WO2019066437A1 - Antigène de protéine f du virus respiratoire syncytial (rsv) modifié soluble - Google Patents

Antigène de protéine f du virus respiratoire syncytial (rsv) modifié soluble Download PDF

Info

Publication number
WO2019066437A1
WO2019066437A1 PCT/KR2018/011324 KR2018011324W WO2019066437A1 WO 2019066437 A1 WO2019066437 A1 WO 2019066437A1 KR 2018011324 W KR2018011324 W KR 2018011324W WO 2019066437 A1 WO2019066437 A1 WO 2019066437A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
rsv
amino acid
protein
protein antigen
Prior art date
Application number
PCT/KR2018/011324
Other languages
English (en)
Korean (ko)
Inventor
김은솜
권태우
김학
서기원
이수진
김진설
김창신
함동수
김훈
Original Assignee
에스케이바이오사이언스 주식회사
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020180113291A external-priority patent/KR20190038358A/ko
Application filed by 에스케이바이오사이언스 주식회사 filed Critical 에스케이바이오사이언스 주식회사
Publication of WO2019066437A1 publication Critical patent/WO2019066437A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses

Definitions

  • the present invention relates to RSV protein antigens and / or RSV immunostimulatory compositions for the treatment and / or prevention of respiratory syncytial virus (RSV) infections. More particularly, the present invention relates to a soluble RSV F protein antigen, an RSV immunogenic composition comprising the same, a RSV vaccine, and a method for inducing RSV immunity using the same.
  • RSV respiratory syncytial virus
  • Respiratory syncytial virus is a worldwide prevalent virus that causes respiratory illnesses and is a major cause of serious respiratory infections in infants and young children. Nearly all infants (> 95%) are infected within 2 years of age. In the United States, infections cause infections close to 100% in 2-3 year-old infants, and there are hundreds of direct signs / indirect signs associated with RSV, Was reportedly dead. It is also known that infants are the main infections, but infect respiratory diseases of patients with weakened immune system and elderly people and cause fatal respiratory diseases. It is the second most common cause of respiratory illness after influenza, but the annual death rate from RSV per 100,000 children under 1 year of age is 1.3 to 2.5 times higher than that of influenza. In 2002, WHO reported that 64,000 people were infected with RSV every year, of which 160,000 died.
  • RSV is resistant to external environments such as telephone and door knockers, which survive for six hours on a hard surface, which makes it possible to continuously infect and spread rapidly, which is very likely to outbreak in hospitals and community settings .
  • Symptoms develop from about 4-6 days after exposure to RSV virus.
  • Symptoms such as fever, cough, and wheezing develop from about 3 days after the onset of the first symptom, starting with runny nose and anorexia.
  • Clinical manifestations range from relatively mild symptoms to otitis media, apnea of premature infants, asthma, pneumonia, and bronchiolitis.
  • RSV belongs to the order of Mononegavirales, Orthopneumovirus (genus) of Pneumoviridae (family). Viruses similar to RSV include measles virus, mumps virus causing parotitis, and parainfluenza types 1, 2, and 3.
  • RSV is a medium-sized virus of the order of 120-200 nm, and has a genome of linear, negative strand RNA consisting of about 15,000 nucleotides.
  • RSV is divided into serotypes A and B by G proteins.
  • the RSV genome encodes 10 proteins and is composed of non-structural proteins and structural proteins.
  • Nonstructural proteins are nonstructural (NS) 1, nonstructural (NS) 2, nucleocapsid (N), phosphoprotein (P) and viral polymerase (L).
  • NS nonstructural
  • N nucleocapsid
  • P phosphoprotein
  • L viral polymerase
  • matrix (M) protein, fusion (F) protein and glyco (G) protein form the envelope of the virus.
  • F protein and G protein form a spike.
  • RSV F protein is an important component that fuses with cell membrane at the early stage of virus entry and is known to be a major target of vaccine and antiviral drug due to its high antigenicity.
  • the F protein of RSV changes its structure from "pre-fusion" type to "post-fusion” type during entry into cells during the infection process.
  • the F protein consists of about 574 amino acids and can be divided into F2 (about 20 kDa) and F1 (about 50 kDa) subunits, where the total F protein is F0.
  • Subunit Furin cleavage between F2 and F1 is cleaved by F2 and F1 by furin protease.
  • furin cleavage recognizes the amino acid sequences of the two sites "RARR” and “KKRKRR", and Arginine (R) acts as an important core amino acid.
  • the F1 subunit is called the HRA (fusion domain) at the N-terminal side and the trans-membrane domain (HRB) at the C-terminal side. Two or more heptad in the HRA and HRB regions, It is confirmed that it forms. Fusion of the F protein and cell-virus fusion occurs when the hinge portion of the curved HRA is expanded. It has also been found that trimer formation is induced by purine cleavage with removal of the p27 portion.
  • RSV is widely prevalent in many parts of the world and is a particularly dangerous virus for infants and young children, but there is no vaccine available.
  • the soluble RSV F protein obtainable with the present invention can provide the immunogen that is required for this requirement.
  • the present invention is directed to providing recombinant soluble respiratory cell fusion virus (RSV) fusion (F) polypeptides.
  • RSV soluble respiratory cell fusion virus
  • F fusion
  • the soluble RSV F polypeptides of the present invention comprise at least one epitope specific for the RSV F protein.
  • the polypeptide is a modified form of the fusion peptide region.
  • the polypeptide comprises one or two or more amino acid modifications.
  • the present invention also relates to the use of a soluble RSV F polypeptide, a nucleic acid molecule and / or a composition for inducing immunogenicity, and its use in inducing an animal and human immunological response to a soluble RSV F protein, particularly its use as a vaccine .
  • the present invention also relates to a nucleic acid molecule encoding the RSV F polypeptide and / or a vector comprising the nucleic acid molecule and / or a polypeptide transcribed from the nucleic acid molecule in a specific context for use as a vaccine, ≪ / RTI >
  • the present invention relates to a method of inducing a neutralizing anti-respiratory cell fusion virus (RSV) F protein antibody in a subject, comprising the step of administering to the subject a nucleic acid molecule encoding the RSV F polypeptide, and / A vector comprising a nucleic acid molecule, and / or a polypeptide transcribed from a nucleic acid molecule.
  • RSV neutralizing anti-respiratory cell fusion virus
  • protein or " polypeptide " used herein refers to a collection of amino acids that are encoded and generated in a particular nucleic acid. Here, the aggregate means a unit consisting of two, three, four, or more amino acids.
  • protein or “ polypeptide " may be understood as meaning "antigen” or "immunogen”.
  • antigen refers to any substance that induces immunogenicity.
  • antigen includes macromolecular molecules composed of proteins or proteins.
  • a large protein refers to a large number of protein sequences and protein aggregates, including polymer antigens and virus-like particles (VLPs).
  • immunogenicity induction refers to both cellular immunity and humoral immunity.
  • antigen includes, for example, all and some foreign material that has penetrated into the body, and induction of cellular immunity by the term “ antigen " may mean generation of antigen-specific antibodies.
  • Antigen-specific antibodies include neutralizing antibodies that neutralize re-infiltrated foreign material.
  • mutated refers to any nucleic acid and / or polypeptide that forms a modified nucleic acid or polypeptide Lt; / RTI > Mutations may include, for example, point mutations, deletions or insertions of single or multiple residues in a polynucleotide, and include changes in the protein-coding region of the gene as well as changes in the protein, such as but not limited to control or promoter sequences - Contains changes in the outer region of the coding sequence.
  • the genetic change may be any form of mutation.
  • a mutation can constitute a point mutation, a frame-shifting mutation, an insertion or deletion of all or part of a gene.
  • the mutation occurs naturally.
  • the mutation is the result of an artificial mutation pressure.
  • the term " vaccine” refers to a preparation of an induced antigenic determinant used to induce the formation of antibodies or immunity against dead or weakened pathogen or pathogen. Vaccines are given to provide immunity against diseases caused by various kinds of viruses, for example, influenza and the like.
  • the term " vaccine” also refers to a suspension or solution of an immunogen (e. G., A modified or mutated RSV F protein) administered to a vertebrate animal to cause protective immunity, i. E. it means.
  • the present invention provides vaccine compositions that are immunogenic and capable of providing protection against diseases associated with infection.
  • mutations in RSV F proteins are the result of genetic engineering.
  • &Quot about “ includes all values that provide the same effect and result as a reference value. However, “ about “ gives meaning only to the reference value. Also, the scope encompassed by the term “ about “ may vary depending on the scope or characteristics of the content (reference value) in which the term is included. Thus, depending on the context, “ about “ means, for example, ⁇ 15%, ⁇ 10%, ⁇ 5%, ⁇ 4%, ⁇ 3%, ⁇ 2%, ⁇ 1%, or less than 1% .
  • the present invention provides recombinant RSV F proteins containing the amino acid sequence that has been modified compared to the wild-type RSV F protein.
  • the inventors of the present invention found that the modified RSV F proteins of the present invention increase expression of RSV F proteins and improve immunogenicity compared to wild type RSV F proteins.
  • the RSV F proteins are human RSV F proteins.
  • the recombinant respiratory syncytial virus (RSV) F protein antigen provides a RSV F protein antigen wherein the 525-574 position of the amino acid sequence of SEQ ID NO: 1 is deleted.
  • the RSV F protein antigen comprises SEQ ID NO: 2.
  • the RSV F protein antigen may further comprise at least one amino acid substitution at the fusion peptide position of the amino acid sequence of SEQ ID NO: 2.
  • the RSV F protein antigen may be substituted with a hydrophilic amino acid at amino acid positions 137-145 of SEQ ID NO: 2, and preferably the hydrophobic amino acid may be substituted with an amino acid " A ". That is, the RSV F protein antigen may be a RSV F protein antigen modified with AAGAAAGAG amino acid sequence FLGFLLGVG at amino acid sequence 137-145 of SEQ ID NO: 2.
  • the RSV F protein antigen may be substituted with a non-polar amino acid at one or more polar amino acids at amino acid positions 137-145 of SEQ ID NO: 2.
  • the RSV F protein antigen may be an RSV F protein antigen modified from the amino acid sequence FLGFLLGVG of amino acid sequence 137-145 of SEQ ID NO: 2 by QNGQNNGSG.
  • the RSV F protein antigen may be a RSV F protein antigen modified from the amino acid sequence FLGFLLGVG at position 137-145 of SEQ ID NO: 2 by NSGNSSGGG.
  • the RSV F protein antigen may be a RSV F protein antigen modified with the amino acid sequence FLGFLLGVG of amino acid sequence 137-145 of SEQ ID NO: 2 by TLSKKRKRR.
  • the present invention provides recombinant soluble respiratory syncytial virus (RSV) fusion (F) polypeptides comprising a mutated or modified amino acid sequence compared to the wild type RSV F protein.
  • RSV respiratory syncytial virus
  • F fusion polypeptides comprising a mutated or modified amino acid sequence compared to the wild type RSV F protein.
  • the present invention specifically provides the use of a novel recombinant RSV F protein obtained as a RSV immunogen, obtained through mutation or modification of one or more amino acid sequences of the wild-type RSV F protein.
  • the present invention provides an immunogen for new forms of RSV infection.
  • the inventors of the present invention have confirmed that the amino acid sequence of a specific region of the F protein among the RSV structural proteins can be modified to provide excellent immunogenicity, thus completing the present invention.
  • the RSV F protein in which a part of the fusion peptide (F protein) of the wild-type respiratory syncytial virus (RSV) comprising SEQ ID NO: 1 is deleted can be represented by SEQ ID NO: 2,
  • the RSV F protein of SEQ ID NO: 2 may be provided as an RSV immunogen in the body or as an antigen for RSV immunization.
  • the RSV F protein of SEQ ID NO: 2 comprises a deletion of amino acid sequence positions 525-574 of the wild-type RSV F protein of SEQ ID NO: 1.
  • the amino acid sequence position 525-574 of SEQ ID NO: 2 can be understood as a site including a transmembrane domain and a cytosol region of the F protein.
  • RSV immunogens of the present invention are those in which the RSV F protein of SEQ ID NO: 2, in which the amino acid sequence positions 525-574 of the wild type RSV F protein of SEQ ID NO: 1 is deleted, undergoes one or more sequence variations (amino acids or nucleic acids) , Excellent immunogenicity (i.e., high antibody affinity), and can provide body stability.
  • the RSV immunogen is capable of providing a soluble F protein when the amino acid sequence positions 525-574 of the wild-type RSV F protein of SEQ ID NO: 1 are deleted.
  • the RSV immunogen comprises an RSV F protein of SEQ ID NO: 2 in which the amino acid sequence positions 525-574 of the wild-type RSV F protein of SEQ ID NO: 1 are deleted is one or more sequence variants (amino acids or nucleic acids) When present, soluble F protein can be provided.
  • the position at which the RSV F protein of SEQ ID NO: 2 undergoes one or more sequence variations can be understood as the fusion peptide region of the RSV F protein. Mutations in the fusion peptide region have confirmed that the recombinant RSV F protein can work with excellent immunogenicity.
  • the fusion peptide moiety is at position 137-145 of SEQ ID NO: 2, more preferably at position 137-144 of SEQ ID NO: 2.
  • the inventors of the present invention confirmed that the deletion at positions 137-144 of SEQ ID NO: 2 may contribute to the stabilization of the RSV F protein, but the synergistic effect of the antibody may be insignificant.
  • the amino acid of the fusion peptide was replaced with another amino acid (substitution)
  • substitution there was a difference in the immune response, and it was confirmed that it could act as an immunity of excellent effect.
  • the inventors of the present invention have developed a recombinant RSV F protein immunogen that can provide excellent antibody titer through one or more sequence variations of the fusion peptide of the RSV F protein through long term studies.
  • the RSV F protein immunogen can be used for prevention and treatment of RSV infection, and can be preferably provided as a vaccine for prevention of RSV infection.
  • one embodiment of the present invention provides a RSV immunogen that mutates or modifies a partial sequence of a fusion peptide of RSV F protein.
  • One embodiment of the present invention is a method for the production of a RSV F protein of SEQ ID NO: 2 wherein the RSV F protein comprises one or more mutations in the amino acid sequence positions 137-145 and / or the RSV F protein of SEQ ID NO: 2 corresponding to the amino acid sequence at positions 137-145 , A recombinant soluble RSV F protein immunogen comprising one or more mutations in the nucleotide sequence of SEQ ID NO: 24.
  • soluble respiratory cell fusion virus (RSV) fusion (F) polypeptide More specifically, it is intended to provide a recombinant soluble respiratory cell fusion virus (RSV) fusion (F) polypeptide.
  • the soluble RSV F polypeptides of one embodiment of the invention comprise at least one epitope specific for the RSV F protein.
  • the polypeptide is a modified form of the fusion peptide region.
  • the polypeptide comprises one or two or more amino acid modifications.
  • the present invention also relates to the use of a soluble RSV F polypeptide, a nucleic acid molecule and / or a composition for inducing immunogenicity, and its use in inducing an animal and human immunological response to a soluble RSV F protein, particularly its use as a vaccine .
  • the present invention also relates to a nucleic acid molecule encoding the RSV F polypeptide and / or a vector comprising the nucleic acid molecule and / or a polypeptide transcribed from the nucleic acid molecule in a specific context for use as a vaccine, ≪ / RTI >
  • the present invention relates to a method of inducing a neutralizing anti-respiratory cell fusion virus (RSV) F protein antibody in a subject, comprising the step of administering to the subject a nucleic acid molecule encoding the RSV F polypeptide, and / A vector comprising a nucleic acid molecule, and / or a polypeptide transcribed from a nucleic acid molecule.
  • RSV neutralizing anti-respiratory cell fusion virus
  • the variant in which the amino acid sequence positions 525-574 of the wild-type RSV F protein is deleted can be understood as an RSV F protein consisting of SEQ ID NO: 2.
  • the inventors of the present invention have confirmed that a fusion peptide of the RSV F protein of SEQ ID NO: 2, specifically a mutation at an amino acid position of 137-145 can increase antibody titer.
  • repeated infections can be reduced and the number of inoculations can be reduced.
  • the inventors of the present invention have found that a mutation or mutation induced at the RSV F protein antigen and the specific amino acid position of the antigen of SEQ ID NO: 2 increases the expression of RSV F protein in the host cell and significantly increases immunogenicity Respectively.
  • the RSV F protein antigen may comprise a fusion peptide of SEQ ID NO: 2, more preferably at least one amino acid substituted at amino acid positions 137-145 of SEQ ID NO: 2.
  • the amino acid sequence 137-145 of SEQ ID NO: 2 can be understood as a fusion peptide portion of the RSV F protein.
  • the amino acid substitution at amino acid positions 137-145 of SEQ ID NO: 2 may include one or more mutations or mutations of the amino acids corresponding to the position.
  • the RSV F proteins may comprise three or more modifications or mutations in the amino acids corresponding to that position.
  • substitution can be understood as meaning a modification or mutation of an amino acid. That is, the substitution may be understood to include all cases in which a part of the amino acid sequence of SEQ ID NO: 2 is changed. For example, all cases in which one or more amino acid sequences of the amino acid sequence of SEQ ID NO: 2 are altered.
  • At least one hydrophobic amino acid at amino acid positions 137-145 of SEQ ID NO: 2 may be substituted with a hydrophilic amino acid.
  • the RSV F protein of SEQ ID NO: 2 is a variation of one or more hydrophobic portions of the fusion peptide portion into hydrophilic portions.
  • the invention comprises a RSV F protein antigen (SEQ ID NO: 3, FP1) modified with AAGAAAGAG by substituting A with the hydrophobic portion of the FLGFLLGVG sequence at positions 137-145 of SEQ ID NO:
  • the RSV F protein of SEQ ID NO: 2 is a mutation in which the polar portion of the fusion peptide portion is transformed into a nonpolar side chain. More specifically, the present invention encompasses a RSV F protein antigen (SEQ ID NO: 4, FP3) that has been modified from the FLGFLLGVG sequence at positions 137-145 of SEQ ID NO: 2 to QNGQNNGSG. In another embodiment, the RSV F protein antigen (SEQ ID NO: 5, FP4) is obtained by modifying the FLGFLLGVG sequence of amino acid sequence 137-145 of SEQ ID NO: 2 with NSGNSSGGG.
  • the inventors of the present invention have found that the RSV F protein mutated at amino acid positions 137-145 of SEQ ID No. 2, preferably FP1 (SEQ ID No. 3), FP3 (SEQ ID No. 4) and FP4 (SEQ ID No. 5) Can serve as an excellent immunogen for the purpose of Mutations may occur in the amino acid sequence at positions 137-145 of SEQ ID NO: 2, but as known to those of ordinary skill in the art, substitution, deletion, and insertion of amino acids constituting the protein may result in completely different functions and effects of the protein .
  • the present invention provides an optimized recombinant RSV F protein that is expected to have the best immunogenicity among the mutations induced particularly at positions 137-145 of SEQ ID NO: 2.
  • the RSV F protein antigen according to an embodiment of the present invention is a soluble protein, and in one embodiment of the present invention, provides a soluble RSV F protein antigen and / or an immunogenic composition comprising the RSV F protein antigen.
  • the RSV F protein antigen is selected from the group consisting of SEQ ID NO: 3, 4, 5, 6, 7, 8, 9, 10, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: Or more of the above protein antigens.
  • the RSV F protein antigen may comprise at least one protein antigen selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: More preferably, in one embodiment, the RSV F protein antigen may comprise at least one protein antigen selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 4, and SEQ ID NO: 5.
  • the RSV F protein antigen is selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: SEQ ID NO: 34, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: Or a protein antigen encoded by the above base sequence.
  • the RSV F protein antigen may be a protein antigen encoded by any one or more of the nucleotide sequences selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 and SEQ ID NO: More preferably, the RSV F protein antigen may be a protein antigen encoded by at least one base sequence selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, and SEQ ID NO: 27.
  • Additional mutations that maintain the stabilization of the F1 portion of the wild RSV F protein further include at least one additional mutation selected from the group consisting of (a) to (g) based on SEQ ID NO: 2.
  • RSV F protein antigens of the present invention are based on SEQ ID NO: 2
  • the RSV F protein antigen is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • the amino acid corresponding to position 140 of SEQ ID NO: 2 is substituted with W, the amino acid corresponding to 163 is substituted with Q, the amino acid corresponding to 188 is substituted with Q, the amino acid corresponding to 189 is substituted with L No. 15);
  • amino acid corresponding to position 163 of SEQ ID NO: 2 is substituted with Q, the amino acid corresponding to 505 is replaced with W (SEQ ID NO: 20);
  • An amino acid corresponding to position 188 of SEQ ID NO: 2 is substituted with Q, an amino acid corresponding to 189 is substituted with L, and an amino acid corresponding to 505 is substituted with W (SEQ ID NO: 21);
  • amino acid corresponding to position 163 of SEQ ID NO: 2 may be substituted with Q and the amino acid corresponding to 487 may be substituted with L (SEQ ID NO: 22).
  • the present invention provides a RSV immunogenic composition and / or vaccine comprising said RSV F protein.
  • RSV F proteins other than the human RSV F protein (SEQ ID NO: 1), including variants corresponding to those described above.
  • RSV F proteins may include, but are not limited to, A strains of human RSV, B strains of human RSV, strains of bovine RSV, and RSV F proteins from strains of avian RSV.
  • this includes using known methods of protein processing and recombinant DNA technology to enhance or modify the properties of the mutated RSV F proteins mentioned above.
  • Genes that encode the proteins are modified by codonification, and modifications of the nucleotides are known to those skilled in the art.
  • Various forms of mutagenesis can be used to produce and / or isolate variant nucleic acids encoding the protein molecules and / or further modify / mutate the proteins of the invention.
  • mutagenesis include, but are not limited to, site-specific mutagenesis, random point mutagenesis, homologous recombination (DNA shuffling), mutagenesis using uracil containing template, oligonucleotide-induced mutagenesis, phosphorothioate-modified DNA mutagenesis, DNA (gapped-duplex DNA), and the like.
  • Other suitable methods include point mismatch repair, repair-deficient host strains, restriction-selection and restriction-mutagenesis using purification, deletion mutagenesis, total gene synthesis, double-strand breakage Recovery, and the like.
  • mutagenesis involving a chimeric structure is also encompassed by the present invention.
  • mutagenesis can be guided by known information, such as sequences, sequence comparisons, physical properties, crystal structures, etc., of molecules that have been generated pre-existing or of naturally occurring molecules with acquired or mutated.
  • the present invention provides a pharmaceutical pack or kit comprising one or more containers filled with one or more components of the vaccine formulations of the present invention.
  • the invention provides a vaccine or antigenic composition that induces an immunity to an infection or at least one disease symptom thereof in a mammal, comprising the step of adding an effective dose of a modified or mutated RSV F protein to the agent Thereby providing a method of preparing the composition.
  • the infection is a RSV infection.
  • the modified or mutated RSV F protein of the present invention is useful for producing compositions that stimulate an immune response that provides immunity or substantial immunity to an infectious agent.
  • the invention provides a method of inducing immunity against a subject's infection or at least one disease symptom thereof, comprising administering at least one effective dose of a modified or mutated RSV F protein.
  • the invention provides a method of inducing substantial immunity to a subject's RSV viral infection or at least one disease symptom, comprising administering at least one effective dose of a modified or mutated RSV F protein.
  • compositions of the present invention can induce substantial immunity in a vertebrate animal (e.g., a human) when administered to a vertebrate animal.
  • a vertebrate animal e.g., a human
  • the invention provides a method of inducing a substantial immunity to a subject ' s RSV viral infection or at least one disease symptom, comprising administering at least one effective dose of a modified or mutated RSV F protein do.
  • the method may further comprise purifying the expressed RSV F protein or fragment thereof.
  • the recombinant viral vector may be, for example, a phage, a plasmid, a virus or a retroviral vector, and preferably a viral vector may be used.
  • the vector is a recombinant baculovirus vector.
  • the expression constructs will further include a ribosome binding site for translation, at the site for transcription initiation, for termination and at the transcribed region.
  • the coding portion of the transcripts expressed by the constructs will preferably initially contain a translation initiation codon and a termination codon appropriately located at the end of the polypeptide to be translated.
  • the expression vectors comprise at least one selectable marker. These markers include neomycin resistance genes for dihydrofolate reductase, G418 or eukaryotic cell cultures, and tetracycline, kanamycin, or resistance genes for E. coli and other bacterial cultures .
  • Baculoviridae for example, Virus-Autographa californica nucleopolyhedrovirus
  • Adenoviridae e.g., canine adenovirus-canine adenovirus
  • Hepadnaviridae E.g., Hepadnaviridae, e.g., aviepadnavirus
  • Vacciniaviridae e.g., modified vaccinia Ankara virus
  • Parvoviridae e. G.
  • a virus vector selected from the group consisting of a virus, a virus, an autonomous parvovirus, and the like.
  • the baculovirus is selected from the group consisting of Autographa californica nucleoside varicella virus or a modified virus strain; Or a Bombyx mori nucleoside viral virus strain or a modified viral strain thereof.
  • Bacterial vectors may also be used.
  • Exemplary bacterial vectors include pQE70, pQE60 and pQE-9, p BlueScript vector, phage script vector, pNH8A, pNH16a, pNH18A, pNH46A, ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5.
  • pFastBacl pWINEO pSV2CAT, pOG44, pXTl, pSG, pSVK3, pBPV, pMSG, and pSVL are preferred.
  • Eukaryotic host cells may include yeast, insect, avian, plant, small nematode (or nematode), and mammalian host cells.
  • Non-limiting examples of insect cells are, for example, Trichoplusiani cells such as Spodoptera frugiperda (Sf) cells such as Sf9, Sf21, and High Five cells, and Drosophila S2 cells.
  • yeast Trichoplusiani cells
  • Sf Spodoptera frugiperda
  • fungal (including yeast) host cells include S. cerevisiae, Kluyveromyces lactis (K. lactis), C. albicans and C.
  • mammalian cells include human embryonic kidney lineage, Chinese hamster ovary cell lineage, Vero cell line (African green monkey lineage), MRC cell line (human lung fibroblast cell lineage), and MDCK cells (Madin-darby canine kidney cell lineage). Other cells of the African claw frog (Xenopus laevis oocyte) or amphibians can also be used.
  • Prokaryotic host cells include, for example, bacterial cells such as E. coli, B. subtilis and mycobacteria.
  • An embodiment of the present invention is a method for producing a protein having the amino acid sequence of SEQ ID NO: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, RSV comprising at least one protein selected from the group consisting of SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: Provide a preventive vaccine.
  • RSV preventive vaccine comprising any one or more proteins selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO:
  • One embodiment of the present invention is directed to an isolated nucleic acid molecule comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 25, 26, 27, 28, 29, 30, 31, 32, , SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43 and SEQ ID NO: Protein-containing RSV vaccine.
  • one embodiment of the present invention provides a RSV preventive vaccine comprising a protein encoded by any one or more nucleic acids selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 and SEQ ID NO: More preferably, an embodiment of the present invention provides a RSV preventive vaccine comprising a protein encoded by any one or more nucleic acids selected from the group consisting of SEQ ID NO: 25, SEQ ID NO: 26 and SEQ ID NO: 27.
  • the vaccine may further comprise adjuvants or immunostimulants.
  • adjuvants or immunostimulants for example, an aluminum adjuvant.
  • the immunogenic compositions of the present invention include any pharmaceutical material that does not itself induce a harmful immune response to the vertebrate to which the composition is administered and may be any suitable diluent that can be administered with the RSV F polypeptide Or a pharmaceutically acceptable carrier comprising an excipient.
  • pharmaceutically acceptable refers to those listed in the United States Pharmacopoeia, European Pharmacopoeia, or other commonly recognized pharmacopoeia for use in vertebrates and more specifically in humans.
  • the RSV F immunogen of the invention is administered in an effective amount or amount (as defined above) sufficient to stimulate an immune response against one or more strains of the RSV virus.
  • Such compositions can be used as vaccine and / or immunogenic compositions for inducing a protective immune response in vertebrates.
  • the composition may contain other RSV F proteins or fragments thereof.
  • the concentration of the immunogen is at least about 10 ⁇ g / mL, about 20 ⁇ g / mL, about 30 ⁇ g / mL, about 40 ⁇ g / mL, about 50 ⁇ g / mL, About 100 ⁇ g / mL, about 200 ⁇ g / mL, or about 500 ⁇ g / mL.
  • the concentration of the immunogen is from about 10 ⁇ g / mL to about 1 mg / mL, or from about 20 ⁇ g / mL to about 500 ⁇ g / mL, or from about 30 ⁇ g / mL to about 100 ⁇ g / mL, To about 50 [mu] g / mL.
  • the concentration of the immunogen may be comprised between 10 ⁇ g / mL and 200 ⁇ g / mL.
  • the pharmaceutical formulations disclosed herein comprise a RSV F protein, predominantly a spike protein; And a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical agent comprises a purified, high affinity antibody produced in an animal to which the immunogen is administered.
  • Pharmaceutically acceptable carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, sterile isotonic aqueous buffer and combinations thereof.
  • Pharmaceutically acceptable carriers, diluents and other excipients are provided in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. current edition).
  • the formulation should be appropriate for the mode of administration.
  • the formulation is suitable for administration to humans, preferably sterile, not particulate and / or pyrogenic. If desired, the composition may contain minor amounts of wetting or emulsifying agents or pH buffering agents.
  • the composition may be in the form of a solid, liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release preparation or powder, such as a lyophilized powder suitable for recombination.
  • Oral preparations may include standard carriers such as mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the components of the immunogenic vaccine formulations.
  • the kit comprises two containers, one comprising an RSV F immunogen and the other comprising an antigen adjuvant.
  • Announcements in the form prescribed by governmental bodies governing the manufacture, use or sale of medicinal or biological products may be incorporated into such container (s), and such announcements may be made by the institution of manufacture, use or sale for human administration Indicates approval.
  • the agent may be packaged in a sealed container such as an ampoule or sachette indicating the amount of the composition.
  • the composition is supplied as a liquid, and in another embodiment, is supplied as a dry sterile lyophilized powder or water-removing concentrate in a sealed container, for administration to a subject, for example, with water or saline It can be reconstituted to an appropriate concentration.
  • the composition will preferably contain about 1 ⁇ g, about 5 ⁇ g, about 10 ⁇ g, about 20 ⁇ g, about 25 ⁇ g, about 30 ⁇ g, about 50 ⁇ g, about 100 ⁇ g, about 125 ⁇ g, about 150 ⁇ g, As a dry sterile lyophilized powder in an airtight container at a unit dose of about 200 ⁇ g.
  • the unit dose of the composition may be about 1 microgram (e.g., about 0.08 microgram, about 0.04 microgram, about 0.2 microgram, about 0.4 microgram, about 0.8 microgram, about 0.5 microgram, about 0.25 microgram, ) Or greater than about 125 ⁇ g (eg, greater than about 150 ⁇ g, greater than about 250 ⁇ g, or greater than about 500 ⁇ g).
  • Such doses can be measured as [mu] g of total RSV F protein (e.g., spike protein or fragment thereof).
  • the immunogen of the present invention should be administered within about 12 hours, preferably within about 6 hours, within about 5 hours, within about 3 hours, or within about 1 hour after reconstitution with lyophilized powder.
  • the RSV F protein immunogenic composition is supplied in liquid form in a sealed container representing the amount and concentration of the RSV F protein composition.
  • the liquid form of the immunogenic composition of the invention is at least about 50 ⁇ g / ml, more preferably at least about 100 ⁇ g / ml, at least about 200 ⁇ g / ml, at least 500 ⁇ g / ml, or at least 1 mg / ml And is supplied to the sealed container.
  • the vaccine or immunogenic composition of the invention may be administered to an animal to induce an immune response against RSV.
  • the animal is vulnerable to RSV infection.
  • the animal is a human.
  • administration of the immunogen induces substantial immunity against at least one RSV strain, isolate, clade and / or species.
  • administration of an immunogen induces substantial immunity against at least two RSV strains, isolates, clades and / or species.
  • the dosage can be adjusted within this range based on, for example, age, physical condition, body weight, age, food, time of administration, and other clinical factors.
  • the invention includes a method of formulating a vaccine or immunogenic composition that results in substantial immunity to an infection of the subject or at least one symptom thereof, including the step of adding an effective amount of an immunogen to the formulation.
  • Stimulation of substantial immunity by a single dose is preferred, but additional doses may be administered via the same or different routes to achieve the desired effect. In neonates and infants, for example, multiple administrations may be necessary to induce a sufficient level of immunity. Administration can be continued at intervals over the childhood period if necessary to maintain adequate protection against infection.
  • a method of inducing substantial immunity against a viral infection or at least one symptom thereof in a subject comprises administering at least one effective amount of an RSV F protein, or fragment or aggregate thereof.
  • Methods of administering the vaccine and / or immunogenic agent include parenteral administration (e.g., endothelium, intramuscular, intravenous and subcutaneous), epidural and mucosal (e.g., nasal and oral or pulmonary routes or suppositories) But is not limited thereto.
  • parenteral administration e.g., endothelium, intramuscular, intravenous and subcutaneous
  • epidural and mucosal e.g., nasal and oral or pulmonary routes or suppositories
  • the composition is administered intramuscularly, intravenously, subcutaneously, orally or intradermally.
  • the composition may be administered orally or parenterally by absorption through, for example, injection or transient injection, epithelium or mucosal (e.g., oral mucosa, colon, conjunctiva, nasopharyngeal, pharyngeal, vaginal, urinary, bladder, May be administered by any convenient route and may be administered with other biologically active substances.
  • administration via the nasal passage or other mucosal route may result in a substantially higher antibody or other immune response than other routes of administration.
  • the nasal or other mucosal pathways of administration of the immunogenic composition and / or vaccine may induce antibodies or other immune responses that will induce cross protection against other strains of the virus. Administration may be systemic or local.
  • the prophylactic vaccine preparation is administered systemically by subcutaneous or intramuscular injection using a needle and injection or by a needle-free injection device.
  • the vaccine preparation is administered into the nasal cavity by drops, large particle aerosols (greater than about 10 microns), or by injection into the conduit. Any of the above pathways of delivery may cause an immune response while nasal administration provides an increased effect of inducing mucosal immunity at the site of penetration of the virus.
  • the vaccine and / or immunogenic agent is administered in a manner that targets mucosal tissue to cause an immune response to the site of immunization.
  • mucosal tissues such as gut associated lymphoid tissue (GALT) may be targets for immunization by using oral administration of a composition containing an immunogen adjuvant with specific targeting properties.
  • Other mucosal tissues such as nasopharyngeal lymphoid tissue (NALT) and bronchial-associated lymphoid tissue (BALT) may also be targets.
  • the vaccine and / or immunogenic agent may be administered according to a dosing schedule such as administration of the initial vaccine composition followed by intensification of the administration.
  • the second dose of the composition is administered at any time between two weeks to one year after the initial administration, preferably about 1, about 2, about 3, about 4, about 5 to about 6 months.
  • the third dose may also be administered after the second dose and after about 3 months to about 2 years, preferably about 4, about 5, or about 6 months, or about 7 months to about 1 year after the first dose.
  • the third dose may be administered orally when there is no specific immunoglobulin in the subject's serum and / or urine or mucosal secretions after a second dose, or when a small amount of specific immunoglobulin is detected.
  • the second dose is administered about one month after the first dose and the third dose is administered about six months after the first dose.
  • the second dose is administered about 6 months after the first dose.
  • an immunogen comprising a RSV F protein can be administered as part of a combination therapy.
  • the RSV F protein or fragment thereof or a collection thereof may be formulated with other immunogenic compositions and / or antiviral agents.
  • Dosages of the pharmaceutical preparations can be determined, for example, by first determining the effective dose to induce a prophylactic or therapeutic immune response by measuring the serum titer of a virus-specific immunoglobulin or by measuring the inhibition rate of antibodies in serum samples or urine samples or mucosal secretions Can be easily determined by those skilled in the art.
  • human clinical studies can be performed by those skilled in the art to determine a desirable effective amount for humans. These clinical studies are routine and well known in the art. The exact dose to be used will depend on the route of administration.
  • an effective amount can be estimated from a dose-response curve derived from an in vitro or animal testing system.
  • the immunity of certain compositions can be improved by using nonspecific stimulators of the immune response, known as antigen-adjuvants.
  • Antigen adjuvants have been used to experimentally improve the general increase in immunity against unknown immunogens (e. G., U.S. Patent No. 4,877,611).
  • Immunization protocols have used antigenic adjuvants to stimulate responses for many years, and antigenic adjuvants are well known to those skilled in the art.
  • Some antagonists affect the way antigens are present. For example, an immune response increases when protein antigens are immersed in alum. Emulsification of the antigen prolongs the antigen delivery period.
  • Antigen adjuvants may be included. Suitable antigenic adjuvants include those described in Vogel et al., &Quot; A Compendium of Vaccine Adjuvants and Excipients (2nd Edition), which are incorporated by reference in their entirety for all purposes. Other exemplary antigenic adjuvants include the complete Freund ' (Non-specific irritant of immune response containing dead Mycobacterium tuberculosis), incomplete Freund's adjuvant and aluminum hydroxide adjuvant. Other antagonists include GMCSP, BCG, aluminum hydroxide, thur-MDP and nor- (MDP), CGP (MTP-PE), lipid A, montanide ISA 206 and monophosphoryl lipid A (MPL).
  • Bacteria, MPL, trehalulose dimycolate (TDM) and (CWS) RIBI is considered to contain three components extracted from the cell wall skeleton (CWS) in a 2% squalene / tween 80 emulsion.
  • MF-59, Novasom®, and MHC antigens may also be used.
  • an adjuvant is a pouch sila melanoma lipid vesicles (paucilamellar lipid vesicle) substantially with a two to ten bilayers arranged in a rectangular cover type separated by aqueous layers surrounding a large amorphous central cavity of the lipid bilayer is removed.
  • the adjuvant effect is achieved by the use of a material such as alum, and is used as a 0.05 to about 0.1% solution in phosphate buffered saline.
  • the immunogen may be prepared with a linear mixture of a synthetic polymer of sugars (Carbopol®) used in about 0.25% solution.
  • An example is the muramyldipeptide (N-acetylmuramyl-L-alanyl-D-isoglutamine [MDP]), a bacterial peptidoglycan.
  • MDP muramyldipeptide
  • hemocyanin and hemoeryritin can be used.
  • Hemostatic from the keyhole limpet (KLH) is preferred in certain embodiments, although mollusks and arthropod hemoshihenian and hemoeryritin can be used.
  • a variety of polysaccharide adjuvants may be used.
  • the use of pneumococcal polysaccharide antigen adjuvants for antibody responses in mice has been disclosed (Yin et al, 1989).
  • Doses that do not produce an optimal response or inhibition should be used as directed (Yin et al, 1989).
  • Polyamine modifications of the polysaccharides are particularly like chitin and chitosan, including chitin and deacetylated chitin.
  • the muramyldipeptide lipophilic daiskaride-tripeptide derivatives disclosed for use in artificial liposomes have been formed from phosphatidyl choline and phosphatidyl glycerol.
  • suitable adjuvants include bipolar surface active agents, such as saponin and derivatives such as QS21 (Cambridge Biotech).
  • Saponin-based antigen adjuvants include those which contain substrate A and substrate C alone and in combination. Nonionic block copolymer surfactants (Rabinovich et al, 1994) can be used. Oligonucleotides are another useful group of adjuvants (Yamamoto et al, 1988).
  • Another group of antigen adjuvants are the decrypted endotoxins such as the purified decoded endotoxin of U.S. Patent No. 4,866,034. These purified endotoxins are effective in causing an adjuvant response in vertebrates.
  • the detoxified endotoxin can bind other antagonists to produce a multivalent-antigen adjuvant preparation.
  • Alkyl lysophospholipids ALP
  • BCG BCG
  • antigen-adjuvant that can be conjugated to a vaccine comprising biotin (including biotinylated derivatives).
  • biotin including biotinylated derivatives.
  • One particular antigen-reinforcing agent that specifically contemplates use is teichoic acid derived from gram-cells. This includes lipoteichoic acid (LTA), ribitol teico acids (RTA) and glycerol teico acids (GTA). Active forms of such synthetic counterparts can also be used (Takada et al, 1995).
  • antigen adjuvants that are not conventionally used in humans can still be used in other vertebrates, for example, when it is desired to generate antibodies or subsequently to obtain active T cells.
  • the toxicity or other adverse effects that may arise from cells such as those that can occur using an antigen reinforcement or, for example, non-irradiated tumor cells, are independent of this environment.
  • Other methods of inducing an immune response can be accomplished by formulating the immunogen of the invention with an " immunostimulant ". These are the body's own chemical messengers (cytokines) to increase the response of the immune system.
  • Immunostimulants are immunosuppressive, immune-enhancing and inflammatory cytokines, such as interleukins (e.g., IL-1, IL-2, IL-3, IL-4, IL- Cain, lymphokine and chemokine; Other immune stimuli such as growth factors (e.g., granulocyte-macrophage (GM) -colony stimulating factor (CSF)) and macrophage inflammatory factors, Flt3 ligand, B7.1, B7.2, The immunostimulatory molecules may be administered to the same formulation as an immunogen or may be administered separately. [0064] Expression vectors encoding proteins or proteins may be administered to produce an immunostimulatory effect.
  • interleukins e.g., interleukins (e.g., IL-1, IL-2, IL-3, IL-4, IL- Cain, lymphokine and chemokine
  • Other immune stimuli such as growth factors (e.g., granulocyte-macrophage (GM)
  • the saponin-based antigen adjuvant may be present in a range with the following lower limits: about 0.2 ⁇ g, about 0.4 ⁇ g, about 0.6 ⁇ g, about 0.8 ⁇ g, about 1 ⁇ g, about 2 ⁇ g, about 3 ⁇ g Saponin-based antigen-adjuvant is administered at a dosage of at least about 1 mg / kg, preferably at least about 10 mg / kg, about 4 mg / kg, about 5 mg / About 15 ⁇ g, about 20 ⁇ g, about 25 ⁇
  • the saponin-based adjuvant is from about 5 ⁇ g to about 20 ⁇ g or from about 1 ⁇ g to about 10 ⁇ g.
  • Such doses are particularly suitable in mice and can be adjusted for human use based on a typical mouse weight of 20 g versus a human body weight of about 60 kg.
  • the RSV F protein, fragment or aggregate thereof more preferably SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID NO: 11, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: , And SEQ ID NO: 23, or a vaccine comprising the RSV F immunoconjugate composition, wherein the RSV F immunogenic composition comprises at least one protein selected from the group consisting of SEQ ID NO: 23 and SEQ ID NO: 23.
  • the method comprises administering a RSV F immunogenic composition comprising a protein selected from the group consisting of SEQ ID NO: 3, SEQ ID NO: 4, and SEQ ID NO: 5 or a vaccine comprising the same , A method of inducing preventive immunity against RSV F infection.
  • a method of inducing an immune response to RSV comprising administering to a subject a composition comprising the RSV F protein antigen.
  • the method can reduce or prevent RSV infection.
  • the present invention provides immunogenic compositions for the prevention and / or treatment of RSV infection.
  • the present invention is able to achieve a more improved RSV F protein expression in the host cell than the wild-type RSV F protein through mutation of the RSV F protein.
  • the present invention can achieve high immunogenicity of the RSV F protein. That is, an antigen prepared through the sequence variation of the RSV F protein of the present invention (and / or the RSV F protein expression base sequence variation) can provide excellent neutralizing antibody titer and achieve excellent immunogenicity.
  • the present invention can provide a stabilized and soluble RSV F protein.
  • the present invention is applicable to a variety of cell lines and can provide a safe RSV immunogenic composition, more preferably a vaccine.
  • the RSV immunogen completed through the variation of the present invention can achieve a long duration of immune maintenance in the human body.
  • the RSV immunogen completed through the variation of the present invention is less likely to cause repeated infections.
  • the RSV immunogen which is completed through the mutation of the present invention, can induce excellent immunogenicity due to low interference with blood antibodies in the body.
  • FIG. 1 is a schematic diagram showing the variation of the RSV F protein mainly performed in the present invention.
  • FIG. 1 is a schematic diagram showing the variation of the RSV F protein mainly performed in the present invention.
  • FIG. 2 is an experiment for confirming the expression amount of the RSV F protein mutation performed in the present invention. This experiment measured the reactivity with palivizumab binding to the site II epitope. It was confirmed that the mutations tried in the present invention are superior to the mutation forms known in the art.
  • Fig. 3 shows results of western blotting and coomassie staining of the proteins expressed and purified through the mutations performed in the present invention.
  • 3-a is the result of confirming F0 and F1 using ⁇ ME.
  • 3-b is the result of confirming FP1 and FP4 among the final purified mutant proteins.
  • FIG. 4 is a transmission electron microscopy photograph of RSV F protein prepared according to the present invention.
  • FIGS. 5A and 5B are graphs showing the results of measurement of total antibody titers to examine the induction of the mouse immunogenicity of the RSV F protein prepared according to the present invention.
  • the total antibody titers are the values determined by ELISA analysis.
  • GMT is the Geometric Mean Titer.
  • 1ug + Adjuvant " 10ug + Adjuvant”
  • 30ug + Adjuvant are substances adsorbed to 1ug, 10ug and 30ug of RSV F protein, respectively, in aluminum adjuvant.
  • the PBS treatment time was 1.
  • FIG. 6 is a schematic diagram of an experimental result for confirming the concentration of a specific antibody capable of competing with palivizumab binding to a site II epitope in mouse serum in which immunity was induced by the RSV F protein prepared in the present invention.
  • FIGS. 7A and 7B are graphs showing experimental results for confirming the concentration of a specific antibody capable of competing with palivizumab binding to a site II epitope in a mouse serum immunized with the RSV F protein prepared according to the present invention.
  • FIG. The result value means a GMT value which suppresses 50% of palivizumab.
  • FIGS. 8A and 8B are experiments to confirm the inhibition of the infection of the wild-type RSV virus in the mouse serum in which immunity was induced by the RSV F protein prepared according to the present invention.
  • the wild-type virus used here is RSV A2 (ATCC. VR-1540). Through the neutralizing immunity test, it was confirmed that the RSV F protein produced by the present invention inhibited the infection of the wild-type RSV A2.
  • FIG. 9 shows the screening results of soluble F protein of FP1, 3, 4, and 6.
  • Variations in the position of the fusion peptide of the present invention may serve as excellent RSV immunogen, Respectively.
  • the mutation mutated the fusion peptide portion (amino acid sequence 137-145).
  • the RSV F protein is a mutation that transforms the hydrophobic portion of the fusion peptide portion into a hydrophilic portion. Specifically, the hydrophobic part of the sequence FLGFLLGVG of the fusion peptide was substituted with A and transformed into AAGAAAGAG (SEQ ID NO: 3). For the purpose of the present invention, the optimum effect can be obtained as a result of substitution with A among hydrophilic amino acids.
  • the polar portion of FLGFLLGVG in the fusion peptide portion was modified to a nonpolar side chain, and the modified fusion peptide portion was modified to QNGQNNGSG and NSGNSSGGG (SEQ ID NO: 4 and SEQ ID NO: 5, respectively).
  • the FLGFLLGVG portion of the fusion peptide portion was modified to TLSKKRKRR (SEQ ID NO: 6).
  • Additional mutations that maintain the stabilization of the F1 portion of the wild RSV F protein further include at least one additional mutation selected from the group consisting of (a) to (g) based on SEQ ID NO: 2.
  • RSV F protein antigens of the present invention are based on SEQ ID NO: 2
  • the RSV F protein antigen is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • the amino acid corresponding to position 140 of SEQ ID NO: 2 is substituted with W, the amino acid corresponding to 163 is substituted with Q, the amino acid corresponding to 188 is substituted with Q, the amino acid corresponding to 189 is substituted with L No. 15);
  • amino acid corresponding to position 163 of SEQ ID NO: 2 is substituted with Q, the amino acid corresponding to 505 is replaced with W (SEQ ID NO: 20);
  • An amino acid corresponding to position 188 of SEQ ID NO: 2 is substituted with Q, an amino acid corresponding to 189 is substituted with L, and an amino acid corresponding to 505 is substituted with W (SEQ ID NO: 21);
  • amino acid corresponding to position 163 of SEQ ID NO: 2 may be substituted with Q and the amino acid corresponding to 487 may be substituted with L (SEQ ID NO: 22).
  • this includes using known methods of protein processing and recombinant DNA technology to enhance or modify the properties of the mutated RSV F proteins mentioned above.
  • Genes that encode the proteins are modified by codonification, and modifications of the nucleotides are known to those skilled in the art.
  • Various forms of mutagenesis can be used to produce and / or isolate variant nucleic acids encoding the protein molecules and / or further modify / mutate the proteins of the invention.
  • mutagenesis include, but are not limited to, site-specific mutagenesis, random point mutagenesis, homologous recombination (DNA shuffling), mutagenesis using uracil containing template, oligonucleotide-induced mutagenesis, phosphorothioate-modified DNA mutagenesis, DNA (gapped-duplex DNA), and the like.
  • Other suitable methods include point mismatch repair, repair-deficient host strains, restriction-selection and restriction-mutagenesis using purification, deletion mutagenesis, total gene synthesis, double-strand breakage Recovery, and the like.
  • mutagenesis involving a chimeric structure is also encompassed by the present invention.
  • mutagenesis can be guided by known information, such as sequences, sequence comparisons, physical properties, crystal structures, etc., of molecules that have been generated pre-existing or of naturally occurring molecules with acquired or mutated.
  • the gene mutation for the F protein described in the previous example was confirmed by expressing the protein using 293FT cells.
  • the candidate substance plasmid to which each mutation was applied was inserted into a vector containing a eukaryotic promoter (promoter) and the degree of expression was confirmed.
  • the vector used in one embodiment is pcDNA3.1. It will be appreciated by those skilled in the art that not only pcDNA3.1 but also other eukaryotic expression vectors can be used as the vector.
  • ELISA was used for the evaluation of the expression level.
  • the antibody used was palivizumab, which binds to RSV site II. As can be seen in FIG. 2, the screening results of the expressed proteins for the applied mutations showed a higher level of expression than the mutations of the known patents.
  • lysis Medium & Pellet
  • lysis Medium
  • lysis Medium
  • TMB acts as an electron donor to reduce hydrogen peroxide and the like by peroxidase such as horseradish peroxidase (HRP).
  • HRP horseradish peroxidase
  • RSV-B F (SEQ ID NO: 1) The codons of the proteins were optimized and the antigens obtained through the gene mutation described above were prepared.
  • the described RSV F gene was cloned into RSV F protein recombinant baculovirus (AcMNPV) and then inoculated into insect cells, Sf9.
  • Sf9 insect cells The normal cell concentration of Sf9 insect cells should not exceed 3.00E6 / ml.
  • Sf9 insect cells are cultured in sterile 125ml / 250ml / 500ml / 1L spinner flask or 5L / 50L / 100L bioreactor.
  • serum-free serum-insect cell-specific medium containing no serum was used for the culture.
  • the serum-free insect cell-specific medium may include Insect Express (Lonza).
  • the RSV F recombinant baculovirus for RSV F protein production is inoculated between 0.01-0.8 MOI (multiplicity of infection), wherein the concentration of Sf9 insect cells is in the range of 5.00E5-1.50E6, the initial stage of the exponential phase Lt; / RTI >
  • the RSV F recombinant baculovirus used in this example harvests at 60-90 hours after inoculation, with viability between 75-98%.
  • the reason why low-level viruses are inoculated to low-growth cells and harvested at relatively high viability is to minimize the target protein cleavage by proteases exiting the cells in the apoptosis stage and to reduce the amount of virus inoculum used It is to minimize.
  • the Sf9 insect cells obtained after the inoculation of the recombinant baculovirus of Example 3 are centrifuged at 3000-8000xg for 1-50 minutes to separate the cell sedimentation layer and the media supernatant. Remove the separated cell sedimentation layer and secure media supernatant. Infection of the recombinant baculovirus can occur effectively when the cells are in the early-log phase of growth and are in the 1.0E5-7.0E6 cell / ml concentration, preferably 5.00E5-2.50E6 cells / ml concentration.
  • RSV F RSV F
  • affinity chromatography affinity chromatography
  • affinity chromatography affinity chromatography
  • the crude extract was further purified by passing it through anion exchange chromatography, lentile lectin affinity and cation exchange chromatography.
  • RSV F RSV F
  • affinity chromatography affinity chromatography
  • affinity chromatography as known in the art.
  • the crude extract was further purified by passing it through anion exchange chromatography, lentile lectin affinity and cation exchange chromatography
  • a sample of the same volume of each purification step was diluted with 2xSDS sample buffer containing ⁇ ME (beta-mercaptoethanol), filled with 15-20 ⁇ l of each SDS-gel, electrophoresed, and stained with Coomassie stain Lt; / RTI > to stain whole proteins.
  • SDS-gel was transferred to the membrane and further analyzed by WB (Western blot) using anti-single or multiple RSV F-specific antibodies.
  • mice Animal experiments using mouse as the antigen obtained in this example were carried out.
  • the mouse used in the experiment can be used as an antigen used in the animal experiment itself and can also be used in combination with an immunostimulant such as an aluminum adjuvant.
  • the adjuvant may include, for example, aluminum or calcium salts, in particular inorganic salts such as hydroxide, phosphoric acid, calcium phosphate and the like.
  • Aluminum or Alum-based adjuvants are currently the most commonly used adjuvants in human vaccines. Therefore, adjuvants, which are proven to be stable and effective enough to be used as a standard for developing and evaluating new adjuvants, to be.
  • alum adjuvants aluminum hydroxide (Al (OH) 3 ) and aluminum phosphate (AlPO4) are typical examples. These two alum adjuvants exhibit different physical and adjuvant properties.
  • aluminum hydroxide is most widely used in aluminum adsorption vaccine. In this example, aluminum hydroxide was used as an immunity enhancer, and aluminum hydroxide was used at a level of 180 ug / once administration.
  • the concentration of RSV F antigen administered in this example was administered at different concentrations of 1 ug / ml, 10 ug / ml and 30 ug / ml, and the administration frequency was administered twice.
  • the dosing schedules in this example were administered 2 times after 2 weeks of the first administration, and blood sampling was performed at 2 weeks and 4 weeks after the first administration.
  • the immunogenicity inducing effect of the RSV F antigen obtained in this example was confirmed. Identification of the immunogenicity induced in this example was performed by total antibody measurement, PCA measurement, and neutralizing antibody measurement.
  • total antibody titers to identify antigen-specific IgG antibody titers were determined using ELISA.
  • 100 ng of RSV F antigen was coated on a 96-well plate and incubated overnight at 4 ° C.
  • the incubated plate is washed three times with buffer containing 0.05% tween 20, then incubated with buffer solution containing 5% skim milk for 2 hours and washed again with buffer solution containing 0.05% tween 20.
  • the obtained mouse blood is centrifuged to obtain a serum sample, the prepared serum sample is diluted to 1/20 of the original concentration, and then serial dilution is performed four times to make the final concentration 1/327680 times.
  • Diluted serum samples are plated on 100 ⁇ l / well plates, incubated at room temperature for 2 hours, and washed three times with buffer containing 0.05% tween 20.
  • 100 ⁇ l of goat anti-mouse IgG antibody (Invitrogen, US) combined with 1: 5000 diluted HRP-conjugated goat anti-mouse IgG was added to the washed plate, incubated at room temperature for 1 hour, Wash three times with the solution.
  • TMB substrate 3,3A, 5,5A-tetramethylbenzidine supra
  • KPL TMB stop solution
  • a competitive ELISA was performed using the palivizumab antigen that binds to the site II epitope to confirm that the expressed / purified RSV F immunogen induces a neutralizing antibody corresponding to the site II epitope of the RSV F protein .
  • 200 ng of RSV F antigen was coated on a 96-well plate and incubated overnight at 4 ° C. The incubated plate is washed three times with buffer containing 0.05% tween20, incubated with buffer solution containing 5% skim milk for 2 hours, and washed again with buffer solution containing 0.05% tween20.
  • the obtained mouse blood is centrifuged to obtain a serum sample, the prepared serum sample is diluted to 1/10 of the original concentration, and then serial dilution is performed twice to make the final concentration 1/1280 times.
  • Biotin-conjugated paribizumab is diluted to 50 ng per well and serial dilutions are performed in duplicate to achieve a final concentration of 390 pg per well. Diluted serum samples and biotin-conjugated paribisuram are incubated for 2 hours and washed three times with buffer containing 0.05% tween20.
  • Hep2 cells were seeded in a 24-well plate at a rate of 3.5E5 per well and cultured at 37 ° C in 5% CO 2 for one day. Serum samples from animal studies are diluted to 1/20 of the original concentration and serial dilutions are performed in triplicate to make the final concentration 1/4860. 50 ⁇ l of the diluted serum sample and 50 ⁇ l of RSV virus (40 pfu) are mixed and incubated at room temperature for 1 hour, then inoculated onto Hep2 cells of the plate seeded the day before. After 5 days of inoculation, the cells are cultured at 37 ° C and 5% CO 2 . After incubation, the plates are stained with neutral red and neutralized. The neutralization value was calculated to provide a value (ND50) that protects 50% of the virus. (Fig. 8)
  • the present invention provides a soluble RSV.
  • the soluble RSV may be provided as a vaccine composition capable of preventing RSV infection.

Abstract

La présente invention concerne une protéine F du virus respiratoire syncytial (RSV) soluble et comprend des compositions immunogènes telles que des vaccins pour le traitement et/ou la prévention d'une infection par le RSV.
PCT/KR2018/011324 2017-09-29 2018-09-21 Antigène de protéine f du virus respiratoire syncytial (rsv) modifié soluble WO2019066437A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2017-0128003 2017-09-29
KR20170128003 2017-09-29
KR10-2018-0113291 2018-09-20
KR1020180113291A KR20190038358A (ko) 2017-09-29 2018-09-20 가용성 변형 호흡기 융합세포 바이러스 (rsv) f 단백질 항원

Publications (1)

Publication Number Publication Date
WO2019066437A1 true WO2019066437A1 (fr) 2019-04-04

Family

ID=65901591

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/011324 WO2019066437A1 (fr) 2017-09-29 2018-09-21 Antigène de protéine f du virus respiratoire syncytial (rsv) modifié soluble

Country Status (1)

Country Link
WO (1) WO2019066437A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3875471A4 (fr) * 2018-11-01 2022-06-01 SK Bioscience Co., Ltd. Protéine f recombinée du virus respiratoire syncytial et composition de vaccin la contenant
CN115850396A (zh) * 2022-12-22 2023-03-28 北京吉诺卫生物科技有限公司 一种rsv纳米颗粒疫苗及其制备方法与应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009128951A2 (fr) * 2008-04-18 2009-10-22 Vaxinnate Corporation Compositions de protéines du virus respiratoire syncytial et leurs méthodes d'utilisation
US20140024076A1 (en) * 2011-01-28 2014-01-23 Medimmune, Llc. Expression Of Soluble Viral Fusion Glycoproteins In Mammalian Cells
US20140271699A1 (en) * 2013-03-13 2014-09-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Prefusion rsv f proteins and their use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009128951A2 (fr) * 2008-04-18 2009-10-22 Vaxinnate Corporation Compositions de protéines du virus respiratoire syncytial et leurs méthodes d'utilisation
US20140024076A1 (en) * 2011-01-28 2014-01-23 Medimmune, Llc. Expression Of Soluble Viral Fusion Glycoproteins In Mammalian Cells
US20140271699A1 (en) * 2013-03-13 2014-09-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Prefusion rsv f proteins and their use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MCLELLAN, JASON S. ET AL.: "Structure-based Design of a Fusion Glycoprotein Vaccine for Respiratory Syncytial Virus", SCIENCE, vol. 342, 1 November 2013 (2013-11-01), pages 592 - 598, XP055391357 *
PIERANTONI, ANGIOLO ET AL.: "Mucosal Delivery of a Vectored RSV Vaccine Is Safe and Elicits Protective Immunity in Rodents and Nonhuman Primates", MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT, vol. 2, no. 15018, 2015, pages 1 - 11, XP055524844 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3875471A4 (fr) * 2018-11-01 2022-06-01 SK Bioscience Co., Ltd. Protéine f recombinée du virus respiratoire syncytial et composition de vaccin la contenant
CN115850396A (zh) * 2022-12-22 2023-03-28 北京吉诺卫生物科技有限公司 一种rsv纳米颗粒疫苗及其制备方法与应用
CN115850396B (zh) * 2022-12-22 2024-02-06 北京吉诺卫生物科技有限公司 一种rsv纳米颗粒疫苗及其制备方法与应用

Similar Documents

Publication Publication Date Title
WO2011043584A2 (fr) Vaccin protéique contre le virus de la grippe aviaire hautement pathogène, issu de plantes transgéniques et son procédé de préparation
WO2018074884A1 (fr) Protéine recombinante et son utilisation
WO2019209079A1 (fr) Molécules d'acide nucléique dans lesquelles des séquences de régulation d'expression sont insérées, vecteur d'expression comprenant les molécules d'acide nucléique et leur utilisation pharmaceutique
WO2010147268A1 (fr) Vaccin contre le cancer du col de l'uterus
WO2020091529A1 (fr) Protéine f recombinée du virus respiratoire syncytial et composition de vaccin la contenant
WO2019039891A1 (fr) Anticorps monoclonal pour la protéine de spicule du coronavirus du syndrome respiratoire du moyen-orient, et son utilisation
WO2019066437A1 (fr) Antigène de protéine f du virus respiratoire syncytial (rsv) modifié soluble
WO2019203599A1 (fr) Polysaccharides capsulaires de streptococcus pneumoniae et conjugués immunogènes de ceux-ci
WO2021158073A1 (fr) Anticorps de fusion permettant la présentation d'un épitope d'antigène de lymphocyte t dérivé d'un antigène ou d'un peptide le contenant sur une surface cellulaire, et composition le comprenant
KR20180058206A (ko) 중동호흡기증후군 코로나바이러스 s 단백질 면역원 조성물 및 이의 제작 방법
WO2020005028A1 (fr) Composition vaccinale pour prévenir ou traiter des maladies provoquées par une infection virale à syndrome de fièvre grave avec thrombocytopénie (sfts)
WO2019182417A1 (fr) Protéine ns1 dérivée du virus de la fièvre jaune, anticorps monoclonal se liant de manière spécifique à celle-ci, et utilisation associée
WO2020251284A1 (fr) Fragments protéiques p72, p104, p205 dérivés du virus de la peste porcine africaine et leur utilisation
WO2019212312A1 (fr) Vaccin chimère contre le virus zika
WO2014142515A1 (fr) Vaccin à sous-unité de circovirus porcin (pcv2) mettant en œuvre des cellules entières de levure recombinantes et procédé pour la fabrication d'un tel vaccin
WO2020246750A2 (fr) Composition pharmaceutique contenant un adjuvant d'acide nucléique stabilisé
WO2019194393A1 (fr) Nanoparticules dans lesquelles un peptide antigène et un adjuvant sont liés à un auto-assemblage de ferritine, et leur utilisation
WO2022119380A1 (fr) Nouveau variant d'eca2 et utilisation associée
WO2022019665A1 (fr) Peptide pour agent immunothérapeutique
WO2023055154A1 (fr) Souche de vaccin rsv atténué vivant recombiné et son procédé de production
WO2022050521A1 (fr) Variant du domaine de liaison au récepteur dérivé du coronavirus ayant une capacité de liaison réduite à l'ac2, et composition vaccinale le comprenant
WO2020197346A2 (fr) Composition d'adjuvant de vaccin antiviral contenant un composant dérivé du thé vert
WO2024014943A1 (fr) Composition de rappel de vaccin contre le sars-cov-2
WO2023101381A1 (fr) Plate-forme de vaccin pour produire des particules de type virus de la fièvre aphteuse
WO2022035246A1 (fr) Adjuvant immunitaire comprenant un polypeptide dérivé du virus de l'hépatite b

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18862512

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18862512

Country of ref document: EP

Kind code of ref document: A1