Multicomponent immunogenic composition for the prevention of beta-hemolytic streptococcal (BHS) disease

Information

  • Patent Grant
  • 9127050
  • Patent Number
    9,127,050
  • Date Filed
    Tuesday, July 16, 2013
    10 years ago
  • Date Issued
    Tuesday, September 8, 2015
    8 years ago
Abstract
A number of β-hemolytic streptococci polynucleotides and polypeptides, particularly Streptococcus pyogenes polypeptides and polynucleotides, are described. Two or more of the polypeptides of the invention can be formulated for use as immunogenic compositions. Also disclosed are methods for immunizing against and reducing infection caused by β-hemolytic streptococci.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. The ASCII copy is named 09531.365US2_SL.txt and is 45,669 bytes in size.


FIELD OF THE INVENTION

This invention relates generally to β-hemolytic streptococcal (BHS) polypeptides and polynucleotides, particularly Streptococcus pyogenes polypeptides and polynucleotides, and their use in multicomponent immunogenic compositions to prevent BHS disease. More specifically, the invention relates to polypeptides of Streptococcus pyogenes which are surface localized. The invention further relates to immunogenic compositions, and methods for immunizing against and reducing β-hemolytic streptococcal infection comprising combinations of two or more of polypeptides.


BACKGROUND OF THE INVENTION

Traditional phenotypic criteria for classification of streptococci include both hemolytic reactions and Lancefield serological groupings. However, with taxonomic advances, it is now known that unrelated species of β-hemolytic (defined as the complete lysis of sheep erythrocytes in agar plates) streptococci (BHS) may produce identical Lancefield antigens and that strains genetically related at the species level may have heterogeneous Lancefield antigens. In spite of these exceptions to the traditional rules of streptococcal taxonomy, hemolytic reactions and Lancefield serological tests can still be used to divide streptococci into broad categories as a first step in identification of clinical isolates. Ruoff, K. L., R. A. Whiley, and D. Beighton. 1999. Streptococcus. In P. R. Murray, E. J. Baron, M. A. Pfaller, F. C. Tenover, and R. H. Yolken (eds.), Manual of Clinical Microbiology. American Society of Microbiology Press, Washington D.C.


β-hemolytic isolates with Lancefield group A, C, or G antigen can be subdivided into two groups: large-colony (>0.5 mm in diameter) and small-colony (<0.5 mm in diameter) formers. Large-colony-forming group A (Streptococcus pyogenes), C, and G strains are “pyogenic” streptococci replete with a variety of effective virulence mechanisms. Streptococcus agalactiae (group B) is still identified reliably by its production of Lancefield group B antigen or other phenotypic traits.


Similarities between BHS species include not only virulence factors, but also disease manifestations. Included in the latter are pneumonia, arthritis, abscesses, rhinopharyngitis, metritis, puerperal sepsis, neonatal septicemia, wound infections, meningitis, peritonitis, cellulitis, pyoderma, necrotizing fasciitis, toxic shock syndrome, septicemia, infective endocarditis, pericarditis, glomerulonephritis, and osteomyelitis.



Streptococcus pyogenes are Gram-positive diplococci that colonize the pharynx and skin of humans, sites that then serve as the primary reservoir for this organism. An obligate parasite, this bacterium is transmitted by either direct contact of respiratory secretions or by hand-to-mouth. The majority of Streptococcus pyogenes infections are relatively mild illnesses, such as pharyngitis or impetigo. Currently, there are anywhere from twenty million to thirty-five million cases of pharyngitis alone in the U.S., costing about $2 billion for physician visits and other related expenses. Additionally, nonsuppurative sequelae such as rheumatic fever, scarlet fever, and glomerulonephritis result from Streptococcus pyogenes infections. Globally, acute rheumatic fever (ARF) is the most common cause of pediatric heart disease (1997. Case definitions for Infectious Conditions Under Public Health Surveillance. CDC.).


From the initial portals of entry, pharynx, and skin, Streptococcus pyogenes can disseminate to other parts of the body where bacteria are not usually found, such as the blood, deep muscle and fat tissue, or the lungs, and can cause invasive infections. Two of the most severe but least common forms of invasive Streptococcus pyogenes disease are necrotizing fasciitis and streptococcal toxic shock syndrome (STSS). Necrotizing fasciitis (described in the media as “flesh-eating bacteria”) is a destructive infection of muscle and fat tissue. STSS is a rapidly progressing infection causing shock and injury to internal organs such as the kidneys, liver, and lungs. Much of this damage is due to a toxemia rather than localized damage due to bacterial growth.


In 1995, invasive Streptococcus pyogenes infections and STSS became mandated reportable diseases. In contrast to the millions of individuals that acquire pharyngitis and impetigo, the U.S. Centers for Disease Control and Prevention (CDC) mandated case reporting indicates that in 1997 there were from 15,000 to 20,000 cases of invasive Streptococcus pyogenes disease in the United States, resulting in over 2,000 deaths (1997. Case definitions for Infectious Conditions Under Public Health Surveillance. CDC.). Other reports estimate invasive disease to be as high as 10-20 cases per 100,000 individuals per year (Stevens, D. L. 1995. Streptococcal toxic-shock syndrome: spectrum of disease, pathogenesis, and new concepts in treatment. Emerg Infect Dis. 1:69-78). More specifically, of the 15,000 to 20,000 cases of invasive disease, 1,100 to 1,500 are cases of necrotizing fasciitis and 1,000 to 1,400 are cases of STSS, with a 20% and 60% mortality rate, respectively. Also included in serious invasive disease are cases of myositis, which carries a fatality rate of 80% to 100%. An additional 10% to 15% of individuals die with other forms of invasive group A streptococcal disease. These numbers have increased since case reporting was initiated in 1995 and reflect a general trend that has occurred over the past decade or two. Additionally, it is commonly agreed that the stringency of the case definitions results in lower and, thus, misleading numbers, in that many cases are successfully resolved due to early diagnosis and treatment before the definition has been met.


While Streptococcus pyogenes remains sensitive to penicillin and its derivatives, treatment does not necessarily eradicate the organism. Approximately 5% to 20% of the human population are carriers depending on the season (Stevens, D. L. 1995. Streptococcal toxic-shock syndrome: spectrum of disease, pathogenesis, and new concepts in treatment. Emerg Infect Dis. 1:69-78), despite antibiotic therapy. The reasons for this are not totally clear and may involve a variety of mechanisms. In cases of serious invasive infections, treatment often requires aggressive surgical intervention. For those cases involving STSS or related disease, clindamycin (a protein synthesis inhibitor) is the preferred antibiotic as it penetrates tissues well and prevents exotoxin production. There are reports of some resistance to tetracycline, sulfa, and most recently, erythromycin. Clearly, there remains a need for compositions to prevent and treat β-hemolytic infection.


Numerous virulence factors have been identified for Streptococcus pyogenes, some secreted and some surface localized. Although it is encapsulated, the capsule is composed of hyaluronic acid and is not suitable as a candidate antigen for inclusion in immunogenic compositions, since it is commonly expressed by mammalian cells and is nonimmunogenic (Dale, J. B., R. G. Washburn, M. B. Marques, and M. R. Wessels. 1996. Hyaluronate capsule and surface M protein in resistance to opsonization of group A streptococci. Infect Immun. 64:1495-501). The T antigen and Group Carbohydrate are other candidates, but may also elicit cross-reactive antibodies to heart tissue. Lipoteichoic acid is present on the surface of Streptococcus pyogenes, but raises safety concerns similar to LPS.


The most abundant surface proteins fall into a family of proteins referred to as M or “M-like” proteins because of their structural similarity. While members of this class have similar biological roles in inhibiting phagocytosis, they each have unique substrate binding properties. The best characterized protein of this family is the helical M protein. Antibodies directed to homologous M strains have been shown to be opsonic and protective (Dale, J. B., R. W. Baird, H. S. Courtney, D. L. Hasty, and M. S. Bronze. 1994. Passive protection of mice against group A streptococcal pharyngeal infection by lipoteichoic acid. J Infect Dis. 169:319-23, Dale, J. B., M. Simmons, E. C. Chiang, and E. Y. Chiang. 1996. Recombinant, Ellen, R. P., and R. J. Gibbons. 1972. M protein-associated adherence of Streptococcus pyogenes to epithelial surfaces: prerequisite for virulence. Infect Immun. 5:826-830). Complicating the use of M protein as a candidate antigen is the fact that there have been approximately 100 different serotypes of M protein identified with several more untyped. Typically, the Class 1 M serotypes, exemplified by serotypes M1, M3, M6, M12, and M18, are associated with pharyngitis, scarlet fever, and rheumatic fever and do not express immunoglobulin binding proteins. Class II M serotypes, such as M2 and M49, are associated with the more common localized skin infections and the sequelae glomerulonephritis, and do express immunoglobulin binding proteins (Podbielski, A., A. Flosdorff, and J. Weber-Heynemann. 1995. The group A streptococcal virR49 gene controls expression of four structural vir regulon genes. Infect Immun. 63:9-20). It is important to note that there is little, if any, heterologous cross-reactivity of antibodies to M serotypes. Equally important is the role these antibodies play in rheumatic fever. Specific regions of M protein elicit antibodies that cross react with host heart tissue, causing or at least correlating with cellular damage (Cunningham, M. W., and A. Quinn. 1997. Immunological crossreactivity between the class I epitope of streptococcal M protein and myosin. Adv Exp Med Biol. 418:887-921, Quinn, A., K. Ward, V. A. Fischetti, M. Hemric, and M. W. Cunningham. 1998. Immunological relationship between the class I epitope of streptococcal M protein and myosin. Infect Immun. 66:4418-24).


M and M-like proteins belong to a large family of surface localized proteins that are defined by the sortase-targeted LPXTG motif (Mazmanian, S. K., G. Liu, H. Ton-That, and O. Schneewind. 1999. Staphylococcus aureus sortase, an enzyme that anchors surface proteins to the cell wall. Science. 285:760-3, Ton-That, H., G. Liu, S. K. Mazmanian, K. F. Faull, and O. Schneewind. 1999. Purification and characterization of sortase, the transpeptidase that cleaves surface proteins of Staphylococcus aureus at the LPXTG motif. Proc Natl Acad Sci USA. 96:12424-12429). This motif, located near the carboxy-terminus of the protein, is first cleaved by sortase between the threonine and glycine residues of the LPXTG motif. Once cleaved, the protein is covalently attached via the carboxyl of threonine to a free amide group of the amino acid cross-bridge in the peptidoglycan, thus permanently attaching the protein to the surface of the bacterial cell. Included in this family of sortase-targeted proteins are the C5a peptidase (Chen, C. C., and P. P. Cleary. 1989. Cloning and expression of the streptococcal C5a peptidase gene in Escherichia coli: linkage to the type 12 M protein gene. Infect. Immun. 57:1740-1745, Chmouryguina, I., A. Suvorov, P. Ferrieri, and P. P. Cleary. 1996. Conservation of the C5a peptidase genes in group A and B streptococci. Infect. Immun. 64:2387-2390), adhesins for fibronectin (Courtney, H. S., Y. Li, J. B. Dale, and D. L. Hasty. 1994. Cloning, sequencing, and expression of a fibronectin/fibrinogen-binding protein from group A streptococci. Infect Immun. 62:3937-46, Fogg, G. C., and M. G. Caparon. 1997. Constitutive expression of fibronectin binding in Streptococcus pyogenes as a result of anaerobic activation of rofA. J Bacteriol. 179:6172-80, Hanski, E., and M. Caparon. 1992. Protein F, a fibronectin-binding protein, is an adhesion of the group A streptococcus Streptococcus pyogenes. Proc Natl Acad Sci., USA. 89:6172-76, Hanski, E., P. A. Horwitz, and M. G. Caparon. 1992. Expression of protein F, the fibronectin-binding protein of Streptococcus pyogenes JRS4, in heterologous streptococcal and enterococcal strains promotes their adherence to respiratory epithelial cells. Infect Immun. 60:5119-5125), vitronectin, and type IV collagen, and other M-like proteins that bind plasminogen, IgA, IgG, and albumin (Kihlberg, B. M., M. Collin, A. Olsen, and L. Bjorck. 1999. Protein H, an antiphagocytic surface protein in Streptococcus pyogenes. Infect Immun. 67:1708-14).


Numerous secreted proteins have been described, several of which are considered to be toxins. Most Streptococcus pyogenes isolates from cases of serious invasive disease and streptococcal toxic shock syndrome (STSS) produce streptococcal pyogenic exotoxins (SPE) A and C (Cockerill, F. R., 3rd, R. L. Thompson, J. M. Musser, P. M. Schlievert, J. Talbot, K. E. Holley, W. S. Harmsen, D. M. Ilstrup, P. C. Kohner, M. H. Kim, B. Frankfort, J. M. Manahan, J. M. Steckelberg, F. Roberson, and W. R. Wilson. 1998. Molecular, serological, and clinical features of 16 consecutive cases of invasive streptococcal disease. Southeastern Minnesota Streptococcal Working Group. Clin Infect Dis. 26:1448-58). Other pyogenic exotoxins have also been identified in the genomic Streptococcus pyogenes sequence completed at the University of Oklahoma, submitted to GenBank and assigned accession number AE004092, and have been characterized (Proft, T., S. Louise Moffatt, C. J. Berkahn, and J. D. Fraser. 1999. Identification and Characterization of Novel Superantigens from Streptococcus pyogenes. J Exp Med. 189:89-102). Other toxins such as Toxic Shock Like Syndrome toxin, Streptococcal Superantigen (Reda, K. B., V. Kapur, D. Goela, J. G. Lamphear, J. M. Musser, and R. R. Rich. 1996. Phylogenetic distribution of streptococcal superantigen SSA allelic variants provides evidence for horizontal transfer of ssa within Streptococcus pyogenes. Infect Immun. 64:1161-5), and Mitogenic Factor (Yutsudo, T., K. Okumura, M. Iwasaki, A. Hara, S. Kamitani, W. Minamide, H. Igarashi, and Y. Hinuma. 1994. The gene encoding a new mitogenic factor in a Streptococcus pyogenes strain is distributed only in group A streptococci. Infection and Immunity. 62:4000-4004) play lesser-defined roles in disease. Streptolysin O could also be considered a possible candidate antigen, because it causes the release of IL-β release. In addition, a variety of secreted enzymes have also been identified that include the Cysteine protease (Lukomski, S., C. A. Montgomery, J. Rurangirwa, R. S. Geske, J. P. Banish, G. J. Adams, and J. M. Musser. 1999. Extracellular cysteine protease produced by Streptococcus pyogenes participates in the pathogenesis of invasive skin infection and dissemination in mice. Infect Immun. 67:1779-88, Matsuka, Y. V., S. Pillai, S. Gubba, J. M. Musser, and S. B. Olmsted. 1999. Fibrinogen cleavage by the Streptococcus pyogenes extracellular cysteine protease and generation of antibodies that inhibit enzyme proteolytic activity. Infect Immun. 67:4326-33), Streptokinase (Huang, T. T., H. Malke, and J. J. Ferretti. 1989. The streptokinase gene of group A streptococci: cloning, expression in Escherichia coli, and sequence analysis. Mol Microbiol. 3:197-205, Nordstrand, A., W. M. McShan, J. J. Ferretti, S. E. Holm, and M. Norgren. 2000. Allele substitution of the streptokinase gene reduces the nephritogenic capacity of group A streptococcal strain NZ131. Infect Immun. 68:1019-25), and Hyaluronidase (Hynes, W. L., A. R. Dixon, S. L. Walton, and L. J. Aridgides. 2000. The extracellular hyaluronidase gene (hylA) of Streptococcus pyogenes. FEMS Microbiol Lett. 184:109-12, Hynes, W. L., L. Hancock, and J. J. Ferretti. 1995. Analysis of a second bacteriophage hyaluronidase gene from Streptococcus pyogenes: evidence for a third hyaluronidase involved in extracellular enzymatic activity. Infect Immun. 63:3015-20).


Given the number of known virulence factors produced by Streptococcus pyogenes, it is clear that an important characteristic for a successful β-hemolytic streptococcal immunogenic composition would be its ability to stimulate a response that would prevent or limit colonization early in the infection process. This protective response would either block adherence and/or enhance the clearance of cells through opsonophagocytosis. Antibodies to M protein have been shown to be opsonic and provide a mechanism to overcome the antiphagocytic properties of the protein (Jones, K. F., and V. A. Fischetti. 1988. The importance of the location of antibody binding on the M6 protein for opsonization and phagocytosis of group A M6 streptococci. J Exp Med. 167:1114-23) in much the same way that anti-serotype to B capsular antibodies have demonstrated protection from disease caused by Haemophilus influenzae B (Madore, D. V. 1998. Characterization of immune response as an indicator of Haemophilus influenzae type b vaccine efficacy. Pediatr Infect Dis J. 17:S207-10). In addition, antibodies specific to Protein F have been shown to block adherence and internalization by tissue culture cells (Molinari, G., S. R. Talay, P. Valentin-Weigand, M. Rohde, and G. S. Chhatwal. 1997. The fibronectin-binding protein of Streptococcus pyogenes, SfbI, is involved in the internalization of group A streptococci by epithelial cells. Infect Immun. 65:1357-63).


There remains a need to develop immunogenic compositions and methods to prevent or ameliorate infections caused by β-hemolytic streptococci, including groups A, B, C and G. There also remains a need to provide immunogenic compositions which provide immunity to a broad range of BHS bacteria.


SUMMARY OF THE INVENTION

To meet these and other needs, and in view of its purposes, the present invention provides immunogenic compositions for the protecting of susceptible mammals against colonization or infection by β-hemolytic streptococci including Group A, B, C, and/or D streptocci, including those from Streptococcus pyogenes. These immunogenic compositions comprise a mixture of two or more polypeptides as described more fully below. The invention also provides methods of preventing or ameliorating such coloziation, in a susceptible mammal by administering an effective amount of the immunogenic composition to generate antibodies to the specific polypeptides contained within the immunogenic composition. The invention further provides Streptococcus pyogenes polypeptides and polynucleotides, recombinant materials, and methods for their production. Another aspect of the invention relates to methods for using such Streptococcus pyogenes polypeptides and polynucleotides. The polypeptides and polynucleotides can also be used in the manufacture of a medicament for preventing or ameliorating an infection caused by β-hemolytic streptococci.


The polypeptides utilized in the immunogenic compositions of the invention include isolated polypeptides comprising at least one of an amino acid sequence of any of FIG. 2, 4, 6, 8, or 10. The invention also includes amino acid sequences that have at least 90% identity to any of the foregoing amino acid sequences, and mature polypeptides of these. The invention further includes immunogenic fragments and biological equivalents of these polypeptides. Also provided are antibodies that immunospecifically bind to the polypeptides of the invention.


The polynucleotides of the invention include isolated polynucleotides that comprise nucleotide sequences that encode a polypeptide of the invention. These polynucleotides include isolated polynucleotides comprising at least one of a nucleotide sequence of any of FIG. 1, 3, 5, 7, or 9, and also include other nucleotide sequences that, as a result of the degeneracy of the genetic code, also encode a polypeptide of the invention. The invention also includes isolated polynucleotides comprising a nucleotide sequence that has at least 90% identity to a nucleotide sequence that encodes a polypeptide of the invention, and isolated polynucleotides comprising a nucleotide sequences that has at least 90% identity to any of the foregoing nucleotide sequences. In addition, the isolated polynucleotides of the invention include nucleotide sequences that hybridize under stringent hybridization conditions to a nucleotide sequence that encodes a polypeptide of the invention, nucleotide sequences that hybridize under stringent hybridization conditions to a nucleotide sequence of any of the foregoing sequences, and nucleotide sequences that are fully complementary to these polynucleotides. Furthermore, the invention includes expression vectors and host cells comprising these polynucleotides.


The invention also provides immunogenic compositions which comprise an immunogenic amount of at least two or more components (selected from SCP (FIG. 2 (SEQ ID NO:2) and the peptides coded for by ORF 554 (peptidylpropyl isomerase (FIG. 4 (SEQ ID NO:4)), ORF 1218 (hypothetical protein (FIG. 6 (SEQ ID NO:6)), ORF 1358 (putative adhesion protein (FIG. 8 (SEQ ID NO:8)), and ORF 2459 (surface lipoprotein (FIG. 10 (SEQ ID NO:10)) each of which comprises a polypeptide of the invention in an amount effective to prevent or ameliorate a β-hemolytic streptococcal colonization or infection in a susceptible mammal. Each component may comprise the polypeptide itself, or may comprise the polypeptide and any other substance (e.g., one or more chemical agents, proteins, etc.) that can aid in the prevention and/or amelioration of β-hemolytic streptococcal colonization or infection. These immunogenic compositions can further comprise at least a portion of the polypeptide, optionally conjugated or linked to a peptide, polypeptide, or protein, or to a polysaccharide.


The invention also includes methods of protecting a susceptible mammal against β-hemolytic streptococcal colonization or infection. In one embodiment, the method comprises administering to a mammal an effective amount of a two or more immunogenic composition comprising an immunogenic amount of a polypeptide of the invention, which amount is effective to prevent or ameliorate β-hemolytic streptococcal colonization or infection in the susceptible mammal. Such combinations of components, it has been found, are effective to provide such protection to a broad range of groups, and generally provide a greater immune response than the individual components administered separately. The immunogenic compositions of the invention can be administered by any conventional route, for example, by subcutaneous or intramuscular injection, oral ingestion, or intranasally.


The invention further provides immunogenic compositions. In one embodiment, the immunogenic composition comprises at least one polypeptide of the invention. In another embodiment, the immunogenic composition comprises at least one polynucleotide of the invention.


It is to be understood that the foregoing general description and the following detailed description are exemplary, but are not restrictive, of the invention.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 presents the nucleic acid sequence encoding for C5a peptidase (“SCP”; SEQ ID NO:1).



FIG. 2 presents the amino acid sequence of SCP (SEQ ID NO:2).



FIG. 3 presents the nucleic acid sequence of ORF 554 encoding for peptidylpropyl isomerase (SEQ ID NO:3).



FIG. 4 presents the amino acid sequence of peptidylpropyl isomerase (SEQ ID NO:4).



FIG. 5 presents the nucleic acid sequence of ORF 1218 encoding for a hypothetical protein (SEQ ID NO:5).



FIG. 6 presents the amino acid sequence of a hypothetical protein (SEQ ID NO:6).



FIG. 7 presents the nucleic acid sequence of ORF 1358 encoding for a putative adhesion protein (SEQ ID NO:7).



FIG. 8 presents the amino acid sequence of a putative adhesion protein (SEQ ID NO:8).



FIG. 9 presents the nucleic acid sequence of ORF 2459 encoding for a surface lipoprotein (SEQ ID NO:9).



FIG. 10 presents the amino acid sequence of a surface lipoprotein (SEQ ID NO:10).



FIG. 11 graphically presents percentage killing compared to media of the three component (“Trivax”=SCP, peptidylpropyl isomerase (ORF 554), and putative adhesion protein (ORF 1358)) and one component (“554”=peptidylpropyl isomerase (ORF 554)) immunogenic compositions examined in Example 2.



FIGS. 12-16 graphically demonstrate the passive immunity transfer results of Example 3. CFUs=colony forming units.





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides immunogenic compositions to prevent or ameliorate infections caused by β-hemolytic streptococci, including groups A, B, C and G. Two or more of the polypeptides enumerated herein are combined together to make an immunogenic composition.


Specifically, in one embodiment, an immunogenic composition of this invention comprises a mixture of two or more polypeptides, each polypeptide encoded by a nucleic acid sequence having at least 90% identity to a nucleic acid sequence selected from the group consisting of:


(a) C5a peptidase (“SCP”) (FIG. 1 (SEQ ID NO:1));


(b) open reading frame (“ORF”) 554 (FIG. 3 (SEQ ID NO:3));


(c) ORF 1218 (FIG. 5 (SEQ ID NO:5));


(d) ORF 1358 (FIG. 7 (SEQ ID NO:7)); and


(e) ORF 2459 (FIG. 9 (SEQ ID NO:9)).


In another embodiment, an immunogenic composition of this invention comprises a mixture of two or more polypeptides, each polypeptide having at least 90% identity to an amino acid sequence selected from the group consisting of:


(a) SCP (FIG. 2 (SEQ ID NO:2));


(b) peptidylpropyl isomerase (FIG. 4 (SEQ ID NO:4));


(c) hypothetical protein (FIG. 6 (SEQ ID NO:6));


(d) putative adhesion protein (FIG. 8 (SEQ ID NO:8)); and


(e) surface lipoprotein (FIG. 10 (SEQ ID NO:10)).


In yet another embodiment, an immunogenic composition of this invention comprises a mixture of:


(a) an SCP polypeptide encoded by a nucleic acid sequence having at least 90% identity to the nucleic acid sequence of FIG. 1 (SEQ ID NO:1);


(b) a peptidylpropyl isomerase polypeptide encoded by a nucleic acid sequence having at least 90% identity to the nucleic acid sequence of FIG. 3 (SEQ ID NO:3); and


(c) at least one other polypeptide encoded by a nucleic acid sequence having at least 90% identity to an nucleic acid sequence selected from the group consisting of (i) FIG. 5 (SEQ ID NO:5); (ii) FIG. 7 (SEQ ID NO:7); and (iii) FIG. 9 (SEQ ID NO:9).


In still another embodiment, an immunogenic composition of this invention comprises a mixture of:


(a) an SCP polypeptide having at least 90% identity to the amino acid sequence of FIG. 2 (SEQ ID NO:2);


(b) a peptidylpropyl isomerase polypeptide having at least 90% identity to the amino acid sequence of FIG. 4 (SEQ ID NO:4); and


(c) at least one other polypeptide having at least 90% identity to an amino acid sequence of the group consisting of (i) FIG. 6 (SEQ ID NO:6); (ii) FIG. 8 (SEQ ID NO:8); and (iii) FIG. 10 (SEQ ID NO:10).


The terms “polynucleotide”, “nucleic acid” and “nucleic acid fragment” are used interchangeably herein. These terms encompass nucleotides connected by phosphodiester linkages. A “polynucleotide” may be a ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) polymer that is single- or double-stranded, that optionally contains synthetic, non-natural or altered nucleotide bases. A polynucleotide in the form of a polymer of DNA may comprise one or more segments of cDNA, genomic DNA, synthetic DNA, or mixtures thereof. Nucleotide bases are indicated herein by a single letter code: adenine (A), guanine (G), thymine (T), cytosine (C), inosine (I) and uracil (U).


The streptococcal polynucleotides described herein may be obtained using standard cloning and screening techniques. These polynucleotides may be obtained, for example, from genomic DNA, from a cDNA library derived from mRNA, from a genomic DNA library, or can be synthesized using well known and commercially available techniques, such as e.g. by PCR from a cDNA library or via RT-PCR (reverse transcription-polymerase chain reaction).


There are several methods available and well known to those skilled in the art to obtain full-length cDNAs or to extend short cDNAs, such as e.g. those based on the method of rapid amplification of cDNA ends (RACE). See Frohman et al., Proc. Natl. Acad. Sci. USA 85, 8998-9002, 1988. Recent modifications of the technique, exemplified by the MARATHON™ technology (Clontech Laboratories Inc.) for example, have significantly simplified the search for longer cDNAs. In the MARATHON™ technology, cDNAs have been prepared from mRNA extracted from a chosen tissue and an “adaptor” sequence ligated onto each end. Nucleic acid amplification (PCR) is then carried out to amplify the “missing” 5′ end of the cDNA using a combination of gene specific and adaptor specific oligonucleotide primers. The PCR reaction is then repeated using “nested” primers, that is, primers designed to anneal within the amplified product (typically an adaptor specific primer that anneals further 3′ in the adaptor sequence and a gene specific primer that anneals further 5′ in the known gene sequence). The products of this reaction can then be analyzed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or by carrying out a separate full-length PCR using the new sequence information for the design of the 5′ primer.


The term “recombinant” means, for example, that a polynucleotide is made by an artificial combination of two or more otherwise separated polynucleotide segments, e.g., by chemical synthesis or by the manipulation of isolated polynucleotides using genetic engineering techniques. A “recombinant DNA construct” comprises any of the isolated polynucleotides of the present invention operably linked to at least one regulatory element.


Orthologues and allelic variants of the streptococcal polynucleotides can readily be identified using methods well known in the art. Allelic variants and orthologs of the polynucleotides can comprise a nucleotide sequence that is typically at least about 90-95% or more identical to any one or more of the nucleotide sequences shown in odd numbered FIGS. 1-9 (odd numbered SEQ ID NO:'S 1-9), or fragments thereof. The allelic variants and orthologs of these polynucleotides can encode a polypeptide that comprises an amino acid sequence that is at least 90% identical to the amino acid sequence set forth in any one or more of even numbered FIGS. 2-10 (even numbered SEQ ID NO:'S 2-10). Such polynucleotides can readily be identified as being able to hybridize under stringent conditions, to any one or more of the polynucleotides having a nucleotide sequence set forth in FIGS. 1-9 (odd numbered SEQ ID NO:'S 1-9), or fragments thereof.


It is well understood by one skilled in the art that many levels of sequence identity are useful in identifying related polynucleotide and polypeptide sequences. Sequence alignments and percent identity calculations can be performed using the MEGALIGN™ program of the LASERGENE™ bioinformatics computing suite (DNASTAR Inc., Madison, Wis.). Multiple alignment of the sequences can be performed using the Clustal method of alignment (Higgins and Sharp, Gene, 73(1):237-44, 1988) with the default parameters of e.g. GAP PENALTY=10 and GAP LENGTH PENALTY=10. Default parameters for pairwise alignments using the Clustal method can be e.g. KTUPLE 1, GAP PENALTY=3, WINDOW=5 and DIAGONALS SAVED=5.


A polypeptide sequence of the invention may be identical to the recited sequence, that is, 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the % identity is less than 100%. Such alterations include at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion. The alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference amino acid sequence or in one or more contiguous groups within the reference amino acid sequence.


Thus, the invention also provides isolated polypeptides having sequence identity to the amino acid sequences contained in therecited sequences. Depending on the particular sequence, the degree of sequence identity is preferably greater than 90% (e.g., 90%, 95%, 97%, 99% or more). These homologous proteins include mutants and allelic variants.


“Identity,” as known in the art, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. “Identity” and “similarity” can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988). Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al. 1984), BLASTP, BLASTN, and FASTA (Altschul, S. F., et al., 1990. The BLASTX program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al., NCBI NLM NIH Bethesda, Md. 20894; Altschul, S., et al., 1990). The well known Smith Waterman algorithm may also be used to determine identity.


For example, the number of amino acid alterations for a given % identity can be determined by multiplying the total number of amino acids in one of even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10) by the numerical percent of the respective percent identity (divided by 100) and then subtracting that product from said total number of amino acids in the one of even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10), or:

na≦xa−(xa·y),

wherein na is the number of amino acid alterations, xa is the total number of amino acids in the one of even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10), and y is, for instance, 0.90 for 90%, 0.95 for 95%, 0.97 for 97% etc., and wherein any non-integer product of Xa and y is rounded down to the nearest integer prior to subtracting it from Xa.


The present invention also contemplates isolated polypeptides that are substantially conserved across strains of β-hemolytic streptococci. Further, isolated polypeptides that are substantially conserved across strains of β-hemolytic streptococci and that are effective in preventing or ameliorating a β-hemolytic streptococcal colonization or infection in a susceptible subject are also contemplated by the present invention. As used herein, the term “conserved” refers to, for example, the number of amino acids that do not undergo insertions, substitution and/or deletions as a percentage of the total number of amino acids in a protein. For example, if a protein is 90% conserved and has, for example, 263 amino acids, then there are 237 amino acid positions in the protein at which amino acids do not undergo substitution. Likewise, if a protein is 95% conserved and has, for example, about 280 amino acids, then there are 14 amino acid positions at which amino acids may undergo substitution and 266 (i.e., 280 minus 14) amino acid positions at which the amino acids do not undergo substitution. According to an embodiment of the present invention, the isolated polypeptide is preferably at least about 90% conserved across the strains of β-hemolytic streptococci, more preferably at least about 95% conserved across the strains, even more preferably at least about 97% conserved across the strains, and most preferably at least about 99% conserved across the strains, without limitation.


Modifications and changes can be made in the structure of the polypeptides and still obtain polypeptides having β-hemolytic streptococci and/or Streptococcus pyogenes activity and/or antigenicity. For example, certain amino acids can be substituted for other amino acids in a sequence without appreciable loss of activity and/or antigenicity. Because it is the interactive capacity and nature of a polypeptide that defines that polypeptide's biological functional activity, certain amino acid sequence substitutions can be made in a polypeptide sequence (or, of course, its underlying DNA coding sequence) and nevertheless obtain a polypeptide with like properties.


The invention includes any isolated polypeptide which is a biological equivalent that provides the desired reactivity as described herein. The term “desired reactivity” refers to reactivity that would be recognized by a person skilled in the art as being a useful result for the purposes of the invention. Examples of desired reactivity are described herein, including without limitation, desired levels of protection, desired antibody titers, desired opsonophagocytic activity and/or desired cross-reactivity, such as would be recognized by a person skilled in the art as being useful for the purposes of the present invention. The desired opsonophagocytic activity is indicated by a percent killing of bacteria as measured by decrease in colony forming units (CFU) in OPA versus a negative control. Without being limited thereto, the desired opsonophagocytic activity is preferably at least about 15%, more preferably at least about 20%, even more preferably at least about 40%, even more preferably at least about 50% and most preferably at least about 60%.


The invention includes polypeptides that are variants of the polypeptides comprising an amino acid sequence of even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10). “Variant” as the term is used herein, includes a polypeptide that differs from a reference polypeptide, but retains essential properties. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical (i.e., biologically equivalent). A variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, or deletions in any combination. A substituted or inserted amino acid residue may or may not be one encoded by the genetic code. A variant of a polypeptide may be a naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polypeptides may be made by direct synthesis or by mutagenesis techniques.


In making such changes, the hydropathic index of amino acids can be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a polypeptide is generally understood in the art (Kyte & Doolittle, 1982). It is known that certain amino acids can be substituted for other amino acids having a similar hydropathic index or score and still result in a polypeptide with similar biological activity. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. Those indices are listed in parentheses after each amino acid as follows: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5).


It is believed that the relative hydropathic character of the amino acid residue determines the secondary and tertiary structure of the resultant polypeptide, which in turn defines the interaction of the polypeptide with other molecules, such as enzymes, substrates, receptors, antibodies, antigens, and the like. It is known in the art that an amino acid can be substituted by another amino acid having a similar hydropathic index and still obtain a functionally equivalent polypeptide. In such changes, the substitution of amino acids whose hydropathic indices are within +/−2 is preferred, those which are within +/−1 are particularly preferred, and those within +/−0.5 are even more particularly preferred.


Substitution of like amino acids can also be made on the basis of hydrophilicity, particularly where the biological functional equivalent polypeptide or peptide thereby created is intended for use in immunological embodiments. U.S. Pat. No. 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a polypeptide, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the polypeptide.


As detailed in U.S. Pat. No. 4,554,101, incorporated herein by reference, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); proline (−0.5±1); threonine (−0.4); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); and tryptophan (−3.4). It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent and in particular, an immunologically equivalent, polypeptide. In such changes, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred.


As outlined above, amino acid substitutions are generally, therefore, based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions which take various of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine, and isoleucine. As shown in Table I below, suitable amino acid substitutions include the following:












TABLE 1







Original
Exemplary Residue



Residue
Substitution









Ala
Gly; Ser



Arg
Lys



Asn
Gln; His



Asp
Glu



Cys
Ser



Gln
Asn



Glu
Asp



Gly
Ala



His
Asn; Gln



Ile
Leu; Val



Leu
Ile; Val



Lys
Arg



Met
Met; Leu; Tyr



Ser
Thr



Thr
Ser



Trp
Tyr



Tyr
Trp; Phe



Val
Ile; Leu











Thus, the invention includes functional or biological equivalents of the polypeptides of the sequences in even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10) that contain one or more amino acid substitutions.


Biological or functional equivalents of a polypeptide can also be prepared using site-specific mutagenesis. Site-specific mutagenesis is a technique useful in the preparation of second generation polypeptides, or biologically, functionally equivalent polypeptides, derived from the sequences thereof, through specific mutagenesis of the underlying DNA. As noted above, such changes can be desirable where amino acid substitutions are desirable. The technique further provides a ready ability to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA. Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Typically, a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.


In general, the technique of site-specific mutagenesis is well known in the art. As will be appreciated, the technique typically employs a phage vector which can exist in both a single-stranded and double-stranded form. Typically, site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector which includes within its sequence a DNA sequence which encodes all or a portion of the Streptococcus pyogenes polypeptide sequence selected. An oligonucleotide primer bearing the desired mutated sequence is prepared, for example, by well known techniques (e.g., synthetically). This primer is then annealed to the single-stranded vector, and extended by the use of enzymes, such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected which include recombinant vectors bearing the mutation. Commercially available kits provide the necessary reagents.


The polypeptides and polypeptide antigens of the invention are understood to include any polypeptide comprising substantial sequence similarity, structural similarity, and/or functional similarity to a polypeptide comprising an amino acid sequence of any of even numbered FIGS. 2-10 (SEQ ID NO:'S 2, 4, 6, 8 and 10). In addition, a polypeptide or polypeptide antigen of the invention is not limited to a particular source. Thus, the invention provides for the general detection and isolation of the polypeptides from a variety of sources.


The polypeptides of the invention may be in the form of the “mature” protein or may be a part of a larger protein such as a fusion protein. It is often advantageous to include an additional amino acid sequence which contains, for example, secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production.


The term “immunogenic composition” as used herein refers to any type of biological agent in an administratable form capable of stimulating an immune response in a subject inoculated with the immunogenic composition. An immune response may include induction of antibodies and/or induction of a T-cell response. The term “protection,” when used in reference to an immunogenic composition, refers herein to the amelioration (either partial or complete) of any of the symptoms associated with the disease or condition in question. Thus, protection of subjects from infection by a Streptococcus species such as S. dysgalactiae (including the subspecies Dysgalactiae and Equisimilis) by the present immunogenic compositions generally results in a diminishing of bacterial growth and/or one or more of the clinical symptoms associated with streptococcal infection, including arthritis, endocarditis, meningitis, polyserositis, bronchopneumonia, meningitis, permanent hearing loss and septic shock.


The methods disclosed herein may include inducing an immune response against one or more pathogens that include a species of Streptococcus (e.g., Streptococcus dysgalactiae, S. dysgalactiae sub. Equisimilis, S. dysgalactiae sub. Dysgalactiae, S. pyogenes, S. agalactiae, S. anginosus, S. constellatus, S. equisimilis and S. intermedius.) For example, the methods may include inducing polyclonal antibody production against one or more streptococcal pathogens such as e.g. S. dysgalactiae sub. Equisimilis.


As discussed above, immunogenic compositions comprise two or more polypeptides of the invention. To do so, one or more polypeptides are adjusted to an appropriate concentration and can be formulated with any suitable adjuvant, diluent, pharmaceutically acceptable carrier, or any combination thereof. As used herein the phrase “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, excipients and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Physiologically acceptable vehicles may be used as carriers and/or diluents. A pharmaceutically acceptable vehicle is understood to designate a compound or a combination of compounds entering into a pharmaceutical or immunogenic composition which does not cause side effects and which makes it possible, for example, to facilitate the administration of the active compound, to increase its life and/or its efficacy in the body, to increase its solubility in solution or alternatively to enhance its preservation. These pharmaceutically acceptable vehicles are well known and will be adapted by persons skilled in the art according to the nature and the mode of administration of the active compound chosen. These include, but are not limited to, water, Ringer's solution, an appropriate isotonic medium, glycerol, ethanol and other conventional solvents, phosphate buffered saline, and the like.


Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, polyethylene glycol and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, and the like. In many cases, isotonic agents are included in the composition, for example, sugars, polyalcohols such as manitol, sorbitol and/or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the polypeptides in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Immunogenic compositions as described herein also comprise, in certain embodiments, one or more adjuvants. An adjuvant is a substance that enhances the immune response when administered together with an immunogen or antigen. A number of cytokines or lymphokines have been shown to have immune modulating activity, and thus are useful as adjuvants, including, but not limited to, the interleukins 1-α, 1-β, 2, 4, 5, 6, 7, 8 and 10, 12 (see, e.g., U.S. Pat. No. 5,723,127), 13, 14, 15, 16, 17 and 18 (and its mutant forms); the interferons-α, β and γ; granulocyte-macrophage colony stimulating factor (GM-CSF) (see, e.g., U.S. Pat. No. 5,078,996 and ATCC Accession Number 39900); macrophage colony stimulating factor (M-CSF); granulocyte colony stimulating factor (G-CSF); and the tumor necrosis factors α and β. Still other adjuvants that are useful with the immunogenic compositions described herein include chemokines, including without limitation, MCP-1, MIP-1α, MIP-1β, and RANTES; adhesion molecules, such as a selectin, e.g., L-selectin, P-selectin and E-selectin; mucin-like molecules, e.g., CD34, GlyCAM-1 and MadCAM-1; a member of the integrin family such as LFA-1, VLA-1, Mac-1 and p150.95; a member of the immunoglobulin superfamily such as PECAM, ICAMs, e.g., ICAM-1, ICAM-2 and ICAM-3, CD2 and LFA-3; co-stimulatory molecules such as CD40 and CD40L; growth factors including vascular growth factor, nerve growth factor, fibroblast growth factor, epidermal growth factor, B7.2, PDGF, BL-1, and vascular endothelial growth factor; receptor molecules including Fas, TNF receptor, Flt, Apo-1, p55, WSL-1, DR3, TRAMP, Apo-3, AIR, LARD, NGRF, DR4, DR5, KILLER, TRAIL-R2, TRICK2, and DR6; and Caspase (ICE).


Suitable adjuvants used to enhance an immune response further include, without limitation, MPL™ (3-O-deacylated monophosphoryl lipid A, Corixa, Hamilton, Mont.), which is described in U.S. Pat. No. 4,912,094. Also suitable for use as adjuvants are synthetic lipid A analogs or aminoalkyl glucosamine phosphate compounds (AGP), or derivatives or analogs thereof, which are available from Corixa (Hamilton, Mont.), and which are described in U.S. Pat. No. 6,113,918. One such AGP is 2-[(R)-3-Tetradecanoyloxytetradecanoylamino]ethyl 2-Deoxy-4-O-phosphono-3-O—[(R)-3-tetradecanoyoxytetradecanoyl]-2-[(R)-3-tetradecanoyloxytetradecanoyl-amino]-b-D-glucopyranoside, which is also known as 529 (formerly known as RC529). This 529 adjuvant is formulated as an aqueous form (AF) or as a stable emulsion (SE).


Still other adjuvants include muramyl peptides, such as N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanine-2-(1′-2′ dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-ethylamine (MTP-PE); oil-in-water emulsions, such as MF59 (U.S. Pat. No. 6,299,884) (containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE) formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton, Mass.)), and SAF (containing 10% Squalene, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion); aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate; Amphigen; Avridine; L121/squalene; D-lactide-polylactide/glycoside; pluronic polyols; killed Bordetella; saponins, such as Stimulon™ QS-21 (Antigenics, Framingham, Mass.), described in U.S. Pat. No. 5,057,540, ISCOMATRIX (CSL Limited, Parkville, Australia), described in U.S. Pat. No. 5,254,339, and immunostimulating complexes (ISCOMS); Mycobacterium tuberculosis; bacterial lipopolysaccharides; synthetic polynucleotides such as oligonucleotides containing a CpG motif (e.g., U.S. Pat. No. 6,207,646); IC-31 (Intercell AG, Vienna, Austria), described in European Patent Nos. 1,296,713 and 1,326,634; a pertussis toxin (PT) or mutant thereof, a cholera toxin or mutant thereof (e.g., U.S. Pat. Nos. 7,285,281, 7,332,174, 7,361,355 and 7,384,640); or an E. coli heat-labile toxin (LT) or mutant thereof, particularly LT-K63, LT-R72 (e.g., U.S. Pat. Nos. 6,149,919, 7,115,730 and 7,291,588).


The polypeptide can also include at least a portion of the polypeptide, optionally conjugated or linked to a peptide, polypeptide, or protein, or to a polysaccharide. It is also anticipated that the immunogenic compositions can contain other components, such as polysaccharides, alone or conjugated to proteins which can elicit an immune response.


Various tests are used to assess the in vitro immunogenicity of the polypeptides comprising the immunogenic compositions of the invention. For example, an in vitro opsonic assay is conducted by incubating together a mixture of Streptococcus sp. cells, heat inactivated serum containing specific antibodies to the polypeptide in question, and an exogenous complement source. Opsonophagocytosis proceeds during incubation of freshly isolated polymorphonuclear cells (PMN's) and the antibody/complement/Streptococcus sp. cell mixture. Bacterial cells that are coated with antibody and complement are killed upon opsonophagocytosis. Colony forming units (cfu) of surviving bacteria that escape from opsonophagocytosis are determined by plating the assay mixture. Titers are reported as the reciprocal of the highest dilution that gives ≧50% bacterial killing, as determined by comparison to assay controls. Specimens that demonstrate less than 50% killing at the lowest serum dilution tested (1:8), are reported as having an opsonophagocytosis antibody (OPA) titer of 4. The method described above is a modification of Gray's method (Gray, Conjugate Vaccines Supplement, p. 694-697, 1990).


A test serum control, which contains test serum plus bacterial cells and heat inactivated complement, is included for each individual serum. This control is used to assess whether the presence of antibiotics or other serum components are capable of killing the bacterial strain directly (i.e. in the absence of complement or PMN's). A human serum with known opsonic titer is used as a positive human serum control. The opsonic antibody titer for each unknown serum is calculated as the reciprocal of the initial dilution of serum giving 50% cfu reduction compared to the control without serum.


A whole cell ELISA assay can also be used to assess in vitro immunogenicity and surface exposure of the polypeptide antigen, wherein the bacterial strain of interest is coated onto a plate, such as a 96 well plate, and test sera from an immunized animal is reacted with the bacterial cells. If any antibody specific for the test polypeptide antigen is reactive with a surface exposed epitope of the polypeptide antigen, it can be detected by standard methods known to one skilled in the art. A similar approach is to monitor the antigen on the cell surface using Flow Cytometry and antigen specific antibodies.


Any polypeptide demonstrating the desired in vitro activity may then be tested in an in vivo animal challenge model. In some embodiments, immunogenic compositions are used in the immunization of an animal (e.g., a mouse) by methods and routes of immunization known to those of skill in the art (e.g., intranasal, parenteral, intramuscular, oral, rectal, vaginal, transdermal, intraperitoneal, intravenous, subcutaneous, etc.). Following immunization of the animal with a Streptococcal immunogenic composition, the animal is challenged with one or more Streptococcal species and assayed for resistance to Streptococcus spp. infection.


Combination immunogenic compositions are provided by including two or more of the polypeptides of the invention, as well as by combining one or more of the polypeptides of the invention with one or more known Streptococcus pyogenes polypeptides, including, but not limited to, the M proteins, adhesins, and the like.


Once formulated, the immunogenic compositions of the invention can be administered directly to the subject, delivered ex vivo to cells derived from the subject, or in vitro for expression of recombinant proteins. For delivery directly to the subject, administration may be by any conventional form, such as intranasally, parenterally, orally, intraperitoneally, intravenously, subcutaneously, or topically applied to any mucosal surface such as intranasal, oral, eye, lung, vaginal, or rectal surface, such as by an aerosol spray.


It is advantageous to formulate oral or parenteral compositions in unit dosage form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.


Injectable preparations, for example sterile injectable aqueous or oleaginous suspensions, are formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.


For parenteral administration, immunogenic compositions of the invention can be administered as injectable dosages in a physiologically acceptable diluent with a pharmaceutically acceptable carrier that can be a sterile liquid such as water oils, saline, glycerol, or ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, surfactants, pH buffering substances and the like can be present in compositions. Other components can include those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, and mineral oil. In general, glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.


Typically, compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared. The preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above (see Langer, Science 249: 1527 (1990) and Hanes, Advanced Drug Delivery Reviews 28:97 (1997)). The immunogenic compositions of this invention can be administered in the form of a depot injection or implant preparation, which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.


The subjects are generally human. An immunologically effective amount of the immunogenic composition in an appropriate number of doses is administered to the subject to elicit an immune response. Immunologically effective amount, as used herein, means the administration of that amount to a mammalian host (preferably human), either in a single dose or as part of a series of doses, sufficient to at least cause the immune system of the individual treated to generate an immune response that reduces the clinical impact of the bacterial infection. The term “immune response” or “immunological response” includes the development of a humoral (antibody-mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) response. Protection may be conferred by a single dose of the immunogenic composition, or may require the administration of several doses, in addition to booster doses at later times to maintain protection. This may range from a minimal decrease in bacterial burden to prevention of the infection. Ideally, the treated individual will not exhibit the more serious clinical manifestations of the β-hemolytic streptococcal infection. The dosage amount can vary depending upon specific conditions of the individual, such as age and weight. This amount can be determined in routine trials by means known to those skilled in the art.


In prophylactic applications, immunogenic compositions are administered to a subject susceptible to, or otherwise at risk of, beta hemolytic streptococcal infection in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histologic and/or behavioral symptoms of disease associated with the infection, its complications and intermediate pathological phenotypes presenting during development of the disease. In therapeutic applications, compositions are administered to a patient suspected of, or already suffering from such a disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease (biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease.


It has been observed that there is no single peptide sequence that provides protection for all strains of BHS, including groups A, B, C, and G. As shown in Table II (presented in Example 1 below), below, each antigen provides an immune response against a subset of these groups.


Generally, any combination of two or more surface-expressed antigens from BHS will be expected to provide the enhanced immune response described above. Such could include the antigens discussed above BHS capsular antigens, M protein, ABC transporter, or any other surface exposed antigen. However, it has been found that the following antigens exhibit particularly beneficial properties for the production of immunogenic compositions:


SCP (C5a Peptidase)


peptidylpropyl isomerase (encoded by ORF 554)


putative adhesion protein (encoded by ORF 1358)


surface lipoprotein (encoded by ORF 2459)


hypothetical protein (encoded by ORF 1218)


Combinations of two or more of these antigens into a single multicomponent immunogenic composition provide enhanced protection against one or more groups of BHS and produce an enhanced immune response to them.


EXAMPLES

The following examples are illustrative and the present invention is not intended to be limited thereto.


Example 1
Antibody Binding

The binding of antibodies to bacteria, a process known as opsonization, can lead to uptake and killing of the bacteria by phagocytic cells. Screening of such antibodies is used in to determine the effectiveness of antibodies raised against particular serotypes in killing bacteria expressing or not expressing that serotype on the surface.


For each serotype screened, antibodies were raised in mice against the antibodies encoded for by the recited ORF. The antibodies were then screened against various BHS strains. Screening of the antibodies was performed by fluorescence activated cell sorting (FACS). Briefly, heat killed streptococci were incubated with a recited antibody on ice for 45 minutes, followed by two washes. The streptococci were then incubated with a goat anti-mouse-Alexa-488 antibody (Molecular Probes, Eugene, Oreg.) for 30 minutes on ice, followed by two washes. Cells thus treated were run on a FACS machine (e.g. see DeMaster et al., Infect. Immun., 70(1): 350-359, 2002.) The results are summarized in Table 2.


In the course of screening those anti-beta hemolytic-streptococcal antisera and monoclonal antibodies against various beta hemolytic streptococcal (BHS) strains, it was noted that some antisera and antibodies are cross-reactive against many BHS strains, including members of Streptococcus pyogenes (Group A streptococci), Streptococcus agalactiae (Group B streptococci) and Group C and Group G streptococci (which include the streptococcal species Streptococcus anginosus, Streptococcus constellatus, Streptococcus intermedius, Streptococcus dysgalactiae sub. Equisimilis and Streptococcus dysgalactiae sub. Dysgalactiae). This cross-reactivity also means that the polypeptides recited or encoded by the relevant ORF may be used in an immunogenic composition to induce an immune response effective to protect against infection by Group A or Group B Streptococcus, as well as by Group C or Group G Streptococcus.
























strain #
Genus/Species
pre
554
1224
1358
1818
2459
SCP
1218
mIgG1
C5a-1484-16
C5a-1522-13







GAR 1165

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 1199

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 1251

Streptococcus pyogenes


+
+
+
+
+
+
+

+/−



GAR 1278

Streptococcus pyogenes


+
+
+
+
+
+
+/−

+/−
+/−


GAR 1362

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 1439

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 1530

Streptococcus pyogenes


+
+
+
+
+
+
+


+/−


GAR 1566

Streptococcus pyogenes


+
+/−
+
+
+
+
+/−

+/−
+/−


GAR 1672

Streptococcus pyogenes


+
+/−
+
+
+
+
+

+
+


GAR 1839

Streptococcus pyogenes


+
+
+
+
+
+
+/−

+
+


GAR 1923

Streptococcus pyogenes


+
+
+
+
+
+
+/−

+/−
+/−


GAR 2107

Streptococcus pyogenes


+
+/−
+
+
+
+
+

+
+


GAR 2330

Streptococcus pyogenes


+
+
+
+
+
+
+

+/−
+/−


GAR 2646

Streptococcus pyogenes


+
+/−
+
+
+
+
+

+
+


GAR 2650

Streptococcus pyogenes


+
+
+
+
+
+
+


+/−


GAR 2869

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 3104

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 3549

Streptococcus pyogenes


+
+
+
+
+
+
+

+



GAR 3784

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 4029

Streptococcus pyogenes


+
+
+
+
+/−
+
+

+



GAR 4030

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+/−


GAR 4230

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 4773

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 4983

Streptococcus pyogenes


+
+/−
+
+
+
+
+/−

+
+


GAR 4987

Streptococcus pyogenes


+
+/−
+
+
+
+
+

+
+


GAR 5861

Streptococcus pyogenes


+
+/−
+
+
+/−
+
+/−

+
+


GAR 5991

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GAR 6084

Streptococcus pyogenes


+
+/−
+
+
+
+
+

+
+


GAR 7055

Streptococcus pyogenes


+
+
+
+
+
+
+

+
+


GS20

Streptococcus pyogenes


+
+/−
+
+
+
+
+/−
NT
NT
NT


GS21

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS22

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS23

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS24

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS25

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS26

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS29

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS30

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS31

Streptococcus pyogenes


+/−

+/−
+/−
+/−
+

NT
NT
NT


GS32

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS33

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS34

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS35

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS36

Streptococcus pyogenes


+/−

+/−
+/−
+/−
+

NT
NT
NT


GS37

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS38

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS39

Streptococcus pyogenes


+
+/−
+
+
+
+
+/−
NT
NT
NT


GS40

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS41

Streptococcus pyogenes


+

+
+
+
+
+/−
NT
NT
NT


GS42

Streptococcus pyogenes


+
+/−
+/−
+
+/−
+
+
NT
NT
NT


GS43

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS44

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS45

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS46

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS47

Streptococcus pyogenes


+
+/−
+/−
+
+
+
+
NT
NT
NT


GS48

Streptococcus pyogenes


+

+/−
+/−
+/−
+
+/−
NT
NT
NT


GS49

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS50

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS51

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS52

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS53

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS54

Streptococcus pyogenes


+/−
+/−
+/−
+
+/−
+
+
NT
NT
NT


GS55

Streptococcus pyogenes


+
+/−
+
+
+
+
+/−
NT
NT
NT


GS56

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS57

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS58

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS59

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS60

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS61

Streptococcus pyogenes


+
+/−
+
+
+/−
+
+
NT
NT
NT


GS62

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS63

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS64

Streptococcus pyogenes


+
+/−
+
+
+
+
+
NT
NT
NT


GS65

Streptococcus pyogenes


+
+
+
+
+
+
+
NT
NT
NT


GS66

Streptococcus pyogenes


+
+/−
+
+
+/−
+
+
NT
NT
NT


GAR 1

Streptococcus agalactiae


+
+
+
+
+
+
+/−





GAR 1012

Streptococcus agalactiae


+/−




+






GAR 1023

Streptococcus agalactiae














GAR 1049

Streptococcus agalactiae














GAR 10895

Streptococcus agalactiae





+/−

+






GAR 1192

Streptococcus agalactiae


+/−
+/−
+/−
+
+/−
+






GAR 127

Streptococcus agalactiae





+/−

+






GAR 12790

Streptococcus agalactiae







+/−






GAR 1305

Streptococcus agalactiae







+/−






GAR 131

Streptococcus agalactiae







+/−






GAR 1355

Streptococcus agalactiae







+/−






GAR 1446

Streptococcus agalactiae







+
+/−





GAR 1494

Streptococcus agalactiae





+/−

+
+/−





GAR 154

Streptococcus agalactiae


+
+
+
+
+
+/−
+/−





GAR 176

Streptococcus agalactiae







+






GAR 18

Streptococcus agalactiae


+
+
+
+
+
+/−






GAR 1844

Streptococcus agalactiae







+
+/−





GAR 1931

Streptococcus agalactiae





+

+
+





GAR 2369

Streptococcus agalactiae




+/−
+/−
+/−
+






GAR 252

Streptococcus agalactiae







+/−






GAR 2533

Streptococcus agalactiae














GAR 2682

Streptococcus agalactiae


+




+






GAR 2717

Streptococcus agalactiae







+/−






GAR 2723

Streptococcus agalactiae








+/−





GAR 2724

Streptococcus agalactiae







+/−
+/−





GAR 2842

Streptococcus agalactiae


+/−




+/−






GAR 287

Streptococcus agalactiae





+/−

+






GAR 3003

Streptococcus agalactiae














GAR 3751

Streptococcus agalactiae







+
+/−

+/−



GAR 381

Streptococcus agalactiae





+/−

+/−






GAR 3830

Streptococcus agalactiae







+
+





GAR 4131

Streptococcus agalactiae


+/−




+
+





GAR 4293

Streptococcus agalactiae




+/−
+
+/−
+
+/−





GAR 4398

Streptococcus agalactiae








+





GAR 462

Streptococcus agalactiae














GAR 4837

Streptococcus agalactiae


+/−




+
+/−





GAR 54

Streptococcus agalactiae







+


+/−



GAR 562

Streptococcus agalactiae


+
+/−
+
+
+/−
+
+/−





GAR 6016

Streptococcus agalactiae


+
+/−
+
+
+
+






GAR 614

Streptococcus agalactiae


+
+
+/−
+
+
+
+/−





GAR 63

Streptococcus agalactiae


+
+/−
+
+
+
+
+/−





GAR 6332

Streptococcus agalactiae


+/−
+/−
+
+
+
+
+/−

+
+/−


GAR 6387

Streptococcus agalactiae


+
+/−
+/−
+/−
+/−
+
+

+/−



GAR 6505

Streptococcus agalactiae


+/−
+/−
+/−
+/−
+
+
+/−

+
+


GAR 67

Streptococcus agalactiae





+

+






GAR 864

Streptococcus agalactiae


+/−

+/−
+/−
+/−
+






GAR 967

Streptococcus agalactiae







+/−






GS19
GGS

+/−
+/−
+/−
+
+/−
+
+
NT
NT
NT


GS27
GGS

+/−
+
+/−
+
+/−
+
+/−
NT
NT
NT


ATCC 33397

Streptococcus anginosus


+/−
+/−
+/−
+/−
+/−
+/−






ATCC 33397

Streptococcus anginosus














GAR 10823

Streptococcus anginosus


+/−
+
+/−
+/−
+/−
+






GAR 1272

Streptococcus anginosus







+/−






GAR 1370

Streptococcus anginosus














GAR 1425

Streptococcus anginosus


+/−
+/−
+/−
+/−

+/−






GAR 1592

Streptococcus anginosus














GAR 1595

Streptococcus anginosus



+/−



+/−






GAR 2044

Streptococcus anginosus



+/−










GAR 2523

Streptococcus anginosus



+/−

+/−

+/−






GAR 2565

Streptococcus anginosus



+/−

+/−
+/−
+/−
+/−





GAR 2697

Streptococcus anginosus


+/−
+
+/−
+/−
+/−
+/−






GAR 2822

Streptococcus anginosus


+/−



+/−

+/−





GAR 3091

Streptococcus anginosus



+/−

+/−

+/−






GAR 3560

Streptococcus anginosus


+
+
+
+/−
+
+/−






GAR 3576

Streptococcus anginosus




+/−









GAR 3858

Streptococcus anginosus


+/−
+/−
+/−
+/−
+/−
+/−






GAR 3938

Streptococcus anginosus








+/−





GAR 4133

Streptococcus anginosus




+/−
+/−

+
+/−





GAR 4158

Streptococcus anginosus


+

+
+/−
+
+/−






GAR 4234

Streptococcus anginosus




+
+/−
+/−
+/−






GAR 4426

Streptococcus anginosus


+/−

+
+/−
+
+
+





GAR 4680

Streptococcus anginosus


+/−

+
+/−
+/−
+/−
+/−





GAR 4834

Streptococcus anginosus



+/−
+/−









GAR 4896

Streptococcus anginosus


+
+/−
+
+/−
+
+






GAR 5093

Streptococcus anginosus




+

+
+/−
+/−





GAR 5094

Streptococcus anginosus


+/−
+/−
+
+/−
+/−
+/−
+/−





GAR 5675

Streptococcus anginosus



+/−

+/−

+/−






GAR 5776

Streptococcus anginosus


+
+
+
+
+
+
+/−





GAR 5831

Streptococcus anginosus



+
+/−
+
+/−
+
+/−





GAR 6187

Streptococcus anginosus


+/−
+/−
+/−
+/−


+/−





GAR 6590

Streptococcus anginosus


+/−
+/−
+/−
+/−
+/−
+/−






GAR 7000

Streptococcus anginosus


+/−
+
+/−
+
+/−
+






GAR 7023

Streptococcus anginosus


+/−
+/−

+/−

+/−
+/−





GAR 7190

Streptococcus anginosus







+/−






GAR 7214

Streptococcus anginosus


+
+
+/−
+/−
+/−
+
+/−





GAR 7468

Streptococcus anginosus





+

+/−






GAR 7818

Streptococcus anginosus


+
+
+
+
+
+
+/−

NT



GAR 8620

Streptococcus anginosus


+
+
+
+
+/−
+


NT



GAR 8693

Streptococcus anginosus


+/−





+/−





GAR 8722

Streptococcus anginosus




+/−









GAR 8736

Streptococcus anginosus



+/−

+/−








GAR 8954

Streptococcus anginosus


+/−
+/−
+/−
+/−

+/−






ATCC 27823

Streptococcus constellatus


+/−




+/−






GAR 1235

Streptococcus constellatus














GAR 1384

Streptococcus constellatus


+

+/−
+/−
+/−
+/−
+





GAR 1811

Streptococcus constellatus


+
+
+
+
+
+






GAR 2421

Streptococcus constellatus


+/−
+/−
+/−
+/−
+/−
+/−






GAR 3145

Streptococcus constellatus



+/−




+/−





GAR 3355

Streptococcus constellatus














GAR 4048

Streptococcus constellatus


+/−

+/−









GAR 4083

Streptococcus constellatus


+/−

+
+/−
+
+/−






GAR 4861

Streptococcus constellatus


+
+/−
+
+
+
+/−






GAR 4870

Streptococcus constellatus


+/−

+
+
+/−
+
+/−





GAR 5757

Streptococcus constellatus




+/−






+/−


GAR 6129

Streptococcus constellatus


+
+/−
+/−
+/−
+/−
+/−






GAR 6147

Streptococcus constellatus







+/−






GAR 6258

Streptococcus constellatus


+/−
+/−
+
+
+
+






GAR 7224

Streptococcus constellatus


+
+/−
+
+
+/−
+
+/−





GAR 7369

Streptococcus constellatus


+
+
+
+
+/−
+






ATCC 12394

Streptococcus dysgalactiae


+
+
+/−
+
+/−
+


+
+


ATCC 12394

Streptococcus dysgalactiae


+
+
+
+
+/−
+


+/−
+/−


ATCC 43078

Streptococcus dysgalactiae














ATCC 43078

Streptococcus dysgalactiae














GAR 3868

Streptococcus dysgalactiae


+/−
+/−
+/−
+/−
+
+
+/−





GAR 4272

Streptococcus dysgalactiae


+
+
+
+/−
+
+






ATCC 35666

Streptococcus dysgalactiae


+
+
+
+
+
+
+/−






sub. equisimilis


BAA-338

Streptococcus dysgalactiae



+/−
+/−
+/−
+
+
+/−






sub. equisimilis


GAR 3015

Streptococcus equisimilis


+
+/−
+
+/−
+/−
+
+

+
+


ATCC 27335

Streptococcus intermedius


+
+/−
+
+/−
+/−

+/−





ATCC 27335

Streptococcus intermedius


+
+/−
+
+/−

+/−
+/−





GAR 2407

Streptococcus intermedius


+/−
+/−

+/−








GS28
unk

+
+
+
+
+
+
+
NT
NT
NT


GS67
GGS/GCS

+
+
+
+
+/−
+
+
NT
NT
NT


GS68
GGS/GCS

+/−

+/−

+/−
+
+/−
NT
NT
NT


GS69
GGS/GCS

+/−
+/−



+
+
NT
NT
NT


GS70
GGS/GCS

+/−
+/−
+/−

+/−
+
+/−
NT
NT
NT


GS71
GGS/GCS

+
+/−
+
+
+
+
+
NT
NT
NT


GS72
GGS/GCS

+
+
+
+
+
+
+
NT
NT
NT


GS73
GGS/GCS

+/−




+/−
+/−
NT
NT
NT


GS74
GGS/GCS








NT
NT
NT


GS75
GGS/GCS

+/−
+/−
+/−
+/−
+
+
+
NT
NT
NT


GS77
GGS/GCS

+
ND
+
+/−
+/−
+
+
NT
NT
NT


GS78
GGS/GCS

+/−
+/−
+
+/−
+/−
+
+/−
NT
NT
NT


GS79
GGS/GCS

+/−

+/−
+/−

+

NT
NT
NT


GS80
GGS/GCS





+/−
+
+
NT
NT
NT


GS81
GGS/GCS

+
+
+/−
+
+
+
+/−
NT
NT
NT


GS82
GGS/GCS

+/−
+/−
+/−
+
+/−
+
+/−
NT
NT
NT


GS83
GGS/GCS

+
+
+
+
+
+
+
NT
NT
NT


GS84
GGS/GCS








NT
NT
NT


GS85
GGS/GCS

+/−
+/−

+/−
+/−

+/−
NT
NT
NT


GS86
GGS/GCS

+/−

+/−
+/−
+/−
+
+/−
NT
NT
NT


GS88
GGS/GCS

+
+/−
+
+
+
+/−
+
NT
NT
NT


GS89
GGS/GCS

+/−
+/−



+
+
NT
NT
NT


GS90
GGS/GCS


+/−
+/−
+
+
+
+
NT
NT
NT


GS91
GGS/GCS

+/−
+/−
+/−
+
+/−
+
+
NT
NT
NT


GS92
GGS/GCS

+
+/−
+/−
+/−
+/−
+/−
+/−
NT
NT
NT


GS93
GGS/GCS

+
+
+
+
+
+
+
NT
NT
NT


GS94
GGS/GCS

+
+
+
+/−
+
+
+
NT
NT
NT





In Table 2, the symbol “+” means that the antibodies react to the antigen at least three-fold over background; the symbol “+/−” means that the antibodies react to the antigen between two-fold and three-fold over background; and the sysmbol “−” means that the detection of antibody signal is at or below background.






Example 2
Use of a Three Component Immunogenic Composition to Produce Immune Sera

A trivalent immunogenic composition consisting of SCP, the polypeptide encoded by ORF 554, and the polypeptide encoded by ORF 1358 adjuvanted with aluminum phosphate was prepared. and the immunogenic composition was used to produce hyperimmune rabbit serum by three subcutaneous inoculations separated by 2-4 weeks, followed by exsanguination; a monovalent immunogenic composition consisting of similarly adjuvanted polypeptide encoded by ORF 554 was used as a control. The sera were screened for opsonophagocytic activity (OPA) against S. pyogenes SF370 at various dilutions. Briefly, the bacteria were incubated with 10 ul of sera for one hour in the presence of complement (baby rabbit complement), and then diluted 1:10 and plated on blood agar plates. The results are presented in FIG. 11.


As shown, it can be seen that the Trivax elicits increased opsonophagocytic activity than the 554 immunogenic composition, which is indicative of a much better killing of the bacteria.


Example 3
Passive Immunity Transfer

Antibodies were raised against each of the following antigens as described above: SCP and polypeptides encoded by ORFs 554, 1358, 2459, and 1218. These antibodies were then injected into infant rats without fully functional immune systems. The treated rats are then subsequently challenged with S. pyogenes, and recovered bacteria were counted four hours post-challenge. The negative control was PBS, and the positive human control was 385 sera.


The results are shown in FIGS. 12-16. Briefly, the results demonstrated that antibodies elicited by each of the antigens significantly reduced bacteremia in the infant rats.


Although illustrated and described above with reference to specific embodiments, the invention is nevertheless not intended to be limited to the details shown. Rather, various modifications may be made in the details within the scope and range of equivalents of the claims and without departing from the spirit of the invention.

Claims
  • 1. An immunogenic composition comprising an effective amount of a first polypeptide encoded by a nucleic acid set forth as SEQ ID NO:1 and an effective amount of a second polypeptide encoded by a nucleic acid set forth as SEQ ID NO:7.
  • 2. The immunogenic composition of claim 1, which further comprises a physiologically-acceptable vehicle.
  • 3. The immunogenic composition of claim 1, which further comprises an effective amount of an adjuvant.
  • 4. The immunogenic composition of claim 1 wherein each polypeptide is capable of generating an antibody that specifically recognizes said polypeptide, and wherein the amount of said immunogenic composition is effective to prevent or ameliorate colonization or infection by β-hemolytic streptococci in a susceptible animal.
  • 5. The immunogenic composition of claim 4, which further comprises a physiologically-acceptable vehicle.
  • 6. The immunogenic composition of claim 4, which further comprises an effective amount of an adjuvant.
  • 7. The immunogenic composition of claim 4, wherein the β-hemolytic streptococci is Group A streptococci.
  • 8. The immunogenic composition of claim 7, wherein the Group A streptococci is Streptococcus pyogenes.
  • 9. An immunogenic composition comprising an effective amount of a first isolated polypeptide having an amino acid sequence of SEQ ID NO:2 and an effective amount of a second isolated polypeptide having an amino acid sequence of SEQ ID NO:8.
  • 10. The immunogenic composition of claim 9, which further comprises a physiologically-acceptable vehicle.
  • 11. The immunogenic composition of claim 9, which further comprises an effective amount of an adjuvant.
  • 12. The immunogenic composition of claim 9, wherein each polypeptide is capable of generating an antibody that specifically recognizes said polypeptide, and wherein the amount of said immunogenic composition is effective to prevent or ameliorate colonization or infection by β-hemolytic streptococci in a susceptible animal.
  • 13. The immunogenic composition of claim 12, which further comprises a physiologically-acceptable vehicle.
  • 14. The immunogenic composition of claim 12, which further comprises an effective amount of an adjuvant.
  • 15. The immunogenic composition of claim 12, wherein the β-hemolytic streptococci is Group A streptococci.
  • 16. The immunogenic composition of claim 15, wherein the Group A streptococci is Streptococcus pyogenes.
  • 17. A method of treating a susceptible animal against colonization or infection by β-hemolytic streptococci comprising administering to the animal an effective amount of the immunogenic composition of claim 1, wherein each polypeptide is capable of generating an antibody specific to said polypeptide, and wherein the amount of said immunogenic composition is effective to ameliorate colonization or infection by β-hemolytic streptococci in the susceptible animal.
  • 18. The method of claim 17, wherein the immunogenic composition is administered by subcutaneous injection, by intramuscular injection, by oral ingestion, intranasally, or combinations thereof.
  • 19. The method of claim 17 wherein the β-hemolytic streptococci is Group A streptococci.
  • 20. The method of claim 19 wherein the Group A streptococci is Streptococcus pyogenes.
  • 21. The method of claim 17 wherein the animal is a human.
  • 22. A method of treating a susceptible animal against colonization or infection by β-hemolytic streptococci comprising administering to the animal an effective amount of the immunogenic composition of claim 9, wherein each polypeptide is capable of generating an antibody specific to said polypeptide, and wherein the amount of said immunogenic composition is effective to ameliorate colonization or infection by β-hemolytic streptococci in the susceptible animal.
  • 23. The method of claim 22, wherein the immunogenic composition is administered by subcutaneous injection, by intramuscular injection, by oral ingestion, intranasally, or combinations thereof.
  • 24. The method of claim 22 wherein the β-hemolytic streptococci is Group A streptococci.
  • 25. The method of claim 24 wherein the Group A streptococci is Streptococcus pyogenes.
  • 26. The method of claim 22 wherein the animal is a human.
  • 27. The immunogenic composition of claim 1, further an effective amount of a polypeptide encoded by (a) a nucleic acid set forth as SEQ ID NO: 3, SEQ ID NO: 5,(b) a nucleic acid set forth as SEQ ID NO: 9, or(c) a nucleic acid set forth as SEQ ID NO: 5 and a nucleic acid set forth as SEQ ID NO: 9.
  • 28. The immunogenic composition of claim 27, which further comprises a physiologically-acceptable vehicle.
  • 29. The immunogenic composition of claim 27, which further comprises an effective amount of an adjuvant.
  • 30. The immunogenic composition of claim 27 wherein each polypeptide is capable of generating an antibody that specifically recognizes said polypeptide, and wherein the amount of said immunogenic composition is effective to prevent or ameliorate colonization or infection by β-hemolytic streptococci in a susceptible animal.
  • 31. The immunogenic composition of claim 30, which further comprises a physiologically-acceptable vehicle.
  • 32. The immunogenic composition of claim 30, which further comprises an effective amount of an adjuvant.
  • 33. The immunogenic composition of claim 30, wherein the β-hemolytic streptococci is Group A streptococci.
  • 34. The immunogenic composition of claim 33, wherein the Group A streptococci is Streptococcus pyogenes.
  • 35. The immunogenic composition of claim 9, further an effective amount of a polypeptide having (a) an amino acid sequence of SEQ ID NO: 6,(b) an amino acid sequence of SEQ ID NO: 10, or(c) an amino acid sequence of SEQ ID NO: 6 and an amino acid sequence of SEQ ID NO: 10.
  • 36. The immunogenic composition of claim 35, which further comprises a physiologically-acceptable vehicle.
  • 37. The immunogenic composition of claim 35, which further comprises an effective amount of an adjuvant.
  • 38. The immunogenic composition of claim 35 wherein each polypeptide is capable of generating an antibody that specifically recognizes said polypeptide, and wherein the amount of said immunogenic composition is effective to prevent or ameliorate colonization or infection by β-hemolytic streptococci in a susceptible animal.
  • 39. The immunogenic composition of claim 38, which further comprises a physiologically-acceptable vehicle.
  • 40. The immunogenic composition of claim 38, which further comprises an effective amount of an adjuvant.
  • 41. The immunogenic composition of claim 38, wherein the β-hemolytic streptococci is Group A streptococci.
  • 42. The immunogenic composition of claim 41, wherein the Group A streptococci is Streptococcus pyogenes.
  • 43. A method of treating a susceptible animal against colonization or infection by β-hemolytic streptococci comprising administering to the animal an effective amount of the immunogenic composition of claim 27, wherein each polypeptide is capable of generating an antibody specific to said polypeptide, and wherein the amount of said immunogenic composition is effective to ameliorate colonization or infection by β-hemolytic streptococci in the susceptible animal.
  • 44. The method of claim 43, wherein the immunogenic composition is administered by subcutaneous injection, by intramuscular injection, by oral ingestion, intranasally, or combinations thereof.
  • 45. The method of claim 43 wherein the β-hemolytic streptococci is Group A streptococci.
  • 46. The method of claim 45 wherein the Group A streptococci is Streptococcus pyogenes.
  • 47. The method of claim 43 wherein the animal is a human.
  • 48. A method of treating a susceptible animal against colonization or infection by β-hemolytic streptococci comprising administering to the animal an effective amount of the immunogenic composition of claim 35, wherein each polypeptide is capable of generating an antibody specific to said polypeptide, and wherein the amount of said immunogenic composition is effective to ameliorate colonization or infection by β-hemolytic streptococci in the susceptible animal.
  • 49. The method of claim 48, wherein the immunogenic composition is administered by subcutaneous injection, by intramuscular injection, by oral ingestion, intranasally, or combinations thereof.
  • 50. The method of claim 48 wherein the β-hemolytic streptococci is Group A streptococci.
  • 51. The method of claim 50 wherein the Group A streptococci is Streptococcus pyogenes.
  • 52. The method of claim 48 wherein the animal is a human.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. Ser. No. 12/612,399, filed 4 Nov. 2009, which claims priority from U.S. Provisional Application No. 61/111,485, filed 5 Nov. 2008. The entire contents of these applications are incorporated by reference herein in their entirety.

US Referenced Citations (51)
Number Name Date Kind
4152411 Schall, Jr. May 1979 A
4346074 Gilmour et al. Aug 1982 A
4376110 David et al. Mar 1983 A
4454121 Beachey Jun 1984 A
4554101 Hopp Nov 1985 A
4650764 Temin et al. Mar 1987 A
4666829 Glenner et al. May 1987 A
4673574 Anderson Jun 1987 A
4695562 Beachey et al. Sep 1987 A
4772584 Cleary et al. Sep 1988 A
4784948 Scott et al. Nov 1988 A
4797368 Carter et al. Jan 1989 A
4837151 Stocker Jun 1989 A
4902506 Anderson et al. Feb 1990 A
4912094 Myers et al. Mar 1990 A
4989289 Bargellini Feb 1991 A
5057540 Kensil et al. Oct 1991 A
5078996 Conlon, III et al. Jan 1992 A
5097020 Anderson et al. Mar 1992 A
5124153 Beachey et al. Jun 1992 A
5139941 Muzyczka et al. Aug 1992 A
5162226 Beachey et al. Nov 1992 A
5168062 Stinski Dec 1992 A
5360897 Anderson et al. Nov 1994 A
5399346 Anderson et al. Mar 1995 A
5459127 Feigner et al. Oct 1995 A
5580859 Feigner et al. Dec 1996 A
5589466 Feigner et al. Dec 1996 A
5593972 Weiner et al. Jan 1997 A
5643576 Johnston et al. Jul 1997 A
5679654 Tzianabos et al. Oct 1997 A
5723127 Scott et al. Mar 1998 A
5848547 Hite Dec 1998 A
6100380 Green et al. Aug 2000 A
6113918 Johnson et al. Sep 2000 A
6127170 Boutin Oct 2000 A
6168911 Lelental et al. Jan 2001 B1
6168943 Rose Jan 2001 B1
6207646 Krieg et al. Mar 2001 B1
6270775 Cleary Aug 2001 B1
6355255 Cleary et al. Mar 2002 B1
6951653 Cleary et al. Oct 2005 B2
7256265 Cleary et al. Aug 2007 B2
7635483 Cleary et al. Dec 2009 B2
7638136 Meinke et al. Dec 2009 B2
7838010 Bensi et al. Nov 2010 B2
8563001 Dodge et al. Oct 2013 B2
20040052801 Cleary et al. Mar 2004 A1
20050136068 Cleary et al. Jun 2005 A1
20060153879 Cleary et al. Jul 2006 A1
20070208028 Conn et al. Sep 2007 A1
Foreign Referenced Citations (65)
Number Date Country
2012311 Jun 2003 CA
0125023 Nov 1984 EP
0171496 Feb 1986 EP
0173494 Mar 1986 EP
0178220 Apr 1986 EP
0184187 Jun 1986 EP
0185573 Jun 1986 EP
0272929 Jun 1988 EP
0371199 Jun 1990 EP
0453242 Oct 1991 EP
0488528 Jun 1992 EP
1075841 Feb 2001 EP
WO 8601533 Mar 1986 WO
WO 8602269 Apr 1986 WO
WO 8907150 Aug 1989 WO
WO 8909063 Oct 1989 WO
WO 8909064 Oct 1989 WO
WO 9002806 Mar 1990 WO
WO 9118088 Nov 1991 WO
WO 9119740 Dec 1991 WO
WO 9202612 Feb 1992 WO
WO 9205263 Apr 1992 WO
WO 9219265 Nov 1992 WO
WO 9309239 May 1993 WO
WO 9314198 Jul 1993 WO
WO 9318157 Sep 1993 WO
WO 9321220 Oct 1993 WO
WO 9406421 Mar 1994 WO
WO 9406465 Mar 1994 WO
WO 9412649 Jun 1994 WO
WO 9421807 Sep 1994 WO
WO 9426914 Nov 1994 WO
WO 9428152 Dec 1994 WO
WO 9428938 Dec 1994 WO
WO 9502697 Jan 1995 WO
WO 9507358 Mar 1995 WO
WO 9518863 Jul 1995 WO
WO 9521931 Aug 1995 WO
WO 9522617 Aug 1995 WO
WO 9526411 Oct 1995 WO
WO 9528494 Oct 1995 WO
WO 9528960 Nov 1995 WO
WO 9617823 Jun 1996 WO
WO 9622378 Jun 1996 WO
WO 9625508 Aug 1996 WO
WO 9639036 Dec 1996 WO
WO 9719182 May 1997 WO
WO 9726008 Jul 1997 WO
WO 9802697 Jan 1998 WO
WO 9813501 Apr 1998 WO
WO 9901157 Jan 1999 WO
WO 9901158 Jan 1999 WO
WO 9901175 Jan 1999 WO
WO 9927944 Jun 1999 WO
WO 0018434 Apr 2000 WO
WO 0037648 Jun 2000 WO
WO 0234771 May 2002 WO
WO 02077183 Oct 2002 WO
WO 02083859 Oct 2002 WO
WO 2004078907 Sep 2004 WO
WO 2005076010 Aug 2005 WO
WO 2006042027 Apr 2006 WO
WO 2006073396 Jul 2006 WO
WO 2009155476 Dec 2009 WO
WO 2009155484 Dec 2009 WO
Non-Patent Literature Citations (214)
Entry
Accession No. A35066 (1987).
Alexander, et al., “Amino acid changes affecting the activity of pneumolysin alter the behaviour of pneumococci in pneumonia”, Microbial Pathogenesis, 24(3):167-174 (1998).
Alm, R.A., et al., “Genomic-sequence comparison of two unrelated isolates of the human gastric pathogen Helicobacter pylori”, Nature 379:176-180 (1999) [published erratum appears in Nature 379(6721):719 (1999)].
Altschul, S.F., et al., “Protein database searches for multiple alignments”, Proc. Natl. Acad. Sci. USA, 87:5509-5513 (1990).
Altschul, S.F., et al., “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Research, 25(17):3389-3402 (1997).
Anderson, T.F., et al., “Techniques for the Preservation of Three-Diminsional Structure in Preparing Specimens for the Electron Microscope”, Transactions of The New York Academy of Sciences, 13(4):130-134 (1951).
Anderson, E.T., et al., “Processing, stability, and kinetic parameters of C5a peptidase from Streptococcus pyogenes”, Eur. J. Biochem., 269:4839-4851 (2002).
Ausubel, F.M., et al., “Current Protocols in Molecular Biology”, John Wiley & Sons, Inc., vol. 1, Sections 2.10 and 6.3-6.4 (1995).
Banks, D.J., et al., “Progress toward Characterization of the Group A Streptococcus Metagenome: Complete Genome Sequence of a Macrolide-Resistant Serotype M6 Strain”, Journal of Infectious Diseases, 190(4):727-738 (2004).
Bateman, A., et al., “The Pfam Protein Families Database”, Nucleic Acids Research, 28(1):263-266 (2000).
Beard, C.W., et al., “Transcription Mapping of Mouse Adenovirus Type 1 Early Region 3”, Virology, 175:81-90 (1990).
Bender, M.A., et al., “Evidence that the Packaging Signal of Moloney Murine Leukemia Virus Extends into the gag Region”, Journal of Virology, 61(5):1639-1646 (1987).
Benson, G., “Tandem repeats finder: a program to analyze DNA sequences”, Nucleic Acids Research, 27(2):573-580 (1999).
Beres, S.B., et al., “Molecular genetic anatomy of inter- and intraserotype variation in the human bacterial pathogen group A Streptococcus”, PNAS, 103(18):7059-7064 (2006).
Berg, A., et al., “Streptococcal Cysteine Proteinase Releases Biologically Active Fragments of Streptococcal Surface Proteins”, The Journal of Biological Chemistry, 270(17):9862-9867 (1995).
Bessen, D., et al., “Influence of Intranasal Immunization with Synthetic Peptides Corresponding to Conserved Epitopes of M Protein on Mucosa! Colonization by Group A Streptococci”, Infection and Immunity, 56(10):2666-2672 (1988).
Bessen, D., et al., “Synthetic Peptide Vaccine Against Mucosa! Colonization by Group A Streptococci”, The Journal of Immunology, 145(4):1251-1256 (1990).
Better, M., et al., “Escherichia coli Secretion of an Active Chimeric Antibody Fragment”, Science, 240:1041-1043 (1988).
Booth, S.A., et al., “Dapsone Suppresses Integrin-Mediated Neutrophil Adherence Function”, The Journal of Investigative Dermatology, 98(2):135-140 (1992).
Boyle, M.D.P., et al., “Measurement of Leukocyte Chemataxis in Vivo”, Methods in Enzymology, 162:101-114 (1988).
Bronze, M.S., et al., “Protective Immunity Evoked by Locally Admisistered Group A Streptococcal Vaccines in Mice”, The Journal of Immunology, 141(8):2767-2770 (1988).
Bronze, M.S., et al., “Epitopes of Group A Streptococcal M Protein that Envoke Cross-Protective Local Immune Responses”, The Journal of Immunology, 148(3):888-893 (1992).
Brown, C.K., et al., “Structure of the streptococcal cell wall C5a peptidase”, PNAS, 102(51):18391-18396 (2005).
Brummer, E., et al., “Immunological Activation of Polymorphonuclear Neutrophils for Fungal Killing: Studies With Murine Cells and Blastomyces dermatitidis In Vitro”, Journal of Leukocyte Biology, 36:505-520 (1984).
Cabilly, S., et al., “Generation of antibody activity from immunoglobulin polypeptide chains produced in Escherichia coli”, Proc. Natl. Acad. Sci. USA, 81:273-3277 (1984).
Carrillo, H., et al., “The Multiple Sequence Alignment Problem in Biology”, SIAM J. Appl. Math., 48(5):1073-1082 (1988).
Carter, P., et al., “Dissecting the catalytic triad of a serine protease”, Nature, 332:564-568 (1988).
Chen, C.C., et al., “Cloning and Expression of the Streptococcal C5a Peptidase Gene in Escherichia coli: Linkage to the Type 12 M Protein Gene”, Infection and Immunity, 57(6):1740-1745 (1989).
Chen, C.C., et al., “Complete Nucleotide Sequence of the Streptococcal C5a Peptidase Gene of Streptococcus pyogenes”, The Journal of Biological Chemistry, 265(6):3161-3167 (1990).
Cheng, Q., et al., “Antibody Against Surface-Bound C5a Peptidase is Opsonic and Initiates Macrophage Killing of Group B Streptococci”, Infection and Immunity, 69(4):2302-2308 (2001).
Cheng, Q., et al., “The Group B Streptococcal C5a peptidase is Both a Specific Protease and an Invasin”, Infection and Immunity, 70(5):2408-2413 (2002).
Cheng Qi et al., “Immunization with C5a peptidase or peptidase-type III polysaccharide conjugate vaccines enhances clearance of group B streptococci from lungs of infected mice”, Infection and Immunity, vol. 70, No. 11, 6409-6415 (2002).
Chmouryguina, I., et al., “Conservation of the C5a peptidase genes in group A and B streptococci”, Infection and Immunity, 64(7):2387-2390 (1996).
Clark, J.M., et al., “A New Method for Quantitation of Cell-Mediated Immunity in the Mouse”, Journal of the Reticuloendothelial Society, 25(3):255-267 (1979).
Cleary, P., et al., “A Streptococcal Inactivator of Chemotaxis: A new Virulence Factor Specific to Group A Streptococci”, Recent Advances in Streptococci and Streptococcal Diseases, Y. Kimura, S. Kotami and Y. Shiokawa, eds., Reedbooks Ltd, Berkshire, England, pp. 179-180 (1984).
Cleary, P.P., et al., “Virulent human strains of group G streptococci express a C5a peptidase enzyme similar to that produced by group A streptococci”, Infection and Immunity, 59(7):2305-2310 (1991).
Cleary, P.P., et al., “Similarity between the group B and A Streptococcal C5a peptidase genes”, Infection and Immunity, 60(10):4239-4244 (1992).
Cleary, P.P., et al., “Streptococcal C5a peptidase is a highly specific endopeptidase”, Infection and Immunity, 60(12):5219-5223 (1992).
Cleary, P.P., et al., “Immunization with C5a peptidase from either group A or B streptococci enhances clearance of group A streptococci from intranasally infected mice”, Vaccine, 22(31-32):4332-4341 (2004).
Cockerill, F.R., et al., “Molecular, Serological, and Clinical Features of 16 Consecutive Cases of Invasive Streptococcal Disease”, Clin. Infect. Dis., 26(6):1448-1458 (1998).
Coid, C.R., “Escherichia coli and group B streptococcal infections in experimental animals”, Ciba Found Symp., 77:103-118 (1979) (Abstract only).
Courtney, H.S., et al., “Analysis of the Role of M24 Protein in Group A Streptococcal Adhesion and Colonization by Use of 0-Interposon Mutagenesis”, Infection and Immunity, 62(11):4868-4873 (1994).
Courtney, H.S., et al., “Cloning, sequencing, and expression of a fibronectin/fibrinogen-binding protein from group A streptococci”, Infection and Immunity, 62(9):3937-3946 (1994).
Cserzo, M., et al., “Prediction of transmembrane a-helices in prokaryotic membrane proteins: the dense alignment surface method”, Protein Engineering, 10(6):673-676 (1997).
Cunningham, M.W., et al., “Immunological Crossreactivity Between the Class I Epitope of Streptococcal M Protein and Myosin”, Advances in Experimental Medicine and Biology, 418:887-892 (1997).
Curiel, D.T., et al., “High-Efficiency Gene Transfer Mediated by Adenovirus Coupled to DNA-Polylysine Complexes”, Human Gene Therapy, 3:147-154 (1992).
Dale, J.B., et al., “Passive Protection of Mice against Group A Streptococcal Pharyngeal Infection by Lipoteichoic Acid”, Journal of Infectious Diseases, 169(2):319-323 (1994).
Dale, J.B., et al., “Recombinant, octavalent group A streptococcal M protein vaccine”, Vaccine, 14(10):944-948 (1996).
Dale, J.B., et al., “Hyaluronate capsule and surface M protein in resistance to opsonization of groupA streptococci”, Infection and Immunity, 64(5):1495-1501 (1996).
Dale J. B. et al., “Group A and Group B streptoccal vaccine development a round table presentation”, Advance in Experimental Medicine and Biology, vol. 418, 863-868 (Jan. 1997).
Database EMBL “Online”, “Streptococcus pyogenes M1 GAS”, Section 136 of 167 of the complete genome, XP02344849 (Apr. 16, 2001).
Database EMBL “Online”, “Streptococcus pyogenes M1 GAS”, Section 63 of 167 of the complete genome, XP002363053 (Apr. 16, 2001).
Database EMBL EBI: “S. pyogenes protein export PrtM precursor (prtM) gene” database accession No. AF387738 (Jul. 4, 2001).
Davis, L.G., et al., “Basic Methods in Molecular Biology”, Elsevier Science Publishing Co., Inc. (1986).
Devereux, J., et al., “A comprehensive set of sequence analysis programs for the VAX”, Nucleic Acids Research, 12(1):387-395 (1984).
Eddy, S.R., “Hidden Markov models”, Cur. Opin. Struct. Bio., 6:361-365 (1996).
Efstratiou, A., et al., “Outbreaks of human infection caused by pyogenic streptococci of Lancefield groups C and G”, J. Med. Microbiol., 29(3):207-219 (1989).
Ellen, R.P., et al., “M Protein-Associated Adherence of Streptococcus pyogenes to Epithelial Surfaces: Prerequisite for Virulence”, Infection and Immunity, 5(5):826-830 (1972).
Eng, J.K., et al., “An Approach to Correlate Tandem Mass Spectral Data of peptides with Amino Acid Sequences in a Protein Database”, American Society for Mass Spectrometry, 5:976-989 (1994).
Feldman, C., et al., “Pneumolysin Induces the Salient Histologic Features of Pneumococcal Infection in the Rat Lung In Vivo”, Am. J. Respir. Cell. Mol. Biol., 5(5):416-423 (1991).
Feldman, R.G., et al., “Solid-phase antigen density and avidity of antibodies detected in anti-group B streptococcal type III IgG enzyme immunoassays”, J. Immunol. Methods, 170:37-45 (1994).
Felgner, P.L., et al., “Cationic liposome-mediated transfection”, Nature, 337:387-388 (1989).
Felgner, P.L., et al., “Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure”, Proc. Natl. Acad. Sci. USA, 84:7413-7417 (1987).
Fenderson, P.G., et al., “Tropomyosin Shares Immunologic Epitopes with Group A Streptococcal M Proteins”, The Journal of Immunology, 142(7):2475-2481 (1989).
Ferretti, J.J., et al., “Complete genome sequence of an M1 strain of Streptococcus pyogenes”, PNAS, 98(8):4658-4663 (2001).
Fischetti, V.A., “Streptococcal M protein: molecular design and biological behavior”, Clinical Microbiology Reviews, 2(3):285-314 (1989).
Fischetti, V.A., et al., “Protection Against Streptococcal Pharyngeal Colonization with a Vaccinia: M Protein Recombinant”, Science, 244:1487-1490 (1989).
Fischetti, V.A., et al., “Conservation of a hexapeptide sequence in the anchor region of surface proteins from Gram-positive cocci”, Molecular Microbiology, 4(9):1603-1605 (1990).
Fischetti, V.A., et al., “Protection against streptococcal pharynogeal colonization with vaccines composed of M protein conserved regions”, Immunobiology of Proteins and Peptides VI, M.Z. Atassi, ed. (Plenum Press, New York, NY) pp. 159-167 (1991).
Fogg, G.C., et al., “Constitutive Expression of Fibronectin Binding in Streptococcus pyogenes as a Result of Anaerobic Activation of rofA”, Journal of Bacteriology, 179(19):6172-6180 (1997).
Foster, T.J., et al., “Surface protein adhesins of Staphylococcus aureus”, Trends in Microbiology, 6(12):484-488 (1998).
Fraser, C.M., et al., “Genomic sequence of a Lyme disease spirochaete, Borrelia burgdorferi”, Nature, 390:580-586 (1997).
Friedman, H., et al., “Immunoadjuvanticity of Endotoxins and Nontoxic Derivatives for Normal and Leukemic Immunocytes”, Advances in Experimental Medicine and Biology, 256:525-535 (1990).
Graham, F.L., et al., “Characteristics of a Human Cell Line Transformed by DNA from Human Adenovirus Type 5”, J. Gen. Virol., 36:59-72 (1977).
Graham, F.L., “Covalently closed circles of human adenovirus DNA are infectious”, The EMBO Journal, 3(12):2917-2922 (1984).
Gribskov, M., et al., eds., “Sequence Analysis Primer”, Stockton Press, New York (1991).
Griffin, H.G., et al., “Chapter 1: Computer Analysis of Sequence Data”, Methods in Molecular Biology, Humana Press, New Jersey, pp. 1-8 (1994).
Hacker, J., et al., “Pathogenicity islands of virulent bacteria: structure, function and impact on microbial evolution”, Molecular Microbiology, 23(6):1089-1097 (1997).
Hanski, E., et al., “Expression of Protein F, the Fibronectin-Binding protein of Streptococcus pyogenes JRS4, in Heterologous Streptococcal and Enterococcal Strains Promotes Their Adherence to Respiratory Epithelial Cells”, Infection and Immunity, 60(12):5119-5125 (1992).
Hanski, E., et al., “Protein F, a fibronectin-binding protein, is an adhesin of the group Astreptococcus Streptococcus pyogenes”, Proc. Natl. Acad. Sci. USA, 89:6172-6176 (1992).
Hernandez-Sanchez, J., et al., “A bar minigene-mediated inhibition of protein synthesis involves accumulation of peptidyl-tRNA and starvation for tRNA”, The EMBO Journal, 17(13):3758-3765 (1998).
Hill, H.R., et al., “Group B Streptococci Inhibit the Chemotactic Activity of the Fifth Component of Complement”, The Journal of Immunology, 141(10):3551-3556 (1988).
Holmgren, J., et al., “Bacterial Enteric Infections and Vaccine Development”, Mucosal Immunology II: Clinical Applications, 21(2):283-302 (1992).
Hope-Simpson, R., “Streptococcus pyogenes in the Throat: A study in a small population, 1962-1975”, J. Hyg. Camb., 87(1):109-129 (1981).
Houghten et al. (Vaccines, 1986, Edited by Fred Brown: Cold Spring Harbor Laboratory).
Huang, T.-T., “The streptokinase gene of group A streptococci: cloning, expression in Escherichia coli, and sequence analysis”, Molecular Microbiology, 3(2):197-205 (1989).
Hynes, W.L., et al., “Analysis of a Second Bacteriophage Hyaluronidase Gene from Streptococcus pyogenes: Evidence for a Third Hyaluronidase Involved in Extracellular Enzymatic Activity”, Infection and Immunity, 63(8):3015-3020 (1995).
Hynes, W.L., et al., “The extracellular hyaluronidase gene (hylA) of Streptococcus pyogenes”, FEMS Microbiology Letters, 184:109-112 (2000).
International Search Report dated Feb. 25, 2010.
International Search Report mailed Jan. 21, 2010 for Intl Appl. No. PCT/US2009/047886.
International Search Report mailed Jan. 21, 2010 for Intl Appl. No. PCT/US2009/047902.
Isberg, R.R., et al., “Binding and internalization of microorganisms by integrin receptors”, Trends in Microbiology, 2(1):10-14 (1994).
Ji, Y., et al., “C5a peptidase alters clearance and trafficking of a group A streptococci by infected mice”, Infection and Immunity, 64(2):503-510 (1996).
Ji, Y., et al, “Intranasal immunization with C5a peptidase prevents nasopharyngeal colonization of mice by the group A Streptococcus”, Infection and Immunity, 65(6):2080-2087 (1997).
Jones, K. F., et al., “The importance of the location of antibody binding on the M6 protein for opsonization and phagocytosis of group A M6 streptococci”, J Exp Med. 167:1114-1123 (1988).
Kafri, T., et al., “A Packaging Cell Line for Lentivirus Vectors”, Journal of Virology, 73(1):576-584 (1999).
Kaplitt, M.G., et al., “Expression of a Functional Foreign Gene in Adult Mammalian Brain following in Vivo Transfer via a Herpes Simplex Virus Type 1 Defective Viral Vector”, Molecular and Cellular Neurosciences, 2:320-330 (1991).
Kapur, V., et al., “Vaccination with streptococcal extracellular cysteine protease (interleukin-113 convertase) protects mice against challenge with heterologous group A streptococci”, Microbial Pathogenesis, 16(6):443-450 (1994).
Kihlberg, B.M., et al., “Protein H, an Antiphagocytic Surface Protein in Streptococcus pyogenes”, Infection and Immunity, 67(4):1708-1714 (1999).
Koebnik, R., “Proposal for a peptidoglycan-associating alpha-helical motif in the C-terminal regions of some bacterial cell-surface proteins”, Molecular Microbiology, 16(6):1269-1270 (1995).
Kohler, G., et al., “Continuous cultures of fused cells secreting antibody of predefined specificity”, Nature, 256:495-497 (1975).
Kuo, M.-L., et al., “Efficient Gene Transfer Into Primary Murine Lymphocytes Obviating the Need for Drug Selection”, Blood, 82(3):845-852 (1993).
Laemmli, U.K., “Cleavage of Structural Proteins during the Assembly of the Head of Bacteriophage T4”, Nature, 227:680-685 (1970).
Lazar, E., et al., “Transforming growth factor a: Mutation of aspartic acid 47 and leucine 48 results in different biological activities”, Mol. Cell. Biol., 8(3):1247-1252 (1988).
Lebkowski, J.S., et al., “Adeno-Associated Virus: a Vector System for Efficient Introduction and Integration of DNA into a Variety of Mammalian Cell Types”, Molecular and Cellular Biology, 8(10):3988-3996, (1988).
Lederman, S., et al., “A Single Amino Acid Substitution in a Common African Allele of the CD4 Molecule Ablates Binding of the Monoclonal Antibody, OKT4”, Molecular Immunology, 28(11):1171-1181 (1991).
Lee, P.K., et al., “Quantification and Toxicity of Group A Streptococcal Pyrogenic Exotoxins in an Animal Model of Toxic Shock Syndrome-Like Illness”, Journal of Clinical Microbiology, 27(8):1890-1892 (1989).
Lesk, A.M., ed., “Computational Molecular Biology: Sources and Methods for Sequence Analysis”, Oxford Univ. Press, New York (1988).
Levrero, M., et al., “Defective and nondefective adenovirus vectors for expressing foreign genes in vitro and in vivo”, Gene, 101:195-202 (1991).
Li, C.H., et al., “13-Endorphin omission analogs: Dissociation of immunoreactivity from other biological activities”, Proc. Natl. Acad. Sci. USA, 77(6):3211-3214 (1980).
Liu, A.Y., “Chimeric mouse-human IgG1 antibody that can mediate lysis of cancer cells”, Proc. Natl. Acad. Sci. USA, 84:3439-3443 (1987).
Loessner, M.J., et al., “Evidence for a Holin-Like protein Gene Fully Embedded Out of Frame in the Endolysin Gene of Staphylococcus aureus Bacteriophage 187”, 181(15):4452-4460 (1999).
Lukashin, A.V., et al., “GeneMark.hmm: new solutions for gene finding”, Nucleic Acids Research, 26(4):1107-1115 (1998).
Lukomski, S., et al., “Extracellular Cysteine Protease Produced by Streptococcus pyogenes Participates in the Pathogenesis of Invasive Skin Infection and Dissemination in Mice”, Infection and Immunity, 67(4):1779-1788 (1999).
Machy, P., et al., “Gene transfer from targeted liposomes to specific lymphoid cells by electroporation”, Proc. Natl. Acad. Sci. USA, 85:8027-8031 (1988).
Madore, D. V. “Characterization of immune response as an indicator of Haemophilus influenzae type b vaccine efficacy”, Pediatr Infect Dis J. 17:S207-10 (1998).
Maione Domenico et al., “Identification of a universal Group B Streptococcus vaccine by multiple genome screen”, Science, American Association for the Advancement of Science, vol. 309, No. 5731, 148-150 (Jul. 2005).
Mann, R., et al., “Construction of a Retrovirus Packaging Mutant and Its Use to Produce Helper-Free Defective Retrovirus”, Cell, 33:153-159 (1983).
Markowitz, D., et al., “A safe packaging line for gene transfer: separating viral genes on two different plasmids”, Journal of Virology, 62(4):1120-1124 (1988).
Martin, T., et al., “The Effect of Type-Specific Polysaccharide Capsule on the Clearance of Group B Streptococci from the Lungs of Infant and Adult Rats”, The Journal of Infectious Diseases, 165(2):306-314 (1992).
Massell, B.F., et al., “Rheumatic Fever Following Streptococcal Vaccination: Report of Three Cases”, JAMA, 207(6):1115-1119 (1989).
Matsuka, Y.V., et al., “Fibrinogen Cleavage by the Streptococcus pyogenes Extracellular Cysteine Protease and Generation of Antibodies That Inhibit Enzyme Proteolytic Activity”, Infection and Immunity, 67(9):4326-4333 (1999).
Mazmanian, S.K., et al., “Staphylococcus aureus Sortase, an Enzyme that Anchors Surface Proteins to the Cell Wall”, 285:760-763 (1999).
McAtee, C.P., et al., “Identification of Potential Diagnostic and Vaccine Candidates of Helicobacter pylori by Two-Dimensional Gel Electrophoresis, Sequence Analysis, and Serum Profiling”, Clinical and Diagnostic Laboratory Immunology, 5(4):537-542 (1998).
McAtee, C.P., et al., “Identification of Potential Diagnostic and Vaccine Candidates of Helicobacter pylori by “Proteome” Technologies”, Helicobacter, 3(3):163-169 (1998).
McAtee, C.P., et al., “Characterization of a Helicobacter pylori vaccine candidate by proteome techniques”, Journal of Chromatography B, 714:325-333 (1998).
McCormick, D., et al., “Human Gene Therapy: The First Round”, Nature Biotechnology, 3(8):689-693 (1985).
McGhee, J.R., et al., “Mucosal Immunity to Vaccines: Current Concepts for Vaccine Development and Immune Response Analysis”, Genetically Engineered Vaccines, J.E. Ciardi et al., eds., Plenum Press, New York, pp. 3-12 (1992).
McGhee, J., et al., “New Perspective in Mucosa! Immunity with Emphasis on Vaccine Development”, Seminars in Hematology, 30(14):3-15 (1993).
McMillan D. J. et al., “Identification and assessment of new vaccine candidates for group A streptococcal infections”, Vaccine, Butterworth Scientific, vol. 22, No. 21-22, (Jul. 2004).
Medaglini, D., et al., “Mucosal and systemic immune responses to a recombinant protein expressed on the surface of the oral commensal bacterium Streptococcus gordonii after oral colonization”, Proc. Natl. Acad. Sci. USA, 92(15):6868-6872 (1995).
Mejlhede, N., et al., “Ribosomal-1 Frameshifting during Decoding of Bacillus subtilis cdd Occurs at the Sequence CGA AAG”, Journal of Bacteriology, 181(9):2930-2937 (1999).
Miller, A.D., et al., “Improved Retroviral Vectors for Gene Transfer and Expression”, BioTechniques, 7(9):980-990 (1989).
Mir, L.M., et al., “Long-term, high level in vivo gene expression after electric pulse-mediated gene transfer into skeletal muscle”, C.R. Acad. Sci. Paris, Life Sciences, 321:893-899 (1998).
Mitchell, T.J., et al., “Complement activation and antibody binding by pneumolysin via a region of the toxin homologous to a human acute-phase protein”, Mol. Microbiol., 5(8):1883-1888 (1991).
MMWR (Morbidity and Mortality Weekly Report), “Case Definitions for Infectious Conditions Under Public Health Surveillance”, vol. 46, No. RR-10 (May 2, 1997).
Molinari, G., S. R. et al., “The fibronectin-binding protein of Streptococcus pyogenes, Sfbl, is involved in the internalization of group A streptococci by epithelial cells”, Infect Immun. 65:1357-63 (1997).
Morrison, S.L., et al., “Chimeric human antibody molecules: Mouse antigen-binding domains with human constant region domains”, Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984).
Mountzouros, K.T., et al., “Detection of Complement-Mediated Antibody-Dependent Bactericidal Activity in a Fluorescence-Based Serum Bactericidal Activity in a Fluorescence-Based Serum Bactericidal Assay for Group B Neisseria meningitidis”, Journal of Clinical Microbiology, 38(8):2878-2884 (2000).
Nakai, K., et al., “Expert system for Predicting Protein Localization Sites in Gram-Negative Bacteria”, Proteins: Structure, Function, and Genetics, 11:95-110 (1991).
Naldini, L., “Lentiviruses as gene transfer agents for delivery to non-dividing cells”, Current Opinion in Biotechnology, 9:457-463 (1998).
Navarre, W.W., et al., “Surface proteins of Gram-Positive Bacteria and Mechanisms of Their Targeting to the Cell Wall Envelope”, Microbiology and Molecular Biology Reviews, 63(1):174-229 (1999).
NCBI Genbank, “High-affinity zinc uptake system protein znuA precursor [Streptococcus pyogenes MGAS10394]”, Accession No. AAT86698, accessed Aug. 19, 2010.
NCBI Genbank, “Putative protease maturation protein [Streptococcus pyogenes]”, Accession No. NP—269488, accessed Jun. 27, 2008.
NCBI Genbank, “Zinc ABC transporter, zinc-binding adhesion liprotein [Streptococcus agalactiae 2603V/R]”, Accession No. NP—687564, accessed Aug. 19, 2010.
Nielsen, H., et al., “Identification of prokaryotic and eukaryotic signal peptides and prediction of their cleavage sites”, Protein Engineering, 10(1):1-6 (1997).
Nizet, V., et al., “Genetic Locus for Streptolysin S Production by Group A Streptococcus”, Infection and Immunity, 68(7):4245-4254 (2000).
Nordstrand, A., et al., “Allele Substitution of the Streptokinase Gene Reduces the Nephritogenic Capacity of Group A Streptococcal Strain NZ131”, Infection and Immunity, 68(3):1019-1025 (2000).
O'Connor, S., et al., “In Vivo Streptococcus pyogenes C5a Peptidase Activity: Analysis Using Transposon- and Nitrosoguanidine-Induced Mutants”, The Journal of Infectious Diseases, 156(3):495-504 (1987).
O'Connor, S., et al., “The Human Antibody Response to Streptococcal C5a Peptidase”, The Journal of Infectious Diseases, 163:109-116 (1991).
Olmsted, S.B., et al., “High-Resolution Visualization by Field Emission Scanning Electron Microscopy of Enterococcus faecalis Surface Proteins Encoded by the Pheromone-Inducible Conjugative Plasm id pCF10”, Journal of Bacteriology, 175(19):6229-6237 (1993).
Park, J., et al., “DIVCLUS: an automatic method in the GEANFAMMER package that finds homologous domains in single- and multi-domain proteins”, Bioinformatics, 14(2):144-150 (1998).
Parkhill, J., et al., “Complete DNA sequence of a serogroup A strain of Neisseria meningitidis Z2491”, Nature, 404:502-506 (2000).
Pierschbacher, M.D., et al., “Influence of Sterochemistry of the Sequence Arg-Gly-Asp-Xaa on Binding Specificity in Cell Adhesion”, The Journal of Biological Chemistry, 262(36):17294-17298 (1987).
Pizza, M., et al., “Identification of Vaccine Candidates Against Serogroup B Meningococcus by Whole-Genome Sequencing”, Science, 287(5459):1816-1820 (2000).
Podbielski, A., et al., “The Group A streptococcal virR49 Gene Controls Expression of Four Structural vir Regulon Genes”, Infection and Immunity, 63(1):9-20 (1995).
Proft, T., et al., “Identification and Characterization of Novel Superantigens from Streptococcus pyogenes”, J. Exp. Med., 189(1):89-101 (1999).
Pugsley, A.P., “The Complete General Secretory Pathway in Gram-Negative Bacteria”, Microbiological Reviews, 57(1):50-108 (1993).
Quinn, A., et al., “Immunological Relationship between the Class I Epitope of Streptococcal M Protein and Myosin”, Infection and Immunity, 66(9):4418-4424 (1998).
Raeder, R., et al., “Properties of IgG-Binding Proteins Expressed by Streptococcus pyogenes Isolates Are Predictive of Invasive Potential”, The Journal of Infectious Diseases, 173(4):888-895 (1996).
Reda, K.B., et al., “Phylogenetic Distribution of Streptococcal Superantigen SSA Allelic Variants Provides Evidence for Horizontal Transfer of ssa within Streptococcus pyogenes”, Infection and Immunity, 64(4):1161-1165 (1996).
Robles, G., et al., “Antibodies against extracellular products of group A Streptococcus: Diagnostic importance in acute rheumatic fever”, Arch. Inst. Cardiol. Mex., 65(2):115-119 (1995) (Abstract only).
Rossi, F., et al., “Engineered Idiotypes: Immunochemical Analysis of Antigenized Antibodies expressing a Conformationally Constrained Arg-Gly-Asp Motif”, Molecular Immunology, 32(5):341-346 (1995).
Rudinger, J., “Chapter 1: Characteristics of the amino acids as components of a peptide hormone sequence”, Peptide Hormones, J.A. Parsons, ed., University Park Press, 6 pages (1976).
Ruoff, K.L., et al., “Chapter 17: Streptococcus”, Manual of Clinical Microbiology, P.R. Murray, et al., eds., ASM Press, Washington, DC (7th Ed.), pp. 283-296 (1999).
Sahagan, B.G., et al., “A Genetically Engineered Murine/Human Chimeric Antibody Retains Specificity for Human Tumor-Associated Antigen”, The Journal of Immunology, 137(3):1066-1074 (1986).
Salzberg, S.L., et al., “Microbial gene identification using interpolated Markov models”, Nucleic Acids Research, 26(2):544-548 (1998).
Sambrook, J., et al., “Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, New York (2nd Ed.), Chapters 9 and 11 (1989).
Samulski, R.J., et al., “A Recombinant Plasmid from Which an Infectious Adeno-Associated Virus Genome Can Be Excised In Vitro and Its Use to Study Viral Replication”, Journal of Virology, 61(10):3096-3101 (1987).
Samulski, R.J., et al., “Helper-Free Stocks of Recombinant Adeno-Associated Viruses: Normal Integration Does Not Require Viral Gene Expression”, Journal of Virology, 63(9):3822-3828 (1989).
Scott, T.A., et al., eds., “The Concise Encyclopedia: Biochemistry and Molecular Biology” (3rd Ed.), Walter de Gruyter Inc., New York, p. 489 (1996).
Japanese Office Action and translation thereof, JP App. No. 2000-586920, 6 pages, mailing date Jun. 28, 2011.
Siezen, R.J., et al., “Homology modeling and protein engineering strategy of subilases, the family of subtilisin-like serine proteinases”, Protein Engineering, 4(7):719-737 (1991).
Siezen, R.J., et al., “Subtilases: The superfamily of subtilisin-like serine proteases”, Protein Science, 6(3):501-523 (1997).
Smith, D.W., ed., “Biocomputing: Informatics and Genome Projects”, Academic Press, Inc., New York (1994).
Smoot, J.C., et al., “Genome sequence and comparative microarray analysis of serotype M18 group A Streptococcus strains associated with acute rheumatic fever outbreaks”, PNAS, 99(7):4668-4673 (2002).
Sonnenberg, M.G., et al., “Definition of Mycobacterium tuberculosis Culture Filtrate Proteins by Two-Dimensional Polyacrylamide Gel Electrophoresis, N-Terminal Amino Acid Sequencing, and Electrospray Mass Spectrometry”, Infection and Immunity, 65(11):4515-4524 (1997).
Sonnhammer, E.L.L., et al., “Pfam: A Comprehensive Database of Protein Domain Families Based on Seed Alignments”, Proteins: Structure, Function, and Genetics, 28:405-420 (1997).
Springer, T., et al., “Mac-1: a macrophage differentiation antigen Identified by monoclonal antibody”, European Journal of Immunology, 9(4):301-306 (1979).
Sriskandan, S., et al., “Streptococcal Pyrogenic Exotoxin A Release, Distribution, and Role in Murine Model of Fasciitis and Multiorgan Failure Due to Streptococcus pyogenes”, Journal of Infectious Diseases, 173(6):1399-1407 (1996).
Sriskandan, S., et al., “The Role of Nitric Oxide in Experimental Murine Sepsis Due to Pyrogenic Exotoxin A-Producing Streptococcus pyogenes”, Infection and Immunity, 65(5):1767-1772 (1997).
Stafslien, D.K., et al., “Site Directed Mutagenesis of the Streptococcal C5a Peptidase”, Abstracts of the 98th General Meeting of the American Society for Microbiology (Atlanta, GA), Abstract B-21, p. 59 (May 17-21, 1998).
Stafslien, D.K., et al., “Characterization of the Streptococcal C5a Peptidase using a C5a-Green Fluorescent Protein Fusion Protein Substrate”, Journal of Bacteriology, 182(11):3254-3258 (2000).
Stevens, D.L., “Invasive Group A Streptococcus Infections”, Clinical Infectious Diseases, 14(1):2-11 (1992).
Stevens, D.L., “Streptococcal Toxic-Shock Syndrome: Spectrum of Disease, Pathogenesis, and New Concepts in Treatment”, Emerg. Infect. Dis., 1(3):69-78 (1995).
Stockbauer, K.E., et al., “A natural variant of the cysteine protease virulence factor of group A Streptococcus with an arginine-glycine-aspartic acid (RGD) motif preferentially binds human integrins ανβ3 and αllbβ3”, Proc. Natl. Acad. Sci. USA, 96:242-247 (1999).
Stratford-Perricaudet, L.D., et al., “Widespread Long-term Gene Transfer to Mouse Skeletal Muscles and Heart”, J. Clin. Invest., 90:626-630 (1992).
Sumby, P., et al., “Evolutionary Origin and Emergence of a Highly Successful Clone of Serotype M1 Group A Streptococcus Involved Multiple Horizontal Gene Transfer Events”, J. Infect. Dis., 192:771-782 (2005).
Sun, L.K., et al., “Chimeric antibody with human constant regions and mouse variable regions directed against carcinoma-associated antigen 17-1A”, Proc. Natl. Acad. Sci. USA, 84:214-218 (1987).
Suvorov, A.N., et al., “C5a Peptidase Gene from Group B Streptococci”, Genetics and Molecular Biology of Streptococci, Lactococci, and Enterococci, G.M. Dunny, L.L. McKay & P.P. Cleary, eds., American Society for Microbiology, Washington, D.C., pp. 230-232 (1991).
Third Party Observations under Article 115 EPC, European Patent Application No. 02762074.9, published as EP1421098 (W002/083859), Wyeth, 2 pages (submitted on Jul. 4, 2008).
Ton-That, H., et al., “Purification and characterization of sortase, the transpeptidase that cleaves surface proteins of Staphylococcus aureus at the LPXTG motif”, PNAS, 96(22):12424-12429 (1999).
Ulmer, J.B., et al., “Heterologous Protection Against Influenza by Injection of DNA Encoding a Viral Protein”, Science, 259:1745-1749 (1993).
Von Heijne, G., “Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit”, Academic Press, Inc., New York (1987).
Vugia, D.J., et al., “Invasive group A streptococcal infections in children with varicella in Southern California”, Pediatr. Infect. Dis. J., 15(2):146-150 (1996).
Wahl, R.L., et al., “Improved Radioimaging and Tumor Localization with Monoclonal F(ab′)2”, J. Nucl. Med. 24:316-325 (1983).
Weldingh, K., et al., “Two-Dimensional Electrophoresis for Analysis of Mycobacterium tuberculosis Culture Filtrate and Purification and Characterization of Six Novel Proteins”, Infection and Immunity, 66(8):3492-3500 (1998).
Wessels, M.R., et al., “Critical role of the group A streptococcal capsule in pharyngeal colonization and infection in mice”, Proc. Natl. Acad. Sci. USA, 91(25):12238-12242 (1994).
Wexler, D.E., et al., “Purification and Characteristics of the Streptococcal Chemotactic Factor Inactivator”, Infection and Immunity, 50(3):757-764 (1985).
Wexler, D.E., et al., “Mechanism of action of the group A streptococcal C5a inactivator”, Proc. Natl. Acad. Sci. USA, 82(23):8144-8148 (1985).
Williams, R.S., et al., “Introduction of foreign genes into tissues of living mice by DNA-coated microprojectiles”, Proc. Natl. Acad. Sci. USA, 88:2726-2730 (1991).
Wilson, J.M., et al., “Hepatocyte-directed Gene Transfer in Vivo Leads to Transient Improvement of Hypercholesterolemia in Low Density Lipoprotein Receptor-deficient Rabbits”, The Journal of Biological Chemistry, 267(2):963-967 (1992).
Wu, G.Y., et al., “Receptor-mediated in Vitro Gene Transformation by a Soluble DNA Carrier System”, The Journal of Biological Chemistry, 262(10):4429-4432 (1987).
Wu, G.Y., et al., “Receptor-mediated Gene Delivery and Expression in Vivo”, The Journal of Biological Chemistry, 263(29):14621-14624 (1988).
Yutsudo, T., et al., “The Gene Encoding a New Mitogenic Factor in a Streptococcus pyogenes Strain Is Distributed Only in Group A Streptococci”, Infection and Immunity, 62(9):4000-4004 (1994).
Zufferey, R., et al., “Self-Inactivating Lentivirus Vector for Safe and Efficient In Vivo Gene Delivery”, Journal of Virology, 72(12):9873-9880 (1998).
Akgun et al., “Ligands that interact with putative MOR-mGluR5 heteromer in mice with inflammatory pain produce potent antinociception”, PNAS vol. 110 (28), 11595-11599 (2013).
Fischer et al., “Increased efficacy of μ-opioid agonist-induced antinociception by metabotropic glutamate receptor antagonists in C57BL/6 mice: comparison with (−)-6-phosphonomethyl-deca-hydroisoquinoline-3-carboxylic acid (LY235959)”, Psychopharmacology, 198, 271-278 (2008).
Gabra et al., “mGluR5 antagonists that block calcium mobilization in vitro also reverse (s)-3,5-DHPG-induced hyperalgesia and morphine antinociceptive tolerance in vivo”, Brain Research 1187, 58-66 (2008).
Lee et al., “Pharmacological Profiles of Oligomerized μ-Opioid Receptors”, Cells 2, 689-714 (2013).
Patent Cooperation Treaty, International Searching Authority, Search Report and Written Opinion for PCT/US2014/015395, 12 pages, Jun. 10, 2014.
Lei et al., “Identification of new candidate vaccine antigens made by Streptococcus pyogenes: purification and characterization of 16 putative extracellular lipoproteins”, The Journal of Infectious Diseases 189, 79-89 (2003).
Maione et al., “Identification of a universal Group B Streptococcus vaccine by multiple genome screen”, Science 309, 148-150 (2005).
Reid et al., “Postgenomic analysis of four novel antigens of Group A Streptococcus: growth phase-dependent gene transcription and human serologic response”, Journal of Bacteriology, 184 (22), 6316-6324 (2002).
Related Publications (1)
Number Date Country
20140037669 A1 Feb 2014 US
Provisional Applications (1)
Number Date Country
61111485 Nov 2008 US
Divisions (1)
Number Date Country
Parent 12612399 Nov 2009 US
Child 13943484 US