol. 83, No. 23, pp. 12601-12610 (Dec. 2009).
Konz et al, “Serotype Specificity of Adenovirus Purification Using Anion-Exchange Chromatography,” Human Gene Therapy, vol. 16, pp. 1346-1353 (Nov. 2005).
Konz et al, “Scaleable Purification of Adenovirus Vectors,” Methods in Molecular Biology, vol. 434, No. 2, pp. 13-23 (2008).
Krarup et al, “A Highly Stable Prefusion RSV F Vaccine Derived from Structural Analysis of the Fusion Mechanism,” Nature Communications, vol. 6, pp. 1-11 (Sep. 2015).
Nan et al, “Development of an Ad7 cosmid system and generation of an Ad7LE1LE3HIVMN env/rev recombinant virus,” Gene Therapy, vol. 10, pp. 326-336 (2003).
Pemberton et al, “Cytoxic T Cell Specificity for Respiratory Syncytial Virus Proteins: Fusion Protein is an Important Target Antigen,” Journal of General Virology, vol. 68, pp. 2177-2182 (1987).
Solabomi et al, “The Oligomerization Domain of C4-Binding Protein (C4bp) Acts as an Adjuvant, and the Fusion Protein Comprised of the 19-Kilodalton Merozoite Surface Protein 1 Fused with the Murine C4bp Domain Protects Mice against Malaria,” Infection and Immunity, vol. 76, No. 8, pp. 3817-3823 (Aug. 2008).
Vogels et al, “Replication-Deficient Human Adenovirus Type 35 Vectors for Gene Transfer and Vaccination: Efficient Human Cell Infection and Bypass of Preexisting Adenovirus Immunity,” Journal of Virology, vol. 77, No. 15, pp. 8263-8271 (Aug. 2003).
Yu et al., “Single Intranasal Immunization with Recombinant Adenovirus-Based Vaccine Induces Protective Immunity against Respiratory Syncytial Virus Infection,” Journal of Virology, vol. 82, No. 5, pp. 2350-2357 (Mar. 2008).
Int'l Search Report and Written Opinion dated Jun. 12, 2017 in Int'l Application No. PCT/EP2017/057957.
Krause et al, “A Broadly Neutralizing Human Monclonal Antibody That Recognizes a Conserved, Novel Epitope on the Globular Head of the Influenza H1N1 Virus Hemagglutinin,” Journal of Virology, vol. 85, No. 20, pp. 10905-10908 (Oct. 2011).
McLellan et al, “Structure of Respiratory Syncytial Virus Fusion Glycoprotein in the Postfusion Conformation Reveals Preservation of Neutralizing Epitopes,” Journal of Virology, vol. 85, No. 15, pp. 7788-7796 (Aug. 2011).
Database Geneseq (online) “RSV fusion protein N671 S215P, RSV CL57-v224, fibritin, SEQ: 74”, XP002761983, retrieved from EBI accession No. GSP:BBP75438, Database accession No. BBP75438 sequence.
Neuzil, “Progress toward a Respiratory Syncytial Virus Vaccine”, Clinical and Vaccine Immunology, vol. 23, pp. 186-188, 2016.
Int'l Search Report and Written Opinion dated Sep. 8, 2018 in Int'l Application No. PCT/EP2018/062604.
Bangari et al., “Development of nonhuman adenoviruses as vaccine vectors”, Vaccine, 24(7), pp. 849-862, 2006.
D. Roymans et al: “Binding of a potent small-molecule inhibitor of six-helix bundle formation requires interactions with both heptad-repeats of the RSV fusion protein”, Proceedings of the National Academy of Sciences, vol. 107, No. 1, Dec. 4, 2009 (Dec. 4, 2009), pp. 308-313, XP055708078,ISSN: 0027-8424, DOI: 10.1073/pnas.0910108106.
Related Publications (1)
Number Date Country
20220288186 A1 Sep 2022 US
Continuations (2)
Number Date Country
Parent 16946485 Jun 2020 US
Child 17664290 US
Parent 16091257 US
Child 16946485 US