WO2000037105A2 - Streptococcus pneumoniae proteins and immunogenic fragments for vaccines - Google Patents

Streptococcus pneumoniae proteins and immunogenic fragments for vaccines Download PDF

Info

Publication number
WO2000037105A2
WO2000037105A2 PCT/US1999/030390 US9930390W WO0037105A2 WO 2000037105 A2 WO2000037105 A2 WO 2000037105A2 US 9930390 W US9930390 W US 9930390W WO 0037105 A2 WO0037105 A2 WO 0037105A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
amino acids
seq
sequence
fragment
Prior art date
Application number
PCT/US1999/030390
Other languages
French (fr)
Other versions
WO2000037105A3 (en
Inventor
Leslie S. Johnson
Scott Koenig
John E. Adamou
Original Assignee
Medimmune, Inc.
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22347343&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2000037105(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Medimmune, Inc. filed Critical Medimmune, Inc.
Priority to EP99967460A priority Critical patent/EP1140157B1/en
Priority to EP09001206.3A priority patent/EP2050464B1/en
Priority to DE69940439T priority patent/DE69940439D1/en
Priority to JP2000589215A priority patent/JP4689044B2/en
Priority to AU23731/00A priority patent/AU776828B2/en
Priority to DK99967460T priority patent/DK1140157T3/en
Priority to CA2355364A priority patent/CA2355364C/en
Publication of WO2000037105A2 publication Critical patent/WO2000037105A2/en
Publication of WO2000037105A3 publication Critical patent/WO2000037105A3/en
Priority to AU2004242430A priority patent/AU2004242430C1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • C07K14/3156Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci from Streptococcus pneumoniae (Pneumococcus)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1275Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Streptococcus (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • This invention relates generally to the field of bacterial antigens and their use, for example, as immunogenic agents in humans and animals to stimulate an immune response. More specifically, it relates to the vaccination of mammalian species with a polypeptide comprising at least one conserved histidine triad residue (HxxHxH) and at least one helix-forming polypeptide obtained from Streptococcus pneumoniae as a mechanism for stimulating production of antibodies that protect the vaccine recipient against infection by a wide range of serotypes of pathogenic S. pneumoniae. Further, the invention relates to antibodies against such polypeptides useful in diagnosis and passive immune therapy with respect to diagnosing and treating such pneumococcal infections.
  • the present invention relates to the prevention and treatment of pneumococcal infections such as infections of the middle ear, nasopharynx, lung and bronchial areas, blood, CSF, and the like, that are caused by pneumococcal bacteria.
  • Streptococcus pneumoniae is a gram positive bacteria which is a major causative agent in invasive infections in animals and humans, such as sepsis, meningitis, otitis media and lobar pneumonia (Tuomanen et al. New Engl. J. Med. 322: 1 280-1 284 (1 995)).
  • pneumococci readily bind to non-inflamed human epithelial cells of the upper and lower respiratory tract by binding to eukaryotic carbohydrates in a lectin-like manner (Cundell et al., Micro. Path. 17:361 -374 (1 994)).
  • Conversion to invasive pneumococcal infections for bound bacteria may involve the local generation of inflammatory factors which may activate the epithelial cells to change the number and type of receptors on their surface (Cundell et al., Nature, 377:435-438 (1 995)).
  • inflammatory factors which may activate the epithelial cells to change the number and type of receptors on their surface
  • PAF platelet activating factor
  • one such receptor, platelet activating factor (PAF) is engaged by the pneumococcal bacteria and within a very short period of time (minutes) from the appearance of PAF, pneumococci exhibit strongly enhanced adherence and invasion of tissue.
  • Certain soluble receptor analogs have been shown to prevent the progression of pneumococcal infections (Idanpaan-Heikkila et al., J. Inf. D/ ' s.
  • vaccines and vaccine compositions that include polypeptides obtained from S. pneumoniae and/or variants of said polypeptides and/or active fragments of such polypeptides.
  • the active fragments include a histidine triad residue(s) and/or coiled coil regions of such polypeptides.
  • percent identity when referring to a sequence, means that a sequence is compared to a claimed or described sequence from an alignment of the sequence to be compared (the "Compared
  • C is the number of differences between the Reference Sequence and the Compared Sequence over the length of the alignment between the Compared Sequence and the Reference Sequence wherein (i) each base or amino acid in the Reference Sequence that does not have an aligned base or amino acid in the Compared Sequence and (ii) each gap in the Reference Sequence and (iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, each being a difference; and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid.
  • isolated in the context of the present invention with respect to polypeptides and/or polynucleotides means that the material is removed from its original environment (e.g. , the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living organism is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the co-existing materials in the natural system, is isolated.
  • Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • the polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
  • FIGS 1 A-1 C report the results of three experiments using different preparations of SP36.
  • the results demonstrate that active immunization with recombinant SP36 derived from pneumococcal strain Norway serotype 4 is able to protect mice from death in a model of pneumococcal sepsis using a heterologous strain, SJ2 (serotype 6B).
  • SJ2 heterologous strain 6B
  • one hundred percent of the mice immunized with SP36 survived for the 1 4-day observation period following challenge with approximately 500 cfu of pneumococci, while eighty to one hundred percent of sham-immunized mice (injected with PBS and adjuvant) died during the same period.
  • Figures 2A-2B show that passive administration of rabbit antiserum raised against Sp36 derived from Norway type 4 was able to protect mice in the pneumococcal sepsis model using two heterologous strains.
  • Figure 2A shows that one hundred percent of the mice immunized with the SP36 antiserum survived the 21 -day observation period after challenge with 1 72 CFU of strain SJ2 (serotype 6B). Eighty percent of the mice immunized with a control serum (rabbit anti-FimC) died by day 8, and ninety percent died by day 1 2.
  • Figure 2B shows that 90 percent of the mice immunized with the Sp36 antiserum survived the 8-day observation after challenge with 862 CFU of strain EF6796 (serotype 6A). Ninety percent of the mice immunized with a control serum (collected before immunization) died by day 5.
  • Figure 3 is a western blot demonstrating the ability of antisera raised against recombinant Sp36 derived from strain Norway type 4 to react with Sp36 of heterologous strains. Total cell lysates were immunoblotted with mouse antisera to Sp36. A band representing Sp36 protein was detected in all 23 S. pneumoniae strains tested, which included isolates from each of the 23 pneumococcal serotypes represented in the current polysaccharide vaccine.
  • Figure 4 is a Southern blot showing that the Sp36 gene from Norway type 4 hybridizes with genomic DNA from 24 other pneumococcal strains, indicating the presence of similar sequences in all these strains.
  • Figure 5 is a western blot showing the reactivity of patient sera with Sp36.
  • Sp36 (either full-length, panel A; N-terminal half, panel B; or C- terminal half, panel C) was electrophoresed by SDS-PAGE and transferred to nitrocellulose.
  • Patient sera collected soon after the onset of illness (acute serum, lanes A) or eight to 30 days later (convalescent serum, lanes C) were used to probe the blots. For patients 2, 3, and 5, convalescent serum reacted more strongly with Sp36 than did the corresponding acute serum.
  • Figure 6 is an amino acid alignment comparison of four related pneumococcal proteins, namely Sp36A (PhtA; SEQ ID NO:8), Sp36B (PhtB; SEQ ID NO: 1 0), Sp36D (PhtD; SEQ ID NO:4), Sp36E (PhtE; SEQ ID NO:6), respectively. Dashes in a sequence indicate gaps introduced to maximize the sequence similarity. Amino acid residues that match are boxed.
  • Figure 7 is a nucleotide alignment comparison of four related pneumococcal genes, namely Sp36A (PhtA; SEQ ID NO:9), Sp36B (PhtB; SEQ ID NO: 1 1 ), Sp36D (PhtD; SEQ ID NO:5), Sp36E (PhtE; SEQ ID NO:7), respectively. Dashes in a sequence indicate gaps introduced to maximize the sequence similarity.
  • FIG. 8 shows the results of immunization of mice with PhtD
  • mice were immunized subcutaneously
  • PhtD protein (1 5 ⁇ g in 50 ⁇ l PBS emulsified in 50 ⁇ l complete Freund's
  • mice received PBS with 81 9 amino acid residues, starting with the cysteine (residue 20).
  • a group of 1 0 sham-immunized mice received PBS with 81 9 amino acid residues, starting with the cysteine (residue 20).
  • strains The LD 50 for strain EF5668 was determined to be ⁇ 5 CFU. Survival
  • mice Data are presented as the percent survival for a total of 1 0 mice per
  • a vaccine generally in the form of a composition, that includes at least one polypeptide that is at least 90% identical to (c) a polypeptide sequence comprising amino acids 1 -81 9 of SEQ ID NO:4 or (ii) a polypeptide sequence comprising amino acids 1 -460 of SEQ ID NO:6 or an active fragment of the foregoing.
  • a vaccine generally in the form of a composition, that includes an active fragment of a polypeptide that is at least 90% identical to (i) a polypeptide comprising amino acids 1 -800 of SEQ ID NO:8 or (ii) a polypeptide comprising amino acids 1 -800 of SEQ ID NO: 10.
  • active fragment means a fragment that includes one or more histidine triad residues and/or one or more coiled coil regions.
  • a "histidine triad residue” is the portion of the polypeptide that has the sequence HxxHxH wherein H is histidine and x is an amino acid other than histidine
  • a coiled coil region is the region predicted by "Coils” algorithm: Lupas, A., Van Dyke, M., and Stock, J. (1 991 ) Predicting Coiled Coils from Protein Sequences, Science 252: 1 1 62-1 1 64.
  • the active fragment includes both one or more histidine triad residues and at least one coiled coil region of the applicable polypeptide sequence. In accordance with another embodiment, the active fragment includes at least two histidine triad residues.
  • the active fragment that includes at least one histidine triad residue or at least one coiled-coil region of the applicable polypeptide includes at least about ten percent of the applicable polypeptide and no more than about 85% of the applicable polypeptide.
  • the polypeptide of SEQ ID NO:4 includes five histidine triad residues, as follows:
  • polypeptide of SEQ ID NO:6 includes five histidine triad residues, as follows:
  • polypeptide of SEQ ID NO:4 includes two coiled-coil regions (amino acids 1 20-140 and amino acids 750-772) and the polypeptide of SEQ ID NO:6 includes one coiled-coil region (amino acids 1 1 9-1 52).
  • the polypeptide of SEQ ID NO: 8 includes the following regions:
  • HxxHxH amino acids 63-68, 1 89-1 94, 309-314, 550-555, 634-639.
  • Coiled-coils amino acids 1 1 8-145, 406-434, 462-493, 724-751 .
  • a vaccine of the type hereinabove described is administered for the purpose of preventing or treating infection caused by S. pneumoniae.
  • a vaccine, or vaccine composition, in accordance with the present invention may include one or more of the hereinabove described polypeptides or active fragments thereof.
  • polypeptides or active fragments When employing more than one polypeptide or active fragment, such two or more polypeptides and/or active fragments may be used as a physical mixture or as a fusion of two or more polypeptides or active fragments.
  • the fusion fragment or fusion polypeptide may be produced, for example, by recombinant techniques or by the use of appropriate linkers for fusing previously prepared polypeptides or active fragments.
  • polypeptide that is at least 95% identical or at least 97% identical or 100% identical to (i) a polypeptide sequence comprising amino acids 1 to 81 9 of SEQ ID NO:4 or
  • the variation in the polypeptide or fragment is generally in a portion thereof other than the histidine triad residues and the coiled-coil region, although variations in one or more of these regions may be made.
  • the variation in the polypeptide or active fragment is a conservative amino acid substitution, although other substitutions are within the scope of the invention.
  • a polypeptide variant includes variants in which one or more amino acids are substituted and/or deleted and/or inserted.
  • the invention relates to passive immunity vaccines formulated from antibodies against a polypeptide or active fragment of a polypeptide of the present invention.
  • passive immunity vaccines can be utilized to prevent and/or treat pneumococcal infections in patients.
  • a vaccine can be produced from a synthetic or recombinant polypeptide of the present invention or an antibody against such polypeptide.
  • the present invention relates to a method of using one or more antibodies (monoclonal, polyclonal or sera) to the polypeptides of the invention as described above for the prophylaxis and/or treatment of diseases that are caused by pneumococcal bacteria.
  • the invention relates to a method for the prophylaxis and/or treatment of infectious diseases that are caused by S. pneumoniae.
  • the invention relates to a method for the prophylaxis and/or treatment of otitis media, nasopharyngeal, bronchial infections, and the like in humans by utilizing a vaccine of the present invention.
  • vaccines are prepared as injectables, in the form of aqueous solutions or suspensions. Vaccines in an oil base are also well known such as for inhaling. Solid forms which are dissolved or suspended prior to use may also be formulated.
  • Pharmaceutical carriers are generally added that are compatible with the active ingredients and acceptable for pharmaceutical use. Examples of such carriers include, but are not limited to, water, saline solutions, dextrose, or glycerol. Combinations of carriers may also be used.
  • Vaccine compositions may further incorporate additional substances to stabilize pH, or to function as adjuvants, wetting agents, or emulsifying agents, which can serve to improve the effectiveness of the vaccine.
  • Vaccines are generally formulated for parental administration and are injected either subcutaneously or intramuscularly. Such vaccines can also be formulated as suppositories or for oral administration, using methods known in the art.
  • the amount of vaccine sufficient to confer immunity to pathogenic bacteria is determined by methods well known to those skilled in the art. This quantity will be determined based upon the characteristics of the vaccine recipient and the level of immunity required. Typically, the amount of vaccine to be administered will be determined based upon the judgment of a skilled physician. Where vaccines are administered by subcutaneous or intramuscular injection, a range of 50 to 500 ⁇ g purified protein may be given.
  • the present invention is also directed to a vaccine in which a polypeptide or active fragment of the present invention is delivered or administered in the form of a polynucleotide encoding the polypeptide or active fragment, whereby the polypeptide or active fragment is produced in vivo.
  • the polynucleotide may be included in a suitable expression vector and combined with a pharmaceutically acceptable carrier.
  • polypeptides of the present invention can be used as immunogens to stimulate the production of antibodies for use in passive immunotherapy, for use as diagnostic reagents, and for use as reagents in other processes such as affinity chromatography.
  • the present invention provides polynucleotides which encode the hereinabove described polypeptides and active fragments of the invention.
  • the polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA.
  • the DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
  • A an isolated polynucleotide that is at least 90% identical to a polynucleotide sequence encoding (i) a polypeptide comprising amino acids 1 - 81 9 of SEQ ID NO:4 or (ii) a polypeptide comprising amino acids 1 -460 of SEQ ID NO:6, or
  • the polynucleotide is at least 95% identical, preferably at least 97% identical, and even 100% identical to such polynucleotide sequence.
  • polynucleotide encoding a polypeptide encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
  • the present invention further relates to variants of polynucleotides.
  • the variants of the polynucleotides may be a naturally occurring allelic variant of the polynucleotides or a non-naturally occurring variant of the polynucleotides.
  • the variants include variants in which one or more bases are substituted, deleted or inserted.
  • Complements to such coding polynucleotides may be utilized to isolate polynucleotides encoding the same or similar polypeptides. In particular, such procedures are useful to obtain native immunogenic portions of polypeptides from different serotypes of S. pneumoniae, which is especially useful in the production of "chain" polypeptide vaccines containing multiple immunogenic segments.
  • SEQ ID NO:5 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:4 and SEQ ID NO:7 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:6.
  • SEQ ID NO:9 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:8, and SEQ ID NO: 1 1 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO: 10.
  • the polynucleotides encoding the immunogenic polypeptides described above may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptides of the present invention.
  • the marker sequence may be, for example, a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptides fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1 984)).
  • the present invention also relates to vectors which include polynucleotides encoding one or more of the polypeptides of the invention, host cells which are genetically engineered with vectors of the invention and the production of such immunogenic polypeptides by recombinant techniques in an isolated and substantially immunogenically pure form.
  • Host cells are genetically engineered (transduced or transformed or transfected) with the vectors comprising a polynucleotide encoding a polypeptide of the invention.
  • the vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the polynucleotides which encode such polypeptides.
  • the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • Vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies.
  • any other vector may be used as long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by a variety of procedures.
  • the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoter for example, LTR or SV40 promoter, the E. coli. lac or trp, the phage lambda P L promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector also contains a ribosome binding site for translation initiation and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in coli.
  • the vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the proteins.
  • bacterial cells such as E. coli, Streptomyces, Salmonella typhimurium
  • fungal cells such as yeast
  • insect cells such as Drosophila S2 and Spodoptera Sf9
  • animal cells such as CHO, COS or Bowes melanoma
  • adenoviruses plant cells, etc.
  • the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • a promoter operably linked to the sequence.
  • Bacterial pQE70, pQE60, pQE-9 (Qiagen, Inc.), pbs, pD1 0, phagescript, psiX1 74, pbluescript SK, pbsks, pNH8A, pNH16a, pNH1 8A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia).
  • Eukaryotic pWLNEO, pSV2CAT, pOG44, pXT1 , pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia).
  • any other plasmid or vector may be used as long as they are replicable and viable in the host.
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers.
  • Two appropriate vectors are pKK232-8 and pCM7.
  • Particular named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda P R , P L and TRP.
  • Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-l. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • the present invention relates to host cells containing the above-described constructs.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1 986)).
  • constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1 989), the disclosure of which is hereby incorporated by reference.
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription. Examples including the SV40 enhancer on the late side of the replication origin bp 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence.
  • promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), ⁇ - factor, acid phosphatase, or heat shock proteins, among others.
  • PGK 3-phosphoglycerate kinase
  • the heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences.
  • the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter.
  • the vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host.
  • Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 3701 7).
  • cloning vector pBR322 ATCC 3701 7
  • Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, Wl, USA). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
  • the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
  • appropriate means e.g., temperature shift or chemical induction
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, a french press, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.
  • preferred are host cells which secrete the polypeptide of the invention and permit recovery of the polypeptide from the culture media.
  • mammalian cell culture systems can also be employed to express recombinant protein.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981 ), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines.
  • Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
  • the polypeptides can be recovered and/or purified from recombinant cell cultures by well-known protein recovery and purification methods. Such methodology may include ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. In this respect, chaperones may be used in such a refolding procedure. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • polypeptides that are useful as immunogens in the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
  • Procedures for the isolation of the individually expressed polypeptides may be isolated by recombinant expression/isolation methods that are well- known in the art. Typical examples for such isolation may utilize an antibody to a conserved area of the protein or to a His tag or cleavable leader or tail that is expressed as part of the protein structure.
  • polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto.
  • These antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
  • Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1 975, Nature, 256:495- 497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1 983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1 985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • Patent 4,946,778 can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice may be used to express humanized antibodies to immunogenic polypeptide products of this invention.
  • genomic DNA used as target for amplification was isolated from
  • S. pneumoniae Norway strain (serotype 4), the same strain used for genomic sequencing.
  • the complete sequence of the Sp36 gene (SEQ ID NO:9), and its predicted amino acid sequence (SEQ ID NO:8), are given in the Sequence Listing appended hereto. It was noted that the predicted amino acid sequence included a hydrophobic leader sequence followed by a sequence (LSVC) similar to the consensus sequence for Type II signal peptidase (LxxC, in which both x's typically represent small amino acids).
  • Primers (listed as SEQ ID NOS: 1 -3) were designed that would amplify the Sp36 gene and allow its cloning into pQE10 and expression as a histidine-tagged protein lacking the signal sequence for purification by nickel-affinity chromatography.
  • C3H/HeJ mice (10/group) were immunized intraperitoneally (i.p.) with Sp36 protein (15 ⁇ g in 50 ⁇ l PBS emulsified in 50 ⁇ l complete Freund's adjuvant (CFA)).
  • CFA complete Freund's adjuvant
  • a group of 10 sham-immunized mice received PBS with adjuvant.
  • a second immunization of 15 ⁇ g protein with incomplete Freund's adjuvant (IFA) was administered 4 weeks later; the sham group received PBS with IFA.
  • Blood was drawn (retro-orbital bleed) at weeks 3, 6, and 9; and sera from each group were pooled for analysis of anti-Sp36 antibody by ELISA.
  • mice were challenged at week 10 by an i.p. injection of approximately 500 CFU S. pneumoniae strain SJ2 (serotype 6B; provided by P. Flynn, St. Jude Children's Research Hospital, Memphis, TN). In preliminary experiments, the LD 50 of this strain was determined to be approximately 10 CFU. Mice were monitored for 14 days for survival.
  • S. pneumoniae strain SJ2 serotype 6B; provided by P. Flynn, St. Jude Children's Research Hospital, Memphis, TN.
  • the LD 50 of this strain was determined to be approximately 10 CFU. Mice were monitored for 14 days for survival.
  • the three experiments shown in Figures 1 A-1 C used slightly different preparations of recombinant Sp36.
  • the experiments shown in Figure 1 A and 1 B both used Sp36 containing amino acids 20-81 5, but different batches of protein were used in the two experiments.
  • the experiment shown in Figure 1 C used Sp36 containing amino acids 25-81 5.
  • the rabbit was given two boosts of 125 ⁇ g Sp36 in IFA on days 29 and 50 and bled on days 39 and 60.
  • a second rabbit was immunized with a control antigen, E. coli FimC.
  • mice (10 mice/group) were passively immunized by two i.p. injections of 100 ⁇ l of rabbit serum. The first injection was administered twenty-four hours before challenge with 172 cfu of S. pneumoniae strain SJ2, and the second injection was given four hours after challenge.
  • Figure 2 shows the survival of mice after infection with two different strains of pneumococci.
  • Figure 2A shows that of mice injected with 172 cfu of strain SJ2 ( Figure 2A), one hundred percent of the mice immunized with rabbit immune serum raised against Sp36 protein survived the 21 -day observation period. Of the mice immunized with the control serum (anti-FimC), eighty percent died by day 8, and ninety percent died by day 12.
  • Figure 2B shows that of mice injected with 862 cfu of strain EF6796, ninety percent of the mice immunized with rabbit immune serum raised against Sp36 protein survived the 8-day observation period. Of those given a control serum (collected from a rabbit before immunization), ninety percent died by day 8.
  • the 23 pneumococcal strains used in this experiment were obtained from the American Type Culture Collection (Rockville, MD) and include one isolate each of the 23 serotypes in the multivalent pneumococcal vaccine.
  • pneumococci were grown to mid-logarithmic phase (optical density at 620 nm, 0.4 to 0.6) in 2 ml Todd-Hewitt broth with 0.5% yeast extract (Difco, Detroit, ME) at 37°C. Bacteria were harvested by centrifugation and washed twice with water.
  • Pellets were resuspended in 200 ⁇ l lysis buffer (0.01 % sodium dodecyl sulfate, 0.15 M sodium citrate and 0.1 % sodium deoxycholate) and incubated at 37°C for 30 min, then diluted in an equal volume 2x SSC (0.3 M sodium chloride, 0.03 M sodium citrate). Lysates were separated by SDS-PAGE, transferred to nitrocellulose membranes (Bio-Rad Laboratories, Hercules, CA), and probed with antibody in a standard Western blotting procedure. Sera from ten C3H/HeJ mice immunized with Sp36 (as described in Example 1 ) were pooled and used at a dilution of 1 :3000. Bound antibody was detected with peroxidase- conjugated sheep anti- mouse IgG using the chemiluminescence kit from Amersham, Inc. (Cambridge, MA).
  • the mouse anti-Sp36 sera detected two major bands with apparent molecular weights of 97 and 100 kDa in all 23 pneumococcal lysates tested (shown in Figure 3).
  • the Sp36 signals obtained from S. pneumoniae serotypes 1 , 5, 17F and 22F were lower, indicating either that the level of Sp36 expression is reduced in these strains, or that Sp36 in these strains is antigenically different.
  • Genomic DNA was prepared from each of the 23 pneumococcal strains listed in the previous section and also from strain SJ2. DNA was digested with PvuW and BamYW, electrophoresed in an agarose gel and transferred to a nylon membrane. A probe was prepared by amplifying the Sp36 gene from Norway type 4 DNA (as in Example 1 ) and labeling the amplified fragment with fluorescein by the random-priming method, using a kit from Amersham. Hybridization, washing, and exposure of film were carried out as in the protocol supplied by Amersham. Figure 4 shows that the Sp36 probe hybridized with DNA from each of the 24 strains studied. The lane marked "M” contained DNA from lambda phage, digested with Hind ⁇ and labeled with fluorescein, as molecular weight markers.
  • patients 2, 3 and 5 reactivity of the convalescent serum with Sp36 was stronger that that of the corresponding acute serum. The difference between the acute and convalescent sera was particularly evident for reactivity with the C-terminal half of the protein.
  • Table 1 provides the percent identity between the various sequences.

Abstract

A vaccine composition is disclosed that comprises polypeptides and fragments of polypeptides containing histidine triad residues or coiled-coil regions, some of which polypeptides or fragments lie between 80 and 680 residues in length. Also disclosed are processes for preventing infection caused by S. pneumoniae comprising administering of vaccine compositions.

Description

STREPTOCOCCUS PNEUMONIAE / PROTEINS AND
IMMUNOGENIC FRAGMENTS FOR VACCINES
This application is based on U.S. Provisional Application No. 60/1 1 3,048, filed 21 December 1 998, which is hereby incorporated in its entirety.
FIELD OF THE INVENTION
This invention relates generally to the field of bacterial antigens and their use, for example, as immunogenic agents in humans and animals to stimulate an immune response. More specifically, it relates to the vaccination of mammalian species with a polypeptide comprising at least one conserved histidine triad residue (HxxHxH) and at least one helix-forming polypeptide obtained from Streptococcus pneumoniae as a mechanism for stimulating production of antibodies that protect the vaccine recipient against infection by a wide range of serotypes of pathogenic S. pneumoniae. Further, the invention relates to antibodies against such polypeptides useful in diagnosis and passive immune therapy with respect to diagnosing and treating such pneumococcal infections.
In a particular aspect, the present invention relates to the prevention and treatment of pneumococcal infections such as infections of the middle ear, nasopharynx, lung and bronchial areas, blood, CSF, and the like, that are caused by pneumococcal bacteria. BACKGROUND OF THE INVENTION
Streptococcus pneumoniae is a gram positive bacteria which is a major causative agent in invasive infections in animals and humans, such as sepsis, meningitis, otitis media and lobar pneumonia (Tuomanen et al. New Engl. J. Med. 322: 1 280-1 284 (1 995)). As part of the infective process, pneumococci readily bind to non-inflamed human epithelial cells of the upper and lower respiratory tract by binding to eukaryotic carbohydrates in a lectin-like manner (Cundell et al., Micro. Path. 17:361 -374 (1 994)). Conversion to invasive pneumococcal infections for bound bacteria may involve the local generation of inflammatory factors which may activate the epithelial cells to change the number and type of receptors on their surface (Cundell et al., Nature, 377:435-438 (1 995)). Apparently, one such receptor, platelet activating factor (PAF) is engaged by the pneumococcal bacteria and within a very short period of time (minutes) from the appearance of PAF, pneumococci exhibit strongly enhanced adherence and invasion of tissue. Certain soluble receptor analogs have been shown to prevent the progression of pneumococcal infections (Idanpaan-Heikkila et al., J. Inf. D/'s. , 176:704-71 2 (1 997)). A number of various other proteins have been suggested as being involved in the pathogenicity of S. pneumoniae. There remains a need for identifying polypeptides having epitopes in common from various strains of S. pneumoniae in order to utilize such polypeptides as vaccines to provide protection against a wide variety of S. pneumoniae.
SUMMARY OF INVENTION
In accordance with the present invention, there is provided vaccines and vaccine compositions that include polypeptides obtained from S. pneumoniae and/or variants of said polypeptides and/or active fragments of such polypeptides.
The active fragments, as hereinafter defined, include a histidine triad residue(s) and/or coiled coil regions of such polypeptides.
The term "percent identity" or "percent identical," when referring to a sequence, means that a sequence is compared to a claimed or described sequence from an alignment of the sequence to be compared (the "Compared
Sequence") with the described or claimed sequence (the "Reference
Sequence"). The percent identity is determined as follows:
Percent Identity = [1 - (C/R)] 1 00
wherein C is the number of differences between the Reference Sequence and the Compared Sequence over the length of the alignment between the Compared Sequence and the Reference Sequence wherein (i) each base or amino acid in the Reference Sequence that does not have an aligned base or amino acid in the Compared Sequence and (ii) each gap in the Reference Sequence and (iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, each being a difference; and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid.
If an alignment exists between the Compared Sequence and the Reference Sequence in which the Percent Identity as calculated above is about equal to or greater than a specified minimum Percent Identity than the Compared Sequence has the specified minimum Percent Identity to the Reference Sequence even though alignments may exist in which the hereinabove calculated Percent Identity is less than the specified Percent Identity.
"Isolated" in the context of the present invention with respect to polypeptides and/or polynucleotides means that the material is removed from its original environment (e.g. , the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living organism is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the co-existing materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment. The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
BRIEF DESCRIPTION OF DRAWINGS
Figures 1 A-1 C, respectively, report the results of three experiments using different preparations of SP36. The results demonstrate that active immunization with recombinant SP36 derived from pneumococcal strain Norway serotype 4 is able to protect mice from death in a model of pneumococcal sepsis using a heterologous strain, SJ2 (serotype 6B). In each of the three experiments shown, one hundred percent of the mice immunized with SP36 survived for the 1 4-day observation period following challenge with approximately 500 cfu of pneumococci, while eighty to one hundred percent of sham-immunized mice (injected with PBS and adjuvant) died during the same period.
Figures 2A-2B show that passive administration of rabbit antiserum raised against Sp36 derived from Norway type 4 was able to protect mice in the pneumococcal sepsis model using two heterologous strains. Figure 2A shows that one hundred percent of the mice immunized with the SP36 antiserum survived the 21 -day observation period after challenge with 1 72 CFU of strain SJ2 (serotype 6B). Eighty percent of the mice immunized with a control serum (rabbit anti-FimC) died by day 8, and ninety percent died by day 1 2. Figure 2B shows that 90 percent of the mice immunized with the Sp36 antiserum survived the 8-day observation after challenge with 862 CFU of strain EF6796 (serotype 6A). Ninety percent of the mice immunized with a control serum (collected before immunization) died by day 5.
Figure 3 is a western blot demonstrating the ability of antisera raised against recombinant Sp36 derived from strain Norway type 4 to react with Sp36 of heterologous strains. Total cell lysates were immunoblotted with mouse antisera to Sp36. A band representing Sp36 protein was detected in all 23 S. pneumoniae strains tested, which included isolates from each of the 23 pneumococcal serotypes represented in the current polysaccharide vaccine.
Figure 4 is a Southern blot showing that the Sp36 gene from Norway type 4 hybridizes with genomic DNA from 24 other pneumococcal strains, indicating the presence of similar sequences in all these strains. Figure 5 is a western blot showing the reactivity of patient sera with Sp36. Sp36 (either full-length, panel A; N-terminal half, panel B; or C- terminal half, panel C) was electrophoresed by SDS-PAGE and transferred to nitrocellulose. Patient sera collected soon after the onset of illness (acute serum, lanes A) or eight to 30 days later (convalescent serum, lanes C) were used to probe the blots. For patients 2, 3, and 5, convalescent serum reacted more strongly with Sp36 than did the corresponding acute serum.
Figure 6 is an amino acid alignment comparison of four related pneumococcal proteins, namely Sp36A (PhtA; SEQ ID NO:8), Sp36B (PhtB; SEQ ID NO: 1 0), Sp36D (PhtD; SEQ ID NO:4), Sp36E (PhtE; SEQ ID NO:6), respectively. Dashes in a sequence indicate gaps introduced to maximize the sequence similarity. Amino acid residues that match are boxed.
Figure 7 is a nucleotide alignment comparison of four related pneumococcal genes, namely Sp36A (PhtA; SEQ ID NO:9), Sp36B (PhtB; SEQ ID NO: 1 1 ), Sp36D (PhtD; SEQ ID NO:5), Sp36E (PhtE; SEQ ID NO:7), respectively. Dashes in a sequence indicate gaps introduced to maximize the sequence similarity.
Figure 8 shows the results of immunization of mice with PhtD
recombinant protein, which leads to protection from lethal sepsis. C3H/HeJ
(Panel A and B) or Balb/cByJ (Panel C) mice were immunized subcutaneously
with PhtD protein (1 5 μg in 50 μl PBS emulsified in 50 μl complete Freund's
adjuvant (CFA)). The recombinant PhtD protein used in protection
experiments consisted of 81 9 amino acid residues, starting with the cysteine (residue 20). A group of 1 0 sham-immunized mice received PBS with
adjuvant. A second immunization of 1 5 μg protein with incomplete Freund's
adjuvant (IFA) was administered 3 weeks later; the sham group received PBS
with IFA. Blood was drawn (retro-orbital bleed) at week 7; and sera from
each group was pooled for analysis of anti-PhtD antibody by ELISA. Mice
were challenged at week 8 by an intraperitonial (i.p.) injection of
approximately 550 CFU S. pneumoniae strain SJ2, serotype 6B (Panel A),
850 CFU of strain EF6796, serotype 6A (Panel B) or 450 CFU of strain
EF5668, serotype 4 (Panel C). In preliminary experiments, the LD50 for strain
SJ2 and EF6796 were determined to be approximately 1 0 CFU for both
strains. The LD50 for strain EF5668 was determined to be < 5 CFU. Survival
was determined in all groups over the course of 1 5 days following challenge.
Data are presented as the percent survival for a total of 1 0 mice per
experimental group. Two-sample Log-rank test was used for statistical
analysis comparing recombinant Pht immunized mice to sham-immunized mice.
SUMMARY OF THE INVENTION
In accordance with one aspect of the present invention, there is provided a vaccine, generally in the form of a composition, that includes at least one polypeptide that is at least 90% identical to (c) a polypeptide sequence comprising amino acids 1 -81 9 of SEQ ID NO:4 or (ii) a polypeptide sequence comprising amino acids 1 -460 of SEQ ID NO:6 or an active fragment of the foregoing.
In accordance with another aspect of the present invention, there is provided a vaccine, generally in the form of a composition, that includes an active fragment of a polypeptide that is at least 90% identical to (i) a polypeptide comprising amino acids 1 -800 of SEQ ID NO:8 or (ii) a polypeptide comprising amino acids 1 -800 of SEQ ID NO: 10.
The term "active fragment" means a fragment that includes one or more histidine triad residues and/or one or more coiled coil regions. A "histidine triad residue" is the portion of the polypeptide that has the sequence HxxHxH wherein H is histidine and x is an amino acid other than histidine
A coiled coil region is the region predicted by "Coils" algorithm: Lupas, A., Van Dyke, M., and Stock, J. (1 991 ) Predicting Coiled Coils from Protein Sequences, Science 252: 1 1 62-1 1 64.
In accordance with one embodiment, the active fragment includes both one or more histidine triad residues and at least one coiled coil region of the applicable polypeptide sequence. In accordance with another embodiment, the active fragment includes at least two histidine triad residues.
In another embodiment, the active fragment that includes at least one histidine triad residue or at least one coiled-coil region of the applicable polypeptide includes at least about ten percent of the applicable polypeptide and no more than about 85% of the applicable polypeptide. The polypeptide of SEQ ID NO:4 includes five histidine triad residues, as follows:
amino acids 64-69; 1 88-1 93; 296-301 ; 541 -546; and 625-630.
The polypeptide of SEQ ID NO:6 includes five histidine triad residues, as follows:
amino acids 63-68; 1 85-1 90; 289-294, 376-381 ; and 441 -446.
In addition, the polypeptide of SEQ ID NO:4 includes two coiled-coil regions (amino acids 1 20-140 and amino acids 750-772) and the polypeptide of SEQ ID NO:6 includes one coiled-coil region (amino acids 1 1 9-1 52).
The polypeptide of SEQ ID NO: 8 includes the following regions:
HxxHxH: amino acids 63-68, 1 89-1 94, 309-314, 550-555, 634-639. Coiled-coils: amino acids 1 1 8-145, 406-434, 462-493, 724-751 .
In accordance with a further aspect of the invention, a vaccine of the type hereinabove described is administered for the purpose of preventing or treating infection caused by S. pneumoniae.
A vaccine, or vaccine composition, in accordance with the present invention may include one or more of the hereinabove described polypeptides or active fragments thereof. When employing more than one polypeptide or active fragment, such two or more polypeptides and/or active fragments may be used as a physical mixture or as a fusion of two or more polypeptides or active fragments. The fusion fragment or fusion polypeptide may be produced, for example, by recombinant techniques or by the use of appropriate linkers for fusing previously prepared polypeptides or active fragments.
In an embodiment of the invention, there is provided (a) a polypeptide that is at least 95% identical or at least 97% identical or 100% identical to (i) a polypeptide sequence comprising amino acids 1 to 81 9 of SEQ ID NO:4 or
(ii) a polypeptide sequence comprising amino acids 1 -460 of SEQ ID NO:6; or
(b) an active fragment of the polypeptide of (a).
In the case where the polypeptide is a variant of the polypeptide comprising the mature polypeptide of SEQ ID NO:4 or SEQ ID NO:6, or any of the active fragments of the invention, the variation in the polypeptide or fragment is generally in a portion thereof other than the histidine triad residues and the coiled-coil region, although variations in one or more of these regions may be made.
In many cases, the variation in the polypeptide or active fragment is a conservative amino acid substitution, although other substitutions are within the scope of the invention.
In accordance with the present invention, a polypeptide variant includes variants in which one or more amino acids are substituted and/or deleted and/or inserted.
In another aspect, the invention relates to passive immunity vaccines formulated from antibodies against a polypeptide or active fragment of a polypeptide of the present invention. Such passive immunity vaccines can be utilized to prevent and/or treat pneumococcal infections in patients. In this manner, according to a further aspect of the invention, a vaccine can be produced from a synthetic or recombinant polypeptide of the present invention or an antibody against such polypeptide.
In still another aspect the present invention relates to a method of using one or more antibodies (monoclonal, polyclonal or sera) to the polypeptides of the invention as described above for the prophylaxis and/or treatment of diseases that are caused by pneumococcal bacteria. In particular, the invention relates to a method for the prophylaxis and/or treatment of infectious diseases that are caused by S. pneumoniae. In a still further preferred aspect, the invention relates to a method for the prophylaxis and/or treatment of otitis media, nasopharyngeal, bronchial infections, and the like in humans by utilizing a vaccine of the present invention.
Generally, vaccines are prepared as injectables, in the form of aqueous solutions or suspensions. Vaccines in an oil base are also well known such as for inhaling. Solid forms which are dissolved or suspended prior to use may also be formulated. Pharmaceutical carriers are generally added that are compatible with the active ingredients and acceptable for pharmaceutical use. Examples of such carriers include, but are not limited to, water, saline solutions, dextrose, or glycerol. Combinations of carriers may also be used.
Vaccine compositions may further incorporate additional substances to stabilize pH, or to function as adjuvants, wetting agents, or emulsifying agents, which can serve to improve the effectiveness of the vaccine.
Vaccines are generally formulated for parental administration and are injected either subcutaneously or intramuscularly. Such vaccines can also be formulated as suppositories or for oral administration, using methods known in the art. The amount of vaccine sufficient to confer immunity to pathogenic bacteria is determined by methods well known to those skilled in the art. This quantity will be determined based upon the characteristics of the vaccine recipient and the level of immunity required. Typically, the amount of vaccine to be administered will be determined based upon the judgment of a skilled physician. Where vaccines are administered by subcutaneous or intramuscular injection, a range of 50 to 500 μg purified protein may be given.
The present invention is also directed to a vaccine in which a polypeptide or active fragment of the present invention is delivered or administered in the form of a polynucleotide encoding the polypeptide or active fragment, whereby the polypeptide or active fragment is produced in vivo. The polynucleotide may be included in a suitable expression vector and combined with a pharmaceutically acceptable carrier.
In addition, the polypeptides of the present invention can be used as immunogens to stimulate the production of antibodies for use in passive immunotherapy, for use as diagnostic reagents, and for use as reagents in other processes such as affinity chromatography.
In another aspect the present invention provides polynucleotides which encode the hereinabove described polypeptides and active fragments of the invention. The polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
in accordance with another aspect of the present invention, there is provided
(A) an isolated polynucleotide that is at least 90% identical to a polynucleotide sequence encoding (i) a polypeptide comprising amino acids 1 - 81 9 of SEQ ID NO:4 or (ii) a polypeptide comprising amino acids 1 -460 of SEQ ID NO:6, or
(B) a fragment of the polynucleotide of (A) that encodes an active polypeptide fragment or
(C) a polynucleotide that is at least 90% identical to a polynucleotide sequence encoding an active fragment of (i) a polypeptide comprising amino acids 1 to 800 of SEQ ID NO:8 or (ii) a polypeptide comprising amino acids 1 to 800 of SEQ ID NO: 10.
In specific embodiments, the polynucleotide is at least 95% identical, preferably at least 97% identical, and even 100% identical to such polynucleotide sequence.
The term "polynucleotide encoding a polypeptide" encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
The present invention further relates to variants of polynucleotides. The variants of the polynucleotides may be a naturally occurring allelic variant of the polynucleotides or a non-naturally occurring variant of the polynucleotides. The variants include variants in which one or more bases are substituted, deleted or inserted. Complements to such coding polynucleotides may be utilized to isolate polynucleotides encoding the same or similar polypeptides. In particular, such procedures are useful to obtain native immunogenic portions of polypeptides from different serotypes of S. pneumoniae, which is especially useful in the production of "chain" polypeptide vaccines containing multiple immunogenic segments.
SEQ ID NO:5 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:4 and SEQ ID NO:7 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:6. SEQ ID NO:9 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO:8, and SEQ ID NO: 1 1 is a representative example of a polynucleotide encoding the polypeptide of SEQ ID NO: 10. As a result of the known degeneracy of the genetic code, other polynucleotides that encode the polypeptides of SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 and SEQ ID NO: 10 should be apparent to those skilled in the art from the teachings herein.
The polynucleotides encoding the immunogenic polypeptides described above may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptides of the present invention. The marker sequence may be, for example, a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptides fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al., Cell, 37:767 (1 984)).
The present invention also relates to vectors which include polynucleotides encoding one or more of the polypeptides of the invention, host cells which are genetically engineered with vectors of the invention and the production of such immunogenic polypeptides by recombinant techniques in an isolated and substantially immunogenically pure form. Host cells are genetically engineered (transduced or transformed or transfected) with the vectors comprising a polynucleotide encoding a polypeptide of the invention. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the polynucleotides which encode such polypeptides. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
Vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies. However, any other vector may be used as long as it is replicable and viable in the host.
The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art.
The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis. As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the E. coli. lac or trp, the phage lambda PL promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expression.
In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in coli.
The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the proteins.
As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as E. coli, Streptomyces, Salmonella typhimurium; fungal cells, such as yeast; insect cells such as Drosophila S2 and Spodoptera Sf9; animal cells such as CHO, COS or Bowes melanoma; adenoviruses; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.
More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial: pQE70, pQE60, pQE-9 (Qiagen, Inc.), pbs, pD1 0, phagescript, psiX1 74, pbluescript SK, pbsks, pNH8A, pNH16a, pNH1 8A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXT1 , pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host.
Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda PR, PL and TRP. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-l. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
In a further embodiment, the present invention relates to host cells containing the above-described constructs. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M., Battey, I., Basic Methods in Molecular Biology, (1 986)).
The constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1 989), the disclosure of which is hereby incorporated by reference.
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription. Examples including the SV40 enhancer on the late side of the replication origin bp 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), α- factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 3701 7). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, Wl, USA). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period. Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, a french press, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art. However, preferred are host cells which secrete the polypeptide of the invention and permit recovery of the polypeptide from the culture media.
Various mammalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23:175 (1981 ), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
The polypeptides can be recovered and/or purified from recombinant cell cultures by well-known protein recovery and purification methods. Such methodology may include ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. In this respect, chaperones may be used in such a refolding procedure. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
The polypeptides that are useful as immunogens in the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
Procedures for the isolation of the individually expressed polypeptides may be isolated by recombinant expression/isolation methods that are well- known in the art. Typical examples for such isolation may utilize an antibody to a conserved area of the protein or to a His tag or cleavable leader or tail that is expressed as part of the protein structure.
The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1 975, Nature, 256:495- 497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1 983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1 985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
Techniques described for the production of single chain antibodies (U.S.
Patent 4,946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice may be used to express humanized antibodies to immunogenic polypeptide products of this invention.
The invention will be further described with respect to the following examples; however, the scope of the invention is not limited thereby:
Example 1
Active Protection with Anti-Sp36
A. Cloning, expression, and purification of SP36
The genomic DNA used as target for amplification was isolated from
S. pneumoniae Norway strain (serotype 4), the same strain used for genomic sequencing. The complete sequence of the Sp36 gene (SEQ ID NO:9), and its predicted amino acid sequence (SEQ ID NO:8), are given in the Sequence Listing appended hereto. It was noted that the predicted amino acid sequence included a hydrophobic leader sequence followed by a sequence (LSVC) similar to the consensus sequence for Type II signal peptidase (LxxC, in which both x's typically represent small amino acids). Primers (listed as SEQ ID NOS: 1 -3) were designed that would amplify the Sp36 gene and allow its cloning into pQE10 and expression as a histidine-tagged protein lacking the signal sequence for purification by nickel-affinity chromatography. Cloning of the fragment amplified by SEQ ID Nos 1 and 3 would result in a protein containing amino acids 2 through 800 of Sp36; cloning of the fragment amplified by SEQ ID Nos 2 and 3 would result in a protein containing amino acids 7 through 800 of Sp36 (amino acid numbers refer to SEQ ID NO:8).
B. Active Protection With Sp36 Vaccination
In each of the three experiments shown in Figures 1 A-1 C, C3H/HeJ mice (10/group) were immunized intraperitoneally (i.p.) with Sp36 protein (15 μg in 50 μl PBS emulsified in 50 μl complete Freund's adjuvant (CFA)). A group of 10 sham-immunized mice received PBS with adjuvant. A second immunization of 15 μg protein with incomplete Freund's adjuvant (IFA) was administered 4 weeks later; the sham group received PBS with IFA. Blood was drawn (retro-orbital bleed) at weeks 3, 6, and 9; and sera from each group were pooled for analysis of anti-Sp36 antibody by ELISA. Mice were challenged at week 10 by an i.p. injection of approximately 500 CFU S. pneumoniae strain SJ2 (serotype 6B; provided by P. Flynn, St. Jude Children's Research Hospital, Memphis, TN). In preliminary experiments, the LD50 of this strain was determined to be approximately 10 CFU. Mice were monitored for 14 days for survival.
The three experiments shown in Figures 1 A-1 C used slightly different preparations of recombinant Sp36. The experiments shown in Figure 1 A and 1 B both used Sp36 containing amino acids 20-81 5, but different batches of protein were used in the two experiments. The experiment shown in Figure 1 C used Sp36 containing amino acids 25-81 5.
In the experiment shown in Figure 1 A, 9-week sera collected from the ten mice immunized with Sp36 (first batch) had an endpoint ELISA titer of 1 :4,096,000. No anti-Sp36 antibody was detected in sera from sham- immunized mice. One hundred percent of the mice immunized with Sp36 protein survived the challenge (520 cfu of pneumococci) for 14 days. Eighty percent of sham-immunized mice were dead by day 4, and the remainder survived.
In the experiment shown in Figure 1 B, 9-week sera collected from the ten mice immunized with Sp36 (second batch) had an endpoint ELISA titer of > 1 :4, 096, 000. No anti-Sp36 antibody was detected in sera from sham- immunized mice. One hundred percent of the mice immunized with Sp36 protein survived the challenge (510 cfu of pneumococci) for 14 days. Of the sham-immunized mice, eighty percent were dead by day 4, and all died by day 9.
In the experiment shown in Figure 1 C, 9-week sera collected from the ten mice immunized with Sp36 (containing amino acids 25- 81 5) had an endpoint ELISA titer of 1 :4, 096,000. No anti-Sp36 antibody was detected in sera from sham-immunized mice. One hundred percent of the mice immunized with Sp36 protein survived the challenge (510 cfu of pneumococci) for 14 days. Of the sham-immunized mice, ninety percent died by day 4, and all died by day 12. These data demonstrate that immunization of mice with recombinant Sp36 proteins elicits a response capable of protecting against systemic pneumococcal infection and death. This protection was not strain-specific: the recombinant pneumococcal protein was cloned from a serotype 4 strain, while the challenge was with a heterologous strain, SJ2 (serotype 6B).
Example 2
Passive Protection with Anti-Sp36 Antisera
A. Generation of Rabbit Immune Sera
Following collection of preimmune serum, a New Zealand White rabbit was immunized with 250 μg of Sp36 (containing amino acids 20-815) in
CFA. The rabbit was given two boosts of 125 μg Sp36 in IFA on days 29 and 50 and bled on days 39 and 60. A second rabbit was immunized with a control antigen, E. coli FimC.
B. Passive Protection in Mice
C3H/HeJ mice (10 mice/group) were passively immunized by two i.p. injections of 100 μl of rabbit serum. The first injection was administered twenty-four hours before challenge with 172 cfu of S. pneumoniae strain SJ2, and the second injection was given four hours after challenge. Figure 2 shows the survival of mice after infection with two different strains of pneumococci.
Figure 2A shows that of mice injected with 172 cfu of strain SJ2 (Figure 2A), one hundred percent of the mice immunized with rabbit immune serum raised against Sp36 protein survived the 21 -day observation period. Of the mice immunized with the control serum (anti-FimC), eighty percent died by day 8, and ninety percent died by day 12. Figure 2B shows that of mice injected with 862 cfu of strain EF6796, ninety percent of the mice immunized with rabbit immune serum raised against Sp36 protein survived the 8-day observation period. Of those given a control serum (collected from a rabbit before immunization), ninety percent died by day 8.
These data indicate that the protection against pneumococcal infection resulting from immunization with Sp36 is antibody-mediated, since mice can be protected by passive transfer of serum from a hyperimmunized rabbit. As seen in the mouse active challenge experiments described above, serum directed against recombinant Sp36 protein cloned from a serotype 4 strain was protective against challenge with heterologous strains.
Example 3 Conservation of Sp36 Among Strains of S. pneumoniae
A. Western blotting
The 23 pneumococcal strains used in this experiment were obtained from the American Type Culture Collection (Rockville, MD) and include one isolate each of the 23 serotypes in the multivalent pneumococcal vaccine. For total cell lysates, pneumococci were grown to mid-logarithmic phase (optical density at 620 nm, 0.4 to 0.6) in 2 ml Todd-Hewitt broth with 0.5% yeast extract (Difco, Detroit, ME) at 37°C. Bacteria were harvested by centrifugation and washed twice with water. Pellets were resuspended in 200 μl lysis buffer (0.01 % sodium dodecyl sulfate, 0.15 M sodium citrate and 0.1 % sodium deoxycholate) and incubated at 37°C for 30 min, then diluted in an equal volume 2x SSC (0.3 M sodium chloride, 0.03 M sodium citrate). Lysates were separated by SDS-PAGE, transferred to nitrocellulose membranes (Bio-Rad Laboratories, Hercules, CA), and probed with antibody in a standard Western blotting procedure. Sera from ten C3H/HeJ mice immunized with Sp36 (as described in Example 1 ) were pooled and used at a dilution of 1 :3000. Bound antibody was detected with peroxidase- conjugated sheep anti- mouse IgG using the chemiluminescence kit from Amersham, Inc. (Cambridge, MA).
The mouse anti-Sp36 sera detected two major bands with apparent molecular weights of 97 and 100 kDa in all 23 pneumococcal lysates tested (shown in Figure 3). The Sp36 signals obtained from S. pneumoniae serotypes 1 , 5, 17F and 22F were lower, indicating either that the level of Sp36 expression is reduced in these strains, or that Sp36 in these strains is antigenically different.
These data show that Sp36 is antigenically conserved among strains of the 23 pneumococcal serotypes represented in the current polysaccharide vaccine.
B. Southern blotting
Genomic DNA was prepared from each of the 23 pneumococcal strains listed in the previous section and also from strain SJ2. DNA was digested with PvuW and BamYW, electrophoresed in an agarose gel and transferred to a nylon membrane. A probe was prepared by amplifying the Sp36 gene from Norway type 4 DNA (as in Example 1 ) and labeling the amplified fragment with fluorescein by the random-priming method, using a kit from Amersham. Hybridization, washing, and exposure of film were carried out as in the protocol supplied by Amersham. Figure 4 shows that the Sp36 probe hybridized with DNA from each of the 24 strains studied. The lane marked "M" contained DNA from lambda phage, digested with Hind\\\ and labeled with fluorescein, as molecular weight markers.
Example 4
Immunogenicity of Sp36 in Humans
In order to determine whether Sp36 is immunogenic during human pneumococcal infection, sera from patients with culture-proven pneumococcal bacteremia were used in Western blots containing recombinant Sp36 protein. In the experiment shown in Figure 5, sera from five patients (indicated as 1 through 5) were diluted 1 :3000 and used to probe blots containing full-length Sp36, the N-terminal half of Sp36 (preceding the proline-rich region), or the C-terminal half of Sp36 (following the proline-rich region). Lanes labeled A (acute) were probed with serum collected shortly after diagnosis of pneumococcal infection; lanes C (convalescent) were probed with serum collected either one month later (patients 1 , 2, and 3) or eight days after the first serum collection (patients 4 and 5). For patients 2, 3 and 5, reactivity of the convalescent serum with Sp36 was stronger that that of the corresponding acute serum. The difference between the acute and convalescent sera was particularly evident for reactivity with the C-terminal half of the protein.
In additional experiments (not shown), convalescent sera from 23 patients with pneumococcal infections were tested individually for reactivity with full-length Sp36: 20 of the 23 sera were found to bind Sp36 on a Western blot.
These experiments indicate that Sp36 is recognized by the human immune system and suggest that antibodies able to bind the Sp36 protein may be produced during natural S. pneumoniae infection in humans. Since the patients were infected with a variety of pneumococcal strains, these data also support the idea that Sp36 is antigenically conserved.
Example 5
Table 1 provides the percent identity between the various sequences.
Alignment of the predicted amino acid sequences of PhtA, PhtB, PhtD, and PhtE using the MEGALIGN program of Lasergene showed strong N- terminal homology with substantial divergence of the C-termini (Figure 6). The alignment of the nucleotide sequences of the same genes is shown in Figure 7. Amino acid and nucleotide sequences were compared using the identity weighting in a Lipman-Pearson pairwise alignment, in which the number of matching residues is divided by the total of matching residues plus the number of mismatched residues plus the number of residues in gaps. In the table below, the percent identity between each pair of sequences is shown at the intersection of the corresponding row and column.
Example 6
Active Protection with PhtD Vaccination.
Mice immunized with recombinant PhtD derived from strain N4
generated potent antibody titers (reciprocal endpoint titers ranging form
2,048,00 to 4,096,000). Mice immunized with PhtD were protected against
death following intraperitoneal injection with either of three heterologous
strains, SJ2 (serotype 6B; provided by P. Flynn, St. Jude Children's Research Hospital, Memphis, TN), EF6796 (serotype 6A) or EF5668 (serotype 4; both strains provided by D. Briles, University of Alabama, Birmingham). In the experiment shown in Figure 8 (Panel A), all ten of the sham-immunized mice died within 10-days after challenge with virulent pneumococci (strain SJ2), while eighty percent of the PhtD-immunized mice survived the 1 5-day observation period. Immunization with PhtD also protected against a serotype 6A strain, EF6796 (Panel B) and a serotype 4 strain, EF5668 (Panel C). In the experiment shown in Figure 8 (Panel B), all ten of the sham-immunized mice died within 7-days after challenge with virulent pneumococci (strain EF6796), while ninety percent of the PhtD-immunized mice survived the 1 5- day observation period. In the experiment shown in Figure 8 (Panel C), all ten of the sham-immunized mice died within 6-days after challenge with virulent pneumoccoci (strain EF5668), while eight of nine mice immunized with PhtD survived the 1 5-day observation period.
Table 1 . Percent Identities
Percent Identity Between Amino Acid Sequences
PhtA PhtB PhtD PhtE
PhtA — 66.4 63.9 49.5
PhtB — 87.2 49.5
PhtD 49.8
PhtE —
Percent Identity Between Nucleotide Sequences
PhtA PhtB PhtD PhtE
PhtA — 58.3 59.3 47.9
PhtB — 86.4 47.4
PhtD 47.9
PhtE —

Claims

WHAT IS CLAIMED IS:
1 . A vaccine composition comprising:
(a) at least one member selected from the groups consisting of (i) a polypeptide comprising a polypeptide sequence that is at least 90% identical to amino acids 1 -81 9 of SEQ ID NO:4; (ii) a polypeptide comprising a polypeptide sequence that is at least 90% identical to amino acids 1 -460 of SEQ ID NO:6; (iii) a fragment of the polypeptide of (i) that includes at least one of a histidine triad residue or coiled-coil region; (iv) a fragment of the polypeptide of (ii) that includes at least one of a histidine triad residue or a coiled-coil region; (v) a fragment of a polypeptide that is at least 90% identical to the polypeptide sequence comprising amino acids 1 -800 of SEQ ID NO:8, wherein said fragment includes at least one of a histidine triad residue or coiled-coil region wherein said fragment includes at least 80 amino acids and no more than 680 amino acids; and (vi) a fragment of a polypeptide that is at least 90% identical to the polypeptide sequence comprising amino acids 1 -800 of SEQ ID NO: 10, wherein said fragment includes at least one of a histidine triad residue or coiled-coil region wherein said fragment includes at least 80 amino acids and no more than 680 amino acids; and
(b) a pharmaceutically acceptable carrier.
2. A process for preventing infection caused by S. pneumoniae comprising: administering the vaccine of claim 1 .
3. A vaccine composition comprising:
(a) at least one antibody against a member selected from the group consistng of (i) a polypeptide comprising a polypeptide sequence that is at least 90% identical to amino acids 1 -81 9 of SEQ ID NO:4; (ii) a polypeptide comprising a polypeptide sequence that is at least 90% identical to amino acids 1 -460 of SEQ ID NO:6; (iii) a fragment of the polypeptide of (i) that includes at least one of histidine triad residue or coiled-coil region; (iv) a fragment of the polypeptide of (ii) that includes at least one of a histidine triad residue or a coiled-coil region; (v) a fragment of a polypeptide that is at least 90% identical to the polypeptide sequence comprising amino acids 1 - 800 of SEQ ID NO:8, wherein said fragment includes at least one of a histidine triad residue or coiled-coil region wherein said fragment includes at least 80 amino acids and no more than 680 amino acids and (vi) a fragment of a polypeptide that is at least 90% identical to the polypeptide sequence comprising amino acids 1 -800 of SEQ ID NO: 1 0, wherein said fragment includes at least one of a histidine triad residue or coiled-coil region wherein said fragment includes at least 80 amino acids and no more than 680 amino acids.
PCT/US1999/030390 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines WO2000037105A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP99967460A EP1140157B1 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
EP09001206.3A EP2050464B1 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
DE69940439T DE69940439D1 (en) 1998-12-21 1999-12-21 STREPTOCOCCUS PNEUMONIAE PROTEINS AND IMMUNOGENIC FRAGMENTS FOR IMPORTS
JP2000589215A JP4689044B2 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccine use
AU23731/00A AU776828B2 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
DK99967460T DK1140157T3 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
CA2355364A CA2355364C (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
AU2004242430A AU2004242430C1 (en) 1998-12-21 2004-12-22 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11304898P 1998-12-21 1998-12-21
US60/113,048 1998-12-21

Publications (2)

Publication Number Publication Date
WO2000037105A2 true WO2000037105A2 (en) 2000-06-29
WO2000037105A3 WO2000037105A3 (en) 2000-11-09

Family

ID=22347343

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/030390 WO2000037105A2 (en) 1998-12-21 1999-12-21 Streptococcus pneumoniae proteins and immunogenic fragments for vaccines

Country Status (12)

Country Link
US (5) US6582706B1 (en)
EP (2) EP2050464B1 (en)
JP (1) JP4689044B2 (en)
AT (1) ATE422899T1 (en)
AU (1) AU776828B2 (en)
CA (1) CA2355364C (en)
CY (1) CY1109024T1 (en)
DE (1) DE69940439D1 (en)
DK (1) DK1140157T3 (en)
ES (1) ES2322306T3 (en)
PT (1) PT1140157E (en)
WO (1) WO2000037105A2 (en)

Cited By (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001096368A2 (en) * 2000-06-14 2001-12-20 Cytovax Biotechnologies, Inc. Use of coiled-coil structural scaffold to generate structure-specific peptides
WO2002022168A2 (en) * 2000-09-15 2002-03-21 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
WO2003054007A2 (en) * 2001-12-20 2003-07-03 Shire Biochem Inc. Streptococcus antigens
US6676943B1 (en) 1997-04-24 2004-01-13 Regents Of The University Of Minnesota Human complement C3-degrading protein from Streptococcus pneumoniae
JP2004501618A (en) * 2000-06-20 2004-01-22 シャイアー・バイオケム・インコーポレイテッド Streptococcus antigen
US6699703B1 (en) 1997-07-02 2004-03-02 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7074415B2 (en) 2000-06-20 2006-07-11 Id Biomedical Corporation Streptococcus antigens
US7081530B1 (en) 1997-07-02 2006-07-25 sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7128918B1 (en) 1998-12-23 2006-10-31 Id Biomedical Corporation Streptococcus antigens
EP1754717A1 (en) 2005-08-19 2007-02-21 Université de Lausanne Antigenic peptides and their use
EP1801218A2 (en) * 1998-07-27 2007-06-27 Sanofi Pasteur Limited Nucleic acids and proteins from streptococcus pneumoniae
WO2007071707A2 (en) 2005-12-22 2007-06-28 Glaxosmithkline Biologicals Sa Pneumococcal polysaccharide conjugate vaccine
WO2007116028A2 (en) 2006-04-07 2007-10-18 Glaxosmithkline Biologicals S.A. Conjugate vaccines
WO2008028956A1 (en) 2006-09-07 2008-03-13 Glaxosmithkline Biologicals S.A. Vaccine
EP1950302A2 (en) 1998-12-23 2008-07-30 ID Biomedical Corporation Streptococcus antigens
WO2009000826A1 (en) 2007-06-26 2008-12-31 Glaxosmithkline Biologicals S.A. Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
EP2023143A1 (en) * 2007-08-06 2009-02-11 Boehringer Ingelheim Vetmedica Gmbh Immunogenic streptococcus proteins
WO2009111337A1 (en) 2008-03-03 2009-09-11 Irm Llc Compounds and compositions as tlr activity modulators
WO2010125480A1 (en) 2009-04-30 2010-11-04 Coley Pharmaceutical Group, Inc. Pneumococcal vaccine and uses thereof
US20100297133A1 (en) * 2007-07-23 2010-11-25 Martina Ochs Immunogenic Polypeptides and Monoclonal Antibodies
WO2010144734A1 (en) 2009-06-10 2010-12-16 Novartis Ag Benzonaphthyridine-containing vaccines
EP2261358A3 (en) * 1998-12-23 2011-02-23 ID Biomedical Corporation Novel streptococcus antigen
WO2011027257A2 (en) 2009-09-03 2011-03-10 Pfizer Vaccines Llc Pcsk9 vaccine
WO2011027222A2 (en) 2009-09-02 2011-03-10 Novartis Ag Immunogenic compositions including tlr activity modulators
WO2011030218A1 (en) 2009-09-10 2011-03-17 Novartis Ag Combination vaccines against respiratory tract diseases
WO2011049677A1 (en) 2009-09-02 2011-04-28 Irm Llc Compounds and compositions as tlr activity modulators
WO2011057148A1 (en) 2009-11-05 2011-05-12 Irm Llc Compounds and compositions as tlr-7 activity modulators
WO2011075823A1 (en) * 2009-12-22 2011-06-30 Sanofi Pasteur Limited Immunogenic compositions
WO2011084549A1 (en) 2009-12-15 2011-07-14 Novartis Ag Homogeneous suspension of immunopotentiating compounds and uses thereof
EP2351578A1 (en) 2005-06-27 2011-08-03 GlaxoSmithKline Biologicals S.A. Process for manufacturing vaccines
EP2364724A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
WO2011110570A1 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Treatment of streptococcal infections
WO2011110241A1 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Immunogenic composition comprising s. pneumoniae polysaccharides conjugated to carrier proteins
WO2011110531A2 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Conjugation process
WO2011119759A1 (en) 2010-03-23 2011-09-29 Irm Llc Compounds (cystein based lipopeptides) and compositions as tlr2 agonists used for treating infections, inflammations, respiratory diseases etc.
WO2012072769A1 (en) 2010-12-01 2012-06-07 Novartis Ag Pneumococcal rrgb epitopes and clade combinations
US8226953B2 (en) 1998-02-20 2012-07-24 Id Biomedical Corporation Of Quebec Group B Streptococcus antigens
WO2012119972A1 (en) 2011-03-07 2012-09-13 Glaxosmithkline Biologicals S.A. Conjugation process
WO2012131504A1 (en) 2011-03-02 2012-10-04 Pfizer Inc. Pcsk9 vaccine
WO2012156391A1 (en) 2011-05-17 2012-11-22 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
US8324354B2 (en) 2001-02-02 2012-12-04 Stichting Dienst Landbouwkundig Onderzoek Environmentally regulated genes of Streptococcus suis
EP2572726A1 (en) 2007-08-01 2013-03-27 Novartis AG Compositions comprising pneumococcal antigens
EP2612680A1 (en) 2008-04-16 2013-07-10 GlaxoSmithKline Biologicals SA Vaccine
WO2013131983A1 (en) 2012-03-07 2013-09-12 Novartis Ag Adjuvanted formulations of streptococcus pneumoniae antigens
EP2682127A1 (en) 2007-05-02 2014-01-08 GlaxoSmithKline Biologicals S.A. Vaccine
US8632784B2 (en) 1998-07-27 2014-01-21 Sanofi Pasteur Limited Nucleic acids and proteins from Streptococcus pneumoniae
US8668911B2 (en) 2009-05-14 2014-03-11 The Regents Of The University Of Michigan Streptococcus vaccine compositions and methods of using the same
US8728490B2 (en) 1998-07-22 2014-05-20 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
WO2014118305A1 (en) 2013-02-01 2014-08-07 Novartis Ag Intradermal delivery of immunological compositions comprising toll-like receptor agonists
US8962026B2 (en) 2008-09-26 2015-02-24 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
WO2015110940A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
WO2015110941A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2015110942A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
WO2015121783A1 (en) 2014-02-14 2015-08-20 Pfizer Inc. Immunogenic glycoprotein conjugates
EP3017826A1 (en) 2009-03-24 2016-05-11 Novartis AG Combinations of meningococcal factor h binding protein and pneumococcal saccharide conjugates
WO2016113644A1 (en) 2015-01-15 2016-07-21 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2017013548A1 (en) 2015-07-21 2017-01-26 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2017067962A1 (en) 2015-10-21 2017-04-27 Glaxosmithkline Biologicals S.A. Vaccine
WO2017085586A1 (en) 2015-11-20 2017-05-26 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2017216286A1 (en) 2016-06-17 2017-12-21 Glaxosmithkline Biologicals S.A. Immunogenic composition
WO2018134693A1 (en) 2017-01-20 2018-07-26 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2019157509A1 (en) 2018-02-12 2019-08-15 Inimmune Corporation Toll-like receptor ligands
WO2020016322A1 (en) 2018-07-19 2020-01-23 Glaxosmithkline Biologicals Sa Processes for preparing dried polysaccharides
WO2020039359A2 (en) 2018-08-24 2020-02-27 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2020121159A1 (en) 2018-12-12 2020-06-18 Pfizer Inc. Immunogenic multiple hetero-antigen polysaccharide-protein conjugates and uses thereof
WO2020170190A1 (en) 2019-02-22 2020-08-27 Pfizer Inc. Methods for purifying bacterial polysaccharides
WO2020208502A1 (en) 2019-04-10 2020-10-15 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2021021729A1 (en) 2019-07-31 2021-02-04 Sanofi Pasteur Inc. Multivalent pneumococcal polysaccharide-protein conjugate compositions and methods of using the same
WO2021084429A1 (en) 2019-11-01 2021-05-06 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2021165928A2 (en) 2020-02-23 2021-08-26 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2021165847A1 (en) 2020-02-21 2021-08-26 Pfizer Inc. Purification of saccharides
US11160855B2 (en) 2014-01-21 2021-11-02 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2022084852A1 (en) 2020-10-22 2022-04-28 Pfizer Inc. Methods for purifying bacterial polysaccharides
WO2022090893A2 (en) 2020-10-27 2022-05-05 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022097010A1 (en) 2020-11-04 2022-05-12 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2022096590A1 (en) 2020-11-04 2022-05-12 Eligo Bioscience Phage-derived particles for in situ delivery of dna payload into c. acnes population
WO2022137078A1 (en) 2020-12-23 2022-06-30 Pfizer Inc. E. coli fimh mutants and uses thereof
WO2022234405A1 (en) 2021-05-03 2022-11-10 Pfizer Inc. Vaccination against bacterial and betacoronavirus infections
WO2022234416A1 (en) 2021-05-03 2022-11-10 Pfizer Inc. Vaccination against pneumoccocal and covid-19 infections
WO2022249107A2 (en) 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2022249106A2 (en) 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2023135515A1 (en) 2022-01-13 2023-07-20 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2023161817A1 (en) 2022-02-25 2023-08-31 Pfizer Inc. Methods for incorporating azido groups in bacterial capsular polysaccharides
EP4272750A2 (en) 2013-02-07 2023-11-08 Children's Medical Center, Corp. Protein antigens that provide protection against pneumococcal colonization and/or disease
WO2023218322A1 (en) 2022-05-11 2023-11-16 Pfizer Inc. Process for producing of vaccine formulations with preservatives

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2269663A1 (en) * 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
WO1999051266A2 (en) * 1998-04-07 1999-10-14 Medimmune, Inc. Derivatives of pneumococcal choline binding proteins for vaccines
PL355178A1 (en) * 1999-03-19 2004-04-05 Smithkline Beecham Biologicals S.A. Vaccine
US6887480B1 (en) * 1999-06-10 2005-05-03 Medimmune, Inc. Streptococcus pneumoniae proteins and vaccines
US8229903B2 (en) * 2002-12-19 2012-07-24 International Business Machines Corporation Suggesting data interpretations and patterns for updating policy documents
CA2683748C (en) 2007-04-13 2016-02-16 The Board Of Regents Of The University Of Oklahoma Mutants of cholesterol-dependent cytolysins and uses thereof
US20160228500A9 (en) * 2007-07-23 2016-08-11 Martina Ochs Immunogenic Polypeptides and Monoclonal Antibodies
EP2515940A4 (en) 2009-12-22 2013-10-09 Sanofi Pasteur Ltd Immunogenic compositions and related methods
AU2011336366B2 (en) 2010-12-03 2016-05-12 Sanofi Pasteur Limited Composition for immunization against Streptococcus pneumoniae
AU2015349787B2 (en) 2014-11-21 2021-07-29 The Board Of Regents Of The University Of Oklahoma Pneumolysin mutants and methods of use thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998018930A2 (en) * 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
WO1999015675A1 (en) * 1997-09-24 1999-04-01 Regents Of The University Of Minnesota HUMAN COMPLEMENT C3-DEGRADING PROTEINASE FROM $i(STREPTOCOCCUS PNEUMONIAE)
WO2000006737A2 (en) * 1998-07-27 2000-02-10 Microbial Technics Limited Streptococcus pneumoniae proteins and nucleic acid molecules

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5100664A (en) * 1985-09-20 1992-03-31 Cetus Corporation Human IL-2 as a vaccine adjuvant
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6042838A (en) * 1991-02-15 2000-03-28 Uab Research Foundation immunogenic compositions for mucosal administration of pneumococcal surface protein A (PspA)
US5928900A (en) 1993-09-01 1999-07-27 The Rockefeller University Bacterial exported proteins and acellular vaccines based thereon
ATE323721T1 (en) 1996-05-01 2006-05-15 Univ Rockefeller CHOLINE-BINDING PROTEIN USED AS AN PNEUMOCOCCAL VACCINE
WO1997048417A1 (en) 1996-06-21 1997-12-24 Virginia Commonwealth University Vaccine to prevent streptococcal endocarditis
US6800744B1 (en) * 1997-07-02 2004-10-05 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
CN1352691A (en) 1998-09-24 2002-06-05 美国明尼苏达州大学 Human complement C3-degrading polypeptide from i(streptococcus pneumoniae)
DE69938670D1 (en) * 1998-12-23 2008-06-19 Id Biomedical Corp STREPTOCOCCUS ANTIGENE

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998018930A2 (en) * 1996-10-31 1998-05-07 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
WO1999015675A1 (en) * 1997-09-24 1999-04-01 Regents Of The University Of Minnesota HUMAN COMPLEMENT C3-DEGRADING PROTEINASE FROM $i(STREPTOCOCCUS PNEUMONIAE)
WO2000006737A2 (en) * 1998-07-27 2000-02-10 Microbial Technics Limited Streptococcus pneumoniae proteins and nucleic acid molecules

Cited By (196)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6676943B1 (en) 1997-04-24 2004-01-13 Regents Of The University Of Minnesota Human complement C3-degrading protein from Streptococcus pneumoniae
US7081530B1 (en) 1997-07-02 2006-07-25 sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7404958B2 (en) 1997-07-02 2008-07-29 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7098023B1 (en) 1997-07-02 2006-08-29 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7405291B2 (en) 1997-07-02 2008-07-29 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7115731B1 (en) 1997-07-02 2006-10-03 sanofi pasteur limited/ sanofi pasteur limiteé Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7335493B2 (en) 1997-07-02 2008-02-26 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7396532B2 (en) 1997-07-02 2008-07-08 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US6699703B1 (en) 1997-07-02 2004-03-02 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7390493B2 (en) 1997-07-02 2008-06-24 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7388090B2 (en) 1997-07-02 2008-06-17 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7385047B1 (en) 1997-07-02 2008-06-10 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7122368B1 (en) 1997-07-02 2006-10-17 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7381814B1 (en) 1997-07-02 2008-06-03 Sanofi Pasteur Limted Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7326544B2 (en) 1997-07-02 2008-02-05 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7442523B2 (en) 1997-07-02 2008-10-28 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7335494B2 (en) 1997-07-02 2008-02-26 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7074914B1 (en) 1997-07-02 2006-07-11 sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7129340B1 (en) 1997-07-02 2006-10-31 sanofi pasteur limited/sanofi pasteur limitée Nucleic acid and amino acid sequences relating to streptococcus pneumoniae for diagnostics and therapeutics
US7129339B1 (en) 1997-07-02 2006-10-31 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7381816B2 (en) 1997-07-02 2008-06-03 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7135560B1 (en) 1997-07-02 2006-11-14 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7151171B1 (en) 1997-07-02 2006-12-19 sanofi pasteur limited/sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7153952B1 (en) 1997-07-02 2006-12-26 sanofi pasteur limited/sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7381815B2 (en) 1997-07-02 2008-06-03 Sanofi Parker Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7378258B2 (en) 1997-07-02 2008-05-27 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7378514B2 (en) 1997-07-02 2008-05-27 sanofi pasteur limited/sanofi pasteur limitée Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US7338786B2 (en) 1997-07-02 2008-03-04 Sanofi Pasteur Limited Nucleic acid and amino acid sequences relating to Streptococcus pneumoniae for diagnostics and therapeutics
US8580262B2 (en) 1998-02-20 2013-11-12 Id Biomedical Corporation Of Quebec Group B Streptococcus antigens
US8226953B2 (en) 1998-02-20 2012-07-24 Id Biomedical Corporation Of Quebec Group B Streptococcus antigens
US8728490B2 (en) 1998-07-22 2014-05-20 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
USRE45170E1 (en) 1998-07-22 2014-09-30 Stichting Dienst Landbouwkundig Onderzoek Streptococcus suis vaccines and diagnostic tests
EP1801218A2 (en) * 1998-07-27 2007-06-27 Sanofi Pasteur Limited Nucleic acids and proteins from streptococcus pneumoniae
EP1801218A3 (en) * 1998-07-27 2007-10-10 Sanofi Pasteur Limited Nucleic acids and proteins from streptococcus pneumoniae
US8632784B2 (en) 1998-07-27 2014-01-21 Sanofi Pasteur Limited Nucleic acids and proteins from Streptococcus pneumoniae
US7635482B2 (en) 1998-12-23 2009-12-22 Id Biomedical Corporation Streptococcus antigens
EP1950302A2 (en) 1998-12-23 2008-07-30 ID Biomedical Corporation Streptococcus antigens
EP2261358A3 (en) * 1998-12-23 2011-02-23 ID Biomedical Corporation Novel streptococcus antigen
US8211437B2 (en) 1998-12-23 2012-07-03 Id Biomedical Corporation Of Quebec Streptococcus antigens
US7128918B1 (en) 1998-12-23 2006-10-31 Id Biomedical Corporation Streptococcus antigens
EP1950302A3 (en) * 1998-12-23 2010-06-23 ID Biomedical Corporation Streptococcus antigens
WO2001096368A3 (en) * 2000-06-14 2002-05-02 Cytovax Biotechnologies Inc Use of coiled-coil structural scaffold to generate structure-specific peptides
WO2001096368A2 (en) * 2000-06-14 2001-12-20 Cytovax Biotechnologies, Inc. Use of coiled-coil structural scaffold to generate structure-specific peptides
EP2281891A2 (en) 2000-06-20 2011-02-09 ID Biomedical Corporation Streptococcus antigens
US7074415B2 (en) 2000-06-20 2006-07-11 Id Biomedical Corporation Streptococcus antigens
EP2281891A3 (en) * 2000-06-20 2011-08-17 ID Biomedical Corporation Streptococcus antigens
JP2004501618A (en) * 2000-06-20 2004-01-22 シャイアー・バイオケム・インコーポレイテッド Streptococcus antigen
JP2012187110A (en) * 2000-06-20 2012-10-04 Id Biomedical Corp Of Quebec Streptococcus antigen
AU2002238193B2 (en) * 2000-09-15 2005-05-12 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
JP2004508416A (en) * 2000-09-15 2004-03-18 グラクソスミスクライン・バイオロジカルス・ソシエテ・アノニム vaccine
WO2002022168A3 (en) * 2000-09-15 2002-06-27 Smithkline Beecham Biolog Vaccine against streptococcus pneumoniae
JP4903975B2 (en) * 2000-09-15 2012-03-28 グラクソスミスクライン・バイオロジカルス・ソシエテ・アノニム vaccine
US20140099339A1 (en) * 2000-09-15 2014-04-10 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
JP2004508417A (en) * 2000-09-15 2004-03-18 グラクソスミスクライン・バイオロジカルス・ソシエテ・アノニム vaccine
EP2305297A1 (en) 2000-09-15 2011-04-06 GlaxoSmithKline Biologicals s.a. Vaccine against streptococcus pneumoniae
NO337730B1 (en) * 2000-09-15 2016-06-13 Glaxosmithkline Biologicals Sa Immunogenic composition, vaccine containing it, and use and method of preparation.
EP2140878A1 (en) 2000-09-15 2010-01-06 GlaxoSmithKline Biologicals S.A. Vaccine against streptococcus pneumoniae
JP2012006969A (en) * 2000-09-15 2012-01-12 Glaxosmithkline Biologicals Sa Vaccine
WO2002022168A2 (en) * 2000-09-15 2002-03-21 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
CZ305343B6 (en) * 2000-09-15 2015-08-12 Smithkline Beecham Biologicals S. A. Immunogenic formulation, vaccine containing thereof, its use and process for its preparation
EP2314313A1 (en) 2000-09-15 2011-04-27 GlaxoSmithKline Biologicals S.A. Vaccine against streptococcus pneumoniae
EP2305298A1 (en) 2000-09-15 2011-04-06 GlaxoSmithKline Biologicals s.a. Vaccine against streptococcus pneumoniae
KR101268790B1 (en) * 2000-09-15 2013-05-29 글락소스미스클라인 바이오로지칼즈 에스.에이. Vaccine
US20080081050A1 (en) * 2000-09-15 2008-04-03 Philippe Hermand Vaccine against streptococcus pneumoniae
US8324354B2 (en) 2001-02-02 2012-12-04 Stichting Dienst Landbouwkundig Onderzoek Environmentally regulated genes of Streptococcus suis
WO2003054007A3 (en) * 2001-12-20 2003-09-04 Shire Biochem Inc Streptococcus antigens
US7262024B2 (en) 2001-12-20 2007-08-28 Id Biomedical Corporation Streptococcus antigens
WO2003054007A2 (en) * 2001-12-20 2003-07-03 Shire Biochem Inc. Streptococcus antigens
EP2351578A1 (en) 2005-06-27 2011-08-03 GlaxoSmithKline Biologicals S.A. Process for manufacturing vaccines
EP1754717A1 (en) 2005-08-19 2007-02-21 Université de Lausanne Antigenic peptides and their use
EP2364722A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
EP2364723A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
US10039823B2 (en) 2005-12-13 2018-08-07 Glaxosmithkline Biologicals, S.A. Vaccine compositions comprising a saponin adjuvant
US10143745B2 (en) 2005-12-13 2018-12-04 GlacoSmithKline Biologicals, S.A. Vaccine compositions comprising a saponin adjuvant
EP2364720A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
EP2364721A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
EP2364724A1 (en) 2005-12-13 2011-09-14 GlaxoSmithKline Biologicals S.A. Vaccine compositions comprising a saponin adjuvant
US10279033B2 (en) 2005-12-22 2019-05-07 Glaxosmithkline Biologicals Sa Vaccine comprising Streptococcus pneumoniae capsular polysaccharide conjugates
US9884113B2 (en) 2005-12-22 2018-02-06 Glaxosmithkline Biologicals, Sa Pneumoccal polysacchride conjugate vaccine
EP3020411A1 (en) 2005-12-22 2016-05-18 GlaxoSmithKline Biologicals s.a. Vaccine
US9107872B2 (en) 2005-12-22 2015-08-18 Glaxosmithkline Biologicals S.A. Pneumococcal polysaccharide conjugate vaccine
US11400147B2 (en) 2005-12-22 2022-08-02 Glaxosmithkline Biologicals Sa Pneumococcal capsular saccharide conjugate vaccine
WO2007071710A2 (en) 2005-12-22 2007-06-28 Glaxosmithkline Biologicals Sa Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
WO2007071711A2 (en) 2005-12-22 2007-06-28 Glaxosmithkline Biologicals Sa Vaccine
WO2007071707A2 (en) 2005-12-22 2007-06-28 Glaxosmithkline Biologicals Sa Pneumococcal polysaccharide conjugate vaccine
EP2382986A2 (en) 2005-12-22 2011-11-02 GlaxoSmithKline Biologicals s.a. Vaccine against streptococcus pneumoniae
EP2384765A2 (en) 2005-12-22 2011-11-09 GlaxoSmithKline Biologicals S.A. Streptococcus pneumoniae vaccine
US10646564B2 (en) 2005-12-22 2020-05-12 Glaxosmithkline Biologicals S.A. Vaccine
EP2402025A2 (en) 2005-12-22 2012-01-04 GlaxoSmithKline Biologicals S.A. Vaccine
WO2007116028A2 (en) 2006-04-07 2007-10-18 Glaxosmithkline Biologicals S.A. Conjugate vaccines
EP2392346A1 (en) 2006-04-07 2011-12-07 GlaxoSmithKline Biologicals SA Streptococcus pneumoniae vaccine
WO2008028956A1 (en) 2006-09-07 2008-03-13 Glaxosmithkline Biologicals S.A. Vaccine
EP2682127A1 (en) 2007-05-02 2014-01-08 GlaxoSmithKline Biologicals S.A. Vaccine
US9610339B2 (en) 2007-06-26 2017-04-04 Glaxosmithkline Biologicals, S.A. Vaccine comprising Streptococcus pneumoniae capsular polysaccharide conjugates
WO2009000826A1 (en) 2007-06-26 2008-12-31 Glaxosmithkline Biologicals S.A. Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
EP2687228A2 (en) 2007-06-26 2014-01-22 GlaxoSmithKline Biologicals S.A. Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
US9610340B2 (en) 2007-06-26 2017-04-04 Glaxosmithkline Biologicals, S.A. Vaccine comprising Streptococcus pneumoniae capsular polysaccharide conjugates
US8337846B2 (en) * 2007-07-23 2012-12-25 Sanofi Pasteur Limited Immunogenic polypeptides and monoclonal antibodies
US20100297133A1 (en) * 2007-07-23 2010-11-25 Martina Ochs Immunogenic Polypeptides and Monoclonal Antibodies
EP2572726A1 (en) 2007-08-01 2013-03-27 Novartis AG Compositions comprising pneumococcal antigens
CN101821627A (en) * 2007-08-06 2010-09-01 贝林格尔.英格海姆维特梅迪卡有限公司 Immunogenic streptococcus proteins
WO2009020391A1 (en) * 2007-08-06 2009-02-12 Boehringer Ingelheim Vetmedica Gmbh Immunogenic streptococcus proteins
EP2023143A1 (en) * 2007-08-06 2009-02-11 Boehringer Ingelheim Vetmedica Gmbh Immunogenic streptococcus proteins
EP2660604A3 (en) * 2007-08-06 2014-02-12 Boehringer Ingelheim Vetmedica GmbH Immunogenic Streptococcus proteins
WO2009111337A1 (en) 2008-03-03 2009-09-11 Irm Llc Compounds and compositions as tlr activity modulators
EP2612680A1 (en) 2008-04-16 2013-07-10 GlaxoSmithKline Biologicals SA Vaccine
US8962026B2 (en) 2008-09-26 2015-02-24 The Regents Of The University Of Michigan Nanoemulsion therapeutic compositions and methods of using the same
EP3017826A1 (en) 2009-03-24 2016-05-11 Novartis AG Combinations of meningococcal factor h binding protein and pneumococcal saccharide conjugates
WO2010125480A1 (en) 2009-04-30 2010-11-04 Coley Pharmaceutical Group, Inc. Pneumococcal vaccine and uses thereof
US9205143B2 (en) 2009-04-30 2015-12-08 Coley Pharmaceutical Group Inc. Pneumococcal vaccine and uses thereof
US8668911B2 (en) 2009-05-14 2014-03-11 The Regents Of The University Of Michigan Streptococcus vaccine compositions and methods of using the same
WO2010144734A1 (en) 2009-06-10 2010-12-16 Novartis Ag Benzonaphthyridine-containing vaccines
WO2011027222A2 (en) 2009-09-02 2011-03-10 Novartis Ag Immunogenic compositions including tlr activity modulators
WO2011049677A1 (en) 2009-09-02 2011-04-28 Irm Llc Compounds and compositions as tlr activity modulators
EP3358008A1 (en) 2009-09-03 2018-08-08 Pfizer Vaccines LLC Pcsk9 vaccine
US8889144B2 (en) 2009-09-03 2014-11-18 Pfizer Vaccines Llc PCSK9 vaccine
WO2011027257A2 (en) 2009-09-03 2011-03-10 Pfizer Vaccines Llc Pcsk9 vaccine
EP2865752A1 (en) 2009-09-03 2015-04-29 Pfizer Vaccines LLC PCSK9 vaccine
US9481875B2 (en) 2009-09-03 2016-11-01 Pfizer Vaccines Llc PCSK9 vaccine
US9987341B2 (en) 2009-09-03 2018-06-05 Pfizer Vaccines Llc PCSK9 vaccine
WO2011030218A1 (en) 2009-09-10 2011-03-17 Novartis Ag Combination vaccines against respiratory tract diseases
WO2011057148A1 (en) 2009-11-05 2011-05-12 Irm Llc Compounds and compositions as tlr-7 activity modulators
US10046048B2 (en) 2009-12-15 2018-08-14 Glaxosmithkline Biologicals S.A. Homogenous suspension of immunopotentiating compounds and uses thereof
US9408907B2 (en) 2009-12-15 2016-08-09 Glaxosmithkline Biologicals Sa Homogenous suspension of immunopotentiating compounds and uses thereof
WO2011084549A1 (en) 2009-12-15 2011-07-14 Novartis Ag Homogeneous suspension of immunopotentiating compounds and uses thereof
WO2011075823A1 (en) * 2009-12-22 2011-06-30 Sanofi Pasteur Limited Immunogenic compositions
US9314518B2 (en) 2010-03-09 2016-04-19 Glaxosmithkline Biologicals S.A. Treatment of streptococcal infections
WO2011110531A2 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Conjugation process
WO2011110570A1 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Treatment of streptococcal infections
EA023702B1 (en) * 2010-03-09 2016-07-29 Глаксосмитклайн Байолоджикалс С.А. METHOD OF TREATING OR PREVENTING A STREPTOCOCCUS PNEUMONIAE INFECTION AND USE OF PhtX PROTEIN
WO2011110241A1 (en) 2010-03-09 2011-09-15 Glaxosmithkline Biologicals S.A. Immunogenic composition comprising s. pneumoniae polysaccharides conjugated to carrier proteins
AU2011226119B2 (en) * 2010-03-09 2014-04-24 Glaxosmithkline Biologicals S.A. Treatment of Streptococcal infections
EP2815762A2 (en) 2010-03-09 2014-12-24 GlaxoSmithKline Biologicals S.A. Conjugation process of bacterial polysaccharides to carrier proteins
WO2011119759A1 (en) 2010-03-23 2011-09-29 Irm Llc Compounds (cystein based lipopeptides) and compositions as tlr2 agonists used for treating infections, inflammations, respiratory diseases etc.
WO2012072769A1 (en) 2010-12-01 2012-06-07 Novartis Ag Pneumococcal rrgb epitopes and clade combinations
WO2012131504A1 (en) 2011-03-02 2012-10-04 Pfizer Inc. Pcsk9 vaccine
WO2012119972A1 (en) 2011-03-07 2012-09-13 Glaxosmithkline Biologicals S.A. Conjugation process
WO2012156391A1 (en) 2011-05-17 2012-11-22 Glaxosmithkline Biologicals S.A. Vaccine against streptococcus pneumoniae
EP2709658A1 (en) * 2011-05-17 2014-03-26 GlaxoSmithKline Biologicals SA Vaccine against streptococcus pneumoniae
WO2013131983A1 (en) 2012-03-07 2013-09-12 Novartis Ag Adjuvanted formulations of streptococcus pneumoniae antigens
WO2014118305A1 (en) 2013-02-01 2014-08-07 Novartis Ag Intradermal delivery of immunological compositions comprising toll-like receptor agonists
US9827190B2 (en) 2013-02-01 2017-11-28 Glaxosmithkline Biologicals Sa Intradermal delivery of immunological compositions comprising toll-like receptor 7 agonists
EP4272750A2 (en) 2013-02-07 2023-11-08 Children's Medical Center, Corp. Protein antigens that provide protection against pneumococcal colonization and/or disease
WO2015110941A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
US11872274B2 (en) 2014-01-21 2024-01-16 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
EP3957321A2 (en) 2014-01-21 2022-02-23 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
WO2015110942A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
US10918708B2 (en) 2014-01-21 2021-02-16 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
US11426456B2 (en) 2014-01-21 2022-08-30 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
US10105431B2 (en) 2014-01-21 2018-10-23 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
WO2015110940A2 (en) 2014-01-21 2015-07-30 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
US9492559B2 (en) 2014-01-21 2016-11-15 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
EP4286000A2 (en) 2014-01-21 2023-12-06 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
EP3616716A2 (en) 2014-01-21 2020-03-04 Pfizer Inc Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
US11160855B2 (en) 2014-01-21 2021-11-02 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
EP3583947A1 (en) 2014-01-21 2019-12-25 Pfizer Inc Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
EP3607966A1 (en) 2014-01-21 2020-02-12 Pfizer Inc Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2015121783A1 (en) 2014-02-14 2015-08-20 Pfizer Inc. Immunogenic glycoprotein conjugates
US10668164B2 (en) 2014-02-14 2020-06-02 Pfizer Inc. Immunogenic glycoprotein conjugates
EP3443983A1 (en) 2014-02-14 2019-02-20 Pfizer Inc Immunogenic glycoprotein conjugates
US11707529B2 (en) 2014-02-14 2023-07-25 Pfizer Inc. Immunogenic glycoprotein conjugates
US11135279B2 (en) 2015-01-15 2021-10-05 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2016113644A1 (en) 2015-01-15 2016-07-21 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
US10653764B2 (en) 2015-01-15 2020-05-19 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
US10124050B2 (en) 2015-07-21 2018-11-13 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
US11020469B2 (en) 2015-07-21 2021-06-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2017013548A1 (en) 2015-07-21 2017-01-26 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2017067962A1 (en) 2015-10-21 2017-04-27 Glaxosmithkline Biologicals S.A. Vaccine
US10786561B2 (en) 2015-11-20 2020-09-29 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2017085586A1 (en) 2015-11-20 2017-05-26 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2017216286A1 (en) 2016-06-17 2017-12-21 Glaxosmithkline Biologicals S.A. Immunogenic composition
US11413344B2 (en) 2017-01-20 2022-08-16 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2018134693A1 (en) 2017-01-20 2018-07-26 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2019157509A1 (en) 2018-02-12 2019-08-15 Inimmune Corporation Toll-like receptor ligands
WO2020016322A1 (en) 2018-07-19 2020-01-23 Glaxosmithkline Biologicals Sa Processes for preparing dried polysaccharides
WO2020039359A2 (en) 2018-08-24 2020-02-27 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2020121159A1 (en) 2018-12-12 2020-06-18 Pfizer Inc. Immunogenic multiple hetero-antigen polysaccharide-protein conjugates and uses thereof
WO2020170190A1 (en) 2019-02-22 2020-08-27 Pfizer Inc. Methods for purifying bacterial polysaccharides
WO2020208502A1 (en) 2019-04-10 2020-10-15 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2021021729A1 (en) 2019-07-31 2021-02-04 Sanofi Pasteur Inc. Multivalent pneumococcal polysaccharide-protein conjugate compositions and methods of using the same
WO2021084429A1 (en) 2019-11-01 2021-05-06 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2021165847A1 (en) 2020-02-21 2021-08-26 Pfizer Inc. Purification of saccharides
WO2021165928A2 (en) 2020-02-23 2021-08-26 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022084852A1 (en) 2020-10-22 2022-04-28 Pfizer Inc. Methods for purifying bacterial polysaccharides
WO2022090893A2 (en) 2020-10-27 2022-05-05 Pfizer Inc. Escherichia coli compositions and methods thereof
WO2022096596A1 (en) 2020-11-04 2022-05-12 Eligo Bioscience Cutibacterium acnes recombinant phages, method of production and uses thereof
WO2022096590A1 (en) 2020-11-04 2022-05-12 Eligo Bioscience Phage-derived particles for in situ delivery of dna payload into c. acnes population
WO2022097010A1 (en) 2020-11-04 2022-05-12 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
WO2022137078A1 (en) 2020-12-23 2022-06-30 Pfizer Inc. E. coli fimh mutants and uses thereof
WO2022234416A1 (en) 2021-05-03 2022-11-10 Pfizer Inc. Vaccination against pneumoccocal and covid-19 infections
WO2022234405A1 (en) 2021-05-03 2022-11-10 Pfizer Inc. Vaccination against bacterial and betacoronavirus infections
WO2022249107A2 (en) 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2022249106A2 (en) 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2023135515A1 (en) 2022-01-13 2023-07-20 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2023161817A1 (en) 2022-02-25 2023-08-31 Pfizer Inc. Methods for incorporating azido groups in bacterial capsular polysaccharides
WO2023218322A1 (en) 2022-05-11 2023-11-16 Pfizer Inc. Process for producing of vaccine formulations with preservatives

Also Published As

Publication number Publication date
CY1109024T1 (en) 2014-07-02
US20040052781A1 (en) 2004-03-18
DE69940439D1 (en) 2009-04-02
EP2050464B1 (en) 2019-08-07
JP2002532561A (en) 2002-10-02
US20070065458A1 (en) 2007-03-22
DK1140157T3 (en) 2009-06-08
EP1140157B1 (en) 2009-02-18
US7122194B2 (en) 2006-10-17
WO2000037105A3 (en) 2000-11-09
EP2050464A2 (en) 2009-04-22
US6582706B1 (en) 2003-06-24
AU776828B2 (en) 2004-09-23
ATE422899T1 (en) 2009-03-15
EP1140157A2 (en) 2001-10-10
JP4689044B2 (en) 2011-05-25
US20040005331A1 (en) 2004-01-08
PT1140157E (en) 2009-05-06
CA2355364A1 (en) 2000-06-29
EP2050464A3 (en) 2012-04-25
AU2373100A (en) 2000-07-12
CA2355364C (en) 2014-03-18
ES2322306T3 (en) 2009-06-18
US20040001836A1 (en) 2004-01-01

Similar Documents

Publication Publication Date Title
US7122194B2 (en) Vaccine compositions comprising Streptococcus pneumoniae polypeptides having selected structural motifs
EP1185297B1 (en) Streptococcus pneumoniae proteins and vaccines
US6863893B2 (en) Derivatives of choline binding proteins for vaccines
US6689369B2 (en) Immunogenic pneumococcal protein and vaccine compositions thereof
AU2001255772A1 (en) Immunogenic pneumococcal protein and vaccine compositions thereof
US6887480B1 (en) Streptococcus pneumoniae proteins and vaccines
WO2001014421A1 (en) Homologs of a pneumococcal protein and fragments for vaccines
AU2004242430B2 (en) Streptococcus pneumoniae proteins and immunogenic fragments for vaccines
AU2004200125B2 (en) Derivatives of Pneumococcal Choline Binding Proteins for Vaccines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

ENP Entry into the national phase

Ref document number: 2355364

Country of ref document: CA

Ref country code: CA

Ref document number: 2355364

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 23731/00

Country of ref document: AU

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 589215

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1999967460

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999967460

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 23731/00

Country of ref document: AU