US20060165716A1 - Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae - Google Patents

Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae

Info

Publication number
US20060165716A1
US20060165716A1 US11/192,046 US19204605A US2006165716A1 US 20060165716 A1 US20060165716 A1 US 20060165716A1 US 19204605 A US19204605 A US 19204605A US 2006165716 A1 US2006165716 A1 US 2006165716A1
Authority
US
United States
Prior art keywords
gbs
gas
proteins
strain
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/192,046
Inventor
John Telford
Guido Grandi
Peter Lauer
Marirosa Mora
Immaculada Y Ros
Domenico Maione
Guiliano Bensi
Daniela Rinaudo
Vega Masignani
Michelle Barocchi
Rino Rappuoli
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US11/192,046 priority Critical patent/US20060165716A1/en
Assigned to CHIRON CORPORATION reassignment CHIRON CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MASIGNANI, VEGA, MORA, MARIROSA, RAPPUOLI, RINO, TELFORD, JOHN L., Y ROS, IMMACULADA MARGARIT, MAIONE, DOMINICO, GRANDI, GUIDO, BENSI, GIULIANO, RINAUDO, CIRA DANIELA, LAUER, PETER
Assigned to CHIRON CORPORATION reassignment CHIRON CORPORATION RE-RECORD TO CORRECT SEVERAL ASSIGNORS NAMES ON A DOCUMENT PREVIOUSLY RECORDED AT REEL 017376, FRAME 0716. (ASSIGNMENT OF ASSIGNOR'S INTEREST) Assignors: BAROCCHI, MICHELLE, DOMENICO, MAIONE, IMMACULADA, MARGARIT Y ROS, MORA, MARIROSA, RAPPUOLI, RINO, TELFORD, JOHN L., BENSI, GIULIANO, GRANDI, GUIDO, MASIGNANI, VEGA, RINAUDO, CIRA DANIELA, LAUER, PETER
Publication of US20060165716A1 publication Critical patent/US20060165716A1/en
Assigned to NOVARTIS VACCINES AND DIAGNOSTICS, INC. reassignment NOVARTIS VACCINES AND DIAGNOSTICS, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: CHIRON CORPORATION
Priority to US12/906,510 priority patent/US8778358B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/0208Specific bacteria not otherwise provided for
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/05Actinobacteria, e.g. Actinomyces, Streptomyces, Nocardia, Bifidobacterium, Gardnerella, Corynebacterium; Propionibacterium
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/08Clostridium, e.g. Clostridium tetani
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/085Staphylococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/095Neisseria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the identification of adhesin islands within the genome Streptococcus agalactiae (“GBS”) and the use of adhesin island amino acid sequences encoded by these adhesin islands in compositions for the treatment or prevention of GBS infection. Similar sequences have been identified in other Gram positive bacteria.
  • the invention further includes immunogenic compositions comprising adhesin island amino acid sequences of Gram positive bacteria for the treatment or prevention of infection of Gram positive bacteria.
  • Preferred immunogenic compositions of the invention include an adhesin island surface protein which may be formulated or purified in an oligomeric or pilus form.
  • GBS has emerged in the last 20 years as the major cause of neonatal sepsis and meningitis that affects 0.5-3 per 1000 live births, and an important cause of morbidity among older age groups affecting 5-8 per 100,000 of the population.
  • Current disease management strategies rely on intrapartum antibiotics and neonatal monitoring which have reduced neonatal case mortality from >50% in the 1970's to less than 10% in the 1990's. Nevertheless, there is still considerable morbidity and mortality and the management is expensive. 15-35% of pregnant women are asymptomatic carriers and at high risk of transmitting the disease to their babies. Risk of neonatal infection is associated with low serotype specific maternal antibodies and high titers are believed to be protective.
  • invasive GBS disease is increasingly recognized in elderly adults with underlying disease such as diabetes and cancer.
  • the “B” in “GBS” refers to the Lancefield classification, which is based on the antigenicity of a carbohydrate which is soluble in dilute acid and called the C carbohydrate.
  • Lancefield identified 13 types of C carbohydrate, designated A to 0, that could be serologically differentiated.
  • the organisms that most commonly infect humans are found in groups A, B, D, and G.
  • strains can be divided into at least 9 serotypes (Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII) based on the structure of their polysaccharide capsule.
  • serotypes Ia, Ib, II, and III were equally prevalent in normal vaginal carriage and early onset sepsis in newborns.
  • Type V GBS has emerged as an important cause of GBS infection in the USA, however, and strains of types VI and VIII have become prevalent among Japanese women.
  • S. agalactiae is classified as a gram positive bacterium, a collection of about 21 genera of bacteria that colonize humans, have a generally spherical shape, a positive Gram stain reaction and lack endospores.
  • Gram positive bacteria are frequent human pathogens and include Staphylococcus (such as S. aureus ), Streptococcus (such as S. pyogenes (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans ), Enterococcus (such as E. faecalis and E. faecium ), Clostridium (such as C. difficile ), Listeria (such as L. monocytogenes ) and Corynebacterium (such as C. diphtheria ).
  • compositions for providing immunity against disease and/or infection of Gram positive bacteria.
  • the compositions are based on the identification of adhesin islands within Streptococcal genomes and the use of amino acid sequences encoded by these islands in therapeutic or prophylactic compositions.
  • the invention further includes compositions comprising immunogenic adhesin island proteins within other Gram positive bacteria in therapeutic or prophylactic compositions.
  • GBS Adhesin Island 1 a new adhesin island, “GBS Adhesin Island 1”, “AI-1” or “GBS AI-1”, within the genomes of several Group B Streptococcus serotypes and isolates.
  • This adhesin island is thought to encode surface proteins which are important in the bacteria's virulence.
  • surface proteins within GBS Adhesin Islands form a previously unseen pilus structure on the surface of GBS bacteria. Amino acid sequences encoded by such GBS Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of GBS infection.
  • a preferred immunogenic composition of the invention comprises an AI-1 surface protein, such as GBS 80, which may be formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • Electron micrographs depicting some of the first visualizations of this pilus structure in a wild type GBS strain are shown in FIGS. 16, 17 , 49 , and 50 .
  • Applicants have transformed a GBS strain with a plasmid comprising the AI surface protein GBS 80 which resulted in increased production of that AI surface protein.
  • GBS AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“AI-1 proteins”).
  • AI-1 includes polynucleotide sequences encoding for two or more of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648.
  • One or more of the AI-1 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the AI-1 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • AI-1 typically resides on an approximately 16.1 kb transposon-like element frequently inserted into the open reading frame for trmA.
  • One or more of the AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the AI surface proteins of the invention may affect the ability of the GBS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer.
  • one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • AI-1 may encode at least one surface protein.
  • AI-1 may encode at least two surface proteins and at least one sortase.
  • AI-1 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif or other sortase substrate motif.
  • the GBS AI-1 protein of the composition may be selected from the group consisting of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648.
  • GBS AI-1 surface proteins GBS 80 and GBS 104 are preferred for use in the immunogenic compositions of the invention.
  • AI-1 may also include a divergently transcribed transcriptional regulator such as araC (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction). It is believed that araC may regulate the expression of the GBS AI operon. (See Korbel et al., Nature Biotechnology (2004) 22(7): 911-917 for a discussion of divergently transcribed regulators in E. coli ).
  • araC may regulate the expression of the GBS AI operon.
  • a second adhesin island “Adhesin Island-2”, “AI-2” or “GBS AI-2”, has also been identified in numerous GBS serotypes. Amino acid sequences encoded by the open reading frames of AI-2 may also be used in immunogenic compositions for the treatment or prevention of GBS infection.
  • GBS AI-2 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • the GBS AI-2 sequences may be divided into two subgroups.
  • AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406. This collection of open reading frames may be generally referred to as GBS AI-2 subgroup 1.
  • AI-2 may include open reading frames encoding for two or more of 01520, 01521, 01522, 01523, 01523, 01524 and 01525. This collection of open reading frames may be generally referred to as GBS AI-2 subgroup 2.
  • One or more of the AI-2 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the AI-2 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • AI-2 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • AI-2 may encode for at least one surface protein.
  • AI-2 may encode for at least two surface proteins and at least one sortase.
  • AI-2 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • the AI-2 protein of the composition may be selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • AI-2 surface proteins GBS 67, GBS 59, and 01524 are preferred AI-2 proteins for use in the immunogenic compositions of the invention.
  • GBS 67 or GBS 59 is particularly preferred.
  • GBS AI-2 may also include a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB). As in AI-1, rogB is thought to regulate the expression of the AI-2 operon.
  • a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB).
  • rogB is thought to regulate the expression of the AI-2 operon.
  • the GBS AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against GBS infection.
  • the invention may include an immunogenic composition comprising one or more GBS AI-1 proteins and one or more GBS AI-2 proteins.
  • the immunogenic compositions may also be selected to provide protection against an increased range of GBS serotypes and strain isolates.
  • the immunogenic composition may comprise a first and second GBS AI protein, wherein a full length polynucleotide sequence encoding for the first GBS AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second GBS AI protein.
  • each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple GBS serotypes and strain isolates.
  • each antigen is presnt in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) GBS strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5 or more) GBS serotypes.
  • GBS AI-1 Applicants have found that Group B Streptococcus surface exposure of GBS 104 is dependent on the concurrent expression of GBS 80. It is thought that GBS 80 is involved in the transport or localization of GBS 104 to the surface of the bacteria.
  • the two proteins may be oligomerized or otherwise chemically or physically associated. It is possible that this association involves a conformational change in GBS 104 that facilitates its transition to the surface of the GBS bacteria.
  • one or more AI sortases may also be involved in this surface localization and chemical or physical association. Similar relationships are thought to exist within GBS AI-2.
  • the compositions of the invention may therefore include at least two AI proteins, wherein the two AI proteins are physically or chemically associated.
  • the two AI proteins form an oligomer.
  • one or more of the AI proteins are in a hyper-oligomeric form.
  • the associated AI proteins may be purified or isolated from a GBS bacteria or recombinant host cell.
  • compositions for providing prophylactic or therapeutic protection against disease and/or infection of Gram positive bacteria.
  • the compositions are based on the identification of adhesin islands within Streptococcal genomes and the use of amino acid sequences encoded by these islands in therapeutic or prophylactic compositions.
  • the invention further includes compositions comprising immunogenic adhesin island proteins within other Gram positive bacteria in therapeutic or prophylactic compositions.
  • Preferred Gram positive adhesin island proteins for use in the invention may be derived from Staphylococcus (such as S. aureus ), Streptococcus (such as S. agalactiae (GBS), S. pyogenes (GAS), S. pneumonaie, S.
  • the Gram positive adhesin island surface proteins are in oligomeric or hyperologimeric form.
  • adhesin islands within the genomes of several Group A Streptococcus serotypes and isolates. These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fasciitis.
  • post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases.
  • the first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused.
  • the bacteria secretes a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection.
  • the final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart.
  • a general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above.
  • the M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation.
  • the carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci.
  • the amino terminus which extend through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • T-antigen A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen.
  • T-antigen a variable, trypsin-resistant surface antigen
  • Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens.
  • Antisera to define T types is commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • T-antigen T-type 6
  • M6 strain of GAS M6 strain of GAS
  • FCT Fibronectin-binding, Collagen-binding T-antigen
  • the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used.
  • Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms. Immunological recognition of these proteins may allow the host immune response to slow or prevent the bacteria's transition into the more pathogenic later stages of infection.
  • GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix).
  • Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm.
  • Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment is preferable.
  • the invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes.
  • the immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form.
  • the immunogenic compositions of the invention may include one or more GAS AI surface proteins.
  • the invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • Amino acid sequence encoded by such GAS Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • Preferred immunogenic compositions of the invention comprise a GAS AI surface protein which has been formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • GAS Adhesin Islands generally include a series of open reading frames within a GAS genome that encode for a collection of surface proteins and sortases.
  • a GAS Adhesin Island may encode for an amino acid sequence comprising at least one surface protein.
  • the Adhesin Island therefore, may encode at least one surface protein.
  • a GAS Adhesin Island may encode for at least two surface proteins and at least one sortase.
  • a GAS Adhesin Island encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • One or more GAS AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • GAS Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator.
  • the transcriptional regulator may regulate the expression of the GAS AI operon. Examples of transcriptional regulators found in GAS AI sequences include RofA and Nra.
  • the GAS AI surface proteins may bind or otherwise adhere to fibrinogen, fibronectin, or collagen.
  • One or more of the GAS AI surface proteins may comprise a fimbrial structural subunit.
  • One or more of the GAS AI surface proteins may include an LPXTG motif or other sortase substrate motif.
  • the LPXTG motif may be followed by a hydrophobic region and a charged C terminus, which are thought to retard the protein in the cell membrane to facilitate recognition by the membrane-localized sortase. See Barnett, et al., J. Bacteriology (2004) 186 (17): 5865-5875.
  • GAS AI sequences may be generally categorized as Type 1, Type 2, Type 3, or Type 4, depending on the number and type of sortase sequences within the island and the percentage identity of other proteins (with the exception of RofA and cpa) within the island.
  • Schematics of the GAS adhesin islands are set forth in FIG. 51A and FIG. 162 .
  • “GAS Adhesin Island-1 or “GAS AI-1” comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-1 proteins”).
  • GAS AI-1 preferably comprises surface proteins, a srtB sortase and a rofA divergently transcribed transcriptional regulator.
  • GAS AI-1 surface proteins may include a fibronectin binding protein, a collagen adhesion protein and a fimbrial structural subunit.
  • the fimbrial structural subunit also known as tee6
  • the fimbrial structural subunit is thought to form the shaft portion of the pilus like structure
  • the collagen adhesion protein (Cpa) is thought to act as an accessory protein facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule.
  • GAS AI-1 includes polynucleotide sequences encoding for two or more of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161.
  • the GAS AI-1 may also include polynucleotide sequences encoding for any one of CDC SS 410_fimbrial, ISS3650_fimbrial, DSM2071_fimbrial
  • a preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which may be formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • the immunogenic composition of the invention may alternatively comprise an isolated GAS AI-1 surface protein in oligomeric (pilus) form.
  • the oligomer or hyperoligomeric pilus structures comprising GAS AI-1 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-1 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-1 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • GAS AI-1 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-1 may encode for at least one surface protein.
  • GAS AI-1 may encode for at least two surface proteins and at least one sortase.
  • GAS AI-1 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-1 preferably includes a srtB sortase.
  • GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • the GAS AI-1 protein of the composition may be selected from the group consisting of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160 M6_Spy0161, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • GAS AI-1 surface proteins M6_Spy0157 (a fibronectin binding protein), M6_Spy0159 (a collagen adhesion protein, Cpa), M6_Spy0160 (a fimbrial structural subunit, tee6), CDC SS 410_fimbrial (a fimbrial structural subunit), ISS3650_fimbrial (a fimbrial structural subunit), and DSM2071_fimbrial (a fimbrial structural subunit) are preferred GAS AI-1 proteins for use in the immunogenic compositions of the invention.
  • the fimbrial structural subunit tee6 and the collagen adhesion protein Cpa are preferred GAS AI-1 surface proteins.
  • each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine).
  • LPXTG SEQ ID NO: 122
  • LPXSG SEQ ID NO: 1344
  • GAS AI-1 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the GAS AI protein open reading frames, but it transcribed in the opposite direction).
  • rofA a divergently transcribed transcriptional regulator
  • the GAS AI-1 surface proteins may be used alone, in combination with other GAS AI-1 surface proteins or in combination with other GAS AI surface proteins.
  • the immunogenic compositions of the invention include the GAS AI-1 fimbrial structural subunit (tee6) and the GAS AI-1 collagen binding protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-1 fimbrial structural subunit (tee6).
  • GAS Adhesin Island-2 A second GAS adhesion island, “GAS Adhesin Island-2” or “GAS AI-2,” has also been identified in GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-2 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • a preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which may be formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • a preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-2 surface protein in oligomeric (pilus) form.
  • the oligomer or hyperoligomeric pilus structures comprising GAS AI-2 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-2 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-2 proteins”).
  • GAS AI-2 preferably comprises surface proteins, a srtB sortase, a srtC1 sortase and a rofA divergently transcribed transcriptional regulator.
  • GAS AI-2 includes polynucleotide sequences encoding for two or more of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • GAS AI-2 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-2 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • GAS AI-2 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-2 may encode for at least one surface protein.
  • GAS AI-2 may encode for at least two surface proteins and at least one sortase.
  • GAS AI-2 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-2 preferably includes a srtB sortase and a srtC1 sortase.
  • GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • GAS srtC1 sortase may preferentially anchor surface proteins with a V(P/V)PTG (SEQ ID NO: 167) motif.
  • GAS srtC1 may be differentially regulated by rofA.
  • the GAS AI-2 protein of the composition may be selected from the group consisting of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • GAS AI-2 surface proteins GAS15 (Cpa), GAS16 (thought to be a fimbrial protein, M1 — 128), GAS18 (M1_Spy0130), and GAS20 are preferred for use in the immunogenic compositions of the invention.
  • GAS 16 is thought to form the shaft portion of the pilus like structure, while GAS 15 (the collagen adhesion protein Cpa) and GAS 18 are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule.
  • each of these GAS AI-2 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VVXTG (SEQ ID NO: 135), or EVXTG (SEQ ID NO: 136).
  • LPXTG sortase substrate motif such as LPXTG (SEQ ID NO: 122), VVXTG (SEQ ID NO: 135), or EVXTG (SEQ ID NO: 136).
  • GAS AI-2 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the GAS AI protein open reading frames, but it transcribed in the opposite direction).
  • the GAS AI-2 surface proteins may be used alone, in combination with other GAS AI-2 surface proteins or in combination with other GAS AI surface proteins.
  • the immunogenic compositions of the invention include the GAS AI-2 fimbrial protein (GAS 16), the GAS AI-2 collagen binding protein (GAS 15) and GAS 18 (M1_Spy0130). More preferably, the immunogenic compositions of the invention include the GAS AI-2 fimbrial protein (GAS 16).
  • GAS Adhesin Island-3 A third GAS adhesion island, “GAS Adhesin Island-3” or “GAS AI-3,” has also been identified in numerous GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-3 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • a preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which may be formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • a preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-3 surface protein in oligomeric (pilus) form.
  • the oligomer or hyperoligomeric pilus structures comprising GAS AI-3 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-3 proteins”).
  • GAS AI-3 preferably comprises surface proteins, a srtC2 sortase, and a Negative transcriptional regulator (Nra) divergently transcribed transcriptional regulator.
  • GAS AI-3 surface proteins may include a collagen binding protein, a fimbrial protein, and a F2 like fibronectin-binding protein.
  • GAS AI-3 surface proteins may also include a hypothetical surface protein. The fimbrial protein is thought to form the shaft portion of the pilus like structure, while the collagen adhesion protein (Cpa) and the hypothetical surface protein are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule.
  • Preferred AI-3 surface proteins include the fimbrial proein, the collagen binding protein and the hypothetical protein.
  • each of these GAS AI-3 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • LPXTG sortase substrate motif such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-3 includes polynucleotide sequences encoding for two or more of SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, SpyM3 — 0104, Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, Sps0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, spyM18 — 0132, SpyoM0100156, SpyoM0100155, SpyoM0100154, SpyoM01000153, SpyoM01000152, SpyoM01000
  • GAS AI-3 may include open reading frames encoding for two or more of SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, and SpyM3 — 0104.
  • GAS AI-3 may include open reading frames encoding for two or more of Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, and Sps0106.
  • GAS AI-3 may include open reading frames encoding for two or more of orf78, orf79, orf80, orf81, orf82, orf83, and orf84.
  • GAS AI-3 may include open reading frames encoding for two or more of spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, and spyM18 — 0132.
  • GAS AI-3 may include open reading frames encoding for two or more of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149.
  • GAS AI-1 may also include polynucleotide sequences encoding for any one of ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • GAS AI-3 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-3 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • GAS AI-3 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-3 may encode for at least one surface protein.
  • GAS AI-3 may encode for at least two surface proteins and at least one sortase.
  • GAS AI-3 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-3 preferably includes a srtC2 type sortase.
  • GAS srtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • GAS SrtC2 may be differentially regulated by Nra.
  • the GAS AI-3 protein of the composition may be selected from the group consisting of SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, SpyM3 — 0104, Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, Sps0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, spyM18 — 0132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM
  • GAS AI-3 may also include a transcriptional regulator such as Nra.
  • GAS AI-3 may also include a LepA putative signal peptidase I protein.
  • the GAS AI-3 surface proteins may be used alone, in combination with other GAS AI-3 surface proteins or in combination with other GAS AI surface proteins.
  • the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein, the GAS AI-3 collagen binding protein, the GAS AI-3 surface protein (such as SpyM3 — 0102, M3_Sps0104, M5_orf82, or spyM18 — 0130), and fibronectin binding protein PrtF2. More preferably, the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein, the GAS AI-3 collagen binding protein, and the GAS AI-3 surface protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein.
  • GAS AI-3 fimbrial protein examples include SpyM3 — 0100, M3_Sps0102, M5_orf80, spyM18 — 128, SpyoM01000153, ISS3040_fimbrial, ISS3776_fimbrial, ISS4959_fimbrial.
  • GAS AI-3 collagen binding protein Representative examples include SpyM3 — 0098, M3_Sps0100, M5_orf 78, spyM18 — 0126, and SpyoM01000155.
  • GAS AI-3 fibronectin binding protein PrtF2 include SpyM3 — 0104, M3_Sps0106, M5_orf84 and spyM18 — 0132, and SpyoM01000149.
  • GAS Adhesin Island-4 A fourth GAS adhesion island, “GAS Adhesin Island-4” or “GAS AI-4,” has also been identified in GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-4 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • a preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which may be formulated or purified in an oligomeric (pilus) form.
  • the oligomeric form is a hyperoligomer.
  • a preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-4 surface protein in oligomeric (pilus) form.
  • the oligomer or hyperoligomeric pilus structures comprising GAS AI-3 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • the oligomeric or hyperoligomeric pilus structures comprising GAS AI-4 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-4 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-4 proteins”).
  • This GAS adhesin island 4 (“GAS AI-4”) comprises surface proteins, a srtC2 sortase, and a RofA regulatory protein.
  • GAS AI-4 surface proteins within may include a fimbrial protein, F1 and F2 like fibronectin-binding proteins, and a capsular polysaccharide adhesion protein (Cpa).
  • GAS AI-4 surface proteins may also include a hypothetical surface protein in an open reading frame (orf).
  • the fimbral protein (EftLSL) is thought to form the shaft portion of the pilus like structure, while the collagen adhesion protein (Cpa) and the hypothetical protein are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule.
  • each of these GAS AI-4 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-4 includes polynucleotide sequences encoding for two or more of 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, and 19224141.
  • a GAS AI-4 polynucleotide may also include polynucleotide sequences encoding for any one of 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial.
  • One or more of the GAS AI-4 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-4 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • GAS AI-4 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-4 may encode for at least one surface protein.
  • GAS AI-4 may encode for at least two surface proteins and at least one sortase.
  • GAS AI-4 encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-4 includes a SrtC2 type sortase.
  • GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • the GAS AI-4 protein of the composition may be selected from the group consisting of 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • GAS AI-4 surface proteins 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial are preferred proteins for use in the immunogenic compositions of the invention.
  • GAS AI-4 may also include a divergently transcribed transcriptional regulator such as RofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction.
  • RofA a divergently transcribed transcriptional regulator
  • GAS AI-4 may also include a LepA putative signal peptidase I protein and a MsmRL protein.
  • the GAS AI-4 surface proteins may be used alone, in combination with other GAS AI-4 surface proteins or in combination with other GAS AI surface proteins.
  • the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein (EftLSL or 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, or ISS4538_fimbrial), the GAS AI-4 collagen binding protein, the GAS AI-4 surface protein (such as M12 isolate A735 orf 2), and fibronectin binding protein PrtF1 and PrtF2.
  • GAS AI-4 fimbrial protein EftLSL or 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, or ISS4538_f
  • the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein, the GAS AI-4 collagen binding protein, and the GAS AI-4 surface protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein.
  • the GAS AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against GAS infection.
  • the invention may include an immunogenic composition comprising one or more GAS AI-1 proteins and one or more of any of GAS AI-2, GAS AI-3, or GAS AI-4 proteins.
  • the invention includes an immunogenic composition comprising at least two GAS AI proteins where each protein is selected from a different GAS adhesin island.
  • the two GAS AI proteins may be selected from one of the following GAS AI combinations: GAS AI-1 and GAS AI-2; GAS AI-1 and GAS AI-3; GAS AI-1 and GAS AI-4; GAS AI-2 and GAS AI-3; GAS AI-2 and GAS AI-4; and GAS AI 3 and GAS AI-4.
  • the combination includes fimbrial proteins from one or more GAS adhesin islands.
  • the immunogenic compositions may also be selected to provide protection against an increased range of GAS serotypes and strain isolates.
  • the immunogenic composition may comprise a first and second GAS AI protein, wherein a full length polynucleotide sequence encoding for the first GAS AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second GAS AI protein.
  • each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple GAS serotypes and strain isolates.
  • each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) GAS strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) GAS serotypes.
  • Applicants have also identified adhesin islands within the genome of Streptococcus pneumoniae . These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence. Amino acid sequence encoded by such S. pneumoniae Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of S. pneumoniae infection.
  • Preferred immunogenic compositions of the invention comprise a S. pneumoniae AI surface protein which has been formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer.
  • a preferred immunogenic composition of the invention alternatively comprises an isolated S. pneumoniae surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising S. pneumoniae surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • the S. pneumoniae Adhesin Islands generally include a series of open reading frames within a S. pneumoniae genome that encode for a collection of surface proteins and sortases.
  • a S. pneumoniae Adhesin Island may encode for an amino acid sequence comprising at least one surface protein.
  • the S. pneumoniae Adhesin Island may encode for at least two surface proteins and at least one sortase.
  • a S. pneumoniae Adhesin Island encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPTXG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • One or more S. pneumoniae AI surface proteins may participate in the formation of a pilus structure on the surface of the S. pneumoniae bacteria.
  • the S. pneumoniae Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator.
  • the transcriptional regulator may regulate the expression of the S. pneumonaie AI operon.
  • An example of a transcriptional regulator found in S. pneumoniae AI sequences is rlrA.
  • FIG. 137 A schematic of the organization of a S. pneumoniae AI locus is provided in FIG. 137 .
  • the locus comprises open reading frames encoding a transcriptional regulator (rlrA), cell wall surface proteins (rrgA, rrgB, rrgc) and sortases (srt B, srtC, srtD).
  • rlrA transcriptional regulator
  • rrgA cell wall surface proteins
  • srtB, srtC, srtD sortases
  • S. pneumoniae AI sequences may be generally divided into two groups of homology, S. pneuamoniae AI-a and AI-b.
  • S. pneumoniae strains that comprise AI-a include 14 CSR 10, 19A Hungary 6, 23 F Tru 15, 670, 6B Finland 12, and 6B Spain 2.
  • S. pneumoniae AI strains that comprise AI-b include 19F Taiwan 14, 9V Spain 3, 23F Taiwan 15 and TIGR 4.
  • S. pneumoniae AI from TIGR4 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from TIGR4 includes polynucleotide sequences encoding for two or more of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, and SP0468.
  • One or more of the S. pneumoniae AI from TIGR4 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from TIGR4 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae strain 670 AI comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae strain 670 AI includes polynucleotide sequences encoding for two or more of orf1 — 670, orf3 — 670, orf4 — 670, orf5 — 670, orf6 — 670, orf7 — 670, and orf8 — 670.
  • One or more of the S. pneumoniae strain 670 AI polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 670 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 14 CSR10 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”).
  • S. pneumoniae AI proteins include polynucleotide sequences encoding for two or more of ORF2 — 14CSR, ORF3 — 14CSR, ORF4 — 14CSR, ORF5 — 14CSR, ORF6 — 14CSR, ORF7 — 14CSR, and ORF8 — 14CSR.
  • One or more of the S. pneumoniae AI from 14 CSR10 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 14 CSR10 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 19A Hungary 6 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 19A Hungary 6 includes polynucleotide sequences encoding for two or more of ORF2 — 19AH, ORF3 — 1 gAH, ORF4 — 19AH, ORF5 — 19AH, ORF6 — 19AH, ORF7 — 19AH, and ORF8 — 19AH.
  • One or more of the S. pneumoniae AI from 19A Hungary 6 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 19A Hungary 6 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 19F Taiwan 14 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 19F Taiwan 14 includes polynucleotide sequences encoding for two or more of ORF2 — 19FTW, ORF3 — 19FTW, ORF4 — 19FTW, ORF5 — 19FTW, ORF6 — 19FTW, ORF7 — 19FTW, and ORF8 — 19FTW.
  • One or more of the S. pneumoniae AI from 19F Taiwan 14 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 19F Taiwan 14 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 23F Poland 16 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 23F Poland 16 includes polynucleotide sequences encoding for two or more of ORF2 — 23FP, ORF3 — 23FP, ORF4 — 23FP, ORF5 — 23FP, ORF6 — 23FP, ORF7 — 23FP, and ORF8 — 23FP.
  • One or more of the S. pneumoniae AI from 23F Poland 16 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 23F Poland 16 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 23F Taiwan 15 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 23F Taiwan 15 includes polynucleotide sequences encoding for two or more of ORF2 — 23FTW, ORF3 — 23FTW, ORF4 — 23FTW, ORF5 — 23FTW, ORF6 — 23FTW, ORF7 — 23FTW, and ORF8 — 23FTW.
  • One or more of the S. pneumoniae AI from 23F Taiwan 15 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 23F Taiwan 15 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 6B Finland 12 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 6B Finland 12 includes polynucleotide sequences encoding for two or more of ORF2 — 6BF, ORF3 — 6BF, ORF4 — 6BF, ORF5 — 6BF, ORF6 — 6BF, ORF7 — 6BF, and ORF8 — 6BF.
  • One or more of the S. pneumoniae AI from 6B Finland 12 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 6B Finland 12 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 6B Spain 2 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”).
  • S. pneumoniae AI proteins include polynucleotide sequences encoding for two or more of ORF2 — 6BSP, ORF3 — 6BSP, ORF4 — 6BSP, ORF5 — 6BSP, ORF6 — 6BSP, ORF7 — 6BSP, and ORF8 — 6BSP.
  • One or more of the S. pneumoniae AI from 6B Spain 2 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 6B Spain 2 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 9V Spain 3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“ S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 9V Spain 3 includes polynucleotide sequences encoding for two or more of ORF2 — 9VSP, ORF3 — 9VSP, ORF4 — 9VSP, ORF5 — 9VSP, ORF6 — 9VSP, ORF7 — 9VSP, and ORF8 — 9VSP.
  • One or more of the S. pneumoniae AI from 9V Spain 3 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae AI from 9V Spain 3 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae AI may encode for at least one surface protein.
  • the Adhesin Island may encode at least one surface protein.
  • S. pneumoniae AI may encode for at least two surface proteins and at least one sortase.
  • S. pneumoniae AI encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif.
  • the S. pneumoniae AI protein of the composition may be selected from the group consisting of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, SP0468, orf1 — 670, orf5 — 670, orf4 — 670, orf5 — 670, orf6 — 670, orf7 — 670, orf8 — 670, ORF2 — 14CSR, ORF3 — 14CSR, ORF4 — 14CSR, ORF5 — 14CSR, ORF6 — 14CSR, ORF7 — 14CSR, ORF8 — 14CSR, ORF2 — 19AH, ORF3 — 19AH, ORF4 — 19AH, ORF5 — 9AH, ORF6 — 19AH, ORF7 — 19AH, ORF8 — 19AH, ORF2 — 19FTW, ORF3 — 19FTW, ORF4 — 19FTW, ORF5 —
  • S. pneumoniae AI surface proteins are preferred proteins for use in the immunogenic compositions of the invention.
  • the compositions of the invention comprise combinations of two or more S pneumoniae AI surface proteins. Preferably such combinations are selected from two or more of the group consisting of SP0462, SP0463, SP0464, orf3 — 670, orf4 — 670, orf5 — 670, ORF3 — 14CSR, ORF4 — 14CSR, ORF5 — 14CSR, ORF3 — 19AH, ORF4 — 19AH, ORF5 — 19AH, ORF3 — 19FTW, ORF4 — 19FTW, ORF5 — 1 gFTW, ORF3 — 23FP, ORF4 — 23FP, ORF5 — 23FP, ORF3 — 23FTW, ORF4 — 23FTW, ORF5 — 23FTW, ORF3 — 6BF, ORF4 — 6BF, ORF5 — 6BF, ORF5
  • S. pneumoniae AI may also include a transcriptional regulator.
  • the S. pneumoniae AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against S. pneumoniae infection.
  • the invention may include an immunogenic composition comprising one or more S. pneumoniae from TIGR4 AI proteins and one or more S. pneumoniae strain 670 proteins.
  • the immunogenic composition may comprise one or more AI proteins from any one or more of S. pneumoniae strains TIGR4, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 14 CSR 10, 19F Taiwan 14, 23F Taiwan 15, 23F Poland 16, and 670.
  • the immunogenic compositions may also be selected to provide protection against an increased range of S. pneumoniae serotypes and strain isolates.
  • the immunogenic composition may comprise a first and second S. pneumoniae AI protein, wherein a full length polynucleotide sequence encoding for the first S. pneumoniae AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second S. pneumoniae AI protein.
  • each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple S. pneumoniae serotypes and strain isolates.
  • each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) S. pneumoniae strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) S. pneumoniae serotypes.
  • the immunogenic compositions may also be selected to provide protection against an increased range of serotypes and strain isolates of a Gram positive bacteria.
  • the immunogenic composition may comprise a first and second Gram positive bacteria AI protein, wherein a full length polynucleotide sequence encoding for the first Gram positive bacteria AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second Gram positive bacteria AI protein.
  • each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple serotypes and strain isolates of the Gram positive bacteria.
  • each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) Gram positive bacteria strain isolates.
  • each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) Gram positive bacteria serotypes.
  • One or both of the first and second AI proteins may preferably be in oligomeric or hyperoligomeric form.
  • Adhesin island surface proteins from two or more Gram positive bacterial genus or species may be combined to provide an immunogenic composition for prophylactic or therapeutic treatment of disease or infection of two more Gram positive bacterial genus or species.
  • the adhesin island surface proteins may be associated together in an oligomeric or hyperoligomeric structure.
  • the invention comprises adhesin island surface proteins from two or more Streptococcus species.
  • the invention includes a composition comprising a GBS AI surface protein and a GAS adhesin island surface protein.
  • the invention includes a composition comprising a GAS adhesin island surface protein and a S. pneumoniae adhesin island surface protein.
  • One or both of the GAS AI surface protein and the S. pneumoniae AI surface protein may be in oligomeric or hyperoligomeric form.
  • the invention includes a composition comprising a GBS adhesin island surface protein and a S. pneumoniae adhesin island surface protein.
  • the invention comprises an adhesin island surface protein from two or more Gram positive bacterial genus.
  • the invention includes a composition comprising a Streptococcus adhesin island protein and a Corynebacterium adhesin island protein.
  • One or more of the Gram positive bacteria AI surface proteins may be in an oligomeric or hyperoligomeric form.
  • the AI polynucleotides and amino acid sequences of the invention may also be used in diagnostics to identify the presence or absence of GBS (or a Gram positive bacteria) in a biological sample. They may be used to generate antibodies which can be used to identify the presence of absence of an AI protein in a biological sample or in a prophylactic or therapeutic treatment for GBS (or a Gram positive bacterial) infection. Further, the AI polynucleotides and amino acid sequences of the invention may also be used to identify small molecule compounds which inhibit or decrease the virulence associated activity of the AI.
  • FIG. 1 presents a schematic depiction of Adhesin Island 1 (“AI-1”) comprising open reading frames for GBS 80, GBS 52, SAG0647, SAG0648 and GBS 104.
  • AI-1 Adhesin Island 1
  • FIG. 2 illustrates the identification of AI-1 sequences in several GBS serotypes and strain isolates (GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate nem316; GBS serotype II, strain isolate 18RS21; GBS serotype V, strain isolate CJB111; GBS serotype III, strain isolate COH1 and GBS serotype 1a, strain isolate A909).
  • GBS serotype V strain isolate 2603
  • GBS serotype III strain isolate nem316
  • GBS serotype II strain isolate 18RS21
  • GBS serotype V strain isolate CJB111
  • GBS serotype III strain isolate COH1 and GBS serotype 1a, strain isolate A909
  • FIG. 3 presents a schematic depiction of the correlation between AI-1 and the Adhesin Island 2 (“AI-2”) within the GBS serotype V, strain isolate 2603 genome.
  • AI-2 comprises open reading frames for GBS 67, GBS 59, SAG1406, SAG1405 and GBS 150).
  • FIG. 4 illustrates the identification of AI-2 comprising open reading frames encoding for GBS 67, GBS 59, SAG1406, SAG1404 and GBS 150 (or sequences having sequence homology thereto) in several GBS serotypes and strain isolates (GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate NEM316; GBS serotype 1b, strain isolate H36B; GBS serotype V, strain isolate CJB111; GBS serotype II, strain isolate 18RS21; and GBS serotype 1a, strain isolate 515).
  • GBS serotype V strain isolate 2603
  • GBS serotype III strain isolate NEM316
  • GBS serotype 1b strain isolate H36B
  • GBS serotype V strain isolate CJB111
  • GBS serotype II strain isolate 18RS21
  • GBS serotype 1a strain isolate 515
  • FIG. 4 also illustrates the identification of AI-2 comprising open reading frames encoding for 01520 (a sortase), 01521, 01522 (a sortase), 01523 (spb1), 01524 and 01525 (or sequences having sequence homology thereto).
  • FIG. 5 presents data showing that GBS 80 binds to fibronectin and fibrinogen in ELISA.
  • FIG. 6 illustrates that all genes in AI-1 are co-transcribed as an operon.
  • FIG. 7 presents schematic depictions of in-frame deletion mutations within AI-1.
  • FIG. 8 presents FACS data showing that GBS 80 is required for surface localization of GBS 104.
  • FIG. 9 presents FACS data showing that sortases SAG0647 and SAG0648 play a semi-redundant role in surface exposure of GBS 80 and GBS 104.
  • FIG. 10 presents Western Blots of the in-frame deletion mutants probed with anti-GBS80 and anti-GBS 104 antisera.
  • FIG. 11 Electron micrograph of surface exposed pili structures in Streptococcus agalactiae containing GBS 80.
  • FIG. 12 PHD predicted secondary structure of GBS 067.
  • FIGS. 13, 14 and 15 Electron micrograph of surface exposed pili structures of strain isolate COH1 of Streptococcus agalactiae containing a plasmid insert encoding GBS 80.
  • FIGS. 16 and 17 Electron micrograph of surface exposed pili structure of wild type strain isolate COH1 of Streptococcus agalactiae.
  • FIG. 18 Alignment of polynucleotide sequences of AI-1 from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype HI, strain isolates COH1 and NEM316; and serotype 1a, strain isolate A909.
  • FIG. 19 Alignment of polynucleotide sequences of AI-2 from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype 1b, strain isolate H36B; and serotype 1a, strain isolate 515.
  • FIG. 20 Alignment of polynucleotide sequences of AI-2 from serotype V, strain isolate 2603 and serotype III, strain isolate NEM316.
  • FIG. 21 Alignment of polynucleotide sequences of AI-2 from serotype III, strain isolate COH1 and serotype Ia, strain isolate A909.
  • FIG. 22 Alignment of amino acid sequences of AI-1 surface protein GBS 80 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate A909; serotype III, strain isolates COH1 and NEM316.
  • FIG. 23 Alignment of amino acid sequences of AI-1 surface protein GBS 104 from serotype V, strain isolates 2603 and CJB111; serotype III, strain isolates COH1 and NEM316; and serotype II, strain isolate 18RS21.
  • FIG. 24 Alignment of amino acid sequences of AI-2 surface protein GBS 067 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate 515; serotype II, strain isolate 18RS21; serotype Ib, strain isolate H36B; and serotype III, strain isolate NEM316.
  • FIG. 25 Illustrates that GBS closely associates with tight junctions and cross the monolayer of ME180 cervical epithelial cells by a paracellular route.
  • FIG. 26 Illustrates GBS infection of ME180 cells.
  • FIG. 27 Illustrates that GBS 80 recombinant protein does not bind to epithelial cells.
  • FIG. 28 Illustrates that deletion of GBS 80 does not effect the capacity of GBS strain 2603 V/R to adhere and invade ME180 cervical epithelial cells.
  • FIG. 29 Illustrates binding of recombinant GBS 104 protein to epithelial cells.
  • FIG. 30 Illustrates that deletion of GBS 104 in the GBS strain COH1, reduces the capacity of GBS to adhere to ME180 cervical epithelial cells.
  • FIG. 31 Illustrates that GBS 80 knockout mutant strain partially loses the ability to translocate through an epithelial cell monolayer.
  • FIG. 32 Illustrates that deletion of GBS 104, but not GBS 80, reduces the capacity of GBS to invade J774 macrophage-like cell line.
  • FIG. 33 Illustrates that GBS 104 knockout mutant strain translocates through an epithelial monolayer less efficiently than the isogenic wild type.
  • FIG. 34 Negative stained electron micrographs of GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80.
  • FIG. 35 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 10 nm gold particles).
  • FIG. 36 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 10 nm gold particles).
  • FIG. 37 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles).
  • FIG. 38 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 104 antibodies or preimmune sera (visualized with 10 nm gold particles).
  • FIG. 39 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles) and anti-GBS 104 antibodies (visualized with 10 nm gold particles).
  • FIG. 40 Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles) and anti-GBS 104 antibodies (visualized with 10 nm gold particles).
  • FIG. 41 Illustrates that GBS 80 is necessary for polymer formation and GBS104 and sortase SAG0648 are necessary for efficient assembly of pili.
  • FIG. 42 Illustrates that GBS 67 is part of a second pilus and that GBS 80 is polymerized in strain 515.
  • FIG. 43 Illustrates that two macro-molecules are visible in Coh1, one of which is the GBS 80 pilin.
  • FIG. 44 Illustrates pilin assembly.
  • FIG. 45 Illustrates that GBS 52 is a minor component of the GBS pilus.
  • FIG. 46 Illustrates that the pilus is found in the supernatant of a bacterial culture.
  • FIG. 47 Illustrates that the pilus is found in the supernatant of bacterial cultures in all phases.
  • FIG. 48 Illustrates that in Coh1, only the GBS 80 protein and one sortase (sag0647 or sag0648) is required for polymerization.
  • FIG. 49 IEM image of GBS 80 staining of a GBS serotype VIII strain JM9030013 that express pili.
  • FIG. 50 IEM image of GBS 104 staining of a GBS serotype VIII strain JM9030013 that express pili.
  • FIG. 51A Schematic depiction of open reading frames comprising a GAS AI-2 serotype M1 isolate, GAS AI-3 serotype M3, M5, M18, and M49 isolates, a GAS AI-4 serotype M12 isolate, and an GAS AI-1 serotype M6 isolate.
  • FIG. 51B Amino acid alignment of SrtC1-type sortase of a GAS AI-2 serotype M1 isolate, SrtC2-type sortases of serotype M3, M5, M118, and M49 isolates, and a SrtC2-type sortase of a GAS AI-4 serotype M12 isolate.
  • FIG. 52 Amino acid alignment of the capsular polysaccharide adhesion proteins of GAS AI-4 serotype M12 (A735), GAS AI-3 serotype M5 (Manfredo), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI-1 serotype M3, S. pyogenes strain MGAS8232 serotype M3, and GAS AI-2 serotype M1.
  • FIG. 53 Amino acid alignment of F-like fibronectin-binding proteins of GAS AI-4 serotype M12 (A735) and S. pyogenes strain MGAS10394 serotype M6.
  • FIG. 54 Amino acid alignment of F2-like fibronectin-binding proteins of GAS AI-4 serotype M12 (A735), S. pyogenes strain MGAS8232 serotype M3, GAS AI-3 strain M5 (Manfredo), S. pyogenes strain SSI serotype M3, and S. pyogenes strain MGAS315 serotype M3.
  • FIG. 55 Amino acid alignment of fimbrial proteins of GAS AI-4 serotype M12 (A735), GAS AI-3 serotype M5 (Manfredo), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI serotype M3, S. pyogenes strain MGAS8232 serotype M3, and S. pyogenes M1 GAS serotype M1.
  • FIG. 56 Amino acid alignment of hypothetical proteins of GAS AI-4 serotype M12 (A735), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI-1 serotype M3, GAS AI-3 serotype M5 (Manfredo), and S. pyogenes strain MGAS8232 serotype M3.
  • FIG. 57 Results of FASTA homology search for amino acid sequences that align with the collagen adhesion protein of GAS AI-1 serotype M6 (MGAS10394).
  • FIG. 58 Results of FASTA homology search for amino acid sequences that align with the fimbrial structural subunit of GAS AI-1 serotype M6 (MGAS10394).
  • FIG. 59 Results of FASTA homology search for amino acid sequences that align with the hypothetical protein of GAS AI-2 serotype M1 (SF370).
  • FIG. 60 Specifies pilin and E box motifs present in GAS type 3 and 4 adhesin islands.
  • FIG. 61 Illustrates that surface expression of GBS 80 protein on GBS strains COH and JM9130013 correlates with formation of pili structures.
  • Surface expression of GBS 80 was determined by FACS analysis using an antibody that cross-hybridizes with GBS 80. Formation of pili structures was determined by immunogold electron microscopy using gold-labelled anti-GBS 80 antibody.
  • FIG. 62 Illustrates that surface exposure is capsule-dependent for GBS 322 but not for GBS 80.
  • FIG. 63 Illustrates the amino acid sequence identity of GBS 59 proteins in GBS strains.
  • FIG. 64 Western blotting of whole GBS cell extracts with anti-GBS 59 antibodies.
  • FIG. 65 Western blotting of purified GBS 59 and whole GBS cell extracts with anti-GBS 59 antibodies.
  • FIG. 66 FACS analysis of GBS strains CJB111, 7357B, 515 using GBS 59 antiserum.
  • FIG. 67 Illustrates that anti-GBS 59 antibodies are opsonic for CJB111 GBS strain serotype V.
  • FIG. 68 Western blotting of GBS strain JM9130013 total extracts.
  • FIG. 69 Western blotting of GBS stain 515 total extracts shows that GBS 67 and GBS 150 are parts of a pilus.
  • FIG. 70 Western blotting of GBS strain 515 knocked out for GBS 67 expression
  • FIG. 71 FACS analysis of GBS strain 515 and GBS strain 515 knocked out for GBS 67 expression using GBS 67 and GBS 59 antiserum.
  • FIG. 72 Illustrates complementation of GBS 515 knocked out for GBS 67 expression with a construct overexpressing GBS 80.
  • FIG. 73 FACS analysis of GAS serotype M6 for spyM6 — 0159 surface expression.
  • FIG. 74 FACS analysis of GAS serotype M6 for spyM6 — 0160 surface expression.
  • FIG. 75 FACS analysis of GAS serotype M1 for GAS 15 surface expression.
  • FIG. 76 FACS analysis of GAS serotype M1 for GAS 16 surface expression using a first anti-GAS 16 antiserum.
  • FIG. 77 FACS analysis of GAS serotype M1 for GAS 18 surface expression using a first anti-GAS 18 antiserum.
  • FIG. 78 FACS analysis of GAS serotype M1 for GAS 18 surface expression using a second anti-GAS 18 antiserum.
  • FIG. 79 FACS analysis of GAS serotype M1 for GAS 16 surface expression using a second anti-GAS 16 antisera.
  • FIG. 80 FACS analysis of GAS serotype M3 for spyM3 — 0098 surface expression.
  • FIG. 81 FACS analysis of GAS serotype M3 for spyM3 — 0100 surface expression.
  • FIG. 82 FACS analysis of GAS serotype M3 for spyM3 — 0102 surface expression.
  • FIG. 83 FACS analysis of GAS serotype M3 for spyM3 — 0104 surface expression.
  • FIG. 84 FACS analysis of GAS serotype M3 for spyM3 — 0106 surface expression.
  • FIG. 85 FACS analysis of GAS serotype M12 for 19224134 surface expression.
  • FIG. 86 FACS analysis of GAS serotype M12 for 19224135 surface expression.
  • FIG. 87 FACS analysis of GAS serotype M12 for 19224137 surface expression.
  • FIG. 88 FACS analysis of GAS serotype M12 for 19224141 surface expression.
  • FIG. 89 Western blot analysis of GAS 15 expression on GAS M1 bacteria.
  • FIG. 90 Western blot analysis of GAS 15 expression using GAS 15 immune sera.
  • FIG. 91 Western blot analysis of GAS 15 expression using GAS 15 pre-immune sera.
  • FIG. 92 Western blot analysis of GAS 16 expression on GAS M1 bacteria.
  • FIG. 93 Western blot analysis of GAS 16 expression using GAS 16 immune sera.
  • FIG. 94 Western blot analysis of GAS 16 expression using GAS 16 pre-immune sera.
  • FIG. 95 Western blot analysis of GAS 18 on GAS M1 bacteria.
  • FIG. 96 Western blot analysis of GAS 18 using GAS 18 immune sera.
  • FIG. 97 Western blot analysis of GAS 18 using GAS 18 pre-immune sera.
  • FIG. 98 Western blot analysis of M6_Spy0159 expression on GAS bacteria.
  • FIG. 99 Western blot analysis of 19224135 expression on M12 GAS bacteria.
  • FIG. 100 Western blot analysis of 19224137 expression on M12 GAS bacteria.
  • FIG. 101 Full length nucleotide sequence of an S. pneumoniae strain 670 ⁇ l.
  • FIG. 102 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2580.
  • FIG. 103 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2913.
  • FIG. 104 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 3280.
  • FIG. 105 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 3348.
  • FIG. 106 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2719.
  • FIG. 107 Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain SF370.
  • FIG. 108 Western blot analysis of 19224135 and 19224137 in GAS M12 strain 2728.
  • FIG. 109 Western blot analysis of 19224139 in GAS M12 strain 2728 using antisera raised against SpyM3 — 0102.
  • FIG. 110 Western blot analysis of M6_Spy0159 and M6_Spy0160 in GAS M6 strain 2724.
  • FIG. 111 Western blot analysis of M6 Spy0159 and M6 Spy0160 in GAS M6 strain SF370.
  • FIG. 112 Western blot analysis of M6_Spy160 in GAS M6 strain 2724.
  • FIGS. 113 - 115 Electron micrographs of surface exposed GAS 15 on GAS M1 strain SF370.
  • FIGS. 116 - 121 Electron micrographs of surface exposed GAS 16 on GAS M1 strain SF370.
  • FIGS. 122 - 125 Electron micrographs of surface exposed GAS 18 on GAS M1 strain SF370 detected using anti-GAS 18 antisera.
  • FIG. 126 IEM image of a hyperoligomer on GAS M1 strain SF370 detected using anti-GAS 18 antisera.
  • FIGS. 127 - 132 IEM images of oligomeric and hyperoligomeric structures containing M6_Spy0160 extending from the surface of GAS serotype M6 3650.
  • FIGS. 133A and B Western blot analysis of L. lactis transformed to express GBS 80 with anti-GBS 80 antiserum.
  • FIG. 134 Western blot analyses of L. lactis transformed to express GBS AI-1 with anti-GBS 80 antiserum.
  • FIG. 135 Ponceau staining of same acrylamide gel as used in FIG. 134 .
  • FIG. 136A Western blot analysis of sonicated pellets and supernatants of cultured L. lactis transformed to express GBS AI-1 polypeptides using anti-GBS 80 antiserum.
  • FIG. 136B Polyacrylamide gel electrophoresis of sonicated pellets and supernatants of cultured L. lactis transformed to express GBS AI polypeptides.
  • FIG. 137 Depiction of an example S. pneumoniae AI locus.
  • FIG. 138 Schematic of primer hybridization sites within the S. pneumoniae AI locus of FIG. 137 .
  • FIG. 139A The set of amplicons produced from the S. pneumoniae strain TIGR4 AI locus.
  • FIG. 139B Base pair lengths of amplicons produced from FIG. 139A primers in S. pneumoniae strain TIGR4.
  • FIG. 140 CGH analysis of S. pneumoniae strains for the AI locus.
  • FIG. 141 Amino acid sequence alignment of polypeptides encoded by AI orf 2 in S. pneumoniae AI-positive strains.
  • FIG. 142 Amino acid sequence alignment of polypeptides encoded by AI orf 3 in S. pneumoniae AI-positive strains.
  • FIG. 143 Amino acid sequence alignment of polypeptides encoded by AI orf 4 in S. pneumoniae AI-positive strains.
  • FIG. 144 Amino acid sequence alignment of polypeptides encoded by AI orf 5 in S. pneumoniae AI-positive strains.
  • FIG. 145 Amino acid sequence alignment of polypeptides encoded by AI orf 6 in S. pneumoniae AI-positive strains.
  • FIG. 146 Amino acid sequence alignment of polypeptides encoded by AI orf 7 in S. pneumoniae AI-positive strains.
  • FIG. 147 Amino acid sequence alignment of polypeptides encoded by AI orf 8 in S. pneumoniae AI-positive strains.
  • FIG. 148 Diagram comparing amino acid sequences of RrgA in S. pneumoniae strains.
  • FIG. 149 Amino acid sequence comparison of RrgB S. pneumoniae strains.
  • FIG. 150A Sp0462 amino acid sequence.
  • FIG. 150B Primers used to produce a clone encoding the Sp0462 polypeptide.
  • FIG. 151A Schematic depiction of recombinant Sp0462 polypeptide.
  • FIG. 151B Schematic depiction of full-length Sp0462 polypeptide.
  • FIG. 152A Western blot probed with serum obtained from S. pneumoniae -infected patients for Sp0462.
  • FIG. 152B Western blot probed with GBS 80 serum for Sp0462.
  • FIG. 153A Sp0463 amino acid sequence.
  • FIG. 153B Primers used to produce a clone encoding the Sp0463 polypeptide.
  • FIG. 154A Schematic depiction of recombinant Sp0463 polypeptide.
  • FIG. 154B Schematic depiction of full-length Sp0463 polypeptide.
  • FIG. 155 Western blot detection of recombinant Sp0463 polypeptide.
  • FIG. 156 Western blot detection of high molecular weight Sp0463 polymers.
  • FIG. 157A Sp0464 amino acid sequence.
  • FIG. 157B Primers used to produce a clone encoding the Sp0464 polypeptide.
  • FIG. 158A Schematic depiction of recombinant Sp0464 polypeptide.
  • FIG. 158B Schematic depiction of full-length Sp0464 polypeptide.
  • FIG. 159 Western blot detection of recombinant Sp0464 polypeptide.
  • FIG. 160 Amplification products prepared for production of Sp0462, Sp0463, and Sp0464 clones.
  • FIG. 161 Opsonic killing by anti-sera raised against L. lactis expressing GBS AI
  • FIG. 162 Schematic depicting GAS adhesin islands GAS AI-1, GAS AI-2, GAS AI-3 and GAS AI-4.
  • FIGS. 163 A-D Immunoblots of cell-wall fractions of GAS strains with antisera specific for LPXTG proteins of M6_ISS3650 (A), M1_SF370 (B), M5_ISS4883 (C) and M12 — 20010296 (D).
  • FIGS. 163 E-H Immunoblots of cell-wall fractions of deletion mutants M1_SF370 ⁇ 128 (E) M1_SF370 ⁇ 130 (F) M1_SF370 ⁇ SrtC1 (G) and the M1 — 128 deletion strain complemented with plasmid pAM::128 which contains the M1 — 128 gene (H) with antisera specific for the pilin components of M1_SF370.
  • FIGS. 163 I-N Immunogold labelling and transmission electron microscopy of: T6 (I) and Cpa (J) in M6_ISS3650; M1 — 128 in M1_SF370 (K) and deletion strain M1_SF370 ⁇ 128 (N); M5_orf80 in M5_ISS4883 (L); M12_EftLSL.A in M12 — 20010296 (M).
  • FIG. 164 Schematic representation of the FCT region from 7 GAS strains
  • FIGS. 165 A-H Flow cytometry of GAS bacteria treated or not with trypsin and stained with sera specific for the major pilus component. Preimmune staining; black lines, untreated bacteria; green lines and trypsin treated bacteria; blue lines.
  • FIGS. 166 A-C Immunoblots of recombinant pilin components with polyvalent Lancefield T-typing sera. The recombinant proteins are shown above the blot and the sera pool used is shown below the blot.
  • FIGS. 166 D-G Immunoblots of pilin proteins with monovalent T-typing sera. The recombinant proteins are shown below the blot and the sera used above the blot.
  • FIG. 166 H and I Flow cytometry analysis of strain M1_SF370 (H) and the deletion strain M1_SF370 ⁇ 128 (I) with T-typing antisera pool T.
  • FIG. 167 Chart describing the number and type of sortase sequences identified within GAS AIs.
  • FIG. 168 A Immunogold-electronmicroscopy of L. lactis lacking an expression construct for GBS AI-1 using anti-GBS 80 antibodies.
  • FIG. 168 B and C Immunogold-electronmicroscopy detects GBS 80 in oligomeric (pilus) structures on surface of L. lactis transformed to express GBS AI-1
  • FIG. 169 FACS analysis detects expression of GBS 80 and GBS 104 on the surface of L. lactis transformed to express GBS AI-1.
  • FIG. 170 Phase contrast microscopy and immuno-electronmicroscopy shows that expression of GBS AI-1 in L. lactis induces L. lactis aggregation.
  • FIG. 171 Purification of GBS pili from L. lactis transformed to express GBS AI-1.
  • FIG. 172 Schematic depiction of GAS M6 (AI-1), M1 (AI-2), and M12 (AI-4) adhesin islands and portions of the adhesin islands inserted in the pAM401 construct for expression in L. lactis.
  • FIG. 173 A-C Western blot analysis showing assembly of GAS pili in L. lactis expressing GAS AI-2 (M1) (A), GAS AI-4 (M12) (B), and GAS AI-1(M6) (C).
  • FIG. 174 FACS analysis of GAS serotype M6 for M6_Spy0157 surface expression.
  • FIG. 175 FACS analysis of GAS serotype M12 for 19224139 surface expression.
  • FIG. 176 A-E Immunogold electron microscopy using antibodies against M6_Spy0160 detects pili on the surface of M6 strain 2724.
  • FIG. 176 F Immunogold electron microscopy using antibodies against M6_Spy0159 detects M6_Spy0159 surface expression on M6 strain 2724.
  • FIG. 177 A-C Western blot analysis of M1 strain SF370 GAS bacteria individually deleted for M1 — 130, SrtC1, or M1 — 128 using anti-M1 — 130 serum (A), anti-M1 — 128 serum (B), and anti-M1 — 126 serum (C).
  • FIG. 178 A-C Immunogold electron microscopy using antibodies against M1 — 128 to detect surface expression on wildtype strain SF370 bacteria (A), M1 — 128 deleted SF370 bacteria (B), and SrtC1 deleted SF370 bacteria (C).
  • FIG. 179 A-C FACS analysis to detect expression of M1 — 1126 (A), M1 — 128 (B), and M1 — 130 (C) on the surface of wildtype SF370 GAS bacteria.
  • FIG. 179 D-F FACS analysis to detect expression of M1 — 126 (D), M1 — 128 (E), and M1 — 130 (F) on the surface of M1 — 128 deleted SF370 GAS bacteria.
  • FIG. 179 G-I FACS analysis to detect expression of M1 — 126 (G), M1 — 128 (H), and M1 — 130 (I) on the surface of SrtC1 deleted SF370 GAS bacteria.
  • FIG. 180 A and B FACS analysis of wildtype (A) and LepA deletion mutant (B) strains of SF370 bacteria for M1 surface expression.
  • FIG. 181 Western blot analysis detects high molecular weight polymers in S. pneumoniae TIGR4 using anti-RrgB antisera.
  • FIG. 182 Detection of high molecular weight polymers in S. pnuemoniae rlrA positive strains.
  • FIG. 183 Detection of high molecular weight polymers in S. pneumoniae TIGR4 by silver staining and Western blot analysis using anti-RrgB antisera.
  • FIG. 184 Deletion of S. pneumoniae TIGR4 adhesin island sequences interferes with the ability of S. pneumoniae to adhere to A549 alveolar cells.
  • FIG. 185 Negative staining of S. pneumoniae strain TIGR4 showing abundant pili on the bacterial surface.
  • FIG. 186 Negative staining of strain TIGR4 deleted for rrgA-srtD adhesin island sequences showing no pili on the bacterial surface
  • FIG. 187 Negative staining of the TIGR4 mgrA mutant showing abundant pili on the bacterial surface.
  • FIG. 188 Negative staining of the negative control TIGR4 mgrA mutant deleted for adhesin island sequences rrgA-srtD showing no pili on the bacterial surface.
  • FIG. 189 Immuno-gold labelling of S. pneumoniae strain TIGR4 grown on blood agar solid medium using ⁇ -RrgB (5 nm) and ⁇ -RrgC (10 nm). Bar represents 200 nm.
  • FIG. 190 A and B Detection of expression and purification of S. pneumoniae RrgA protein by SDS-PAGE (A) and Western blot analysis (B).
  • FIG. 191 Detection of RrgB by antibodies produced in mice.
  • FIG. 192 Detection of RrgC by antibodies produced in mice.
  • FIG. 193 Purification of S. pneumoniae TIGR 4 pili by a cultivation and digestion method and detection of the purified TIGR4 pili.
  • FIG. 194 Purification of S. pneumoniae TIGR 4 pili by a sucrose gradient centrifugation method and detection of the purified TIGR4 pili.
  • FIG. 195 Purification of S. pneumoniae TIGR 4 pili by a gel filtration method and detection of the purified TIGR4 pili.
  • FIG. 196 Alignment of full length S. pneumoniae adhesin island sequences from ten S. pneumoniae strains.
  • FIG. 197 A Schematic of GBS AI-1 coding sequences.
  • FIG. 197 B Nucleotide sequence of intergenic region between AraC and GBS 80 (SEQ ID NO: 273.
  • FIG. 197 C FACS analysis results for GBS 80 expression in GBS strains having different length polyA tracts in the intergenic region between AraC and GBS 80.
  • FIG. 198 Table comparing the percent identity of surface proteins encoded by a serotype M6 (harbouring a GAS AI-1) adhesin island relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 199 Table comparing the percent identity of surface proteins encoded by a serotype M1 (harbouring a GAS AI-2) adhesin island relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 200 Table comparing the percent identity of surface proteins encoded by serotypes M3, M18, M5, and M49 (harbouring GAS AI-3) adhesin islands relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 201 Table comparing the percent identity of surface proteins encoded by a serotype M12 (harbouring a GAS AI-1) adhesin island-relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 202 GBS 80 recombinant protein does not bind to epithelial cells.
  • FIG. 203 Deletion of GBS 80 protein does not affect the ability of GBS to adhere and invade ME180 cervical epithelial cells.
  • FIG. 204 GBS 80 binds to extracellular matrix proteins.
  • FIG. 205 Deletion of GBS 104 protein, but not GBS 80, reduces the capacity of GBS to invade J774 macrophage-like cells
  • FIG. 206 GBS 104 knockout mutant strains of bacteria translocate through an epithelial monolayer less efficiently that the isogenic wild type strain.
  • FIG. 207 GBS 80 knockout mutant strains of bacteria partially lose the ability to translocate through an epithelial monolayer.
  • FIG. 208 GBS adherence to HUVEC endothelial cells.
  • FIG. 209 Strain growth rate of wildtype, GBS 80-deleted, or GBS 104 deleted COH1 GBS.
  • FIG. 210 Binding of recombinant GBS 104 protein to epithelial cells by FACS analysis.
  • FIG. 211 Deletion of GBS 104 protein in the GBS strain COH1 reduces the ability of GBS to adhere to ME180 cervical epithelial cells.
  • FIG. 212 COH1 strain GBS overexpressing GBS 80 protein has an impaired capacity to translocate through an epithelial monolayer.
  • FIG. 213 Scanning electron microscopy shows that overexpression of GBS 80 protein on COH1 strain GBS enhances the capacity of the COH1 bacteria to form microcolonies on epithelial cells.
  • FIG. 214 Confocal imaging shows that overexpression of GBS 80 proteins on COH1 strain GBS enhances the capacity of the COH1 bacteria to form microcolonies on epithelial cells.
  • FIG. 215 Detection of GBS 59 on the surface of GBS strain 515 by immuno-electron microscopy.
  • FIG. 216 Detection of GBS 67 on the surface of GBS strain 515 by immuno-electron microscopy.
  • FIG. 217 GBS 67 binds to fibronectin.
  • FIG. 218 Western blot analysis shows that deletion of both GBS AI-2 sortase genes abolishes assembly of the pilus.
  • FIG. 219 FACS analysis shows that deletion of both GBS AI-2 sortase genes abolishes assembly of the pilus.
  • FIG. 220 A-C Western blot analysis shows that GBS 59, GBS 67, and GBS 150 form high molecular weight complexes.
  • FIG. 221 A-C Western blot analysis shows that GBS 59 is required for polymer formation of GBS 67 and GBS 150.
  • FIG. 222 FACS analysis shows that GBS 59 is required for surface exposure of GBS 67.
  • FIG. 223 Summary Western blots for detection of GBS 59, GBS 67, or GBS 150 in GBS 515 and GBS 515 mutant strain.
  • FIG. 224 Description of GBS 59 Allelic variants.
  • FIG. 225 GBS 59 is opsonic only against a strain of GBS expressing a homologous GBS 59.
  • FIG. 226 A and B Results of FACS analysis for surface expression of GBS 59 using antibodies specific for different GBS 59 isoforms.
  • FIG. 227 A and B Results of FACS analysis for surface expression of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 on 41 various strains of GBS bacteria.
  • FIG. 228 Results of FACS analysis for surface expression of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 on 41 strains of GBS bacteria obtained from the CDC.
  • FIG. 229 Expected immunogenicity coverage of different combinations of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 across strains of GBS bacteria.
  • FIG. 230 GBS 59 opsonophagocytic activity is comparable to that of a mixture of GBS 80, GBS 104, GBS 322 and GBS 67.
  • FIG. 231 A-C Schematic presentation of example hybrid GBS AIs.
  • FIG. 232 Schematic presentation of an example hybrid GBS AI.
  • FIG. 233 A and B Western blot and FACS analysis detect expression of GBS 80 and GBS 67 on the surface of L. lactis transformed with a hybrid GBS AI.
  • FIG. 234 A-E Hybrid GBS AI cloning strategy.
  • FIG. 235 High magnification of S. pneumoniae strain TIGR4 pili double labeled with ⁇ -RrgB (5 nm) and ⁇ -RrgC (10 nm). Bar represents 100 nm.
  • FIG. 236 Immuno-gold labeling of the S. pneumoniae TIGR4 rrgA-srtD deletion mutant with no visible pili on the surface detectable by ⁇ -RrgB- and ⁇ -RrgC. Bar represents 200 nm.
  • FIG. 237 Variability in GBS 67 amino acid sequences between strains 2603 and H36B.
  • FIG. 238 Strain variability in GBS 67 amino acid sequences of allele I (2603).
  • FIG. 239 Stran variability in GBS 67 amino acid sequence of allele II (H36B).
  • an “Adhesin Island” or “AI” refers to a series of open reading frames within a bacterial genome, such as the genome for Group A or Group B Streptococcus or other gram positive bacteria, that encodes for a collection of surface proteins and sortases.
  • An Adhesin Island may encode for amino acid sequences comprising at least one surface protein.
  • the Adhesin Island may encode at least one surface protein.
  • an Adhesin Island may encode for at least two surface proteins and at least one sortase.
  • an Adhesin Island encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • One or more AI surface proteins may participate in the formation of a pilus structure on the surface of the gram positive bacteria.
  • Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • the transcriptional regulator may regulate the expression of the AI operon.
  • AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“AI-1 proteins”). Specifically, AI-1 includes open reading frames encoding for two or more (i.e., 2, 3, 4 or 5) of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648.
  • One or more of the AI-1 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the AI-1 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • AI-1 typically resides on an approximately 16.1 kb transposon-like element frequently inserted into the open reading frame for trmA.
  • One or more of the AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) motif or other sortase substrate motif.
  • the AI surface proteins of the invention may affect the ability of the GBS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer.
  • one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • AI-1 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • AI-1 may encode for at least one surface protein.
  • AI-1 may encode for at least two surface exposed proteins and at least one sortase.
  • AI-1 encodes for at least three surface exposed proteins and at least two sortases.
  • the AI-1 protein preferably includes GBS 80 or a fragment thereof or a sequence having sequence identity thereto.
  • an LPXTG motif represents an amino acid sequence comprising at least five amino acid residues.
  • the motif includes a leucine (L) in the first amino acid position, a proline (P) in the second amino acid position, a threonine (T) in the fourth amino acid position and a glycine (G) in the fifth amino acid position.
  • the third position, represented by X may be occupied by any amino acid residue.
  • the X is occupied by lysine (K), Glutamate (E), Asparagine (N), Glutamine (Q) or Alanine (A).
  • the X position is occupied by lysine (K).
  • one of the assigned LPXTG amino acid positions is replaced with another amino acid.
  • such replacements comprise conservative amino acid replacements, meaning that the replaced amino acid residue has similar physiological properties to the removed amino acid residue.
  • Genetically encoded amino acids may be divided into four families based on physiological properties: (1) acidic (asparatate and glutamate), (2) basic (lysine, arginine, histitidine), (3) non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophane) and (4) uncharged polar (glycine, asparagines, glutamine, cysteine, serine, threonine, and tyrosine).
  • Phenylalanine, tryptophan and tyrosine are sometimes classified jointly as aromatic amino acids. For example, it is reasonably predictable that an isolated replacement of a leucine with an isoleucine or valine, an asparate with a glutamate, a threonine with a serine, or a similar conservative replacement of an amino acid with a structurally related amino acid will not have a major effect on the biological activity.
  • the first amino acid position of the LPXTG motif may be replaced with another amino acid residue.
  • the first amino acid residue (leucine) is replaced with an alanine (A), valine (V), isoleucine (I), proline (P), phenylalanine (F), methionine (M), glutamic acid (E), glutamine (Q), or tryptophan (Y) residue.
  • the first amino acid residue is replaced with an isoleucine (I).
  • the second amino acid residue of the LPXTG motif may be replaced with another amino acid residue.
  • the second amino acid residue praline (P) is replaced with a valine (V) residue.
  • the fourth amino acid residue of the LPXTG motif may be replaced with another amino acid residue.
  • the fourth amino acid residue (threonine) is replaced with a serine (S) or an alanine (A).
  • an LPXTG motif may be represented by the amino acid sequence XXXXG, in which X at amino acid position 1 is an L, a V, an E, an I, an F, or a Q; X at amino acid position 2 is a P if X at amino acid position 1 is an L, an I, or an F; X at amino acid position 2 is a V if X at amino acid position 1 is a E or a Q; X at amino acid position 2 is a V or a P if X at amino acid position 1 is a V; X at amino acid position 3 is any amino acid residue; X at amino acid position 4 is a T if X at amino acid position 1 is a V, E, I, F, or Q; and X at amino acid position 4 is a T, S, or A if X at amino acid position 1 is an L.
  • the LPXTG motif of a GBS AI protein may be represented by the amino acid sequence XPXTG, in which X at amino acid position 1 is L, I, or F, and X at amino acid position 3 is any amino acid residue.
  • Specific examples of LPXTG motifs in GBS AI proteins may include LPXTG (SEQ ID NO: 122) or IPXTG (SEQ ID NO: 133).
  • the threonine in the fourth amino acid position of the LPXTG motif may be involved in the formation of a bond between the LPXTG containing protein and a cell wall precursor. Accordingly, in preferred LPXTG motifs, the threonine in the fourth amino acid position is not replaced with another amino acid or, if the threonine is replaced, the replacement amino acid is preferably a conservative amino acid replacement, such as serine.
  • the AI surface proteins of the invention may contain alternative sortase substrate motifs such as NPQTN (SEQ ID NO: 142), NPKTN (SEQ ID NO: 168), NPQTG (SEQ ID NO: 169), NPKTG (SEQ ID NO: 170), XPXTGG (SEQ ID NO: 143), LPXTAX (SEQ ID NO: 144), or LAXTGX (SEQ ID NO: 145). (Similar conservative amino acid substitutions can also be made to these membrane motifs).
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the AI surface proteins may be polymerized into pili by sortase-catalysed transpeptidation. (See FIG. 44 .) Cleavage of AI surface proteins by sortase between the threonine and glycine residues of an LPXTG motif yields a thioester-linked acyl intermediate of sortase.
  • Many AI surface proteins include a pilin motif amino acid sequence which interacts with the sortase and LPXTG amino acid sequence. The first lysine residue in a pilin motif can serve as an amino group acceptor of the cleaved LPXTG motif and thereby provide a covalent linkage between AI subunits to form pili.
  • the pilin motif can make a nucleophilic attack on the acyl enzyme providing a covalent linkage between AI subunits to form pili and regenerate the sortase enzyme.
  • pilin motifs may include ((YPKN(X 10 )K; SEQ ID NO: 146), (YPKN(X 9 )K; SEQ ID NO: 147), (YPK(X 7 )K; SEQ ID NO: 148), (YPK(X 11 )K; SEQ ID NO: 149), or (PKN(X 9 )K; SEQ ID NO: 150)).
  • the AI surface proteins of the invention include a pilin motif amino acid sequence.
  • AI surface proteins of the invention will contain an N-terminal leader or secretion signal to facilitate translocation of the surface protein across the bacterial membrane.
  • Group B Streptococci are known to colonize the urinary tract, the lower gastrointestinal tract and the upper respiratory tract in humans.
  • Electron micrograph images of GBS infection of a cervical epithelial cell line (ME180) are presented in FIG. 25 . As shown in these images, the bacteria closely associate with tight junctions between the cells and appear to cross the monolayer by a paracellular route. Similar paracellular invasion of ME180 cells is also shown in the contrast images in FIG. 26 .
  • the AI surface proteins of the invention may effect the ability of the GBS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI-1 surface protein GBS 104 can bind epithelial cells such as ME180 human cervical cells, A549 human lung cells and Caco2 human intestinal cells (See FIGS. 29 and 210 ). Further, deletion of the GBS 104 sequence in a GBS strain reduces the capacity of GBS to adhere to ME180 cervical epithelial cells. (See FIGS. 30 and 211 ). Deletion of GBS 104 also reduces the capacity of GBS to invade J774 macrophage-like cells. (See FIGS. 32 and 205 ). Deletion of GBS 104 also causes GBS to translocate through epithelial monolayers less efficiently. See FIG. 206 . GBS 104 protein therefore appears to bind to ME180 epithelial cells and to have a role in adhesion to epithelial cells and macrophage cell lines.
  • GBS 80 knockout mutant strains also partially lose the ability to translocate through an epithelial monolayer. See FIG. 207 . Deletion of either GBS 80 or GBS 104 in COH1 cells diminishes adherence to HUVEC endothelial cells. See FIG. 208 . Deletion of GBS 80 or GBS 104 in COH1 does not, however, affect growth of COH1 either with ME180 cells or in incubation medium (IM). See FIG. 209 . Both GBS 80 and GBS 104, therefore, appear to be involved in translocation of GBS through epithelial cells.
  • IM incubation medium
  • GBS 80 does not appear to bind to epithelial cells. Incubation of epithelial cells in the presence of GBS 80 protein followed by FACS analysis using an anti-GBS 80 polyclonal antibody did not detect GBS 80 binding to the epithelial cells. See FIG. 202 . Furthermore, deletion of GBS 80 protein does not affect the ability of GBS to adhere and invade ME180 cervical epithelial cells. See FIG. 203
  • one or more of the surface proteins may bind to one or more extracellular matrix (ECM) binding proteins, such as fibrinogen, fibronectin, or collagen.
  • ECM extracellular matrix
  • GBS 80 one of the AI-1 surface proteins, can bind to the extracellular matrix binding proteins fibronectin and fibrinogen. While GBS 80 protein apparently does not bind to certain epithelial cells or affect the capacity of a GBS bacteria to adhere to or invade cervical epithelial cells (See FIGS. 27 and 28 ), removal of GBS 80 from a wild type strain decreases the ability of that strain to translocate through an epithelial cell layer (see FIG. 31 ).
  • GBS 80 may also be involved in formation of biofilms.
  • COH1 bacteria overexpressing GBS 80 protein have an impaired ability to translocate through an epithelial monolayer. See FIG. 212 . These COH1 bacteria overexpressing GBS 80 form microcolonies on epithelial cells. See FIGS. 213 and 214 . These microcolonies may be the initiation of biofilm development.
  • AI Surface proteins may also demonstrate functional homology to previously identified adhesion proteins or extracellular matrix (ECM) binding proteins.
  • GBS 80 a surface protein in AI-1, exhibits some functional homology to FimA, a major fimbrial subunit of a Gram positive bacteria A. naeslundii .
  • FimA is thought to be involved in binding salivary proteins and may be a component in a fimbrae on the surface of A. naeslundii . See Yeung et al. (1997) Infection & Immunity 65:2629-2639; Yeunge et al (1998) J. Bacteriol 66:1482-1491; Yeung et al. (1988) J. Bacteriol 170:3803-3809; and Li et al. (2001) Infection & Immunity 69:7224-7233.
  • diphtheriae pilus proteins and is thought to be involved in sortase catalized oligomerization of the subunits involved in the C. diphtheriae pilus structure.
  • the C. diphtheriae pilin subunit SpaA is thought to occur by sortase-catalyzed amide bond cross-linking of adjacent pilin subunits.
  • the conserved lysine within the SpaA pilin motif might function as an amino group acceptor of cleaved sorting signals, thereby providing for covalent linkages of the C. diphtheria pilin subunits. See FIG. 6 ( d ) of Ton-That et al., Molecular Microbiology (2003) 50(4): 1429-1438.)
  • E box comprising a conserved glutamic acid residue has also been identified in the C. diphtheria pilin associated proteins as important in C. diphtheria pilin assembly.
  • the E box motif generally comprises YxLxETxAPxGY (SEQ ID NO: 152; where x indicates a varying amino acid residue).
  • the conserved glutamic acid residue within the E box is thought necessary for C. diphtheria pilus formation.
  • the AI-1 polypeptides of the immunogenic compositions comprise an E box motif.
  • E box motifs in the AI-1 polypeptides may include the amino acid sequences YxLxExxxxxGY (SEQ ID NO: 153), YxLxExxxPxGY (SEQ ID NO: 154), or YxLxETxAPxGY (SEQ ID NO: 152).
  • the E box motif of the polypeptides may comprise the amino acid sequences YKLKETKAPEGY (SEQ ID NO: 155), YVLKEIETQSGY (SEQ ID NO: 156), or YKLYEISSPDGY (SEQ ID NO: 157).
  • GBS 80 As discussed in more detail below, a pilin motif containing a conserved lysine residue and an E box motif containing a conserved glutamic acid residue have both been identified in GBS 80.
  • FIG. 34 presents electron micrographs of GBS serotype III, strain isolate COH1 with a plasmid insert to facilitate the overexpression of GBS 80. This EM photo was produced with a standard negative stain—no pilus structures are distinguishable.
  • the use of such AI surface proteins in immunogenic compositions for the treatment or prevention of infection against a Gram positive bacteria has not been previously described.
  • FIGS. 11, 16 and 17 show the presence of pilus structures in wild type COH1 Streptococcus agalactiae .
  • FIG. 49 shows that GBS 80 is associated with pili in a wild type clinical isolate of S. agalactiae , JM9030013. (See FIG. 49 .)
  • mutant GBS strains containing a plasmid comprising the GBS 80 sequence resulting in the overexpression of GBS 80 within this mutant The electron micrographs of FIGS. 13-15 are also stained against GBS 80 and reveal long, oligomeric structures containing GBS 80 which appear to cover portions of the surface of the bacteria and stretch far out into the supernatant.
  • FIG. 61 provides FAC analysis of GBS 80 surface levels on bacterial strains COH1 and JM9130013 using an anti-GBS 80 antisera. Immunogold electron microscopy of the COH1 and JM9130013 bacteria using anti-GBS 80 antisera demonstrates that JM9130013 bacteria, which have higher values for GBS 80 surface expression, also form longer pili structures.
  • FIG. 62 provides FACS analysis of capsulated and uncapsulated GBS analyzed with anti-GBS 80 and anti-GBS 322 antibodies. Surface exposure of GBS 80, unlike GBS 322, is not capsule dependent.
  • Adhesin Island surface protein such as GBS 80 appears to be required for pili formation, as well as an Adhesin Island sortase.
  • Pili are formed in Coh1 bacterial clones that overexpress GBS 80, but lack GBS 104, or one of the AI-1 sortases sag0647 or sag0648.
  • pili are not formed in Coh1 bacterial clones that overexpress GBS 80 and lack both sag0647 and sag0648.
  • GBS 80 in GBS strain 515 which lacks an AI-1, also assembles GBS 80 into pili.
  • GBS strain 515 contains an AI-2, and thus AI-2 sortases.
  • the AI-2 sortases in GBS strain 515 apparently polymerize GBS 80 into pili.
  • FIG. 42 Overexpression of GBS 80 in GBS strain 515 cell knocked out for GBS 67 expression also apparently polymerizes GBS 80 into pili. (See FIG. 72 .)
  • GBS 80 appears to be required for GBS AI-1 pili formation
  • GBS 104 and sortase SAG0648 appears to be important for efficent AI-1 pili assembly.
  • high-molecular structures are not assembled in isogenic COH1 strains which lack expression of GBS 80 due to gene disruption and are less efficiently assembled in isogenic COH1 strains which lack the expression of GBS 104 (see FIG. 41 ).
  • This GBS strain comprises high molecular weight pili structures composed of covalently linked GBS 80 and GBS 104 subunits.
  • deleting SAG0648 in COH1 bacteria interferes with assembly of some of the high molecular weight pili structures. Thus, indicating that SAG0648 plays a role in assembly of these pilin species. (See FIG. 41 ).
  • GBS 52 also appears to be a component of the GBS pili. Immunoblots using an anti-GBS 80 antisera on total cell extracts of Coh1 and a GBS 52 null mutant Coh1 reveal a shift in detected proteins in the Coh1 wild type strain relative to the GBS 52 null mutant Coh1 strain. The shifted proteins were also detected in the wild type Coh1 bacteria with an anti-GBS 52 antisera, indicating that the GBS 52 may be present in the pilus. (See FIG. 45 .)
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as GBS 80.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include one or both of a pilin motif comprising a conserved lysine residue and an E box motif comprising a conserved glutamic acid residue.
  • More than one AI surface protein may be present in the oligomeric, pilus-like structures of the invention.
  • GBS 80 and GBS 104 may be incorporated into an oligomeric structure.
  • GBS 80 and GBS 52 may be incorporated into an oligomeric structure, or GBS 80, GBS 104 and GBS 52 may be incorporated into an oligomeric structure.
  • the invention includes compositions comprising two or more AI surface proteins.
  • the composition may include surface proteins from the same adhesin island.
  • the composition may include two or more GBS AI-1 surface proteins, such as GBS 80, GBS 104 and GBS 52.
  • the surface proteins may be isolated from Gram positve bacteria or they may be produced recombinantly.
  • the invention comprises a GBS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more GBS Adhesin Island 1 (“AI-1”) proteins and one or more GBS Adhesin Island 2 (“AI-2”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • AI-1 GBS Adhesin Island 1
  • AI-2 GBS Adhesin Island 2
  • the oligomeric, pilus-like structures of the invention may be combined with one or more additional GBS proteins.
  • the oligomeric, pilus-like structures comprise one or more AI surface proteins in combination with a second GBS protein.
  • the second GBS protein may be a known GBS antigen, such as GBS 322 (commonly referred to as “sip”) or GBS 276.
  • GBS 322 commonly referred to as “sip”
  • GBS 276 GBS antigen
  • Nucleotide and amino acid sequences of GBS 322 sequenced from serotype V isolated strain 2603 V/R are set forth in WO 02/35771 as SEQ ID 8539 and SEQ ID 8540 and in the present specification as SEQ ID NOs: 38 and 39.
  • a particularly preferred GBS 322 polypeptide lacks the N-terminal signal peptide, amino acid residues 1-24.
  • An example of a preferred GBS 322 polypeptide is a 407 amino acid fragment and is shown in SEQ ID NO: 40. Examples of preferred GBS 322 polypeptides are further described in PCTUS04/______, attorney docket number PP20665.002 filed Sep. 15, 2004, hereby incorporated by reference, published as WO 2005/002619.
  • GBS proteins which may be combined with the GBS AI surface proteins of the invention are also described in WO 2005/002619. These GBS proteins include GBS 91, GBS 184, GBS 305, GBS 330, GBS 338, GBS 361, GBS 404, GBS 690, and GBS 691.
  • GBS proteins which may be combined with the GBS AI surface proteins of the invention are described in WO 02/34771.
  • GBS polysaccharides which may be combined with the GBS AI surface proteins of the invention are described in WO 2004/041157.
  • the GBS AI surface proteins of the invention may be combined with a GBS polysaccharides selected from the group consisting of serotype Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII.
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a GBS bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the GBS bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed AI protein.
  • the AI protein is in a hyperoligomeric form. Macromolecular structures associated with oligomeric pili are observed in the supernatant of cultured GBS strain Coh1. (See FIG. 46 .) These pili are found in the supernatant at all growth phases of the cultured Coh1 bacteria. (See FIG. 47 .)
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a GBS bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the GBS bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed Adhesin Island protein.
  • the Adhesin Island protein is in a hyperoligomeric form.
  • the GBS bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • GBS bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the GBS bacteria with a plasmid encoding the AI protein.
  • the plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein.
  • the sequence encoding the AI protein within the GBS bacterial genome may be deleted.
  • the promoter regulating the GBS Adhesin Island may be modified to increase expression.
  • GBS bacteria harbouring a GBS AI-1 may also be adapted to increase AI protein expression by altering the number adenosine nucleotides present at two sites in the intergenic region between AraC and GBS 80.
  • FIG. 197 A which is a schematic showing the organization of GBS AI-1 and FIG. 197 B , which provides the sequence of the intergenic region between AraC and GBS 80 in the AI.
  • the adenosine tracts which applicants have identified as influencing GBS 80 surface expression are at nucleotide positions 187 and 233 of the sequence shown in FIG. 197 B (SEQ ID NO: 273).
  • FACS analysis of these strains using anti GBS 80 antiserum determined that an intergenic region with five adenosines at position 187 and six adenosines at position 233 had higher expression levels of GBS 80 on their surface than other stains. See FIG. 197 C for results obtained from the FACS analysis. Therefore, manipulating the number of adenosines present at positions 187 and 233 of the AraC and GBS 80 intergenic region may further be used to adapt GBS to increase AI protein expression.
  • the invention further includes GBS bacteria which have been adapted to produce increased levels of AI surface protein.
  • the invention includes GBS bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein, such as GBS 80.
  • the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the invention further includes GBS bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface.
  • the GBS bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide.
  • Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria.
  • Increased expression of a leader peptidase in GBS may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation.
  • the GBS bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes ” Infection and Immunity (2004) 72(6):3444-3450).
  • Streptococus gordonii See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine
  • non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis.
  • the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid.
  • the non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria.
  • the AI surface protein may be isolated from cell extracts or culture supernatants.
  • the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • the non-pathogenic Gram positive bacteria may be used to express any of the Gram positive bacterial Adhesin Island proteins described herein, including proteins from a GBS Adhesin Island, a GAS Adhesin Island, or a S pneumo Adhesin Island.
  • the non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein.
  • the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase.
  • the AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with a pathogenic Gram positive bacteria, such as GBS, GAS or Streptococcus pneumoniae .
  • the non-pathogenic Gram positive bacteria may express the Gram positive bacterial Adheshin Island proteins in oligomeric forms that further comprise adhesin island proteins encoded within the genome of the non-pathogenic Gram positive bacteria.
  • L. lactis was transformed with a construct encoding GBS 80 under its own promoter and terminator sequences.
  • the transformed L. lactis appeared to express GBS 80 as shown by Western blot analysis using anti-GBS 80 antiserum. See lanes 6 and 7 of the Western Blots provided in FIGS. 133A and 133B ( 133 A and 133 B are two different exposures of the same Western blot). See also Example 13.
  • L. lactis with a construct encoding GBS AI-1 polypeptides GBS 80, GBS 52, SAG0647, SAG0648, and GBS 104 under the GBS 80 promoter and terminator sequences.
  • These L. lactis expressed high molecular weight structures that were immunoreactive with anti-GBS 80 in immunoblots. See FIG. 134 , lane 2, which shows detection of a GBS 80 monomer and higher molecular weight polymers in total transformed L. lactis extracts.
  • L. lactis is capable of expressing GBS 80 in oligomeric form.
  • the high molecular weight polymers were not only detected in L. lactis extracts, but also in the culture supernatants. See FIG.
  • the GBS AI polypeptides in oligomeric form can be isolated and purified from either L. lactis cell extracts or culture supernatants. These oligomeric forms can, for instance, be isolated from cell extracts or culture supernatants by release by sonication. See FIGS. 136A and B. See also FIG. 171 , which shows purification of GBS pili from whole extracts of L. lactis expressing the GBS AI-1 following sonication and gel filtration on a Sephacryl HR 400 column.
  • FACS analysis of these transformed L. lactis detected cell surface expression of both GBS 80 and GBS 104.
  • the surface expression levels of GBS 80 and GBS 104 on the transformed L. lactis were similar to the surface expression levels of GBS 80 and GBS 104 on GBS strains COH1 and JM9130013, which naturally express GBS AI-1.
  • FIG. 169 for FACS analysis data for L. lactis transformed with GBS AI-1 and wildtype JM9130013 bacteria using anti-GBS 80 and GBS 104 antisera.
  • Table 40 provides the results of FACS analysis of transformed L. lactis , COH1, and JM9130013 bacteria using anti-GBS 80 and anti-GBS 104 antisera. The numbers provided represent the mean fluorescence value difference calculated for immune versus pre-immune sera obtained for each bacterial strain. TABLE 40 FACS analysis of L. lactis and GBS bacteria strains expressing GBS AI-1 Anti-GBS 80 Anti-GBS 104 antiserum antiserum GBS AI-1 transformed 298 251 L.
  • mice with L. lactis transformed with GBS AI-1 were immunized with L. lactis transformed with GBS AI-1.
  • the immunized female mice were bred and their pups were challenged with a dose of GBS sufficient to kill 90% of non-immunized pups.
  • Detailed protocols for intranasal and subcutaneous immunization of mice with transformed L. lactis can be found in Examples 18 and 19, respectively.
  • Table 43 provides data showing that immunization of the female mice with L. lactis expressing GBS AI-1(LL-AI 1) greatly increased survival rate of challenged pups relative to both a negative PBS control (PBS) and a negative L.
  • PBS negative PBS control
  • lactis control (LL 10 E9, which is wild type L. lactis not transformed to express GBS AI-1). TABLE 43 Protection of Mice Immunized with L. lactis expressing GBS AI-1 Immunization Survival Route Antigen Alive/Treated Survival % % Range p value Intraperitoneum Recombinant GBS 80 16/18 89 80-100 ⁇ 0.001 Subcutaneous LL-AI 1 10 E9 40/49 82 70-90 ⁇ 0.001 LL-AI 1 10 E10 50/60 83 60-100 ⁇ 0.001 PBS 4/30 13 0-30 LL 10 E9 3/57 5 0-20 Intranasal LL-AI 1 10 E9 22/60 37 0-100 0.02 LL-AI 1 10 E10 31/49 63 30-90 ⁇ 0.001 LL 10 E9 2/27 7 0-20
  • mice with L. lactis expressing the GBS AI-1 Protection of immunized mice with L. lactis expressing the GBS AI-1 is at least partly due to a newly raised antibody response.
  • Table 46 provides anti-GBS 80 antibody titers detected in serum of the mice immunized with L. lactis expressing the GBS AI-1 as described above. Mice immunized with L. lactis expressing the GBS AI-1 have anti-GBS 80 antibody titres, which are not observed in mice immunized with L. lactis not transformed to express the GBS AI-1. Further, as expected from the survival data, mice subcutaneously immunized with L. lactis transformed to express the GBS AI-1 have significantly higher serum anti-GBS 80 antibody titers than mice intranasally immunized with L.
  • Anti-GBS 80 antibodies of the IgA isotype were specifically detected in various body fluids of the mice subcutaneously or intranasally immunized with L. lactis expressing the GBS AI-1. TABLE 47 Anti-GBS 80 IgA Antibodies Detected in Mouse Tissues Following Immunization with L. lactis Expressing GBS AI-1 Anti-GBS 80 IgA Antibodies Detected in Antigen Immunization route Serum Vaginal Wash Nasal Wash LL 10 E9 0 0 0 LL-AI 1 Subcutaneous 0 25 20 LL-AI 1 Intranasal 140 0 150 GBS 80 Intraperitoneal 60 0
  • opsonophagocytosis assays also demonstrated that at least some of the antiserum produced against the L. lactis expressing GBS AI 1 is opsonic for GBS. See FIG. 161 .
  • a hybrid GBS AI may be a GBS AI-1 with a replacement of the GBS 104 gene with a GBS 67 gene.
  • a schematic of such a hybrid GBS AI is depicted in FIG. 231 A .
  • a hybrid GBS AI may alternatively be a GBS AI-1 with a replacement of the GBS 52 gene with a GBS 59 gene. See the schematic at FIG. 231 B .
  • a hybrid GBS AI may be a GBS AI-1 with a substitution of a GBS 59 polypeptide for the GBS 52 gene and a substitution of the GBS 104 gene for genes encoding GBS 59 and the two GBS AI-2 sortases.
  • Another example of a hybrid GBS AI is a GBS AI-1 with the substitution of a GBS 59 gene for the GBS 52 gene and a GBS 67 for the GBS 104 gene. See the schematic at FIG. 232 .
  • a further example of a hybrid GBS AI is a GBS AI-1 having a GBS 59 gene and genes encoding the GBS AI-2 sortases in place of the GBS 52 gene.
  • hybrid GBS AI is a GBS AI-1 with a substitution of either GBS 52 or GBS 104 with a fusion protein comprising GBS 322 and one of GBS 59, GBS 67, or GBS 150.
  • Some of these hybrid GBS AIs may be prepared as briefly outlined in FIG. 234 A-F.
  • FIG. 231 A Applicants have prepared a hybrid GBS AI having a GBS AI-1 sequence with a substitution of a GBS 67 coding sequence for the GBS 104 gene as depicted in FIG. 231 A . Transformation of L. lactis with the hybrid GBS AI-1 resulted in L. lactis expression of high molecular weight polymers containing the GBS 80 and GBS 67 proteins. See FIG. 233 A , which provides Western blot analysis of L. lactis transformed with the hybrid GBS AI depicted in FIG. 231 A . When L. lactis transformed with the hybrid GBS AI were probed with antibodies to GBS 80 or GBS 67, high molecular weight structures were detected.
  • the oligomeric, pilus-like structures may be produced recombinantly. If produced in a recombinant host cell system, the AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention. Such AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • AI Sortases of the invention will typically have a signal peptide sequence within the first 70 amino acid residues. They may also include a transmembrane sequence within 50 amino acid residues of the C terminus. The sortases may also include at least one basic amino acid residue within the last 8 amino acids. Preferably, the sortases have one or more active site residues, such as a catalytic cysteine and histidine.
  • AI-1 includes the surface exposed proteins of GBS 80, GBS 52 and GBS 104 and the sortases SAG0647 and SAG0648. AI-1 typically appears as an insertion into the 3′ end of the trmA gene.
  • AI-1 may also include a divergently transcribed transcriptional regulator such as araC (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction). It is believed that araC may regulate the expression of the AI operon. (See Korbel et al., Nature Biotechnology (2004) 22(7): 911-917 for a discussion of divergently transcribed regulators in E. coli ).
  • araC may regulate the expression of the AI operon.
  • AI-1 may also include a sequence encoding a rho independent transcriptional terminator (see hairpin structure in FIG. 1 ). The presence of this structure within the adhesin island is thought to interrupt transcription after the GBS 80 open reading frame, leading to increased expression of this surface protein.
  • FIG. 2 A schematic identifying AI-1 within several GBS serotypes is depicted in FIG. 2 .
  • AI-1 sequences were identified in GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate NEM316; GBS serotype II, strain isolate 18RS21; GBS serotype V, strain isolate CJB111; GBS serotype III, strain isolate COH1 and GBS serotype 1a, strain isolate A909. (Percentages shown are amino acid identity to the 2603 sequence). (An AI-1 was not identified in GBS serotype 1b, strain isolate H36B or GBS serotype 1a, strain isolate 515).
  • FIG. 18 An alignment of AI-1 polynucleotide sequences from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype III, strain isolates COH1 and NEM316; and serotype 1a, strain isolate A909 is presented in FIG. 18 .
  • An alignment of amino acid sequences of AI-1 surface protein GBS 80 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate A909; serotype III, strain isolates COH1 and NEM316 is presented in FIG. 22 .
  • AI-1 surface protein GBS 104 from serotype V, strain isolates 2603 and CJB111; serotype III, strain isolates COH1 and NEM316; and serotype II, strain isolate 18RS21 is presented in FIG. 23 .
  • Preferred AI-1 polynucleotide and amino acid sequences are conserved among two or more GBS serotypes or strain isolates.
  • GBS 80 As shown in this figure, the full length of surface protein GBS 80 is particularly conserved among GBS serotypes V (strain isolates 2603 and CJBIII), III (strain isolates NEM316 and COH1), and Ia (strain isolate A909).
  • the GBS 80 surface protein is missing or fragmented in serotypes II (strain isolate 18RS21), Ib (strain isolate H36B) and Ia (strain isolate 515).
  • Polynucleotide and amino acid sequences for AraC are set forth in FIG. 30 .
  • a second adhesin island “Adhesin Island 2” or “AI-2” or GBS AI-2” has also been identified in numerous GBS serotypes.
  • a schematic depicting the correlation between AI-1 and AI-2 within the GBS serotype V, strain isolate 2603 is shown in FIG. 3 .
  • Homology percentages in FIG. 3 represent amino acid identity of the AI-2 proteins to the AI-1 proteins).
  • Alignments of AI-2 polynucleotide sequences are presented in FIGS. 20 and 21 ( FIG. 20 includes sequences from serotype V, strain isolate 2603 and serotype III, strain isolate NEM316.
  • FIG. 21 includes sequences from serotype III, strain isolate COH1 and serotype Ia, strain isolate A909).
  • AI-2 surface protein GBS 067 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate 515; serotype II, strain isolate 18RS21; serotype Ib, strain isolate H36B; and serotype III, strain isolate NEM316 is presented in FIG. 24 .
  • Preferred AI-2 polynucleotide and amino acid sequences are conserved among two or more GBS serotypes or strain isolates.
  • AI-2 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • AI-2 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5 or more) of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406.
  • AI-2 may include open reading frames encoding for two or more of 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • One or more of the surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the GBS AI-2 sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GBS AI-2 may encode for at least one surface protein.
  • AI-2 may encode for at least two surface proteins and at least one sortase.
  • GBS AI-2 encodes for at least three surface proteins and at least two sortases.
  • One or more of the AI-2 surface proteins may include an LPXTG or other sortase substrate motif.
  • One or more of the surface proteins may also typically include pilin motif.
  • the pilin motif may be involved in pili formation. Cleavage of AI surface proteins by sortase between the threonine and glycine residue of an LPXTG motif yields a thioester-linked acyl intermediate of sortase.
  • the first lysine residue in a pilin motif can serve as an amino group acceptor of the cleaved LPXTG motif and thereby provide a covalent linkage between AI subunits to form pili.
  • the pilin motif can make a nucleophilic attack on the acyl enzyme providing a covalent linkage between AI subunits to form pili and regenerate the sortase enzyme.
  • pilin motifs that may be present in the GBS AI-2 proteins include ((YPKN(X 8 )K; SEQ ID NO: 158), (PK(X 8 )K; SEQ ID NO: 159), (YPK(X 9 )K; SEQ ID NO: 160), (PKN(X 8 )K; SEQ ID NO: 161), or (PK(X 10 )K; SEQ ID NO: 162)).
  • One or more of the surface protein may also include an E box motif.
  • the E box motif contains a conserved glutamic acid residue that is believed to be necessary for pilus formation.
  • Some examples of E box motifs may include the amino acid sequences YxLxETxAPxG (SEQ ID NO: 163), YxxxExxAxxGY (SEQ ID NO: 164), YxLxExxxPxDY (SEQ ID NO: 165), or YxLxETxAPxGY (SEQ ID NO: 152).
  • GBS AI-2 may include the surface exposed proteins of GBS 67, GBS 59 and GBS 150 and the sortases of SAG1406 and SAG1405.
  • GBS AI-2 may include the proteins 01521, 01524 and 01525 and sortases 01520 and 01522.
  • GBS 067 and 01524 are preferred AI-2 surface proteins.
  • AI-2 may also include a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB). As in AI-1, rogB is thought to regulate the expression of the AI-2 operon.
  • a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB).
  • rogB is thought to regulate the expression of the AI-2 operon.
  • FIG. 4 A schematic depiction of AI-2 within several GBS serotypes is depicted in FIG. 4 . (Percentages shown are amino acid identity to the 2603 sequence). While the AI-2 surface proteins GBS 59 and GBS 67 are more variable across GBS serotypes than the corresponding AI-1 surface proteins, AI-2 surface protein GBS 67 appears to be conserved in GBS serotypes where the AI-1 surface proteins are disrupted or missing.
  • the AI-1 GBS 80 surface protein is fragmented in GBS serotype II, strain isolate 18RS21.
  • the GBS 67 surface protein has 99% amino acid sequence homology with the corresponding sequence in strain isolate 2603.
  • the AI-1 GBS 80 surface protein appears to be missing in GBS serotype Ib, strain isolate H36B and GBS serotype Ia, strain isolate 515.
  • the GBS 67 surface protein has 97-99% amino acid sequence homology with the corresponding sequence in strain isolate 2603.
  • GBS 67 appears to have two allelic variants, which can be divided according to percent homology with strains 2603 and H36B. See FIGS. 237-239 .
  • GBS 59 of GBS strain isolate 2603 shares 100% amino acid residue homology with GBS strain 18RS21, 62% amino acid sequence homology with GBS strain H36B, 48% amino acid residue homology with GBS strain 515 and GBS strain CJB111, and 47% amino acid residue homology with GBS strain NEM316.
  • the amino acid sequence homologies of the different GBS strains suggest that there are two isoforms of GBS 59. The first isoform appears to include the GBS 59 protein of GBS strains CJB111, NEM316, and 515.
  • the second isoform appears to include the GBS 59 protein of GBS strains 18RS21, 2603, and H36B. (See FIGS. 63 and 224 .) As expected from the variability in GBS 59 isoforms, antibodies specific for the first GBS 59 isoform detect the first but not the second GBS 59 isoform and antibodies specific for the second GBS 59 isoform detect the second but not the first GBS 59 isoform. See FIG. 226A , which shows FACS analysis of 28 GBS strains having a GBS 59 gene detected using PCR for GBS 59 surface expression.
  • GBS 59 isoform 1 ⁇ -Cjb111
  • GBS 59 isoform 2 ⁇ -2603
  • GBS 59 is opsonic only against GBS strains expressing a homologous GBS 59 protein. See FIG. 225 .
  • the immunogenic composition of the invention comprises a first and a second isoform of the GBS 59 protein to provide protection across a wide range of GBS serotypes that express polypeptides from a GBS AI-2.
  • the first isoform may be the GBS 59 protein of GBS strain CJB111, NEM316, or 515.
  • the second isoform may be the GBS 59 protein of GBS strain 18RS21, 2603, or H36B.
  • FIG. 64 shows detection of high molecular weight GBS 59 polymers in whole extracts of GBS strains CJB111, 7357B, COH31, D1363C, 5408, 1999, 5364, 5518, and 515 using antiserum raised against GBS 59 of GBS strain CJB111.
  • FIG. 64 shows detection of high molecular weight GBS 59 polymers in whole extracts of GBS strains CJB111, 7357B, COH31, D1363C, 5408, 1999, 5364, 5518, and 515 using antiserum raised against GBS 59 of GBS strain CJB111.
  • FIG. 65 also shows detection of these high molecular weight GBS 59 polymers in whole extracts of GBS strains D136C, 515, and CJB111 with anti-GBS 59 antiserum. (See also FIG. 220 A for detection of GBS 59 high molecular weight polymers in strain 515.)
  • FIG. 65 confirms the presence of different isoforms of GBS 59. Antisera raised against two different GBS 59 isoforms results in different patterns of immunoreactivity depending on the GBS strain origin of the whole extract.
  • FIG. 65 further shows detection of GBS 59 monomers in purified GBS 59 preparations.
  • GBS 59 is also highly expressed on the surface of GBS strains.
  • GBS 59 was detected on the surface of GBS strains CJB111, DK1, DK8, Davis, 515, 2986, 5551, 1169, and 7357B by FACS analysis using mouse antiserum raised against GBS 59 of GBS CJB111. FACS analysis did not detect surface expression of GBS 59 in GBS strains SMU071, JM9130013, and COH1, which do not contain a GBS 59 gene. (See FIG. 66 .) Further confirmation that GBS 59 is expressed on the surface of GBS is detection of GBS 59 by immuno-electron microscopy on the surface of GBS strain 515 bacteria. See FIG. 215 .
  • GBS 67 and GBS 150 also appear to be included in high molecular weight structures, or pili.
  • FIG. 69 shows that anti-GBS 67 and anti-GBS 150 immunoreact with high molecular weight structures in whole GBS strain 515 extracts. (See also FIG. 220 B and C.) It is also notable in FIG. 69 that the anti-GBS 59 antisera, raised in a mouse following immunization with GBS 59 of GBS strain 2603, does not cross-hybridize with GBS 59 in GBS strain 515. GBS 59 of GBS stain 515 is of a different isotype than GBS 59 of GBS stain 2603. See FIG.
  • FIG. 65 which confirms that GBS 59 antisera raised against GBS strain 2603 does not cross-hybridize with GBS 59 of GBS strain 515.
  • FIG. 70 provides Western blots showing that higher molecular weight structures in GBS strain 515 total extracts immunoreact with anti-GBS 67 and anti-GBS 150 antiserum.
  • anti-GBS 67 antiserum no longer immunoreacts with polypeptides in total extracts, while anti-GBS 150 antiserum is still able to cross-hybridze with high molecular weight structures.
  • GBS 67 cell surface expression is detected on GBS stain 515 cells regardless of GBS 67 expression.
  • GBS 67 while present in pili, appears to be localized around the surface of GBS strain 515 cells. See the immuno-electron micrographs presented in FIG. 216 . GBS 67 binds to fibronectin. See FIG. 217 .
  • GBS AI-2 Formation of pili encoded by GBS AI-2 does require expression of GBS 59.
  • Deletion of GBS 59 from strain 515 bacteria eliminates detection of high molecular weight structures by antibodies that bind to GBS 59 ( FIG. 221 A , lane 3), GBS 67 ( FIG. 221 B , lane 3), and GBS 150 ( FIG. 221 C , lane 3).
  • Western blot analysis of 515 bacteria with a deletion of the GBS 67 gene detects high molecular weight structures using GBS 59 ( FIG. 221 A , lane 2) and GBS 150 ( FIG. 221 C , lane 2) antisera.
  • GBS AI-2 Formation of pili encoded by GBS AI-2 also requires expression of both GBS adhesin island-2 encoded sortases. See FIG. 218 , which provides Western blot analysis of strain 515 bacteria lacking Srt1, Srt2, or both Srt1 and Srt2. Only deletion of both Srt1 and Srt2 abolishes pilus assembly as detected by antibodies that cross-hybridize with each of GBS 59, GBS 67 and GBS 150. The results of the Western blot analysis were verified by FACS, which provided similar results. See FIG. 219 .
  • More than one AI surface protein may be present in the oligomeric, pilus-like structures of the invention.
  • GBS 59 and GBS 67 may be incorporated into an oligomeric structure.
  • GBS 59 and GBS 150 may be incorporated into an oligomeric structure, or GBS 59, GBS 150 and GBS 67 may be incorporated into an oligomeric structure.
  • the invention includes compositions comprising two or more AI surface proteins.
  • the composition may include surface proteins from the same adhesin island.
  • the composition may include two or more GBS AI-2 surface proteins, such as GBS 59, GBS 67 and GBS 150.
  • the surface proteins may be isolated from Gram positve bacteria or they may be produced recombinantly.
  • adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fascilitis.
  • post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases.
  • the first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused.
  • the bacteria secretes a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection.
  • the final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart.
  • a general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above.
  • the M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation.
  • the carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci.
  • the amino terminus which extend through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • T-antigen A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen.
  • T-antigen a variable, trypsin-resistant surface antigen
  • Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens.
  • Antisera to define T types is commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • T-antigen T-type 6
  • M6 strain of GAS M6 strain of GAS
  • FCT Fibronectin-binding, Collagen-binding T-antigen
  • the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used.
  • Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms. Immunological recognition of these proteins may allow the host immune response to slow or prevent the bacteria's transition into the more pathogenic later stages of infection.
  • GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix).
  • Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm.
  • Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment is preferable.
  • the invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes.
  • the immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form.
  • the invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • GAS AI sequences may be generally characterized as Type 1, Type 2, Type 3, and Type 4, depending on the number and type of sortase sequence within the island and the percentage identity of other proteins within the island.
  • Schematics of the GAS adhesin islands are set forth in FIG. 51A and FIG. 162 . In all strains identified so far, the adhesin island region is flanked by highly conserved open reading frames M1 — 123 and M1 — 136. Between three and five genes in each GAS adhesin island code for ECM binding adhesin proteins containing LPXTG motifs.
  • GAS AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-1 proteins”).
  • GAS AI-1 preferably comprises surface proteins, a srtB sortase, and a rofA divergently transcribed transcriptional regulator.
  • GAS AI-1 surface proteins may include a fibronectin binding protein, a collagen adhesion protein and a fimbrial structural subunit.
  • each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine).
  • GAS AI-1 includes open reading frames encoding for two or more (i.e., 2, 3, 4 or 5) of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161.
  • a GAS AI-1 may comprise a polynucleotide encoding any one of CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • the hyper-oligomeric pilus structure of GAS AI-1 appears to be responsible for the T-antigen type 6 classification, and GAS AI-1 corresponds to the FCT region previously identified for tee6.
  • the tee6 FCT region includes open reading frames encoding for a collagen adhesion protein (cpa, capsular polysaccharide adhesion) and a fibronectin binding protein (prtF1).
  • cpa collagen adhesion protein
  • prtF1 fibronectin binding protein
  • Immunoblots of tee6, a GAS AI-1 fimbrial structural subunit corresponding to M6_Spy160 reveal high molecular weight structures indicative of the hyper-oligomeric pilus structures.
  • Immunoblots with antiserum specific for Cpa also recognize a high molecular weight ladder structure, indicating Cpa involvement in the GAS AI-1 pilus structure or formation.
  • Cpa antiserum reveals abundant staining on the surface of the bacteria and occasional gold particles extended from the surface of the bacteria.
  • immunoblots with antiserum specific for PrtF1 recognize only a single molecular species with electrophoretic mobility corresponding to its predicted molecular mass, indicating that PrtF1 may not be associated with the oligomeric pilus structure.
  • a preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • the oligomeric form is a hyperoligomer.
  • Another preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which has been isolated in an oligomeric (pilis) form.
  • the oligomer or hyperoligomeric pilus structures comprising the GAS AI-1 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • GAS AI-1 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-1 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the GAS AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the LPXTG sortase substrate motif of a GAS AI surface protein may be generally represented by the formula XXXXG, wherein X at amino acid position 1 is an L, a V, an E, or a Q, wherein X at amino acid position 2 is a P if X at amino acid position 1 is an L, wherein X at amino acid position 2 is a V if X at amino acid position 1 is a E or a Q, wherein X at amino acid position 2 is a V or a P if X at amino acid position 1 is a V, wherein X at amino acid position 3 is any amino acid residue, wherein X at amino acid position 4 is a T if X at amino acid position 1 is a V, E, or Q, and wherein X at amino acid position 4 is a T, S, or A if X at amino acid position 1 is an L.
  • LPXTG motifs present in GAS AI surface proteins include LPSXG (SEQ ID NO: 134), VVXTG (SEQ ID NO: 135), EVXTG (SEQ ID NO: 136), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138), LPXAG (SEQ ID NO: 139), QVPTG (SEQ ID NO: 140), and FPXTG (SEQ ID NO: 141).
  • the GAS AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer.
  • one or more GAS AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • GAS AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • GAS AI-1 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-1 may encode for at least one surface protein.
  • GAS AI-1 may encode for at least two surface exposed proteins and at least one sortase.
  • GAS AI-1 encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • GAS AI-1 preferably includes a srtB sortase.
  • GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a GAS AI-1 surface protein such as M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, or DSM2071_fimbrial.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively:
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more GAS Adhesin Island 1 (“GAS AI-1”) proteins and one or more GAS Adhesin Island 2 (“GAS AI-2”), GAS Adhesin Island 3 (“GAS AI-3”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the GAS Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • GAS Adhesin Island 1 (“GAS AI-1”) proteins
  • GAS Adhesin Island 2”) GAS Adhesin Island 3
  • GAS Adhesin Island 4 GAS Adhesin Island 4
  • GAS AI-1 may also include a divergently transcribed transcriptional regulator such as RofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • RofA a divergently transcribed transcriptional regulator
  • GAS Adhesin Island 2 A second adhesin island, “GAS Adhesin Island 2” or “GAS AI-2” has also been identified in Group A Streptococcus serotypes and isolates.
  • GAS AI-2 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-2 proteins”).
  • GAS AI-2 proteins include open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, 7, or 8) of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • a preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • the oligomeric form is a hyperoligomer.
  • Another preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which has been isolated in an oligomeric (pilis) form.
  • the oligomer or hyperoligomeric pilus structures comprising the GAS AI-2 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-2 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-2 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the GAS AI-2 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer.
  • one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • GAS AI-2 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-2 may encode for at least one surface protein.
  • GAS AI-2 may encode for at least two surface exposed proteins and at least one sortase.
  • GAS AI-2 encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as GAS 15, GAS 16, or GAS 18.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more GAS Adhesin Island 2 (“GAS AI-2”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 3 (“GAS AI-3”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • GAS AI-2 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • rofA a divergently transcribed transcriptional regulator
  • GAS Adhesin Island 3 A third adhesin island, “GAS Adhesin Island 3” or “GAS AI-3” has also been identified in several Group A Streptococcus serotypes and isolates.
  • GAS AI-3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-3 proteins”).
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, SpyM3 — 0104, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, SPs0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, spyM18 — 0132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, and SpyM3 — 0104.
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, and SPs0106.
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of orf78, orf79, orf80, orf81, orf82, orf83, and orf84.
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, and spyM18 — 0132.
  • GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149.
  • a GAS AI-3 may comprise a polynucleotide encoding any one of ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • GAS AI-3 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-3 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • a preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • the oligomeric form is a hyperoligomer.
  • Another preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which has been isolated in an oligomeric (pilis) form.
  • the oligomer or hyperoligomeric pilus structures comprising the GAS AI-3 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-3 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer.
  • one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • GAS AI-3 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-3 may encode for at least one surface protein.
  • GAS AI-3 may encode for at least two surface exposed proteins and at least one sortase.
  • GAS AI-3 encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine or alanine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyM3 — 0098, SpyM3 — 0100, SpyM3 — 0102, SpyM3 — 0104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM18 — 0126, spyM18 — 0128, spyM18 — 0130, spyM18 — 0132, SpyoM01000155, SpyoM01000153, SpyoM01000151, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • an AI surface protein such as SpyM3 — 0098, SpyM3 — 0100, SpyM3 — 0102, SpyM3 — 0104, SPs0100, SPs0102, SPs0104,
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyM3 — 0098, SpyM3 — 0100, SpyM3 — 0102, and SpyM3 — 0104.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SPs0100, SPs0102, SPs0104, and SPs0106.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as orf78, orf80, orf82, and orf84.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as spyM18 — 0126, spyM18 — 0128, spyM18 — 0130, and spyM18 — 0132.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyoM01000155, SpyoM01000153, SpyoM0000151, and SpyoM1000149.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more GAS Adhesin Island 3 (“GAS AI-3”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 2 (“GAS AI-2”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • GAS AI-3 may also include a transcriptional regulator such as Nra.
  • GAS Adhesin Island 4 or “GAS AI-4” has also been identified in Group A Streptococcus serotypes and isolates.
  • GAS AI-4 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-4 proteins”).
  • GAS AI-4 proteins include open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, 7, or 8) of 19224134, 19224135, 19223136, 19223137, 19224138, 19224139, 19224140, and 19224141.
  • a GAS AI-4 may comprise a polynucleotide encoding any one of 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • GAS AI-4 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the GAS AI-4 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • a preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • the oligomeric form is a hyperoligomer.
  • Another preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which has been isolated in an oligomeric (pilis) form.
  • the oligomer or hyperoligomeric pilus structures comprising the GAS AI-4 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-4 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the AI surface proteins of the invention may effect the ability of the GAS bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer.
  • one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • GAS AI-4 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • GAS AI-4 may encode for at least one surface protein.
  • GAS AI-4 may encode for at least two surface exposed proteins and at least one sortase.
  • GAS AI-4 encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more GAS Adhesin Island 4 (“GAS AI-4”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 2 (“GAS AI-2”), or GAS Adhesin Island 3 (“GAS AI-3”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • GAS Adhesin Island 4 (“GAS AI-4”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 2 (“GAS AI-2”), or GAS Adhesin Island 3 (“GAS AI-3”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • GAS AI-4 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • rofA a divergently transcribed transcriptional regulator
  • the oligomeric, pilus-like structures of the invention may be combined with one or more additional GAS proteins.
  • the oligomeric, pilus-like structures comprise one or more AI surface proteins in combination with a second GAS protein.
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a GAS bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the GAS bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed AI protein.
  • the AI protein is in a hyperoligomeric form.
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a GAS bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the GAS bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed Adhesin Island protein.
  • the Adhesin Island protein is in a hyperoligomeric form.
  • the GAS bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • GAS bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the GAS bacteria with a plasmid encoding the AI protein.
  • the plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein.
  • the sequence encoding the AI protein within the GAS bacterial genome may be deleted.
  • the promoter regulating the GAS Adhesin Island may be modified to increase expression.
  • the invention further includes GAS bacteria which have been adapted to produce increased levels of AI surface protein.
  • the invention includes GAS bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein.
  • the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the invention further includes GAS bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface.
  • the GAS bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of LepA polypeptide, or an equivalent signal peptidase, in the GAS bacteria.
  • Applicants have shown that deletion of LepA in strain SF370 bacteria, which harbour a GAS AI-2, abolishes surface exposure of M and pili proteins on the GAS.
  • Increased levels of LepA expression in GAS are expected to result in increased exposure of M and pili proteins on the surface of GAS.
  • Increased expression of LepA in GAS may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation.
  • the GAS bacteria adapted to have increased levels of LepA expression may additionally be adapted to express increased levels of at least one pili protein.
  • the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes ” Infection and Immunity (2004) 72(6):3444-3450).
  • Streptococus gordonii See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine
  • non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis.
  • the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid.
  • the non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria.
  • the AI surface protein may be isolated from cell extracts or culture supernatants.
  • the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • the non-pathogenic Gram positive bacteria may be used to express any of the GAS Adhesin Island proteins described herein.
  • the non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein.
  • the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase.
  • the AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with pathogenic GAS.
  • L. lactis was transformed with pAM401 constructs encoding entire pili gene clusters of AI-1, AI-2, and AI-4 adhesin islands.
  • the pAM401 is a promoterless high-copy plasmid.
  • the entire pili gene clusters of an M6 (AI-1), M1 (AI-2), and M12 (AI-4) bacteria were inserted into the pAM401 construct.
  • the gene clusters were transcribed under the control their own (M6, M1, or M12) promoter or the GBS promoter that successfully initiated expression of the GBS AI-1 adhesin islands in L. lactis , described above.
  • FIG. 172 provides a schematic depiction of GAS M6 (AI-1), M1 (AI-2), and M12 (AI-4) adhesin islands and indicates the portions of the adhesin island sequences inserted in the pAM401 construct.
  • FIGS. 173 A-C provide results of Western blot analysis of surface protein-enriched extracts of L. lactis transformed with M6 ( FIG. 173 A ), M1 ( FIG. 173 B ), or M12 ( FIG. 173 C ) adhesin island gene clusters using antibodies that bind to the fimbrial structural subunit encoded by each cluster.
  • FIG. 173A at lanes 3 and 4 shows detection of high molecular structures in L.
  • FIG. 173B at lanes 3 and 4 shows detection of high molecular weight structures in L. lactis transformed with an adhesin island pilus gene cluster from an M12 AI-4 using an antibody that binds to fimbrial structural subunit EftLSL.A.
  • FIG. 173C at lane 3 shows detection of high molecular weight structures in L. lactis transformed with an adhesin island pilus gene cluster from an M6 AI-1 using an antibody that binds to fimbrial structural subunit M6_Spy0160.
  • L. lactis is capable of expressing the fimbrial structural subunits encoded by GAS adhesin islands in an oligomeric form.
  • the oligomeric, pilus-like structures may be produced recombinantly. If produced in a recombinant host cell system, the AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention. Such AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • the S. pneumoniae from TIGR4 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae from TIGR4 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, and SP0468.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae from TIGR4 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • the oligomeric form is a hyperoligomer.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae from TIGR4 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • the oligomer or hyperoligomer pilus structures comprising S. pneumoniae surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • One or more of the S. pneumoniae from TIGR4 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae from TIGR4 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae from TIGR4 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae from TIGR4 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells.
  • AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer.
  • one or more S. pneumoniae from TIGR4 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • S. pneumoniae from TIGR4 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae from TIGR4 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae from TIGR4 AI may encode for at least one surface protein.
  • S. pneumoniae from TIGR4 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae from TIGR4 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae from TIGR4 AI surface protein such as SP0462, SP0463, SP0464, or SP0465.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae from TIGR4 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae from TIGR4 AI proteins and one or more S. pneumoniae strain 670 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae from TIGR4 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 670 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 670 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of orf1 — 670, orf3 — 670, orf4 — 670, orf5 — 670, orf6 — 670, orf7 — 670, orf8 — 670.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 670 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 670 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 670 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 670 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 670 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 670 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 670 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 670 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 670 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 670 AI may encode for at least one surface protein.
  • S. pneumoniae strain 670 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 670 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 670 AI surface protein such as orf3 — 670, orf4 — 670, or orf5 — 670.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 670 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 670 AI proteins and one or more S. pneumoniae from TIGR4 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 670 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 14 CSR 10 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 14 CSR 10 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 14CSR, ORF3 — 14CSR, ORF4 — 14CSR, ORF5 — 14CSR, ORF6 — 14CSR, ORF7 — 14CSR, ORF8 — 14CSR.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 14 CSR 10 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 14 CSR 10 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 14 CSR 10 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 14 CSR 10 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 14 CSR 10 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 14 CSR 10 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 14 CSR 10 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 14 CSR 10 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 14 CSR 10 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 14 CSR 10 AI may encode for at least one surface protein.
  • S. pneumoniae strain 14 CSR 10 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 14 CSR 10 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 14 CSR 10 AI surface protein such as orf3_CSR, orf4_CSR, or orf5_CSR.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 14 CSR 10 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 14 CSR 10 AI proteins, and one or more AI proteins of any of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 14 CSR 10 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 19A Hungary 6 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 19A Hungary 6 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 19AH, ORF3 — 19AH, ORF4 — 19AH, ORF5 — 1 gAH, ORF6 — 1 gAH, ORF7 — 19AH, ORF8 — 19AH.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19A Hungary 6 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19A Hungary 6 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 19A Hungary 6 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 19A Hungary 6 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 19A Hungary 6 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 19A Hungary 6 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 19A Hungary 6 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 19A Hungary 6 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 19A Hungary 6 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 19A Hungary 6 AI may encode for at least one surface protein.
  • S. pneumoniae strain 19A Hungary 6 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 19A Hungary 6 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 19A Hungary 6 AI surface protein such as orf3 — 19AH, orf4 — 19AH, or orf5 — 19AH.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 19A Hungary 6 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 19A Hungary 6 AI proteins and one or more AI proteins from one of any one of S. pneumoniae from TIGR4, 670, 14 CSR 10, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI GR4 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 19A Hungary 6 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 19F Taiwan 14 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 19F Taiwan 14 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 19FTW, ORF3 — 19FTW, ORF4 — 19FTW, ORF5 — 19FTW, ORF6 — 19FTW, ORF7 — 19FTW, ORF8 — 19FTW.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 19F Taiwan 14 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 19F Taiwan 14 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • Taiwan 14 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 19F Taiwan 14 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 19F Taiwan 14 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 19F Taiwan 14 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • the S. pneumoniae strain 19F Taiwan 14 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 19F Taiwan 14 ⁇ l may encode for at least one surface protein.
  • S. pneumoniae strain 19F Taiwan 14 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 19F Taiwan 14 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 19F Taiwan 14 AI surface protein such as orf3 — 19FTW, orf4 — 19FTW, or orf5 — 19FTW.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 19F Taiwan 14 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 14 CSR 10, 23F Taiwan 15, or 23F Poland 16, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 19F Taiwan 14 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 23F Tru 16 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 23F Tru 16 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 23FP, ORF3 — 23FP, ORF4 — 23FP, ORF5 — 23FP, ORF6 — 23FP, ORF7 — 23FP, and ORF8 — 23FP.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Tru 16 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Tru 16 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 23F Tru 16 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 23F Tru 16 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 23F Tru 16 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 23F Tru 16 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 23F Tru 16 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 23F Tru 16 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 23F Tru 16 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 23F Poland 16 AI may encode for at least one surface protein.
  • S. pneumoniae strain 23F Poland 16 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 23F Poland 16 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 23F Tru 16 AI surface protein such as orf3 — 23FP, orf4 — 23FP, or orf5 — 23FP.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 23F Tru 16 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 23F Tru 16 AI proteins and one or more AI proteins from any one or more S. pneumoniae strains of TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 14 CSR 10, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 23F Poland 16 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 23F Taiwan 15 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 23F Taiwan 15 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 23FTW, ORF3 — 23FTW, ORF4 — 23FTW, ORF5 — 23FTW, ORF6 — 23FTW, ORF7 — 23F[W, ORF8 — 23FTW.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 23F Taiwan 15 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 23F Taiwan 15 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 23F Taiwan 15 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 23F Taiwan 15 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 23F Taiwan 15 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 23F Taiwan 15 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 23F Taiwan 15 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 23F Taiwan 15 AI may encode for at least one surface protein.
  • S. pneumoniae strain 23F Taiwan 15 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 23F Taiwan 15 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 23F Taiwan 15 AI surface protein such as orf3 — 23FTW, orf4 — 23FTW, or orf5 — 23FTW.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 23F Taiwan 15 AI proteins and one or more AI proteins from any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 14 CSR 10, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 23F Taiwan 15 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 6B Finland 12 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 6B Finland 12 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 6BF, ORF3 — 6BF, ORF4 — 6BF, ORF5 — 6BF, ORF6 — 6BF, ORF7 — 6BF, ORF8 — 6BF.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Finland 12 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Finland 12 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 6B Finland 12 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 6B Finland 12 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 6B Finland 12 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 6B Finland 12 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 6B Finland 12 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 6B Finland 12 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 6B Finland 12 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 6B Finland 12 AI may encode for at least one surface protein.
  • S. pneumoniae strain 6B Finland 12 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 6B Finland 12 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 6B Finland 12 AI surface protein such as orf3 — 6BF, orf4 — 6BF, or orf5 — 6BF.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 6B Finland 12 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 6B Finland 12 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 6B Finland 12 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 6B Spain 2 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 6B Spain 2 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 6BSP, ORF3 — 6BSP, ORF4 — 6BSP, ORF5 — 6BSP, ORF6 — 6BSP, ORF7 — 6BSP, and ORF8 — 6BSP.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Spain 2 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Spain 2 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 6B Spain 2 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 6B Spain 2 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 6B Spain 2 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 6B Spain 2 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 6B Spain 2 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 6B Spain 2 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 6B Spain 2 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 6B Spain 2 AI may encode for at least one surface protein.
  • S. pneumoniae strain 6B Spain 2 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 6B Spain 2 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 6B Spain 2 AI surface protein such as orf3 — 6BSP, orf4 — 6BSP, or orf5 — 6BSP.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 6B Spain 2 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 6B Spain 2 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 14 CSR 10, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 6B Spain 2 AI may also include a transcriptional regulator.
  • the S. pneumoniae strain 9V Spain 3 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases.
  • the S. pneumoniae strain 9V Spain 3 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF2 — 9VSP, ORF3 — 9VSP, ORF4 — 9VSP, ORF5 — 9VSP, ORF6 — 9VSP, ORF7 — 9VSP, and ORF8 — 9VSP.
  • a preferred immunogenic composition of the invention comprises a S. pneumoniae strain 9V Spain 3 AI surface protein which may be formulated or purified in an oligomeric (pilis) form.
  • Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 9V Spain 3 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 9V Spain 3 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF.
  • one or more of the S. pneumoniae strain 9V Spain 3 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 9V Spain 3 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • the S. pneumoniae strain 9V Spain 3 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 9V Spain 3 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 9V Spain 3 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • S. pneumoniae strain 9V Spain 3 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • S. pneumoniae strain 9V Spain 3 AI may encode for at least one surface protein.
  • S. pneumoniae strain 9V Spain 3 AI may encode for at least two surface exposed proteins and at least one sortase.
  • S. pneumoniae strain 9V Spain 3 AI encodes for at least three surface exposed proteins and at least two sortases.
  • the AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 9V Spain 3 AI surface protein such as orf3 — 9VSP, orf4 — 9VSP, or orf5 — 9VSP.
  • the oligomeric, pilus-like structure may comprise numerous units of AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • the invention comprises a S. pneumoniae strain 9V Spain 3 AI protein in oligomeric form, preferably in a hyperoligomeric form.
  • the invention comprises a composition comprising one or more S. pneumoniae strain 9V Spain 3 AI proteins and one or more AI proteins from any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 14 CSR 10, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • S. pneumoniae strain 9V Spain 3 AI may also include a transcriptional regulator.
  • the S. pneumoniae oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an S. pneumoniae AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a S. pneumoniae bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the S. pneumoniae bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed AI protein.
  • the AI protein is in a hyperoligomeric form.
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein.
  • the invention therefore includes a method for manufacturing an S. pneumoniae oligomeric Adhesin Island surface antigen comprising culturing a S. pneumoniae bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the S. pneumoniae bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed Adhesin Island protein.
  • the Adhesin Island protein is in a hyperoligomeric form.
  • the S. pneumoniae bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • S. pneumoniae bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the S. pneumoniae bacteria with a plasmid encoding the AI protein.
  • the plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein.
  • the sequence encoding the AI protein within the S. pneumoniae bacterial genome may be deleted.
  • the promoter regulating the S. pneumoniae Adhesin Island may be modified to increase expression.
  • the invention further includes S. pneumoniae bacteria which have been adapted to produce increased levels of AI surface protein.
  • the invention includes S. pneumoniae bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein.
  • the S. pneumoniae of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the invention further includes S. pneumoniae bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface.
  • the S. pneumoniae bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide.
  • Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria.
  • Increased expression of a leader peptidase in S. pneumoniae may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation.
  • the S. pneumoniae bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes ” Infection and Immunity (2004) 72(6):3444-3450).
  • Streptococus gordonii See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine
  • non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis.
  • the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid.
  • the non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria.
  • the AI surface protein may be isolated from cell extracts or culture supernatants.
  • the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • the non-pathogenic Gram positive bacteria may be used to express any of the S. pneumoniae Adhesin Island proteins described herein.
  • the non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein.
  • the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase.
  • the AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with pathogenic S. pneumoniae.
  • FIGS. 190 A and B, and 193 - 195 provide examples of three methods successfully practiced by applicants to purify pili from S. pneumoniae TIGR4.
  • the Gram positive bacteria AI proteins described herein are useful in immunogenic compositions for the prevention or treatment of Gram positive bacterial infection.
  • the GBS AI surface proteins described herein are useful in immunogenic compositions for the prevention or treatment of GBS infection.
  • the GAS AI surface proteins described herein may be useful in immunogenic compositions for the prevention or treatment of GAS infection.
  • the S. pneumoniae AI surface proteins may be useful in immunogenic cojmpositions for the prevention or treatment of S. pneumoniae infection.
  • Gram positive bacteria AI surface proteins that can provide protection across more than one serotype or strain isolate may be used to increase immunogenic effectiveness.
  • a particular GBS AI surface protein having an amino acid sequence that is at least 50% (i.e., at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%) homologous to the particular GBS AI surface protein of at least 2 (i.e., at least 3, 4, 5, 6, 7, 8, 9, 10, or more) other GBS serotypes or strain isolates may be used to increase the effectiveness of such compositions.
  • fragments of Gram positive bacteria AI surface proteins that can provide protection across more than one serotype or strain isolate may be used to increase immunogenic effectiveness.
  • a fragment may be identified within a consensus sequence of a full length amino acid sequence of a Gram positive bacteria AI surface protein.
  • Such a fragment can be identified in the consensus sequence by its high degree of homology or identity across multiple (i.e, at least 3, 4, 5, 6, 7, 8, 9, 10, or more) Gram positive bacteria serotypes or strain isolates.
  • a high degree of homology is a degree of homology of at least 90% (i.e., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) across Gram positive bacteria serotypes or strain isolates.
  • a high degree of identity is a degree of identity of at least 90% (i.e., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) across Gram positive bacteria serotypes or strain isolates.
  • such a fragment of a Gram positive bacteria AI surface protein may be used in the immunogenic compositions.
  • AI surface protein oligomeric pilus structures may be formulated or purified for use in immunization. Isolated AI surface protein oligomeric pilus structures may also be used for immunization.
  • the invention includes an immunogenic composition comprising a first Gram positive bacteria AI protein and a second Gram positive bacterial AI protein.
  • One or more of the AI proteins may be a surface protein.
  • Such surface proteins may contain an LPXTG motif or other sortase substrate motif.
  • the first and second AI proteins may be from the same or different genus or species of Gram positive bacteria. If within the same species, the first and second AI proteins may be from the same or different AI subtypes. If two AIs are of the same subtype, the AIs have the same numerical designation. For example, all AIs designated as AI-1 are of the same AI subtype. If two AIs are of a different subtype, the AIs have different numerical designations. For example, AI-1 is of a different AI subtype from AI-2, AI-3, AI-4, etc. Likewise, AI-2 is of a different AI subtype from AI-1, AI-3, and AI-4, etc.
  • the invention includes an immunogenic composition comprising one or more GBS AI-1 proteins and one or more GBS AI-2 proteins.
  • One or more of the AI proteins may be a surface protein.
  • Such surface proteins may contain an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) and may bind fibrinogen, fibronectin, or collagen.
  • One or more of the AI proteins may be a sortase.
  • the GBS AI-1 proteins may be selected from the group consisting of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648.
  • the GBS AI-1 proteins include GBS 80 or GBS 104.
  • the GBS AI-2 proteins may be selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • the GBS AI-2 proteins are selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406.
  • the GBS AI-2 proteins may be selected from the group consisting of 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • the GBS AI-2 protein includes GBS 59 or GBS 67.
  • the invention includes an immunogenic composition comprising one or more of any combination of GAS AI-1, GAS AI-2, GAS AI-3, or GAS AI-4 proteins.
  • GAS AI proteins may be a sortase.
  • the GAS AI-1 proteins may be selected from the group consisting of M6_Spy0156, M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • the GAS AI-1 proteins are selected from the group consisting of M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • the GAS AI-2 proteins may be selected from the group consisting of Spy0124, GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • the GAS AI-2 proteins are selected from the group consisting of GAS 15, GAS 16, and GAS 18.
  • the GAS AI-3 proteins may be selected from the group consisting of SpyM3 — 0097, SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, SpyM3 — 0104, SPs0099, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, SPs0106, orf77, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM18 — 0125, spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, spyM18 — 0132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM
  • the GAS AI-3 proteins are selected from the group consisting of SpyM3 — 0097, SpyM3 — 0098, SpyM3 — 0099, SpyM3 — 0100, SpyM3 — 0101, SpyM3 — 0102, SpyM3 — 0103, and SpyM3 — 0104.
  • the GAS AI-3 proteins are selected from the group consisting of SPs0099, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, and SPs0106.
  • the GAS AI-3 proteins are selected from the group consisting of orf77, orf78, orf79, orf80, orf81, orf82, orf83, and orf84.
  • the GAS AI-3 proteins are selected from the group consisting of spyM18 — 0125, spyM18 — 0126, spyM18 — 0127, spyM18 — 0128, spyM18 — 0129, spyM18 — 0130, spyM18 — 0131, and spyM18 — 0132.
  • GAS AI-3 proteins are selected from the group consisting of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149.
  • the GAS AI-4 proteins may be selected from the group consisting of 19224133, 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • the GAS-AI-4 proteins are selected from the group consisting of 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • the invention includes an immunogenic composition comprising one or more of any combination of S. pneumonaie from TIGR4 , S. pneumonaie strain 670 , S. pneumonaie from 19A Hungary 6 , S. pneumonaie from 6B Finland 12 , S. pneumonaie from 6B Spain 2 , S. pneumonaie from 9V Spain 3 , S. pneumonaie from 14 CSR 10 , S. pneumonaie from 19F Taiwan 14 , S. pneumonaie from 23F Taiwan 15, or S. pneumonaie from 23F Poland 16 AI proteins.
  • One or more of the AI proteins may be a surface protein. Such surface proteins may contain an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) and may bind fibrinogen, fibronectin, or collagen.
  • One or more of the AI proteins may be a sortase.
  • the S. pneumonaie from TIGR4 AI proteins may be selected from the group consisting of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, SP0468.
  • the S. pneumonaie from TIGR4 AI proteins include SP0462, SP0463, or SP0464.
  • the S. pneumonaie strain 670 AI proteins may be selected from the group consisting of Orf1 — 670, Orf3 — 670, Orf4 — 670, Orf5 — 670, Orf6 — 670, Orf7 — 670, and Orf8 — 670.
  • the S. pneumonaie strain 670 AI proteins include Orf3 — 670, Orf4 — 670, or Orf5 — 670.
  • the S. pneumonaie from 19A Hungary 6 AI proteins may be selected from the group consisting of ORF2 — 1 gAH, ORF3 — 1 gAH, ORF4 — 19AH, ORF5 — 19AH, ORF6 — 1 gAH, ORF7 — 19AH, or ORF8 — 19AH.
  • the S. pneumonaie from 6B Finland 12 AI proteins may be selected from the group consisting of ORF2 — 6BF, ORF3 — 6BF, ORF4 — 6BF, ORF5 — 6BF, ORF6 — 6BF, ORF7 — 6BF, or ORF8 — 6BF.
  • the S. pneumonaie from 6B Spain 2 AI proteins may be selected from the group consisting of ORF2 — 6BSP, ORF3 — 6BSP, ORF4 — 6BSP, ORF5 — 6BSP, ORF6 — 6BSP, ORF7 — 6BSP, or ORF8 — 6BSP.
  • the S. pneumonaie from 9V Spain 3 AI proteins may be selected from the group consisting of ORF2 — 9VSP, ORF3 — 9VSP, ORF4 — 9VSP, ORF5 — 9VSP, ORF6 — 9VSP, ORF7 — 9VSP, or ORF8 — 9VSP.
  • the S. pneumonaie from 14 CSR 10 AI proteins may be selected from the group consisting of ORF2 — 14CSR, ORF3 — 14CSR, ORF4 — 14CSR, ORF5 — 14CSR, ORF6 — 14CSR, ORF7 — 14CSR, or ORF8 — 14CSR.
  • the S. pneumonaie from 19F Taiwan 14 AI proteins may be selected from the group consisting of ORF2 — 1 gFTW, ORF3 — 19FTW, ORF4 — 19FTW, ORF5 — 19FTW, ORF6 — 19FTW, ORF7 — 19FTW, or ORF8 — 19FTW.
  • the S. pneumonaie from 23F Taiwan 15 AI proteins may be selected from the group consisting of ORF2 — 23FTW, ORF3 — 23FTW, ORF4 — 23FTW, ORF5 — 23FTW, ORF6 — 23FTW, ORF7 — 23FTW, or ORF8 — 23FTW.
  • the S. pneumonaie from 23F Poland 16 AI proteins may be selected from the group consisting of ORF2 — 23FP, ORF3 — 23FP, ORF4 — 23FP, ORF5 — 23FP, ORF6 — 23FP, ORF7 — 23FP or ORF8 — 23FP.
  • the Gram positive bacteria AI proteins included in the immunogenic compositions of the invention can provide protection across more than one serotype or strain isolate.
  • the immunogenic composition may comprise a first AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different serotypes of a Gram positive bacteria.
  • the first AI protein may also be homologous to the amino acid sequence of a third AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different serotypes of a Gram positive bacteria.
  • the first AI protein may also be homologous to the amino acid sequence of a fourth AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different serotypes of a Gram positive bacteria.
  • the GBS AI proteins included in the immunogenic compositions of the invention can provide protection across more than one GBS serotype or strain isolate.
  • the immunogenic composition may comprise a first GBS AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second GBS AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different GBS serotypes.
  • the first GBS AI protein may also be homologous to the amino acid sequence of a third GBS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GBS serotypes.
  • the first AI protein may also be homologous to the amino acid sequence of a fourth GBS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GBS serotypes.
  • the first AI protein may be selected from an AI-1 protein or an AI-2 protein.
  • the first AI protein may be a GBS AI-1 surface protein such as GBS 80.
  • GBS 80 The amino acid sequence of GBS 80 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 80 amino acid sequence from GBS serotype III, strain isolates NEM316 and COH1 and the GBS 80 amino acid sequence from GBS serotype 1a, strain isolate A909.
  • the first AI protein may be GBS 104.
  • the amino acid sequence of GBS 104 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 104 amino acid sequence from GBS serotype III, strain isolates NEM316 and COH1, the GBS 104 amino acid sequence from GBS serotype 1a, strain isolate A909, and the GBS 104 amino acid sequence serotype II, strain isolate 18RS21.
  • Table 12 provides the amino acid sequence identity of GBS 80 and GBS 104 across GBS serotypes Ia, Ib, II, III, V, and VIII.
  • the GBS strains in which genes encoding GBS 80 and GBS 104 were identified share, on average, 99.88 and 99.96 amino acid sequence identity, respectively. This high degree of amino acid identity indicates that an immunogenic composition comprising a first protein of GBS 80 or GBS 104 may provide protection across more than one GBS serotype or strain isolate.
  • GBS 80 GBS 104 Serotype Strains cGH % AA identity cGH % AA identity Ia 090 + 99.79 + 100.00 A909 + 100.00 + 100.00 515 ⁇ ⁇ DK1 ⁇ ⁇ DK8 ⁇ ⁇ Davis ⁇ ⁇ Ib 7357b + 100.00 + H36B ⁇ ⁇ II 18RS21 ⁇ + 100.00 DK21 ⁇ ⁇ III NEM316 + 100.00 + 100.00 COH31 + 100.00 + D136 + 100.00 + M732 + 100.00 + 99.88 COH1 + 99.79 + 99.88 M781 + 99.79 + 99.88 No type CJB110 + 99.37 + 100.00 1169NT ⁇ ⁇ V CJB111 + 100.00 + 100.00 2603 + 100.00 + 100.00 VIII JM130013 + 99.79 + 100.00 SMU014 + 100.00 + total 14/22 99.88 +/ ⁇ 0.19 15/22 99
  • the first AI protein may be an AI-2 protein such as GBS 67.
  • the amino acid sequence of GBS 67 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 67 amino acid sequence from GBS serotype III, strain isolate NEM316, the GBS 67 amino acid sequence from GBS serotype 1b, strain isolate H36B, and the GBS 67 amino acid sequence from GBS serotype II, strain isolate 17RS21.
  • the first AI protein may be an AI-2 protein such as spb1.
  • the amino acid sequence of spb1 from GBS serotype III, strain isolate COH1 is greater than 90% homologous to the spb1 amino acid sequence from GBS serotype Ia, strain isolate A909.
  • the first AI protein may be an AI-2 protein such as GBS 59.
  • the amino acid sequence of GBS 59 from GBS serotype II, strain isolate 18RS21 is 100% homologous to the GBS 59 amino acid sequence from GBS serotype V, strain isolate 2603.
  • the amino acid sequence of GBS 59 from GBS serotype V, strain isolate CJB111 is 98% homologous to the GBS 59 amino acid sequence from GBS serotype III, strain isolate NEM316.
  • compositions of the invention may also be designed to include Gram positive AI proteins from divergent serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of serotypes or strain isolates of a Gram positive bacteria and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • the invention may include an immunogenic composition comprising a first and second Gram positive bacteria AI protein, wherein a polynucleotide sequence encoding for the full length sequence of the first AI protein is not present in a similar Gram positive bacterial genome comprising a polynucleotide sequence encoding for the second AI protein.
  • compositions of the invention may also be designed to include AI proteins from divergent GBS serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of GBS serotypes or strain isolates and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • the invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein a polynucleotide sequence encoding for the full length sequence of the first GBS AI protein is not present in a genome comprising a polynucleotide sequence encoding for the second GBS AI protein.
  • the first AI protein could be GBS 80 (such as the GBS 80 sequence from GBS serotype V, strain isolate 2603).
  • the sequence for GBS 80 in GBS sertoype II, strain isolate 18RS21 is disrupted.
  • the second AI protein could be GBS 104 or GBS 67 (sequences selected from the GBS serotype II, strain isolate 18RS21).
  • the invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein the first GBS AI protein has detectable surface exposure on a first GBS strain or serotype but not a second GBS strain or serotype and the second GBS AI protein has detectable surface exposure on a second GBS strain or serotype but not a first GBS strain or serotype.
  • the first AI protein could be GBS 80 and the second AI protein could be GBS 67.
  • Table 15 there are some GBS serotypes and strains that have surface exposed GBS 80 but that do not have surface exposed GBS 67 and vice versa.
  • An immunogenic composition comprising a GBS 80 and a GBS 67 protein may provide protection across a wider group of GBS strains and serotypes.
  • TABLE 15 Antigen surface exposure of GBS 80, GBS 322, GBS 104, and GBS 67 GBS strains
  • Type GBS 80 GBS 322 GBS 104 GBS 67 DK1* Ia 0 nd 237 478 DK8* 0 213 151 475 Davis* 0 86 271 430 515* 0 227 262 409 090 0 0 0 0 A909 0 0 0 0 2986 0 0 157 397 5551 0 36 384 485 2177 Ib 477 323 328 66 H36B* 0 105 518 444 7357b- 91 102 309 316 2129 57 71 132 0 5518 31 nd 60 28 COH1 III 305 130 305 0 D136C 16 460 226 406 COH31
  • the invention may include an immunogenic composition comprising a first and second Gram positive bacteria AI protein, wherein the polynucleotide sequence encoding the sequence of the first AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second AI protein.
  • the invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein the polynucleotide sequence encoding the sequence of the first GBS AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second GBS AI protein.
  • the first GBS AI protein could be GBS 67 (such as the GBS 67 sequence from GBS serotype 1b, strain isolate H36B). As shown in FIGS.
  • the GBS 67 sequence for this strain is less than 90% homologous (87%) to the corresponding GBS 67 sequence in GBS serotype V, strain isolate 2603.
  • the second GBS AI protein could then be the GBS 80 sequence from GBS serotype V, strain isolate 2603.
  • An example immunogenic composition of the invention may comprise adhesin island proteins GBS 80, GBS 104, GBS 67, and GBS 59, and non-AI protein GBS 322.
  • FACS analysis of different GBS strains demonstrates that at least one of these five proteins is always found to be expressed on the surface of GBS bacteria.
  • FIG. 227 provides the FACS data obtained for surface exposure of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 on each of 37 GBS strains.
  • FIG. 228 provides the FACS data obtained for surface exposure of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 on each of 41 GBS strains obtained from the CDC.
  • each GBS strain had surface expression of at least one of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59.
  • the surface exposure of at least one of these proteins on each bacterial strain indicates that an immunogenic composition comprising these proteins will provide wide protection across GBS strains and serotypes.
  • the surface exposed GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 proteins are also present at high levels as determined by FACS.
  • Table 49 summarizes the FACS results for the initial 70 GBS strains examined for GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 surface expression.
  • a protein was designated as having high levels of surface expression of a protein if a five-fold shift in fluorescence was observed when using antibodies for the protein relative to preimmune control serum.
  • Table 50 details which of the surface proteins is highly expressed on the different GBS serotype. TABLE 50 High Levels of Surface Protein Expression on GBS Serotypes 5-fold shift in fluorescence by FACS GBS 80 GBS 104 GBS 67 GBS 59 GBS 322 Ia + Ib + III 4/36 2/36 22/36 20/36 18/36 II + V 11/25 9/25 21/25 21/25 13/25 Others 2/9 3/9 6/9 5/9 2/9
  • the immunogenic composition of the invention may include GBS 80, GBS 104, GBS 67, and GBS 322. Assuming that protein antigens that are highly accessible to antibodies confer 100% protection with suitable adjuvants, an immunogenic composition containing GBS 80, GBS 104, GBS 67, GBS 59 and GBS 322 will provide protection for 89% of GBS strains and serotypes, the same percentage as an immunogenic composition containing GBS 80, GBS 104, GBS 67, and GBS 322 proteins. See FIG. 229 . However, it may be preferable to include GBS 59 in the composition to increase its immunogenic strength.
  • GBS 59 is highly expressed on the surface two-thirds of GBS bacteria examined by FACS analysis, unlike GBS 80, GBS 104, and GBS 322, which are highly expressed in less than half of GBS bacteria examined.
  • GBS 59 opsonophagocytic activity is also comparable to that of a mix of GBS 322, GBS 104, GBS 67, and GBS 80 proteins. See FIG. 230 .
  • the GAS AI proteins included in the immunogenic compositions of the invention can provide protection across more than one GAS serotype or strain isolate.
  • the immunogenic composition may comprise a first GAS AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second GAS AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different GAS serotypes.
  • the first GAS AI protein may also be homologous to the amino acid sequence of a third GAS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GAS serotypes.
  • the first AI protein may also be homologous to the amino acid sequence of a fourth GAS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GAS serotypes.
  • compositions of the invention may also be designed to include GAS AI proteins from divergent serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of serotypes or strain isolates of a GAS bacteria and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • the first AI protein could be a prtF2 protein (such as the 19224141 protein from GAS serotype M12, strain isolate A735).
  • the sequence for a prtF2 protein is not present in GAS AI types 1 or 2.
  • the second AI protein could be collagen binding protein M6_Spy0159 (from M6 isolate (MGAS10394), which comprises an AI-1) or GAS15 (from M1 isolate (SF370), which comprises an AI-2).
  • the invention may include an immunogenic composition comprising a first and second GAS AI protein, wherein the first GAS AI protein has detectable surface exposure on a first GAS strain or serotype but not a second GAS strain or serotype and the second GAS AI protein has detectable surface exposure on a second GAS strain or serotype but not a first GAS strain or serotype.
  • the invention may include an immunogenic composition comprising a first and second GAS AI protein, wherein the polynucleotide sequence encoding the sequence of the first GAS AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second GAS AI protein.
  • the first and second GAS AI proteins are subunits of the pilus.
  • the first and second GAS AI proteins are selected from the major pilus forming proteins (i.e., M6_Spy0160 from M6 strain 10394, SPy0128 from M1 strain SF370, SpyM3 — 0100 from M3 strain 315, SPs0102 from M3 strain SSI, orf80 from M5 isolate Manfredo, spyM18 — 0128 from M18 strain 8232, SpyoM01000153 from M49 strain 591, 19224137 from M12 strain A735, fimbrial structural subunit from M77 strain ISS4959, fimbrial structural subunit from M44 strain ISS3776, fimbrial structural subunit from M50 strain ISS3776 ISS 4538, fimbrial structural subunit from M12 strain CDC SS635, fimbrial structural subunit from M23 strain DSM2071, fimbrial structural subunit from M6 strain CDC SS410).
  • M6_Spy0160 from M6 strain 10394
  • Table 45 provides the percent identity between the amino acidic sequences of each of the main pilus forming subunits from GAS AI-1, AI-2, AI-3, and AI-4 representative strains (i.e., M6_Spy0160 from M6 strain 10394, SPy0128 from M1 strain SF370, SpyM3 — 0100 from M3 strain 315, SPs0102 from M3 strain SSI, orf80 from M5 isolate Manfredo, spyM18 — 0128 from M18 strain 8232, SpyoM01000153 from M49 strain 591, 19224137 from M12 strain A735, Fimbrial structural subunit from M77 strain ISS4959, fimbrial structural subunit from M44 strain ISS3776, fimbrial structural subunit from M50 strain ISS3776 ISS 4538, fimbrial structural subunit from M12 strain CDC SS635, fimbrial structural subunit from M23 strain DSM2071, fimbrial structural subunit from M6 strain CDC SS410
  • the first main pilus subunit may be selected from bacteria of GAS serotype M6 strain 10394 and the second main pilus subunit may be selected from bacteria of GAS serotype M1 strain 370.
  • the main pilus subunits encoded by these strains of bacteria share only 23% nucleotide identity.
  • An immunogenic composition comprising pilus main subunits from each of these strains of bacteria is expected to provide protection across a wider group of GAS strains and serotypes.
  • main pilus subunits that can be used in combination to provide increased protection across a wider range of GAS strains and serotypes include proteins encoded by GAS serotype M5 Manfredo isolate and serotype M6 strain 10394, which share 23% sequence identity, GAS serotype M18 strain 8232 and serotype M1 strain 370, which share 38% sequence identity, GAS serotype M3 strain 315 and serotype M12 strain A735, which share 61% sequence identity, and GAS serotype M3 strain 315 and serotype M6 strain 10394 which share 25% sequence identity.
  • FIGS. 198-201 provide further tables comparing the percent identity of adhesin island-encoded surface exposed proteins for different GAS serotypes relative to other GAS serotypes harbouring an adhesin island of the same or a different subtype (GAS AI-1, GAS AI-2, GAS AI-3, and GAS AI-4). See also further discussion below.
  • Applicants have discovered that surface exposure of GBS 104 is dependent on the concurrent expression of GBS 80.
  • reverse transcriptase PCR analysis of AI-1 shows that all of the AI genes are co-transcribed as an operon.
  • Applicants constructed a series of mutant GBS containing in frame deletions of various AI-1 genes. (A schematic of the GBS mutants is presented in FIG. 7 ). FACS analysis of the various mutants comparing mean shift values using anti-GBS 80 versus anti-GBS 104 antibodies is presented in FIG. 8 . Removal of the GBS 80 operon prevented surface exposure of GBS 104; removal of the GBS 104 operon did not affect surface exposure of GBS 80.
  • GBS 80 is involved in the transport or localization of GBS 104 to the surface of the bacteria.
  • the two proteins may be oligomerized or otherwise associated. It is possible that this association involves a conformational change in GBS 104 that facilitates its transition to the surface of the GBS bacteria.
  • FIG. 68 shows that polyclonal anti-GBS 104 antibodies (see lane marked ⁇ -104 POLIC.) cross-hybridize with smaller structures than do polyclonal anti-GBS 80 antibodies (see lane marked ⁇ -GBS 80 POLIC.).
  • sortases within the adhesin island also appear to play a role in localization and presentation of the surface proteins.
  • FACS analysis of various sortase deletion mutants showed that removal of sortase SAG0648 prevented GBS 104 from reaching the surface and slightly reduced the surface exposure of GBS 80.
  • sortase SAG0647 and sortase SAG0648 were both knocked out, neither GBS 80 nor GBS 104 were surface exposed. Expression of either sortase alone was sufficient for GBS 80 to arrive at the bacterial surface. Expression of SAG0648, however, was required for GBS 104 surface localization.
  • compositions of the invention may include two or more AI proteins, wherein the AI proteins are physically or chemically associated.
  • the two AI proteins may form an oligomer.
  • the associated proteins are two AI surface proteins, such as GBS 80 and GBS 104.
  • the associated proteins may be AI surface proteins from different adhesin islands, including host cell adhesin island proteins if the AI surface proteins are expressed in a recombinant system.
  • the associated proteins may be GBS 80 and GBS 67.
  • Adhesin Island or “AI” refers to a series of open reading frames within a bacterial genome that encode for a collection of surface proteins and sortases.
  • An Adhesin Island may encode for amino acid sequences comprising at least one surface protein.
  • the Adhesin Island may encode at least one surface protein.
  • an Adhesin Island may encode for at least two surface proteins and at least one sortase.
  • an Adhesin Island encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • One or more AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • Gram positive adhesin islands of the invention preferably include a divergently transcribed transcriptional regulator.
  • the transcriptional regulator may regulate the expression of the AI operon.
  • the invention includes a composition comprising one or more Gram positive bacteria AI surface proteins.
  • AI surface proteins may be associated in an oligomeric or hyperoligomeric structure.
  • Preferred Gram positive adhesin island proteins for use in the invention may be derived from Staphylococcus (such as S. aureus ), Streptococcus (such as S. agalactiae (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans ), Enterococcus (such as E. faecalis and E. faecium ), Clostridium (such as C. difficile ), Listeria (such as L. monocytogenes ) and Corynebacterium (such as C. diphtheria ).
  • Staphylococcus such as S. aureus
  • Streptococcus such as S. agalactiae (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans
  • Enterococcus such as E. faecalis and E. faecium
  • Clostridium such as C
  • Gram positive AI surface protein sequences typically include an LPXTG motif or other sortase substrate motif.
  • Gram positive AI surface proteins of the invention may affect the ability of the Gram positive bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of Gram positive bacteria to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. Gram positive AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • Gram positive AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins.
  • a Gram positive bacteria AI may encode for at least one surface exposed protein.
  • the Adhesin Island may encode at least one surface protein.
  • a Gram positive bacteria AI may encode for at least two surface exposed proteins and at least one sortase.
  • a Gram positive AI encodes for at least three surface exposed proteins and at least two sortases.
  • Gram positive AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase.
  • the sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif.
  • the sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II.
  • the precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • Gram positive bacteria AI surface proteins of the invention will contain an N-terminal leader or secretion signal to facilitate translocation of the surface protein across the bacterial membrane.
  • Gram positive bacteria AI surface proteins of the invention may affect the ability of the Gram positive bacteria to adhere to and invade target host cells, such as epithelial cells.
  • Gram positive bacteria AI surface proteins may also affect the ability of the gram positive bacteria to translocate through an epithelial cell layer.
  • one or more of the Gram positive AI surface proteins are capable of binding to or other associating with an epithelial cell surface.
  • one or more Gram positive AI surface proteins may bind to fibrinogen, fibronectin, or collagen protein.
  • the invention includes a composition comprising oligomeric, pilus-like structures comprising a Gram positive bacteria AI surface protein.
  • the oligomeric, pilus-like structure may comprise numerous units of the AI surface protein.
  • the oligomeric, pilus-like structures comprise two or more AI surface proteins.
  • the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof.
  • the oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif.
  • the oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • Gram positive bacteria AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include one or both of a pilin motif comprising a conserved lysine residue and an E box motif comprising a conserved glutamic acid residue.
  • the oligomeric, pilus like structures may be used alone or in the combinations of the invention.
  • the invention comprises a Gram positive bacteria Adhesin Island in oligomeric form, preferably in a hyperoligomeric form.
  • the oligomeric, pilus-like structures of the invention may be combined with one or more additional Gram positive AI proteins (from the same or a different Gram positive species or genus).
  • the oligomeric, pilus-like structures comprise one or more Gram positive bacteria AI surface proteins in combination with a second Gram positive bacteria protein.
  • the second Gram positive bacteria protein may be a known antigen, and need not normally be associated with an AI protein.
  • the oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing a Gram positive bacteria AI surface protein.
  • the invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a Gram positive bacteria adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the Gram positive bacteria.
  • the AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface.
  • the method may further comprise purification of the expressed Adhesin Island protein.
  • the Adhesin Island protein is in a hyperoligomeric form.
  • Gram positive bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • Gram positive bacteria may be adapted to increase AI protein expression by means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the Gram positive bacteria with a plasmid encoding the AI protein.
  • the plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein.
  • the sequence encoding the AI protein within the Gram positive bacterial genome may be deleted.
  • the promoter regulating the Gram positive Adhesin Island may be modified to increase expression.
  • the invention further includes Gram positive bacteria which have been adapted to produce increased levels of AI surface protein.
  • the invention includes Gram positive bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein.
  • the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • the invention further includes Gram positive bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface.
  • the Gram positive bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide.
  • Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria.
  • Increased expression of a leader peptidase in Gram positive may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation.
  • the Gram positive bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal VaccineMade from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes ” Infection and Immunity (2004) 72(6):3444-3450). It has already been demonstrated, above, that L. lactis expresses GBS and GAS AI polypeptides in oligomeric form and on its surface.
  • the oligomeric, pilus-like structures may be produced recombinantly.
  • the Gram positive bacteria AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention.
  • AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • Gram positive AI Sortases of the invention will typically have a signal peptide sequence within the first 70 amino acid residues. They may also include a transmembrane sequence within 50 amino acid residues of the C terminus. The sortases may also include at least one basic amino acid residue within the last 8 amino acids. Preferably, the sortases have one or more active site residues, such as a catalytic cysteine and histidine.
  • Adhesin island surface proteins from two or more Gram positive bacterial genus or species may be combined to provide an immunogenic composition for prophylactic or therapeutic treatment of disease or infection of two more Gram positive bacterial genus or species.
  • the adhesin island surface proteins may be associated together in an oligomeric or hyperoligomeric structure.
  • the invention comprises an adhesin island surface proteins from two or more Streptococcus species.
  • the invention includes a composition comprising a GBS AI surface protein and a GAS adhesin island surface protein.
  • the invention includes a composition comprising a GAS adhesin island surface protein and a S. pneumoniae adhesin island surface protein.
  • the invention comprises an adhesin island surface protein from two or more Gram positive bacterial genus.
  • the invention includes a composition comprising a Streptococcus adhesin island protein and a Corynebacterium adhesin island protein.
  • adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fasciitis.
  • post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases.
  • the first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused.
  • the bacteria secrete a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection.
  • the final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart.
  • a general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above.
  • the M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation.
  • the carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci.
  • the amino terminus which extends through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • T-antigen A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen.
  • T-antigen a variable, trypsin-resistant surface antigen
  • Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens.
  • Antisera to define T types are commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • T-antigen T-type 6
  • M6 strain of GAS M6 strain of GAS
  • FCT Fibronectin-binding, Collagen-binding T-antigen
  • the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the tee6 gene product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used. See FIG. 163A , last lane labeled “M6 Tee6.”
  • the FCT region in M6_ISS3650 contains two other genes (prtF1 and cpa) predicted to code for surface exposed proteins; these proteins are characterized as containing the cell wall attachment motif LPXTG.
  • Western blot analysis using antiserum specific for PrtF1 detected a single molecular species with electrophoretic mobility corresponding to the predicted molecular mass of the protein and one smaller band of unknown origin.
  • Western blot analysis using antisera specific for Cpa recognized a high molecular weight covalently linked ladder ( FIG. 163A , second lane).
  • Immunogold labelling of Cpa with specific antiserum followed by transmission electron microscopy detected an abundance of Cpa at the cell surface and only occasional structures extending from the cell surface ( FIG. 163J ).
  • FCT region Four classes of FCT region can be discerned by the types and order of the genes contained within the region.
  • the FCT region of strains of types M3, M5, M18 and M49 have a similar organization whereas those of M6, M1 and M12 differ. See FIG. 164 .
  • these four FCT regions correlate to four GAS Adhesin Island types (AI-1, AI-2, AI-3 and AI-4).
  • M1 strain SF370 there are three predicted surface proteins (Cpa (also referred to as M1 — 126 and GAS 15), M1 — 128 (a fimbrial protein also referred to as Spy0128 and GAS 16), and M1 — 130 (also referred to as Spy0130 and GAS 18)) (GAS AI-2).
  • Antisera specific for each surface protein reacted with a ladder of high molecular weight material ( FIG. 163B ).
  • Immunogold staining of M1 strain SF370 with antiserum specific for M1 — 128 revealed pili structures similar to those seen when M6 strain ISS3650 was immunogold stained with antiserum specific for tee6 (See FIG. 1163K ).
  • Antisera specific for surface proteins Cpa and M1 — 130 revealed abundant surface staining and occasional structures extending from the surface of M1 strain SF370 bacteria ( FIG. 163S ).
  • the M1 — 128 protein appears to be necessary for polymerization of Cpa and M1 — 130 proteins. If the M1 — 128 gene in M1_SF370 was deleted, Western blot analysis using antibodies that hybridize to Cpa and M1 — 130 no longer detected high molecular weight ladders comprising the Cpa and M1 — 130 proteins ( FIG. 163 E ). See also FIGS. 177 A-C which provide the results of Western blot analysis of the M1 — 128 ( ⁇ 128) deleted bacteria using anti-M1 — 130 antiserum ( FIG. 177 A ), anti-M1 — 128 antiserum ( FIG. 177 B ), and anti-M1 — 126 antiserum ( FIG. 177 C ).
  • High molecular weight ladders indicative of pilus formation on the surface of M1 strain SF370, could not be detected by any of the three antisera in ⁇ 128 bacteria. If the ⁇ 128 bacteria were transformed with a plasmid containing the gene for M1 — 128, Western blot analysis using antisera specific for Cpa and M1 — 130 again detected high molecular weight ladders ( FIG. 163 H ).
  • FIGS. 177 A-C provide Western blot analysis results of the M1 — 130 deleted ( ⁇ 130) strain SF370 bacteria using anti-M1 — 130 ( FIG. 177 A ), anti-M1 — 128 ( FIG. 177 B ), and anti-M1 — 126 antiserum ( FIG. 177 C ).
  • composition of the pili in GAS resembles that previously described for both C. diphtheria (7, 8) and S. agalactiae (described above) (9) in that each pilus is formed by a backbone component which abundantly stains the pili in EM and is essential for the incorporation of the other components.
  • FIGS. 177 A-C provide Western blot analysis of the SrtC1 deleted ( ⁇ srtC1) strain SF370 bacteria using anti-M1 — 130 ( FIG. 177 A ), anti-M1 — 128 ( FIG. 177 B ), and anti-M1 — 126 antiserum ( FIG. 177 C ). None of the three antisera immunoreacted with high molecular weight structures (pili) in the ⁇ SrtC1 bacteria.
  • FIG. 179 G-I show a shift in fluorescence when antibodies immunoreactive to M1 — 126 ( FIG. 179 G ), M1 — 128 ( FIG. 179 H ), and M1 — 130 ( FIG. 179 I ) are used to detect cell surface protein expression on ⁇ SrtC1 bacteria.
  • SrtC1 deletion prevents pilus formation, but not surface anchoring of proteins involved in pilus formation on the surface of bacteria.
  • Another sortase is possibly involved in anchoring of the proteins to the bacteria surface.
  • Pilus polymerization in C. diphtheriae is also dependent on particular sortase enzyme whose gene resides at the same genetic locus as the pilus components (7, 8).
  • LepA signal peptidase Spy0127
  • LepA deletion mutants ( ⁇ LepA) of strain SF370 fail to assemble pili on the cell surface. Not only are the ⁇ LepA mutants unable to assemble pili, they are also deficient at cell surface M1 expression. See FIG. 180 , which provides a FACS analysis of the wildtype (A) and ⁇ LepA mutant (B) SF370 bacteria using M1 antisera. No shift in fluorescence is observed for the ⁇ LepA mutant bacteria in the presence of M1 immune serum. It is possible that these deletion mutants of LepA will be useful for detecting non-M, non-pili, surface exposed antigens on the surface of GAS, or any Gram positive bacteria. These antigens may also be useful in immunogenic compositions.
  • M5 strain ISS4882 contains genes for four predicted surface exposed proteins (GAS AI-3). Antisera against three of the four products of the FCT region (GAS AI-3) of M5_ISS4883 (Cpa, M5_orf80, M5_orf82) stained high molecular weight ladders in Western blot analysis ( FIG. 163 C ). Long pili were visible when antisera against M5_orf80 was used in immunogold staining followed by electron microscopy ( FIG. 163L ).
  • the M12 strain 20010296 contains genes for five predicted surface exposed proteins.
  • GAS AI-4 Antisera against three of the five products of the FCT region (GAS AI-4) of M12 — 20010296 (Cpa, EftLSL.A, Orf2) stained high molecular weight ladders in Westen blot analysis ( FIG. 163 D ). Long pili were visible when antisera against EftLSL.A were used ( FIG. 163M ).
  • the major pilus forming proteins identified in the four strains studied by applicants share between 23% and 65% amino acid identity in any pairwise comparison, indicating that each pilus may represent a different Lancefield T-antigen.
  • Each pilus is part of a trypsin resistant structure on the GAS bacteria surface, as is the case for the Lancefield T antigens. See FIG. 165 , which provides a FACS analysis of bacteria harboring each of the FCT types that had or had not been treated with trypsin (6).
  • the pili structures identified on the surface of the GAS bacteria were confirmed to be Lancefield T antigens when commercially available T-serotyping sera detected the pili on the surface of bacteria.
  • Western blot analysis was initially performed to determine if polyvalent serum pools (designated T, U, W, X, and Y) could detect recombinant proteins for each of the major pilis components (T6, M1 — 128, M5_orf80 and EftLSL.A) identified in the strains of bacteria discussed above.
  • Pool U which contains the T6 serum, recognized the T6 protein specifically (a surface exposed pilus protein from GAS AI-1) ( FIG. 166 B ).
  • Pool T specifically recognized M1 — 128 (a surface exposed pilus protein from GAS AI-2) ( FIG. 166 A ).
  • Pool W recognized both M5_orf80 and EftLSL.A ( FIG. 166 C ).
  • T6 antigen corresponding to a surface exposed pilus protein from GAS AI-1
  • M1 — 28 antigen T1
  • FIG. 166 D EftLSL.A as antigen T12 (corresponding to a surface exposed pilus protein from GAS AI-4)
  • FIG. 166 G M5_orf80 as a common antigen recognized by the related sera T5, T27 and T44 (corresponding to a surface exposed pilus protein from GAS AI-3).
  • Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms.
  • GAS pili may be involved in formation of biofilms.
  • the GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix).
  • Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm.
  • Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment i.e., before complete biofilm formation) is preferable.
  • the invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes.
  • the immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form.
  • the invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • the invention comprises compositions comprising a first GAS AI protein and a second GAS AI protein wherein the first and second GAS AI proteins are derived from different GAS adhesin islands.
  • the invention includes a composition comprising at least two GAS AI proteins wherein the GAS AI proteins are encoded by the adhesin islands selected from the group consisting of GAS AI-1 and AI-2; GAS AI-1 and GAS AI-3; GAS AI-1 and GAS AI-4; GAS AI-2 and GAS AI-3; GAS AI-2 and GAS AI-4; and GAS AI-3 and GAS AI-4.
  • the two GAS AI proteins are derived from different T-types.
  • FIG. 162 A schematic arrangement of GAS Adhesin Island sequences is set forth in FIG. 162 .
  • the AI region is flanked by the highly conserved open reading frames M1 — 123 and M1-136. Between three and five genes in each locus code for surface proteins containing LPXTG motifs. These surface proteins also all belong to the family of genes coding for ECM binding adhesins.
  • Adhesin island sequences can be identified in numerous M types of Group A Streptococcus . Examples of AI sequences within M1, M6, M3, M5, M12, M18, and M49 serotypes are discussed below.
  • GAS Adhesin Islands generally include a series of open reading frames within a GAS genome that encode for a collection of surface proteins and sortases.
  • a GAS Adhesin Island may encode for amino acid sequences comprising at least one surface protein.
  • a GAS Adhesin Island may encode for at least two surface proteins and at least one sortase.
  • a GAS Adhesin Island encodes for at least three surface proteins and at least two sortases.
  • One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • One or more GAS AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • GAS Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator.
  • the transcriptional regulator may regulate the expression of the GAS AI operon. Examples of transcriptional regulators found in GAS AI sequences include RofA and Nra.
  • the GAS AI surface proteins may bind or otherwise adhere to fibrinogen, fibronectin, or collagen.
  • One or more of the GAS AI surface proteins may comprise a fimbrial structural subunit.
  • One or more of the GAS AI surface proteins may include an LPXTG motif or other sortase substrate motif.
  • the LPXTG motif may be followed by a hydrophobic region and a charged C terminus, which are thought to retard the protein in the cell membrane to facilitate recognition by the membrane-localized sortase. See Barnett, et al., J. Bacteriology (2004) 186 (17): 5865-5875.
  • GAS AI sequences may be generally categorized as Type 1, Type 2, Type 3, or Type 4, depending on the number and type of sortase sequences within the island and the percentage identity of other proteins (with the exception of RofA and cpa) within the island.
  • FIG. 167 provides a chart indicating the number and type of sortase sequences identified within the adhesin islands of various strains and serotypes of GAS.
  • GAS Adhesin Island within M6 serotype is outlined in Table 4 below.
  • This GAS adhesin island 1 (“GAS AI-1”) comprises surface proteins, a srtB sortase and a rofA divergently transcribed transcriptional regulator.
  • GAS AI-1 surface proteins include Spy0157 (a fibronectin binding protein), Spy0159 (a collagen adhesion protein) and Spy0160 (a fimbrial structural subunit).
  • each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine).
  • GAS AI-1 includes a srtB type sortase.
  • GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • M6 isolate M6 isolate
  • Sortase substrate AI-1 sequence sequence or sortase identifier type functional description M6_Spy0156 Transcriptional regulator (rofA)
  • M6_Spy0157 LPXTG Fibronectin-binding protein
  • M6_Spy0158 Reverse transcriptase
  • M6_Spy0159 LPXSG Collagen adhesion protein
  • M6_Spy0160 LPXTG Fimbrial structural subunit
  • M6_Spy0160 appears to be present on the surface of GAS as part of oligomeric (pilus) structures.
  • FIGS. 127-132 present electron micrographs of GAS serotype M6 strain 3650 immunogold stained for M6 Spy0160 using anti-M6 Spy0160 antiserum. Oligomeric or hyperoligmeric structures labelled with gold particles can be seen extending from the surface of the GAS in each of these figures, indicating the presence of multiple M6_Spy0160 polypeptides in the oligomeric or hyperoligomeric structures.
  • 176 A-F present electron micrographs of GAS M6 strain 2724 immunogold stained for M6_Spy0160 using anti-M6_Spy0160 antiserum (FIGS. 176 A-E) or immunogold stained for M6_Spy0159 using anti-M6_Spy0159 antiserum ( FIG. 176 F ). Oligomeric or hyperoligomeric structures labelled with gold particles can again be seen extending from the surface of the M6 strain 2724 GAS bacteria immunogold stained for M6_Spy0160. M6_Spy0159 is also detected on the surface of the M6 strain 2724 GAS.
  • FIG. 73 provides the results of FACS analysis for surface expression of spyM6 — 0159 on each of GAS serotypes M6 2724, M6 3650, and M6 2894. A shift in fluorescence is observed for each GAS serotype when anti-spyM6 — 0159 antiserum is present, demonstrating cell surface expression.
  • FIG. 74 provides the results of FACS analysis for surface expression of spyM6 — 0160 on each of GAS serotypes M6 2724, M6 3650, and M6 2894.
  • anti-spyM6 — 0160 antiserum In the presence of of anti-spyM6 — 0160 antiserum, a shift in fluorescence is observed for each GAS serotype, which demonstrates its cell surface expression.
  • Table 19, below quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-spyM6 — 0160 antiserum, and the change in fluorescence value between the pre-immune and anti-spyM6 — 0160 antiserum.
  • FIG. 98 shows that while pre-immune sera (P ⁇ -0159) does not detect expression of M6_Spy0159 in GAS serotype M6, anti-M6_Spy0159 immune sera (I ⁇ -0159) is able to detect M6_Spy0159 protein in both total GAS M6 extracts (M6 tot) and GAS M6 fractions enriched for cell surface proteins (M6 surf prot).
  • M6_Spy0159 proteins detected in the total GAS M6 extracts or the GAS M6 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that M6_Spy0159 may be in an oligomeric (pilus) form.
  • FIG. 112 shows that while preimmune sera (Preimmune Anti 106) does not detect expression of M6_Spy0160 in GAS serotype M6 strain 2724, anti-M6_Spy0160 immune sera (Anti 160) does in both total GAS M6 strain 2724 extracts (M6 2724 tot) and GAS M6 strain 2724 fractions enriched for surface proteins.
  • the M6_Spy0160 proteins detected in the total GAS M6 strain 2724 extracts or the GAS M6 strain 2724 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that M6_Spy0160 may be in an oligomeric (pilus) form.
  • FIGS. 110 and 111 both further verify the presence of M6_Spy0159 and M6_Spy0160 in higher molecular weight structures on the surface of GAS.
  • FIG. 110 provides a Western blot performed to detect M6_Spy0159 and M6_Spy0160 in GAS M6 strain 2724 extracts enriched for surface proteins. Antiserum raised against either M6_Spy0159 (Anti-159) or M6_Spy0160 (Anti-160) cross-hybridizes with high molecular weight structures (pili) in these extracts.
  • FIG. 111 provides a similar Western blot that verifies the presence of M6_Spy0159 and M6_Spy0160 in high molecular weight structures in GAS M6 strain 3650 extracts enriched for surface proteins.
  • SpyM6 — 0157 (a fibronectin-binding protein) may also be expressed on the surface of GAS serotype M6 bacteria.
  • FIG. 174 shows the results of FACS analysis for surface expression of spyM6 — 0157 on M6 strain 3650. A slight shift in fluorescence is observed, which demonstrates that some spyM6 — 0157 may be expressed on the GAS cell surface.
  • GAS Adhesin Island 2 (“GAS AI-2”)
  • GAS Adhesin Island within M1 serotype is outlined in Table 5 below.
  • This GAS adhesin island 2 (“GAS AI-2”) comprises surface proteins, a SrtB sortase, a SrtC1 sortase and a RofA divergently transcribed transcriptional regulator.
  • GAS AI-2 surface proteins include GAS 15 (Cpa), Spy0128 (thought to be a fimbrial protein) and Spy0130 (a hypothetical protein).
  • each of these GAS AI-2 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VVXTG (SEQ ID NO: 135), or EVXTG (SEQ ID NO: 136).
  • GAS AI-2 includes a srtB type sortase and a srtC1 sortase.
  • GAS SrtB sortases may preferably anchor surface proteins with an LPSTG (SEQ ID NO: 166) motif, particularly where the motif is followed by a serine.
  • GAS SrtC1 sortase may preferentially anchor surface proteins with a V(P/V)PTG (SEQ ID NO: 167) motif.
  • GAS SrtC1 may be differentially regulated by RofA.
  • GAS AI-2 may also include a LepA putative signal peptidase I protein.
  • GAS AI-2 sequence from M1 isolate (SF370) Sortase substrate AI-2 sequence sequence or identifier sortase type functional description SPy0124 rofA regulatory protein GAS15(not VVXTG cpa annotated in SF370) SPy0127 LepA putative signal peptidase I SPy0128 (GAS16) EVXTG hypothetical protein (fimbrial) SPy0129 (GAS17) srtC1 sortase SPy0130 (GAS18) LPXTG hypothetical protein SPy0131 conserved hypothetical protein SPy0133 conserved hypothetical protein SPy0135 (GAS20) srtB sortase (putative fimbrial- associated protein)
  • FIGS. 113-115 present electron micrographs of GAS serotype M1 strain SF370 immunogold stained for GAS 15 using anti-GAS 15 antiserum.
  • FIGS. 116-121 provide electron micrographs of GAS serotype M1 strain SF370 immunogold stained for GAS 16 using anti-GAS 16 antiserum.
  • FIGS. 122-125 present electron micrograph of GAS serotype M1 strain SF370 immunogold stained for GAS 18 using anti-GAS 18 antiserum. Oligomers of these proteins can be seen on the surface of SF370 bacteria in the immuno-gold stained micrographs.
  • FIG. 126 reveals a hyperoligomer on the surface of a GAS serotype M1 strain SF370 bacterium immunogold stained for GAS 18. This long hyperoliogmeric structure comprising GAS 18 stretches far out into the supernatant from the surface of the bacteria.
  • FIG. 75 provides the results of FACS analysis for surface expression of GAS 15 on each of GAS serotypes M1 2719, M1 2580, M1 3280, M1 SF370, M1 2913, and M1 3348. A shift in fluorescence is observed for each GAS serotype when anti-GAS 15 antiserum is present, demonstrating cell surface expression.
  • Table 20 quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-GAS 15 antiserum, and the difference in fluorescence value between the pre-immune and anti-GAS 15 antiserum.
  • TABLE 20 Summary of FACS values for surface expression of GAS 15 Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS immune 15 Change immune 15 Change immune 15 Change 2719 2580 3280 159.46 712.71 553 123.9 682.84 559 217.02 639.69 423 SF370 2913 3348 201.93 722.68 521 121.41 600.45 479 152.09 446.41 294
  • FIGS. 76 and 79 provide the results of FACS analysis for surface expression of GAS 16 on each of GAS serotypes M1 2719, M1 2580, M1 3280, M1 SF370, M1 2913, and M1 3348.
  • the FACS data in FIG. 76 was obtained using antisera was raised against full length GAS 16. In the presence of this anti-GAS 16 antiserum, a shift in fluorescence is observed for each GAS serotype, demonstrating its cell surface expression.
  • SEQ ID NO: 179 GCTACAACAGTTCACGGGGAGACTGTTGTAAACGGAGCCAAACTAACAGT TACAAAAAACCTTGATTTAGTTAATAGCAATGCATTAATTCCAAATACAG ATTTTACATTTAAAATCGAACCTGATACTACTGTCAACGAAGACGGAAAT AAGTTTAAAGGTGTAGCTTTGAACACACCGATGACTAAAGTCACTTACAC CAATTCAGATAAAGGTGGATCAAATACGAAAACTGCAGAATTTGATTTTT CAGAAGTTACTTTTGAAAAACCAGGTGTTTATTATTACAAAGTAACTGAG GAGAAGATAGATAAAGTTCCTGGTGTTTCTTATGATACAACATCTTACAC TGTTCAAGTTCATGTCTTGTGGAATGAAGAGCAACAAAAACCAGTAGCTACTTGTGGAATGAAGCAACAAAAACCAGTAGCTACTTGTGGAATGAAGCAACAAAAACCAGTAGCTACTTGTGGAATGAAGCAACAAAAACCAGTAGCTACTTGTGGAATGAAGAG
  • FIGS. 77 and 78 provide the results of FACS analysis for surface expression of GAS 18 on each of GAS serotypes M12719, M12580, M1 3280, M1 SF370, M12913, and M13348.
  • the antiserum used to obtain the FACS data in each of FIGS. 77 and 78 was different, although each was raised against full length GAS 18.
  • a shift in fluorescence is observed for each GAS serotype, demonstrating its cell surface expression.
  • FIG. 89 shows that while pre-immune sera does not detect GAS M1 expression of GAS 15, anti-GAS 15 immune sera is able to detect GAS 15 protein in both total GAS M1 extracts and GAS M1 proteins enriched for cell surface proteins.
  • the GAS 15 proteins detected in the M1 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that GAS 15 may be in an oligomeric (pilus) form.
  • FIG. 90 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 15 antisera.
  • FIG. 91 provides an additional Western blot identical to that of FIG. 90 , but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIG. 92 provides a Western blot that was probed for GAS 16 protein. While pre-immune sera does not detect GAS M1 expression of GAS 16, anti-GAS 16 immune sera is able to detect GAS 16 protein in GAS M1 extracts enriched for cell surface proteins. The GAS 16 proteins detected in the M1 extracts enriched for surface proteins are present as high molecular weight structures, indicating that GAS 16 may be in an oligomeric (pilus) form.
  • FIG. 93 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 16 antisera.
  • FIG. 94 provides an additional Western blot identical to that of FIG. 93 , but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIG. 95 provides a Western blot that was probed for GAS 18 protein. While pre-immune sera does not detect GAS M1 expression of GAS 18, anti-GAS 18 immune sera is able to detect GAS 18 protein in GAS M1 extracts enriched for cell surface proteins. The GAS 18 proteins detected in the M1 extracts enriched for surface proteins are present as high molecular weight structures, indicating that GAS 18 may be in an oligomeric (pilus) form.
  • FIG. 96 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 18 antisera.
  • FIG. 97 provides an additional Western blot identical to that of FIG. 96 , but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIGS. 102-106 provide additional Western blots to verify the presence of GAS 15, GAS 16, and GAS 18 in high molecular weight structures in GAS.
  • Each Western blot was performed using proteins from a different GAS M1 strain, 2580, 2913, 3280, 3348, and 2719.
  • Each Western blot was probed with antisera raised against each of GAS 15, GAS 16, and GAS 18. As can be seen in FIGS.
  • FIG. 107 provides a similar Western blot performed to detect GAS 15, GAS 16, and GAS 18 proteins in a GAS serotype M1 strain SF370 protein fraction enriched for surface proteins. This Western blot also shows detection of GAS 15 (Anti-15), GAS 16 (Anti-16), and GAS 18 (Anti-18) as high molecular weight structures.
  • GAS Adhesin Island sequences within M3, M5, and M18 serotypes are outlined in Tables 6-8 and 10 below.
  • This GAS adhesin island 3 (“GAS AI-3”) comprises surface proteins, a SrtC2 sortase, and a Negative transcriptional regulator (Nra) divergently transcribed transcriptional regulator.
  • GAS AI-3 surface proteins within include a collagen binding protein, a fimbrial protein, a F2 like fibronectin-binding protein.
  • GAS AI-3 surface proteins may also include a hypothetical surface protein.
  • each of these GAS AI-3 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-3 includes a SrtC2 type sortase.
  • GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • GAS SrtC2 may be differentially regulated by Nra.
  • GAS AI-3 may also include a LepA putative signal peptidase I protein.
  • GAS AI-3 may also include a putative multiple sugar metabolism regulator.
  • GAS AI-3 sequences from M3 isolate (MGAS315) Sortase substrate AI-3 sequence sequence or identifier sortase type Functional description SpyM3_0097 Negative transcriptional regulator (Nra) SpyM3_0098 VPXTG putative collagen binding protein (Cpb) SpyM3_0099 LepA putative signal peptidase I SpyM3_0100 QVXTG conserved hypothetical protein (fimbrial) SpyM3_0101 SrtC2 sortase SpyM3_0102 LPXAG hypothetical protein SpyM3_0103 putative multiple sugar metabolism regulator SpyM3_0104 LPXTG protein F2 like fibronectin-binding protein
  • FIG. 51A A schematic of AI-3 serotypes M3, M5, M 18, and M49 is shown in FIG. 51A .
  • Each contains an open reading frame encoding a SrtC2-type sortase of nearly identical amino acid sequence. See FIG. 52B for an amino acid sequence alignment for each of the SrtC2 amino acid sequences.
  • FIG. 60 indicates the amino acid sequence of the pilin motif and E-box of each of GAS AI-3 serotype M3 MGAS315 (SpyM3 — 0104/21909640), GAS AI-3 serotype M3 SSI (Sps0106/28895018), GAS AI-3 serotype M18 (SpyM18 — 0132/19745307), and GASAI-3 serotype M5 (orf84).
  • FIG. 80 provides the results of FACS analysis for surface expression of SpyM3 — 0098 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM3 — 0098 antiserum is present, demonstrating cell surface expression.
  • FIG. 81 provides the results of FACS analysis for surface expression of SpyM3 — 0100 on each of GAS serotypes M3 2721 and M3 3135.
  • a shift in fluorescence is observed for each GAS serotype when anti-SpyM3 — 0100 antiserum is present, demonstrating cell surface expression.
  • Table 26, below quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM3 — 0100 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM3 — 0100 antiserum.
  • TABLE 26 Summary of FACS values for surface expression of SpyM3_0100 2721 3135 Pre- Anti- Pre- Anti- immune spyM3_0100 Change immune spyM3_0100 Change 110.31 181.91 72 97.87 250.01 152
  • FIG. 82 provides the results of FACS analysis for surface expression of SpyM3 — 0102 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM3 — 0102 antiserum is present, demonstrating cell surface expression.
  • Table 27, below quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM3 — 0102 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM3 — 0102 antiserum.
  • FIG. 82 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SpyM3 — 0102 identified in a different GAS serotype, M6.
  • FACS analysis conducted with the SpyM3 — 0102 antisera was able to detect surface expression of the homologous SpyM3 — 0102 antigen on each of GAS serotypes M6 2724, M6 3650, and M6 2894.
  • SpyM3 — 0102 is also homologous to pilin antigen 19224139 of GAS serotype M12. Antisera raised against SpyM3 — 0102 is able to detect high molecular weight structures in GAS serotype M12 strain 2728 protein fractions enriched for surface proteins, which would contain the 19224139 antigen. See FIG. 109 at the lane labelled M112 2728 surf prot.
  • FIG. 83 provides the results of FACS analysis for surface expression of SpyM3 — 0104 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM3 — 0104 antiserum is present, demonstrating cell surface expression.
  • Table 29, below quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM3 — 0104 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM3 — 0104 antiserum.
  • FIG. 83 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SpyM3 — 0104 identified in a different GAS serotype, M12.
  • FACS analysis conducted with the SpyM3 — 0104 antisera was able to detect surface expression of the homologous SpyM3 — 0104 antigen on GAS serotype M12 2728.
  • Table 30, below quantitatively summarizes the FACS fluorescence values obtained for this GAS serotype in the presence of pre-immune antiserum, anti-SpyM3 — 0104 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM3 — 0104 antiserum.
  • FIG. 84 provides the results of FACS analysis for surface expression of SPs — 0106 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SPs — 0106 antiserum is present, demonstrating cell surface expression.
  • Table 31, below quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SPs — 0106 antiserum, and the difference in fluorescence value between the pre-immune and anti-SPs — 0106 antiserum.
  • FIG. 84 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SPs — 0106 identified in a different GAS serotype, M12.
  • FACS analysis conducted with the SPs — 0106 antisera was able to detect surface expression of the homologous SPs — 0106 antigen on GAS serotype M12 2728.
  • Table 32 below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SPs — 0106 antiserum, and the difference in fluorescence value between the pre-immune and anti-SPs — 0106 antiserum.
  • TABLE 32 Summary of FACS values for surface expression of SPs_0106 in an M12 serotype 2728 Pre-immune Anti-SPs_0106 Change 304.01 254.64 ⁇ 49
  • GAS Adhesin Island 4 (“GAS AI-4”)
  • GAS Adhesin Island sequences within M12 serotype are outlined in Table 11 below.
  • This GAS adhesin island 4 (“GAS AI-4”) comprises surface proteins, a SrtC2 sortase, and a RofA regulatory protein.
  • GAS AI-4 surface proteins within may include a fimbrial protein, an F or F2 like fibronectin-binding protein, and a capsular polysaccharide adhesion protein (Cpa). GAS AI-4 surface proteins may also include a hypothetical surface protein in an open reading frame (orf). Preferably, each of the GAS AI-4 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • LPXTG sortase substrate motif such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-4 includes a SrtC2 type sortase.
  • GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • GAS AI-4 may also include a LepA putative signal peptidase I protein and a MsmRL protein. TABLE 11 GAS AI-4 sequences from M12 isolate (A735) Sortase substrate AI-4 sequence sequence or sortase identifier type Functional description 19224133 RofA regulatory protein 19224134 LPXTG protein F SrtB SrtB (stop codon*) 19224135 VPXTG Cpa 19224136 LepA 19224137 QVXTG EftLSL.A (fimbrial) 19224138 SrtC2 EftLSL.B 19224139 LPXAG Orf2 19224140 MsmRL 19224141 LPXTG protein F2
  • FIG. 51A A schematic of AI- 4 serotype M12 is shown in FIG. 51A .
  • One of the open reading frames encodes a SrtC2-type sortase having an amino acid sequence nearly identical to the amino acid sequence of the SrtC2-type sortase of the AI-3 serotypes described above. See FIG. 52B for an amino acid sequence alignment for each of the SrtC2 amino acid sequences.
  • FIG. 52 is an amino acid alignment of the capsular polysaccharide adhesion protein (cpa) of AI-4 serotype M112 (19224135), GAS AI-3 serotype M5 (ORF78), S. pyogenes strain MGAS315 serotype M3 (21909634), S. pyogenes SSI-1 serotype M3 (28810257), S.
  • cpa capsular polysaccharide adhesion protein
  • FIG. 53 shows that the F-like fibronectin-binding protein encoded by the AI-4 serotype M12 open frame (19224134) shares homology with a F-like fibronectin-binding protein found in S. pyogenes strain MGAS10394 serotype M6 (50913503).
  • FIG. 54 is an amino acid sequence alignment that illustrates that the F2-like fibronectin-binding protein of AI-4 serotype M12 (19224141) shares homology with the F2-like fibronectin-binding protein of S. pyogenes strain MGAS8232 serotype M3 (19745307), GAS AI-3 serotype M5 (ORF84), S. pyogenes strain SSI serotype M3 (28810263), and S. pyogenes strain MGAS315 serotype M3 (21909640).
  • FIG. 55 is an amino acid sequence alignment that illustrates that the fimbrial protein of AI-4 serotype M12 (19224137) shares homology with the fimbrial protein of GAS AI-3 serotype M5 (ORF80), and the hypothetical protein of S. pyogenes strain MGAS315 serotype M3 (21909636), S. pyogenes strain SSI serotype M3 (28810259), S. pyogenes strain MGAS8732 serotype M3 (19745303), and S. pyogenes strain M1 GAS serotype M1 (13621428).
  • FIG. 56 is an amino acid sequence alignment that illustrates that the hypothetical protein of GAS AI-4 serotype M12 (19224139) shares homology with the hypothetical protein of S. pyogenes strain MGAS315 serotype M3 (21909638), S. pyogenes strain SSI-1 serotype M3 (28810261), GAS AI-3 serotype M5 (ORF82), and S. pyogenes strain MGAS8232 serotype M3 (19745305).

Abstract

The invention relates to the identification of a new adhesin islands within the genomes of several Group A and Group B Streptococcus serotypes and isolates. The adhesin islands are thought to encode surface proteins which are important in the bacteria's virulence. Thus, the adhesin island proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against GAS or GBS infection. For example, the invention may include an immunogenic composition comprising one or more of the discovered adhesin island proteins.

Description

    FIELD OF THE INVENTION
  • The invention relates to the identification of adhesin islands within the genome Streptococcus agalactiae (“GBS”) and the use of adhesin island amino acid sequences encoded by these adhesin islands in compositions for the treatment or prevention of GBS infection. Similar sequences have been identified in other Gram positive bacteria. The invention further includes immunogenic compositions comprising adhesin island amino acid sequences of Gram positive bacteria for the treatment or prevention of infection of Gram positive bacteria. Preferred immunogenic compositions of the invention include an adhesin island surface protein which may be formulated or purified in an oligomeric or pilus form.
  • BACKGROUND OF THE INVENTION
  • GBS has emerged in the last 20 years as the major cause of neonatal sepsis and meningitis that affects 0.5-3 per 1000 live births, and an important cause of morbidity among older age groups affecting 5-8 per 100,000 of the population. Current disease management strategies rely on intrapartum antibiotics and neonatal monitoring which have reduced neonatal case mortality from >50% in the 1970's to less than 10% in the 1990's. Nevertheless, there is still considerable morbidity and mortality and the management is expensive. 15-35% of pregnant women are asymptomatic carriers and at high risk of transmitting the disease to their babies. Risk of neonatal infection is associated with low serotype specific maternal antibodies and high titers are believed to be protective. In addition, invasive GBS disease is increasingly recognized in elderly adults with underlying disease such as diabetes and cancer.
  • The “B” in “GBS” refers to the Lancefield classification, which is based on the antigenicity of a carbohydrate which is soluble in dilute acid and called the C carbohydrate. Lancefield identified 13 types of C carbohydrate, designated A to 0, that could be serologically differentiated. The organisms that most commonly infect humans are found in groups A, B, D, and G. Within group B, strains can be divided into at least 9 serotypes (Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII) based on the structure of their polysaccharide capsule. In the past, serotypes Ia, Ib, II, and III were equally prevalent in normal vaginal carriage and early onset sepsis in newborns. Type V GBS has emerged as an important cause of GBS infection in the USA, however, and strains of types VI and VIII have become prevalent among Japanese women.
  • The genome sequence of a serotype V strain 2603 V/R has been published (See Tettelin et al. (2002) Proc. Natl. Acad. Sci. USA, 10.1073/pnas.182380799) and various polypeptides for use a vaccine antigens have been identified (WO 02/34771). The vaccines currently in clinical trials, however, are based primarily on polysaccharide antigens. These suffer from serotype-specificity and poor immunogenicity, and so there is a need for effective vaccines against S. agalactiae infection.
  • S. agalactiae is classified as a gram positive bacterium, a collection of about 21 genera of bacteria that colonize humans, have a generally spherical shape, a positive Gram stain reaction and lack endospores. Gram positive bacteria are frequent human pathogens and include Staphylococcus (such as S. aureus), Streptococcus (such as S. pyogenes (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans), Enterococcus (such as E. faecalis and E. faecium), Clostridium (such as C. difficile), Listeria (such as L. monocytogenes) and Corynebacterium (such as C. diphtheria).
  • It is an object of the invention to provide further and improved compositions for providing immunity against disease and/or infection of Gram positive bacteria. The compositions are based on the identification of adhesin islands within Streptococcal genomes and the use of amino acid sequences encoded by these islands in therapeutic or prophylactic compositions. The invention further includes compositions comprising immunogenic adhesin island proteins within other Gram positive bacteria in therapeutic or prophylactic compositions.
  • SUMMARY OF THE INVENTION
  • Applicants have identified a new adhesin island, “GBS Adhesin Island 1”, “AI-1” or “GBS AI-1”, within the genomes of several Group B Streptococcus serotypes and isolates. This adhesin island is thought to encode surface proteins which are important in the bacteria's virulence. In addition, Applicants have discovered that surface proteins within GBS Adhesin Islands form a previously unseen pilus structure on the surface of GBS bacteria. Amino acid sequences encoded by such GBS Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of GBS infection.
  • A preferred immunogenic composition of the invention comprises an AI-1 surface protein, such as GBS 80, which may be formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Electron micrographs depicting some of the first visualizations of this pilus structure in a wild type GBS strain are shown in FIGS. 16, 17, 49, and 50. In addition, Applicants have transformed a GBS strain with a plasmid comprising the AI surface protein GBS 80 which resulted in increased production of that AI surface protein. The electron micrographs of this mutant GBS strain in FIGS. 13-15 reveal long, hyper-oligomeric structures comprising GBS 80 which appear to cover portions of the surface of the bacteria and stretch far out into the supernatant. These hyper-oligomeric pilus structures comprising a GBS AI surface protein may be purified or otherwise formulated for use in immunogenic compositions.
  • GBS AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“AI-1 proteins”). Specifically, AI-1 includes polynucleotide sequences encoding for two or more of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648. One or more of the AI-1 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the AI-1 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • AI-1 typically resides on an approximately 16.1 kb transposon-like element frequently inserted into the open reading frame for trmA. One or more of the AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. The AI surface proteins of the invention may affect the ability of the GBS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. AI-1 may encode at least one surface protein. Alternatively, AI-1 may encode at least two surface proteins and at least one sortase. Preferably, AI-1 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif or other sortase substrate motif.
  • The GBS AI-1 protein of the composition may be selected from the group consisting of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648. GBS AI-1 surface proteins GBS 80 and GBS 104 are preferred for use in the immunogenic compositions of the invention.
  • In addition to the open reading frames encoding the AI-1 proteins, AI-1 may also include a divergently transcribed transcriptional regulator such as araC (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction). It is believed that araC may regulate the expression of the GBS AI operon. (See Korbel et al., Nature Biotechnology (2004) 22(7): 911-917 for a discussion of divergently transcribed regulators in E. coli).
  • A second adhesin island, “Adhesin Island-2”, “AI-2” or “GBS AI-2”, has also been identified in numerous GBS serotypes. Amino acid sequences encoded by the open reading frames of AI-2 may also be used in immunogenic compositions for the treatment or prevention of GBS infection.
  • GBS AI-2 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525. The GBS AI-2 sequences may be divided into two subgroups. In one embodiment, AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406. This collection of open reading frames may be generally referred to as GBS AI-2 subgroup 1. Alternatively, AI-2 may include open reading frames encoding for two or more of 01520, 01521, 01522, 01523, 01523, 01524 and 01525. This collection of open reading frames may be generally referred to as GBS AI-2 subgroup 2.
  • One or more of the AI-2 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the AI-2 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • One or more of the AI-2 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. AI-2 may encode for at least one surface protein. Alternatively, AI-2 may encode for at least two surface proteins and at least one sortase. Preferably, AI-2 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • The AI-2 protein of the composition may be selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525. AI-2 surface proteins GBS 67, GBS 59, and 01524 are preferred AI-2 proteins for use in the immunogenic compositions of the invention. GBS 67 or GBS 59 is particularly preferred.
  • GBS AI-2 may also include a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB). As in AI-1, rogB is thought to regulate the expression of the AI-2 operon.
  • The GBS AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against GBS infection. For example, the invention may include an immunogenic composition comprising one or more GBS AI-1 proteins and one or more GBS AI-2 proteins.
  • The immunogenic compositions may also be selected to provide protection against an increased range of GBS serotypes and strain isolates. For example, the immunogenic composition may comprise a first and second GBS AI protein, wherein a full length polynucleotide sequence encoding for the first GBS AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second GBS AI protein. In addition, each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple GBS serotypes and strain isolates. Preferably, each antigen is presnt in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) GBS strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5 or more) GBS serotypes.
  • Within GBS AI-1, Applicants have found that Group B Streptococcus surface exposure of GBS 104 is dependent on the concurrent expression of GBS 80. It is thought that GBS 80 is involved in the transport or localization of GBS 104 to the surface of the bacteria. The two proteins may be oligomerized or otherwise chemically or physically associated. It is possible that this association involves a conformational change in GBS 104 that facilitates its transition to the surface of the GBS bacteria. In addition, one or more AI sortases may also be involved in this surface localization and chemical or physical association. Similar relationships are thought to exist within GBS AI-2. The compositions of the invention may therefore include at least two AI proteins, wherein the two AI proteins are physically or chemically associated. Preferably, the two AI proteins form an oligomer. Preferably, one or more of the AI proteins are in a hyper-oligomeric form. In one embodiment, the associated AI proteins may be purified or isolated from a GBS bacteria or recombinant host cell.
  • It is also an object of the invention to provide further and improved compositions for providing prophylactic or therapeutic protection against disease and/or infection of Gram positive bacteria. The compositions are based on the identification of adhesin islands within Streptococcal genomes and the use of amino acid sequences encoded by these islands in therapeutic or prophylactic compositions. The invention further includes compositions comprising immunogenic adhesin island proteins within other Gram positive bacteria in therapeutic or prophylactic compositions. Preferred Gram positive adhesin island proteins for use in the invention may be derived from Staphylococcus (such as S. aureus), Streptococcus (such as S. agalactiae (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans), Enterococcus (such as E. faecalis and E. faecium), Clostridium (such as C. difficile), Listeria (such as L. monocytogenes) and Corynebacterium (such as C. diphtheria). Preferably, the Gram positive adhesin island surface proteins are in oligomeric or hyperologimeric form.
  • For example, Applicants have identified adhesin islands within the genomes of several Group A Streptococcus serotypes and isolates. These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fasciitis. In addition, post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases. The first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused. In the second stage of infection, the bacteria secretes a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection. The final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart. A general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • In order to prevent the pathogenic effects associated with the later stages of GAS infection, an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above. The M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation. The carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci. The amino terminus, which extend through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen. Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens. Antisera to define T types is commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • The gene coding for one form of T-antigen, T-type 6, from an M6 strain of GAS (D741) has been cloned and characterized and maps to an approximately 11 kb highly variable pathogenicity island. Schneewind et al., J. Bacteriol. (1990) 172(6):3310-3317. This island is known as the Fibronectin-binding, Collagen-binding T-antigen (FCT) region because it contains, in addition to the T6 coding gene (tee6), members of a family of genes coding for Extra Cellular Matrix (ECM) binding proteins. Bessen et al., Infection & Immunity (2002) 70(3):1159-1167. Several of the protein products of this gene family have been shown to directly bind either fibronectin and/or collagen. See Hanski et al., Infection & Immunity (1992) 60(12):5119-5125; Talay et al., Infection & Immunity (1992(60(9):3837-3844; Jaffe et al. (1996) 21(2):373-384; Rocha et al., Adv Exp Med Biol. (1997) 418:737-739; Kreikemeyer et al., J Biol Chem (2004) 279(16):15850-15859; Podbielski et al., Mol. Microbiol. (1999) 31(4):1051-64; and Kreikemeyer et al., Int. J. Med Microbiol (2004) 294(2-3):177-88. In some cases direct evidence for a role of these proteins in adhesion and invasion has been obtained.
  • Applicants raised antiserum against a recombinant product of the tee6 gene and used it to explore the expression of T6 in M6 strain 2724. In immunoblot of mutanolysin extracts of this strain, the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used.
  • This pattern of high molecular weight products is similar to that observed in immunoblots of the protein components of the pili identified in Streptococcus agalactiae (described above) and previously in Corynebacterium diphtheriae. Electron microscropy of strain M6 2724 with antisera specific for the product of tee6 revealed abundant surface staining and long pilus like structures extending up to 700 nanometers from the bacterial surface, revealing that the T6 protein, one of the antigens recognized in the original Lancefiled serotyping system, is located within a GAS Adhesin Island (GAS AI-1) and forms long covalently linked pilus structures.
  • Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms. Immunological recognition of these proteins may allow the host immune response to slow or prevent the bacteria's transition into the more pathogenic later stages of infection.
  • In addition, Applicants have discovered that the GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix). Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm. Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment (i.e., before complete biofilm formation) is preferable.
  • The invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes. The immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form. The immunogenic compositions of the invention may include one or more GAS AI surface proteins. The invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • Amino acid sequence encoded by such GAS Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of GAS infection. Preferred immunogenic compositions of the invention comprise a GAS AI surface protein which has been formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer.
  • GAS Adhesin Islands generally include a series of open reading frames within a GAS genome that encode for a collection of surface proteins and sortases. A GAS Adhesin Island may encode for an amino acid sequence comprising at least one surface protein. The Adhesin Island, therefore, may encode at least one surface protein. Alternatively, a GAS Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, a GAS Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more GAS AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • GAS Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator. The transcriptional regulator may regulate the expression of the GAS AI operon. Examples of transcriptional regulators found in GAS AI sequences include RofA and Nra.
  • The GAS AI surface proteins may bind or otherwise adhere to fibrinogen, fibronectin, or collagen. One or more of the GAS AI surface proteins may comprise a fimbrial structural subunit.
  • One or more of the GAS AI surface proteins may include an LPXTG motif or other sortase substrate motif. The LPXTG motif may be followed by a hydrophobic region and a charged C terminus, which are thought to retard the protein in the cell membrane to facilitate recognition by the membrane-localized sortase. See Barnett, et al., J. Bacteriology (2004) 186 (17): 5865-5875.
  • GAS AI sequences may be generally categorized as Type 1, Type 2, Type 3, or Type 4, depending on the number and type of sortase sequences within the island and the percentage identity of other proteins (with the exception of RofA and cpa) within the island. Schematics of the GAS adhesin islands are set forth in FIG. 51A and FIG. 162. “GAS Adhesin Island-1 or “GAS AI-1” comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-1 proteins”). GAS AI-1 preferably comprises surface proteins, a srtB sortase and a rofA divergently transcribed transcriptional regulator. GAS AI-1 surface proteins may include a fibronectin binding protein, a collagen adhesion protein and a fimbrial structural subunit. The fimbrial structural subunit (also known as tee6) is thought to form the shaft portion of the pilus like structure, while the collagen adhesion protein (Cpa) is thought to act as an accessory protein facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule.
  • Specifically, GAS AI-1 includes polynucleotide sequences encoding for two or more of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161. The GAS AI-1 may also include polynucleotide sequences encoding for any one of CDC SS 410_fimbrial, ISS3650_fimbrial, DSM2071_fimbrial
  • A preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which may be formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. The immunogenic composition of the invention may alternatively comprise an isolated GAS AI-1 surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising GAS AI-1 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • One or more of the GAS AI-1 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-1 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • One or more of the GAS AI-1 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-1 may encode for at least one surface protein. Alternatively, GAS AI-1 may encode for at least two surface proteins and at least one sortase. Preferably, GAS AI-1 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-1 preferably includes a srtB sortase. GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • The GAS AI-1 protein of the composition may be selected from the group consisting of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160 M6_Spy0161, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial. GAS AI-1 surface proteins M6_Spy0157 (a fibronectin binding protein), M6_Spy0159 (a collagen adhesion protein, Cpa), M6_Spy0160 (a fimbrial structural subunit, tee6), CDC SS 410_fimbrial (a fimbrial structural subunit), ISS3650_fimbrial (a fimbrial structural subunit), and DSM2071_fimbrial (a fimbrial structural subunit) are preferred GAS AI-1 proteins for use in the immunogenic compositions of the invention. The fimbrial structural subunit tee6 and the collagen adhesion protein Cpa are preferred GAS AI-1 surface proteins. Preferably, each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine).
  • In addition to the open reading frames encoding the GAS AI-1 proteins, GAS AI-1 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the GAS AI protein open reading frames, but it transcribed in the opposite direction).
  • The GAS AI-1 surface proteins may be used alone, in combination with other GAS AI-1 surface proteins or in combination with other GAS AI surface proteins. Preferably, the immunogenic compositions of the invention include the GAS AI-1 fimbrial structural subunit (tee6) and the GAS AI-1 collagen binding protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-1 fimbrial structural subunit (tee6).
  • A second GAS adhesion island, “GAS Adhesin Island-2” or “GAS AI-2,” has also been identified in GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-2 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • A preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which may be formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. A preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-2 surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising GAS AI-2 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-2 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-2 proteins”). GAS AI-2 preferably comprises surface proteins, a srtB sortase, a srtC1 sortase and a rofA divergently transcribed transcriptional regulator.
  • Specifically, GAS AI-2 includes polynucleotide sequences encoding for two or more of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • One or more of the GAS AI-2 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-2 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • One or more of the GAS AI-2 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-2 may encode for at least one surface protein. Alternatively, GAS AI-2 may encode for at least two surface proteins and at least one sortase. Preferably, GAS AI-2 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-2 preferably includes a srtB sortase and a srtC1 sortase. As discussed above, GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine. GAS srtC1 sortase may preferentially anchor surface proteins with a V(P/V)PTG (SEQ ID NO: 167) motif. GAS srtC1 may be differentially regulated by rofA.
  • The GAS AI-2 protein of the composition may be selected from the group consisting of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20. GAS AI-2 surface proteins GAS15 (Cpa), GAS16 (thought to be a fimbrial protein, M1128), GAS18 (M1_Spy0130), and GAS20 are preferred for use in the immunogenic compositions of the invention. GAS 16 is thought to form the shaft portion of the pilus like structure, while GAS 15 (the collagen adhesion protein Cpa) and GAS 18 are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule. Preferably, each of these GAS AI-2 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VVXTG (SEQ ID NO: 135), or EVXTG (SEQ ID NO: 136).
  • In addition to the open reading frames encoding the GAS AI-2 proteins, GAS AI-2 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the GAS AI protein open reading frames, but it transcribed in the opposite direction). The GAS AI-2 surface proteins may be used alone, in combination with other GAS AI-2 surface proteins or in combination with other GAS AI surface proteins. Preferably, the immunogenic compositions of the invention include the GAS AI-2 fimbrial protein (GAS 16), the GAS AI-2 collagen binding protein (GAS 15) and GAS 18 (M1_Spy0130). More preferably, the immunogenic compositions of the invention include the GAS AI-2 fimbrial protein (GAS 16).
  • A third GAS adhesion island, “GAS Adhesin Island-3” or “GAS AI-3,” has also been identified in numerous GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-3 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • A preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which may be formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. A preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-3 surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising GAS AI-3 surface proteins may be purified or otherwise formulated for use in immunogenic compositions. GAS AI-3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-3 proteins”). GAS AI-3 preferably comprises surface proteins, a srtC2 sortase, and a Negative transcriptional regulator (Nra) divergently transcribed transcriptional regulator. GAS AI-3 surface proteins may include a collagen binding protein, a fimbrial protein, and a F2 like fibronectin-binding protein. GAS AI-3 surface proteins may also include a hypothetical surface protein. The fimbrial protein is thought to form the shaft portion of the pilus like structure, while the collagen adhesion protein (Cpa) and the hypothetical surface protein are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule. Preferred AI-3 surface proteins include the fimbrial proein, the collagen binding protein and the hypothetical protein. Preferably, each of these GAS AI-3 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • Specifically, GAS AI-3 includes polynucleotide sequences encoding for two or more of SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, SpyM30104, Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, Sps0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, spyM180132, SpyoM0100156, SpyoM0100155, SpyoM0100154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. In one embodiment, GAS AI-3 may include open reading frames encoding for two or more of SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, and SpyM30104. Alternatively, GAS AI-3 may include open reading frames encoding for two or more of Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, and Sps0106. Alternatively, GAS AI-3 may include open reading frames encoding for two or more of orf78, orf79, orf80, orf81, orf82, orf83, and orf84. Alternatively, GAS AI-3 may include open reading frames encoding for two or more of spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, and spyM180132. Alternatively, GAS AI-3 may include open reading frames encoding for two or more of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149. Alternatively, GAS AI-1 may also include polynucleotide sequences encoding for any one of ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • One or more of the GAS AI-3 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-3 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • One or more of the GAS AI-3 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-3 may encode for at least one surface protein. Alternatively, GAS AI-3 may encode for at least two surface proteins and at least one sortase. Preferably, GAS AI-3 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-3 preferably includes a srtC2 type sortase. GAS srtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail. GAS SrtC2 may be differentially regulated by Nra.
  • The GAS AI-3 protein of the composition may be selected from the group consisting of SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, SpyM30104, Sps0100, Sps0101, Sps0102, Sps0103, Sps0104, Sps0105, Sps0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. GAS AI-3 surface proteins SpyM30098, SpyM3 0100, SpyM3 0102, SpyM30104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM180126, spyM18 0128, spyM180130, spyM180132, SpyoM01000155, SpyoM01000153, SpyoM01000151, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial are preferred GAS AI-3 proteins for use in the immunogenic compositions of the invention.
  • In addition to the open reading frames encoding the GAS AI-3 proteins, GAS AI-3 may also include a transcriptional regulator such as Nra.
  • GAS AI-3 may also include a LepA putative signal peptidase I protein.
  • The GAS AI-3 surface proteins may be used alone, in combination with other GAS AI-3 surface proteins or in combination with other GAS AI surface proteins. Preferably, the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein, the GAS AI-3 collagen binding protein, the GAS AI-3 surface protein (such as SpyM3 0102, M3_Sps0104, M5_orf82, or spyM180130), and fibronectin binding protein PrtF2. More preferably, the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein, the GAS AI-3 collagen binding protein, and the GAS AI-3 surface protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-3 fimbrial protein.
  • Representative examples of the GAS AI-3 fimbrial protein include SpyM3 0100, M3_Sps0102, M5_orf80, spyM18 128, SpyoM01000153, ISS3040_fimbrial, ISS3776_fimbrial, ISS4959_fimbrial.
  • Representative examples of the GAS AI-3 collagen binding protein include SpyM30098, M3_Sps0100, M5_orf 78, spyM180126, and SpyoM01000155.
  • Representative examples of the GAS AI-3 fibronectin binding protein PrtF2 include SpyM30104, M3_Sps0106, M5_orf84 and spyM180132, and SpyoM01000149.
  • A fourth GAS adhesion island, “GAS Adhesin Island-4” or “GAS AI-4,” has also been identified in GAS serotypes. Amino acid sequences encoded by the open reading frames of GAS AI-4 may also be used in immunogenic compositions for the treatment or prevention of GAS infection.
  • A preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which may be formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. A preferred immunogenic composition of the invention alternatively comprises an isolated GAS AI-4 surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising GAS AI-3 surface proteins may be purified or otherwise formulated for use in immunogenic compositions. The oligomeric or hyperoligomeric pilus structures comprising GAS AI-4 surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • GAS AI-4 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-4 proteins”). This GAS adhesin island 4 (“GAS AI-4”) comprises surface proteins, a srtC2 sortase, and a RofA regulatory protein. GAS AI-4 surface proteins within may include a fimbrial protein, F1 and F2 like fibronectin-binding proteins, and a capsular polysaccharide adhesion protein (Cpa). GAS AI-4 surface proteins may also include a hypothetical surface protein in an open reading frame (orf).
  • The fimbral protein (EftLSL) is thought to form the shaft portion of the pilus like structure, while the collagen adhesion protein (Cpa) and the hypothetical protein are thought to act as accessory proteins facilitating the formation of the pilus structure, exposed on the surface of the bacterial capsule. Preferably, each of these GAS AI-4 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • Specifically, GAS AI-4 includes polynucleotide sequences encoding for two or more of 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, and 19224141. A GAS AI-4 polynucleotide may also include polynucleotide sequences encoding for any one of 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial. One or more of the GAS AI-4 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-4 open reading frames may be replaced by a sequence having sequence homology (sequence identity) to the replaced ORF.
  • One or more of the GAS AI-4 surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-4 may encode for at least one surface protein. Alternatively, GAS AI-4 may encode for at least two surface proteins and at least one sortase. Preferably, GAS AI-4 encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • GAS AI-4 includes a SrtC2 type sortase. GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • The GAS AI-4 protein of the composition may be selected from the group consisting of 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial. GAS AI-4 surface proteins 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial are preferred proteins for use in the immunogenic compositions of the invention.
  • In addition to the open reading frames encoding the GAS AI-4 proteins, GAS AI-4 may also include a divergently transcribed transcriptional regulator such as RofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction.
  • GAS AI-4 may also include a LepA putative signal peptidase I protein and a MsmRL protein. The GAS AI-4 surface proteins may be used alone, in combination with other GAS AI-4 surface proteins or in combination with other GAS AI surface proteins. Preferably, the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein (EftLSL or 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, or ISS4538_fimbrial), the GAS AI-4 collagen binding protein, the GAS AI-4 surface protein (such as M12 isolate A735 orf 2), and fibronectin binding protein PrtF1 and PrtF2. More preferably, the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein, the GAS AI-4 collagen binding protein, and the GAS AI-4 surface protein. Still more preferably, the immunogenic compositions of the invention include the GAS AI-4 fimbrial protein.
  • The GAS AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against GAS infection. For example, the invention may include an immunogenic composition comprising one or more GAS AI-1 proteins and one or more of any of GAS AI-2, GAS AI-3, or GAS AI-4 proteins. For example, the invention includes an immunogenic composition comprising at least two GAS AI proteins where each protein is selected from a different GAS adhesin island. The two GAS AI proteins may be selected from one of the following GAS AI combinations: GAS AI-1 and GAS AI-2; GAS AI-1 and GAS AI-3; GAS AI-1 and GAS AI-4; GAS AI-2 and GAS AI-3; GAS AI-2 and GAS AI-4; and GAS AI 3 and GAS AI-4. Preferably the combination includes fimbrial proteins from one or more GAS adhesin islands.
  • The immunogenic compositions may also be selected to provide protection against an increased range of GAS serotypes and strain isolates. For example, the immunogenic composition may comprise a first and second GAS AI protein, wherein a full length polynucleotide sequence encoding for the first GAS AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second GAS AI protein. In addition, each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple GAS serotypes and strain isolates. Preferably, each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) GAS strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) GAS serotypes.
  • Applicants have also identified adhesin islands within the genome of Streptococcus pneumoniae. These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence. Amino acid sequence encoded by such S. pneumoniae Adhesin Islands may be used in immunogenic compositions for the treatment or prevention of S. pneumoniae infection. Preferred immunogenic compositions of the invention comprise a S. pneumoniae AI surface protein which has been formulated or purified in an oligomeric (pilus) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. A preferred immunogenic composition of the invention alternatively comprises an isolated S. pneumoniae surface protein in oligomeric (pilus) form. The oligomer or hyperoligomeric pilus structures comprising S. pneumoniae surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • The S. pneumoniae Adhesin Islands generally include a series of open reading frames within a S. pneumoniae genome that encode for a collection of surface proteins and sortases. A S. pneumoniae Adhesin Island may encode for an amino acid sequence comprising at least one surface protein. Alternatively, the S. pneumoniae Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, a S. pneumoniae Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPTXG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more S. pneumoniae AI surface proteins may participate in the formation of a pilus structure on the surface of the S. pneumoniae bacteria.
  • The S. pneumoniae Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator. The transcriptional regulator may regulate the expression of the S. pneumonaie AI operon. An example of a transcriptional regulator found in S. pneumoniae AI sequences is rlrA.
  • A schematic of the organization of a S. pneumoniae AI locus is provided in FIG. 137. The locus comprises open reading frames encoding a transcriptional regulator (rlrA), cell wall surface proteins (rrgA, rrgB, rrgc) and sortases (srt B, srtC, srtD).
  • S. pneumoniae AI sequences may be generally divided into two groups of homology, S. pneuamoniae AI-a and AI-b. S. pneumoniae strains that comprise AI-a include 14 CSR 10, 19A Hungary 6, 23 F Poland 15, 670, 6B Finland 12, and 6B Spain 2. S. pneumoniae AI strains that comprise AI-b include 19F Taiwan 14, 9V Spain 3, 23F Taiwan 15 and TIGR 4.
  • S. pneumoniae AI from TIGR4 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from TIGR4 includes polynucleotide sequences encoding for two or more of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, and SP0468.
  • One or more of the S. pneumoniae AI from TIGR4 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from TIGR4 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae strain 670 AI comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae strain 670 AI includes polynucleotide sequences encoding for two or more of orf1 670, orf3 670, orf4 670, orf5 670, orf6 670, orf7 670, and orf8 670.
  • One or more of the S. pneumoniae strain 670 AI polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 670 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 14 CSR10 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 14 CSR10 includes polynucleotide sequences encoding for two or more of ORF214CSR, ORF314CSR, ORF414CSR, ORF514CSR, ORF614CSR, ORF714CSR, and ORF814CSR.
  • One or more of the S. pneumoniae AI from 14 CSR10 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 14 CSR10 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 19A Hungary 6 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 19A Hungary 6 includes polynucleotide sequences encoding for two or more of ORF219AH, ORF3 1 gAH, ORF419AH, ORF519AH, ORF619AH, ORF719AH, and ORF819AH.
  • One or more of the S. pneumoniae AI from 19A Hungary 6 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 19A Hungary 6 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 19F Taiwan 14 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 19F Taiwan 14 includes polynucleotide sequences encoding for two or more of ORF219FTW, ORF319FTW, ORF419FTW, ORF519FTW, ORF619FTW, ORF719FTW, and ORF819FTW.
  • One or more of the S. pneumoniae AI from 19F Taiwan 14 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 19F Taiwan 14 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 23F Poland 16 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 23F Poland 16 includes polynucleotide sequences encoding for two or more of ORF223FP, ORF323FP, ORF423FP, ORF523FP, ORF623FP, ORF723FP, and ORF823FP.
  • One or more of the S. pneumoniae AI from 23F Poland 16 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 23F Poland 16 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 23F Taiwan 15 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 23F Taiwan 15 includes polynucleotide sequences encoding for two or more of ORF223FTW, ORF323FTW, ORF423FTW, ORF523FTW, ORF623FTW, ORF723FTW, and ORF823FTW.
  • One or more of the S. pneumoniae AI from 23F Taiwan 15 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 23F Taiwan 15 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 6B Finland 12 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 6B Finland 12 includes polynucleotide sequences encoding for two or more of ORF26BF, ORF36BF, ORF46BF, ORF56BF, ORF66BF, ORF76BF, and ORF86BF.
  • One or more of the S. pneumoniae AI from 6B Finland 12 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 6B Finland 12 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 6B Spain 2 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 6B Spain 2 includes polynucleotide sequences encoding for two or more of ORF26BSP, ORF36BSP, ORF46BSP, ORF56BSP, ORF66BSP, ORF76BSP, and ORF86BSP.
  • One or more of the S. pneumoniae AI from 6B Spain 2 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 6B Spain 2 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • S. pneumoniae AI from 9V Spain 3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“S. pneumoniae AI proteins”). Specifically, S. pneumoniae AI from 9V Spain 3 includes polynucleotide sequences encoding for two or more of ORF29VSP, ORF39VSP, ORF49VSP, ORF59VSP, ORF69VSP, ORF79VSP, and ORF89VSP.
  • One or more of the S. pneumoniae AI from 9V Spain 3 polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae AI from 9V Spain 3 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae AI surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. These sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae AI may encode for at least one surface protein. The Adhesin Island, may encode at least one surface protein. Alternatively, S. pneumoniae AI may encode for at least two surface proteins and at least one sortase. Preferably, S. pneumoniae AI encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif.
  • The S. pneumoniae AI protein of the composition may be selected from the group consisting of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, SP0468, orf1 670, orf5 670, orf4 670, orf5 670, orf6 670, orf7 670, orf8 670, ORF214CSR, ORF314CSR, ORF414CSR, ORF514CSR, ORF614CSR, ORF714CSR, ORF814CSR, ORF219AH, ORF319AH, ORF419AH, ORF59AH, ORF619AH, ORF719AH, ORF819AH, ORF219FTW, ORF319FTW, ORF419FTW, ORF519FTW, ORF619FTW, ORF719FTW, ORF819FTW, ORF223FP, ORF323FP, ORF423FP, ORF523FP, ORF623FP, ORF723FP, ORF823FP, ORF223FTW, ORF323FTW, ORF423FTW, ORF523FTW, ORF623FTW, ORF723FTW, ORF823FTW, ORF26BF, ORF36BF, ORF46BF, ORF56BF, ORF66BF, ORF76BF, ORF86BF, ORF26BSP, ORF36BSP, ORF46BSP, ORF56BSP, ORF66BSP, ORF76BSP, ORF86BSP, ORF29VSP, ORF39VSP, ORF49VSP, ORF59VSP, ORF69VSP, ORF79VSP and, ORF89VSP.
  • S. pneumoniae AI surface proteins are preferred proteins for use in the immunogenic compositions of the invention. In one embodiment, the compositions of the invention comprise combinations of two or more S pneumoniae AI surface proteins. Preferably such combinations are selected from two or more of the group consisting of SP0462, SP0463, SP0464, orf3 670, orf4 670, orf5 670, ORF314CSR, ORF414CSR, ORF514CSR, ORF319AH, ORF419AH, ORF519AH, ORF319FTW, ORF419FTW, ORF5 1 gFTW, ORF323FP, ORF423FP, ORF523FP, ORF323FTW, ORF423FTW, ORF523FTW, ORF36BF, ORF46BF, ORF56BF, ORF36BSP, ORF46BSP, ORF56BSP, ORF39VSP, ORF49VSP, and ORF59VSP.
  • In addition to the open reading frames encoding the S. pneumoniae AI proteins, S. pneumoniae AI may also include a transcriptional regulator.
  • The S. pneumoniae AI proteins of the invention may be used in immunogenic compositions for prophylactic or therapeutic immunization against S. pneumoniae infection. For example, the invention may include an immunogenic composition comprising one or more S. pneumoniae from TIGR4 AI proteins and one or more S. pneumoniae strain 670 proteins. The immunogenic composition may comprise one or more AI proteins from any one or more of S. pneumoniae strains TIGR4, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 14 CSR 10, 19F Taiwan 14, 23F Taiwan 15, 23F Poland 16, and 670.
  • The immunogenic compositions may also be selected to provide protection against an increased range of S. pneumoniae serotypes and strain isolates. For example, the immunogenic composition may comprise a first and second S. pneumoniae AI protein, wherein a full length polynucleotide sequence encoding for the first S. pneumoniae AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second S. pneumoniae AI protein. In addition, each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple S. pneumoniae serotypes and strain isolates. Preferably, each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) S. pneumoniae strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) S. pneumoniae serotypes.
  • The immunogenic compositions may also be selected to provide protection against an increased range of serotypes and strain isolates of a Gram positive bacteria. For example, the immunogenic composition may comprise a first and second Gram positive bacteria AI protein, wherein a full length polynucleotide sequence encoding for the first Gram positive bacteria AI protein is not present in a genome comprising a full length polynucleotide sequence encoding for the second Gram positive bacteria AI protein. In addition, each antigen selected for use in the immunogenic compositions will preferably be present in the genomes of multiple serotypes and strain isolates of the Gram positive bacteria. Preferably, each antigen is present in the genomes of at least two (i.e., 3, 4, 5, 6, 7, 8, 9, 10, or more) Gram positive bacteria strain isolates. More preferably, each antigen is present in the genomes of at least two (i.e., at least 3, 4, 5, or more) Gram positive bacteria serotypes. One or both of the first and second AI proteins may preferably be in oligomeric or hyperoligomeric form.
  • Adhesin island surface proteins from two or more Gram positive bacterial genus or species may be combined to provide an immunogenic composition for prophylactic or therapeutic treatment of disease or infection of two more Gram positive bacterial genus or species. Optionally, the adhesin island surface proteins may be associated together in an oligomeric or hyperoligomeric structure.
  • In one embodiment, the invention comprises adhesin island surface proteins from two or more Streptococcus species. For example, the invention includes a composition comprising a GBS AI surface protein and a GAS adhesin island surface protein. As another example, the invention includes a composition comprising a GAS adhesin island surface protein and a S. pneumoniae adhesin island surface protein. One or both of the GAS AI surface protein and the S. pneumoniae AI surface protein may be in oligomeric or hyperoligomeric form. As a further example, the invention includes a composition comprising a GBS adhesin island surface protein and a S. pneumoniae adhesin island surface protein.
  • In one embodiment, the invention comprises an adhesin island surface protein from two or more Gram positive bacterial genus. For example, the invention includes a composition comprising a Streptococcus adhesin island protein and a Corynebacterium adhesin island protein. One or more of the Gram positive bacteria AI surface proteins may be in an oligomeric or hyperoligomeric form.
  • In addition, the AI polynucleotides and amino acid sequences of the invention may also be used in diagnostics to identify the presence or absence of GBS (or a Gram positive bacteria) in a biological sample. They may be used to generate antibodies which can be used to identify the presence of absence of an AI protein in a biological sample or in a prophylactic or therapeutic treatment for GBS (or a Gram positive bacterial) infection. Further, the AI polynucleotides and amino acid sequences of the invention may also be used to identify small molecule compounds which inhibit or decrease the virulence associated activity of the AI.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 presents a schematic depiction of Adhesin Island 1 (“AI-1”) comprising open reading frames for GBS 80, GBS 52, SAG0647, SAG0648 and GBS 104.
  • FIG. 2 illustrates the identification of AI-1 sequences in several GBS serotypes and strain isolates (GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate nem316; GBS serotype II, strain isolate 18RS21; GBS serotype V, strain isolate CJB111; GBS serotype III, strain isolate COH1 and GBS serotype 1a, strain isolate A909). (An AI-1 was not identified in GBS serotype 1b, strain isolate H36B or GBS serotype 1a, strain isolate 515).
  • FIG. 3 presents a schematic depiction of the correlation between AI-1 and the Adhesin Island 2 (“AI-2”) within the GBS serotype V, strain isolate 2603 genome. (This AI-2 comprises open reading frames for GBS 67, GBS 59, SAG1406, SAG1405 and GBS 150).
  • FIG. 4 illustrates the identification of AI-2 comprising open reading frames encoding for GBS 67, GBS 59, SAG1406, SAG1404 and GBS 150 (or sequences having sequence homology thereto) in several GBS serotypes and strain isolates (GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate NEM316; GBS serotype 1b, strain isolate H36B; GBS serotype V, strain isolate CJB111; GBS serotype II, strain isolate 18RS21; and GBS serotype 1a, strain isolate 515).
  • FIG. 4 also illustrates the identification of AI-2 comprising open reading frames encoding for 01520 (a sortase), 01521, 01522 (a sortase), 01523 (spb1), 01524 and 01525 (or sequences having sequence homology thereto).
  • FIG. 5 presents data showing that GBS 80 binds to fibronectin and fibrinogen in ELISA.
  • FIG. 6 illustrates that all genes in AI-1 are co-transcribed as an operon.
  • FIG. 7 presents schematic depictions of in-frame deletion mutations within AI-1.
  • FIG. 8 presents FACS data showing that GBS 80 is required for surface localization of GBS 104.
  • FIG. 9 presents FACS data showing that sortases SAG0647 and SAG0648 play a semi-redundant role in surface exposure of GBS 80 and GBS 104.
  • FIG. 10 presents Western Blots of the in-frame deletion mutants probed with anti-GBS80 and anti-GBS 104 antisera.
  • FIG. 11: Electron micrograph of surface exposed pili structures in Streptococcus agalactiae containing GBS 80.
  • FIG. 12: PHD predicted secondary structure of GBS 067.
  • FIGS. 13, 14 and 15: Electron micrograph of surface exposed pili structures of strain isolate COH1 of Streptococcus agalactiae containing a plasmid insert encoding GBS 80.
  • FIGS. 16 and 17: Electron micrograph of surface exposed pili structure of wild type strain isolate COH1 of Streptococcus agalactiae.
  • FIG. 18: Alignment of polynucleotide sequences of AI-1 from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype HI, strain isolates COH1 and NEM316; and serotype 1a, strain isolate A909.
  • FIG. 19: Alignment of polynucleotide sequences of AI-2 from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype 1b, strain isolate H36B; and serotype 1a, strain isolate 515.
  • FIG. 20: Alignment of polynucleotide sequences of AI-2 from serotype V, strain isolate 2603 and serotype III, strain isolate NEM316.
  • FIG. 21: Alignment of polynucleotide sequences of AI-2 from serotype III, strain isolate COH1 and serotype Ia, strain isolate A909.
  • FIG. 22: Alignment of amino acid sequences of AI-1 surface protein GBS 80 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate A909; serotype III, strain isolates COH1 and NEM316.
  • FIG. 23: Alignment of amino acid sequences of AI-1 surface protein GBS 104 from serotype V, strain isolates 2603 and CJB111; serotype III, strain isolates COH1 and NEM316; and serotype II, strain isolate 18RS21.
  • FIG. 24: Alignment of amino acid sequences of AI-2 surface protein GBS 067 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate 515; serotype II, strain isolate 18RS21; serotype Ib, strain isolate H36B; and serotype III, strain isolate NEM316.
  • FIG. 25: Illustrates that GBS closely associates with tight junctions and cross the monolayer of ME180 cervical epithelial cells by a paracellular route.
  • FIG. 26: Illustrates GBS infection of ME180 cells.
  • FIG. 27: Illustrates that GBS 80 recombinant protein does not bind to epithelial cells.
  • FIG. 28: Illustrates that deletion of GBS 80 does not effect the capacity of GBS strain 2603 V/R to adhere and invade ME180 cervical epithelial cells.
  • FIG. 29: Illustrates binding of recombinant GBS 104 protein to epithelial cells.
  • FIG. 30: Illustrates that deletion of GBS 104 in the GBS strain COH1, reduces the capacity of GBS to adhere to ME180 cervical epithelial cells.
  • FIG. 31: Illustrates that GBS 80 knockout mutant strain partially loses the ability to translocate through an epithelial cell monolayer.
  • FIG. 32: Illustrates that deletion of GBS 104, but not GBS 80, reduces the capacity of GBS to invade J774 macrophage-like cell line.
  • FIG. 33: Illustrates that GBS 104 knockout mutant strain translocates through an epithelial monolayer less efficiently than the isogenic wild type.
  • FIG. 34: Negative stained electron micrographs of GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80.
  • FIG. 35: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 10 nm gold particles).
  • FIG. 36: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 10 nm gold particles).
  • FIG. 37: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles).
  • FIG. 38: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 104 antibodies or preimmune sera (visualized with 10 nm gold particles).
  • FIG. 39: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles) and anti-GBS 104 antibodies (visualized with 10 nm gold particles).
  • FIG. 40: Electron micrographs of surface exposed pili structures on GBS serotype III, strain isolate COH1, containing a plasmid insert to over-express GBS 80, stained with anti-GBS 80 antibodies (visualized with 20 nm gold particles) and anti-GBS 104 antibodies (visualized with 10 nm gold particles).
  • FIG. 41: Illustrates that GBS 80 is necessary for polymer formation and GBS104 and sortase SAG0648 are necessary for efficient assembly of pili.
  • FIG. 42: Illustrates that GBS 67 is part of a second pilus and that GBS 80 is polymerized in strain 515.
  • FIG. 43: Illustrates that two macro-molecules are visible in Coh1, one of which is the GBS 80 pilin.
  • FIG. 44: Illustrates pilin assembly.
  • FIG. 45: Illustrates that GBS 52 is a minor component of the GBS pilus.
  • FIG. 46: Illustrates that the pilus is found in the supernatant of a bacterial culture.
  • FIG. 47: Illustrates that the pilus is found in the supernatant of bacterial cultures in all phases.
  • FIG. 48: Illustrates that in Coh1, only the GBS 80 protein and one sortase (sag0647 or sag0648) is required for polymerization.
  • FIG. 49: IEM image of GBS 80 staining of a GBS serotype VIII strain JM9030013 that express pili.
  • FIG. 50: IEM image of GBS 104 staining of a GBS serotype VIII strain JM9030013 that express pili.
  • FIG. 51A: Schematic depiction of open reading frames comprising a GAS AI-2 serotype M1 isolate, GAS AI-3 serotype M3, M5, M18, and M49 isolates, a GAS AI-4 serotype M12 isolate, and an GAS AI-1 serotype M6 isolate.
  • FIG. 51B: Amino acid alignment of SrtC1-type sortase of a GAS AI-2 serotype M1 isolate, SrtC2-type sortases of serotype M3, M5, M118, and M49 isolates, and a SrtC2-type sortase of a GAS AI-4 serotype M12 isolate.
  • FIG. 52: Amino acid alignment of the capsular polysaccharide adhesion proteins of GAS AI-4 serotype M12 (A735), GAS AI-3 serotype M5 (Manfredo), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI-1 serotype M3, S. pyogenes strain MGAS8232 serotype M3, and GAS AI-2 serotype M1.
  • FIG. 53: Amino acid alignment of F-like fibronectin-binding proteins of GAS AI-4 serotype M12 (A735) and S. pyogenes strain MGAS10394 serotype M6.
  • FIG. 54: Amino acid alignment of F2-like fibronectin-binding proteins of GAS AI-4 serotype M12 (A735), S. pyogenes strain MGAS8232 serotype M3, GAS AI-3 strain M5 (Manfredo), S. pyogenes strain SSI serotype M3, and S. pyogenes strain MGAS315 serotype M3.
  • FIG. 55: Amino acid alignment of fimbrial proteins of GAS AI-4 serotype M12 (A735), GAS AI-3 serotype M5 (Manfredo), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI serotype M3, S. pyogenes strain MGAS8232 serotype M3, and S. pyogenes M1 GAS serotype M1.
  • FIG. 56: Amino acid alignment of hypothetical proteins of GAS AI-4 serotype M12 (A735), S. pyogenes strain MGAS315 serotype M3, S. pyogenes strain SSI-1 serotype M3, GAS AI-3 serotype M5 (Manfredo), and S. pyogenes strain MGAS8232 serotype M3.
  • FIG. 57: Results of FASTA homology search for amino acid sequences that align with the collagen adhesion protein of GAS AI-1 serotype M6 (MGAS10394).
  • FIG. 58: Results of FASTA homology search for amino acid sequences that align with the fimbrial structural subunit of GAS AI-1 serotype M6 (MGAS10394).
  • FIG. 59: Results of FASTA homology search for amino acid sequences that align with the hypothetical protein of GAS AI-2 serotype M1 (SF370).
  • FIG. 60: Specifies pilin and E box motifs present in GAS type 3 and 4 adhesin islands.
  • FIG. 61: Illustrates that surface expression of GBS 80 protein on GBS strains COH and JM9130013 correlates with formation of pili structures. Surface expression of GBS 80 was determined by FACS analysis using an antibody that cross-hybridizes with GBS 80. Formation of pili structures was determined by immunogold electron microscopy using gold-labelled anti-GBS 80 antibody.
  • FIG. 62: Illustrates that surface exposure is capsule-dependent for GBS 322 but not for GBS 80.
  • FIG. 63: Illustrates the amino acid sequence identity of GBS 59 proteins in GBS strains.
  • FIG. 64: Western blotting of whole GBS cell extracts with anti-GBS 59 antibodies.
  • FIG. 65: Western blotting of purified GBS 59 and whole GBS cell extracts with anti-GBS 59 antibodies.
  • FIG. 66: FACS analysis of GBS strains CJB111, 7357B, 515 using GBS 59 antiserum.
  • FIG. 67: Illustrates that anti-GBS 59 antibodies are opsonic for CJB111 GBS strain serotype V.
  • FIG. 68: Western blotting of GBS strain JM9130013 total extracts.
  • FIG. 69: Western blotting of GBS stain 515 total extracts shows that GBS 67 and GBS 150 are parts of a pilus.
  • FIG. 70: Western blotting of GBS strain 515 knocked out for GBS 67 expression
  • FIG. 71: FACS analysis of GBS strain 515 and GBS strain 515 knocked out for GBS 67 expression using GBS 67 and GBS 59 antiserum.
  • FIG. 72: Illustrates complementation of GBS 515 knocked out for GBS 67 expression with a construct overexpressing GBS 80.
  • FIG. 73: FACS analysis of GAS serotype M6 for spyM6 0159 surface expression.
  • FIG. 74: FACS analysis of GAS serotype M6 for spyM6 0160 surface expression.
  • FIG. 75: FACS analysis of GAS serotype M1 for GAS 15 surface expression.
  • FIG. 76: FACS analysis of GAS serotype M1 for GAS 16 surface expression using a first anti-GAS 16 antiserum.
  • FIG. 77: FACS analysis of GAS serotype M1 for GAS 18 surface expression using a first anti-GAS 18 antiserum.
  • FIG. 78: FACS analysis of GAS serotype M1 for GAS 18 surface expression using a second anti-GAS 18 antiserum.
  • FIG. 79: FACS analysis of GAS serotype M1 for GAS 16 surface expression using a second anti-GAS 16 antisera.
  • FIG. 80: FACS analysis of GAS serotype M3 for spyM30098 surface expression.
  • FIG. 81: FACS analysis of GAS serotype M3 for spyM3 0100 surface expression.
  • FIG. 82: FACS analysis of GAS serotype M3 for spyM3 0102 surface expression.
  • FIG. 83: FACS analysis of GAS serotype M3 for spyM30104 surface expression.
  • FIG. 84: FACS analysis of GAS serotype M3 for spyM30106 surface expression.
  • FIG. 85: FACS analysis of GAS serotype M12 for 19224134 surface expression.
  • FIG. 86: FACS analysis of GAS serotype M12 for 19224135 surface expression.
  • FIG. 87: FACS analysis of GAS serotype M12 for 19224137 surface expression.
  • FIG. 88: FACS analysis of GAS serotype M12 for 19224141 surface expression.
  • FIG. 89: Western blot analysis of GAS 15 expression on GAS M1 bacteria.
  • FIG. 90: Western blot analysis of GAS 15 expression using GAS 15 immune sera.
  • FIG. 91: Western blot analysis of GAS 15 expression using GAS 15 pre-immune sera.
  • FIG. 92: Western blot analysis of GAS 16 expression on GAS M1 bacteria.
  • FIG. 93: Western blot analysis of GAS 16 expression using GAS 16 immune sera.
  • FIG. 94: Western blot analysis of GAS 16 expression using GAS 16 pre-immune sera.
  • FIG. 95: Western blot analysis of GAS 18 on GAS M1 bacteria.
  • FIG. 96: Western blot analysis of GAS 18 using GAS 18 immune sera.
  • FIG. 97: Western blot analysis of GAS 18 using GAS 18 pre-immune sera.
  • FIG. 98: Western blot analysis of M6_Spy0159 expression on GAS bacteria.
  • FIG. 99: Western blot analysis of 19224135 expression on M12 GAS bacteria.
  • FIG. 100: Western blot analysis of 19224137 expression on M12 GAS bacteria.
  • FIG. 101: Full length nucleotide sequence of an S. pneumoniae strain 670 μl.
  • FIG. 102: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2580.
  • FIG. 103: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2913.
  • FIG. 104: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 3280.
  • FIG. 105: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 3348.
  • FIG. 106: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain 2719.
  • FIG. 107: Western blot analysis of GAS 15, GAS 16, and GAS 18 in GAS M1 strain SF370.
  • FIG. 108: Western blot analysis of 19224135 and 19224137 in GAS M12 strain 2728.
  • FIG. 109: Western blot analysis of 19224139 in GAS M12 strain 2728 using antisera raised against SpyM3 0102.
  • FIG. 110: Western blot analysis of M6_Spy0159 and M6_Spy0160 in GAS M6 strain 2724.
  • FIG. 111: Western blot analysis of M6 Spy0159 and M6 Spy0160 in GAS M6 strain SF370.
  • FIG. 112: Western blot analysis of M6_Spy160 in GAS M6 strain 2724.
  • FIGS. 113-115: Electron micrographs of surface exposed GAS 15 on GAS M1 strain SF370.
  • FIGS. 116-121: Electron micrographs of surface exposed GAS 16 on GAS M1 strain SF370.
  • FIGS. 122-125: Electron micrographs of surface exposed GAS 18 on GAS M1 strain SF370 detected using anti-GAS 18 antisera.
  • FIG. 126: IEM image of a hyperoligomer on GAS M1 strain SF370 detected using anti-GAS 18 antisera.
  • FIGS. 127-132: IEM images of oligomeric and hyperoligomeric structures containing M6_Spy0160 extending from the surface of GAS serotype M6 3650.
  • FIGS. 133A and B: Western blot analysis of L. lactis transformed to express GBS 80 with anti-GBS 80 antiserum.
  • FIG. 134: Western blot analyses of L. lactis transformed to express GBS AI-1 with anti-GBS 80 antiserum.
  • FIG. 135: Ponceau staining of same acrylamide gel as used in FIG. 134.
  • FIG. 136A: Western blot analysis of sonicated pellets and supernatants of cultured L. lactis transformed to express GBS AI-1 polypeptides using anti-GBS 80 antiserum.
  • FIG. 136B: Polyacrylamide gel electrophoresis of sonicated pellets and supernatants of cultured L. lactis transformed to express GBS AI polypeptides.
  • FIG. 137: Depiction of an example S. pneumoniae AI locus.
  • FIG. 138: Schematic of primer hybridization sites within the S. pneumoniae AI locus of FIG. 137.
  • FIG. 139A: The set of amplicons produced from the S. pneumoniae strain TIGR4 AI locus.
  • FIG. 139B: Base pair lengths of amplicons produced from FIG. 139A primers in S. pneumoniae strain TIGR4.
  • FIG. 140: CGH analysis of S. pneumoniae strains for the AI locus.
  • FIG. 141: Amino acid sequence alignment of polypeptides encoded by AI orf 2 in S. pneumoniae AI-positive strains.
  • FIG. 142: Amino acid sequence alignment of polypeptides encoded by AI orf 3 in S. pneumoniae AI-positive strains.
  • FIG. 143: Amino acid sequence alignment of polypeptides encoded by AI orf 4 in S. pneumoniae AI-positive strains.
  • FIG. 144: Amino acid sequence alignment of polypeptides encoded by AI orf 5 in S. pneumoniae AI-positive strains.
  • FIG. 145: Amino acid sequence alignment of polypeptides encoded by AI orf 6 in S. pneumoniae AI-positive strains.
  • FIG. 146: Amino acid sequence alignment of polypeptides encoded by AI orf 7 in S. pneumoniae AI-positive strains.
  • FIG. 147: Amino acid sequence alignment of polypeptides encoded by AI orf 8 in S. pneumoniae AI-positive strains.
  • FIG. 148: Diagram comparing amino acid sequences of RrgA in S. pneumoniae strains.
  • FIG. 149: Amino acid sequence comparison of RrgB S. pneumoniae strains.
  • FIG. 150A: Sp0462 amino acid sequence.
  • FIG. 150B: Primers used to produce a clone encoding the Sp0462 polypeptide.
  • FIG. 151A: Schematic depiction of recombinant Sp0462 polypeptide.
  • FIG. 151B: Schematic depiction of full-length Sp0462 polypeptide.
  • FIG. 152A: Western blot probed with serum obtained from S. pneumoniae-infected patients for Sp0462.
  • FIG. 152B: Western blot probed with GBS 80 serum for Sp0462.
  • FIG. 153A: Sp0463 amino acid sequence.
  • FIG. 153B: Primers used to produce a clone encoding the Sp0463 polypeptide.
  • FIG. 154A: Schematic depiction of recombinant Sp0463 polypeptide.
  • FIG. 154B: Schematic depiction of full-length Sp0463 polypeptide.
  • FIG. 155: Western blot detection of recombinant Sp0463 polypeptide.
  • FIG. 156: Western blot detection of high molecular weight Sp0463 polymers.
  • FIG. 157A: Sp0464 amino acid sequence.
  • FIG. 157B: Primers used to produce a clone encoding the Sp0464 polypeptide.
  • FIG. 158A: Schematic depiction of recombinant Sp0464 polypeptide.
  • FIG. 158B: Schematic depiction of full-length Sp0464 polypeptide.
  • FIG. 159: Western blot detection of recombinant Sp0464 polypeptide.
  • FIG. 160: Amplification products prepared for production of Sp0462, Sp0463, and Sp0464 clones.
  • FIG. 161: Opsonic killing by anti-sera raised against L. lactis expressing GBS AI
  • FIG. 162: Schematic depicting GAS adhesin islands GAS AI-1, GAS AI-2, GAS AI-3 and GAS AI-4.
  • FIGS. 163 A-D: Immunoblots of cell-wall fractions of GAS strains with antisera specific for LPXTG proteins of M6_ISS3650 (A), M1_SF370 (B), M5_ISS4883 (C) and M1220010296 (D).
  • FIGS. 163 E-H: Immunoblots of cell-wall fractions of deletion mutants M1_SF370Δ128 (E) M1_SF370Δ130 (F) M1_SF370ΔSrtC1 (G) and the M1 128 deletion strain complemented with plasmid pAM::128 which contains the M1 128 gene (H) with antisera specific for the pilin components of M1_SF370.
  • FIGS. 163 I-N: Immunogold labelling and transmission electron microscopy of: T6 (I) and Cpa (J) in M6_ISS3650; M1 128 in M1_SF370 (K) and deletion strain M1_SF370Δ128 (N); M5_orf80 in M5_ISS4883 (L); M12_EftLSL.A in M1220010296 (M). The strains used are indicated below the panels. Bars=200 nm.
  • FIG. 164: Schematic representation of the FCT region from 7 GAS strains
  • FIGS. 165 A-H: Flow cytometry of GAS bacteria treated or not with trypsin and stained with sera specific for the major pilus component. Preimmune staining; black lines, untreated bacteria; green lines and trypsin treated bacteria; blue lines. M6_ISS3650 stained with sera which recognize the M6 protein (A) or anti-M6_T6 (B), M1_SF370 stained with anti-M1 (C) or anti-M1128 (D), M5_ISS4883 stained with anti-PrtF (E) or anti-M5_orf80 (F) and M1220010296 with anti-M12 (G) or anti-EftLSL.A (H)
  • FIGS. 166 A-C: Immunoblots of recombinant pilin components with polyvalent Lancefield T-typing sera. The recombinant proteins are shown above the blot and the sera pool used is shown below the blot.
  • FIGS. 166 D-G: Immunoblots of pilin proteins with monovalent T-typing sera. The recombinant proteins are shown below the blot and the sera used above the blot.
  • FIG. 166 H and I Flow cytometry analysis of strain M1_SF370 (H) and the deletion strain M1_SF370Δ128 (I) with T-typing antisera pool T.
  • FIG. 167: Chart describing the number and type of sortase sequences identified within GAS AIs.
  • FIG. 168 A: Immunogold-electronmicroscopy of L. lactis lacking an expression construct for GBS AI-1 using anti-GBS 80 antibodies.
  • FIG. 168 B and C: Immunogold-electronmicroscopy detects GBS 80 in oligomeric (pilus) structures on surface of L. lactis transformed to express GBS AI-1
  • FIG. 169: FACS analysis detects expression of GBS 80 and GBS 104 on the surface of L. lactis transformed to express GBS AI-1.
  • FIG. 170: Phase contrast microscopy and immuno-electronmicroscopy shows that expression of GBS AI-1 in L. lactis induces L. lactis aggregation.
  • FIG. 171: Purification of GBS pili from L. lactis transformed to express GBS AI-1.
  • FIG. 172: Schematic depiction of GAS M6 (AI-1), M1 (AI-2), and M12 (AI-4) adhesin islands and portions of the adhesin islands inserted in the pAM401 construct for expression in L. lactis.
  • FIG. 173 A-C: Western blot analysis showing assembly of GAS pili in L. lactis expressing GAS AI-2 (M1) (A), GAS AI-4 (M12) (B), and GAS AI-1(M6) (C).
  • FIG. 174: FACS analysis of GAS serotype M6 for M6_Spy0157 surface expression.
  • FIG. 175: FACS analysis of GAS serotype M12 for 19224139 surface expression.
  • FIG. 176 A-E: Immunogold electron microscopy using antibodies against M6_Spy0160 detects pili on the surface of M6 strain 2724.
  • FIG. 176 F: Immunogold electron microscopy using antibodies against M6_Spy0159 detects M6_Spy0159 surface expression on M6 strain 2724.
  • FIG. 177 A-C: Western blot analysis of M1 strain SF370 GAS bacteria individually deleted for M1 130, SrtC1, or M1 128 using anti-M1 130 serum (A), anti-M1 128 serum (B), and anti-M1 126 serum (C).
  • FIG. 178 A-C: Immunogold electron microscopy using antibodies against M1 128 to detect surface expression on wildtype strain SF370 bacteria (A), M1 128 deleted SF370 bacteria (B), and SrtC1 deleted SF370 bacteria (C).
  • FIG. 179 A-C: FACS analysis to detect expression of M11126 (A), M1128 (B), and M1130 (C) on the surface of wildtype SF370 GAS bacteria.
  • FIG. 179 D-F: FACS analysis to detect expression of M1126 (D), M1128 (E), and M1130 (F) on the surface of M1 128 deleted SF370 GAS bacteria.
  • FIG. 179 G-I: FACS analysis to detect expression of M1126 (G), M1128 (H), and M1130 (I) on the surface of SrtC1 deleted SF370 GAS bacteria.
  • FIG. 180 A and B: FACS analysis of wildtype (A) and LepA deletion mutant (B) strains of SF370 bacteria for M1 surface expression.
  • FIG. 181: Western blot analysis detects high molecular weight polymers in S. pneumoniae TIGR4 using anti-RrgB antisera.
  • FIG. 182: Detection of high molecular weight polymers in S. pnuemoniae rlrA positive strains.
  • FIG. 183: Detection of high molecular weight polymers in S. pneumoniae TIGR4 by silver staining and Western blot analysis using anti-RrgB antisera.
  • FIG. 184: Deletion of S. pneumoniae TIGR4 adhesin island sequences interferes with the ability of S. pneumoniae to adhere to A549 alveolar cells.
  • FIG. 185: Negative staining of S. pneumoniae strain TIGR4 showing abundant pili on the bacterial surface.
  • FIG. 186: Negative staining of strain TIGR4 deleted for rrgA-srtD adhesin island sequences showing no pili on the bacterial surface
  • FIG. 187: Negative staining of the TIGR4 mgrA mutant showing abundant pili on the bacterial surface.
  • FIG. 188: Negative staining of the negative control TIGR4 mgrA mutant deleted for adhesin island sequences rrgA-srtD showing no pili on the bacterial surface.
  • FIG. 189: Immuno-gold labelling of S. pneumoniae strain TIGR4 grown on blood agar solid medium using α-RrgB (5 nm) and α-RrgC (10 nm). Bar represents 200 nm.
  • FIG. 190 A and B: Detection of expression and purification of S. pneumoniae RrgA protein by SDS-PAGE (A) and Western blot analysis (B).
  • FIG. 191: Detection of RrgB by antibodies produced in mice.
  • FIG. 192: Detection of RrgC by antibodies produced in mice.
  • FIG. 193: Purification of S. pneumoniae TIGR 4 pili by a cultivation and digestion method and detection of the purified TIGR4 pili.
  • FIG. 194: Purification of S. pneumoniae TIGR 4 pili by a sucrose gradient centrifugation method and detection of the purified TIGR4 pili.
  • FIG. 195: Purification of S. pneumoniae TIGR 4 pili by a gel filtration method and detection of the purified TIGR4 pili.
  • FIG. 196: Alignment of full length S. pneumoniae adhesin island sequences from ten S. pneumoniae strains.
  • FIG. 197 A: Schematic of GBS AI-1 coding sequences.
  • FIG. 197 B: Nucleotide sequence of intergenic region between AraC and GBS 80 (SEQ ID NO: 273.
  • FIG. 197 C: FACS analysis results for GBS 80 expression in GBS strains having different length polyA tracts in the intergenic region between AraC and GBS 80.
  • FIG. 198: Table comparing the percent identity of surface proteins encoded by a serotype M6 (harbouring a GAS AI-1) adhesin island relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 199: Table comparing the percent identity of surface proteins encoded by a serotype M1 (harbouring a GAS AI-2) adhesin island relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 200: Table comparing the percent identity of surface proteins encoded by serotypes M3, M18, M5, and M49 (harbouring GAS AI-3) adhesin islands relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 201: Table comparing the percent identity of surface proteins encoded by a serotype M12 (harbouring a GAS AI-1) adhesin island-relative to other GAS serotypes harbouring an adhesin island.
  • FIG. 202: GBS 80 recombinant protein does not bind to epithelial cells.
  • FIG. 203: Deletion of GBS 80 protein does not affect the ability of GBS to adhere and invade ME180 cervical epithelial cells.
  • FIG. 204: GBS 80 binds to extracellular matrix proteins.
  • FIG. 205: Deletion of GBS 104 protein, but not GBS 80, reduces the capacity of GBS to invade J774 macrophage-like cells
  • FIG. 206: GBS 104 knockout mutant strains of bacteria translocate through an epithelial monolayer less efficiently that the isogenic wild type strain.
  • FIG. 207: GBS 80 knockout mutant strains of bacteria partially lose the ability to translocate through an epithelial monolayer.
  • FIG. 208: GBS adherence to HUVEC endothelial cells.
  • FIG. 209: Strain growth rate of wildtype, GBS 80-deleted, or GBS 104 deleted COH1 GBS.
  • FIG. 210: Binding of recombinant GBS 104 protein to epithelial cells by FACS analysis.
  • FIG. 211: Deletion of GBS 104 protein in the GBS strain COH1 reduces the ability of GBS to adhere to ME180 cervical epithelial cells.
  • FIG. 212: COH1 strain GBS overexpressing GBS 80 protein has an impaired capacity to translocate through an epithelial monolayer.
  • FIG. 213: Scanning electron microscopy shows that overexpression of GBS 80 protein on COH1 strain GBS enhances the capacity of the COH1 bacteria to form microcolonies on epithelial cells.
  • FIG. 214: Confocal imaging shows that overexpression of GBS 80 proteins on COH1 strain GBS enhances the capacity of the COH1 bacteria to form microcolonies on epithelial cells.
  • FIG. 215: Detection of GBS 59 on the surface of GBS strain 515 by immuno-electron microscopy.
  • FIG. 216: Detection of GBS 67 on the surface of GBS strain 515 by immuno-electron microscopy.
  • FIG. 217: GBS 67 binds to fibronectin.
  • FIG. 218: Western blot analysis shows that deletion of both GBS AI-2 sortase genes abolishes assembly of the pilus.
  • FIG. 219: FACS analysis shows that deletion of both GBS AI-2 sortase genes abolishes assembly of the pilus.
  • FIG. 220 A-C: Western blot analysis shows that GBS 59, GBS 67, and GBS 150 form high molecular weight complexes.
  • FIG. 221 A-C: Western blot analysis shows that GBS 59 is required for polymer formation of GBS 67 and GBS 150.
  • FIG. 222: FACS analysis shows that GBS 59 is required for surface exposure of GBS 67.
  • FIG. 223: Summary Western blots for detection of GBS 59, GBS 67, or GBS 150 in GBS 515 and GBS 515 mutant strain.
  • FIG. 224: Description of GBS 59 Allelic variants.
  • FIG. 225: GBS 59 is opsonic only against a strain of GBS expressing a homologous GBS 59.
  • FIG. 226 A and B: Results of FACS analysis for surface expression of GBS 59 using antibodies specific for different GBS 59 isoforms.
  • FIG. 227 A and B: Results of FACS analysis for surface expression of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 on 41 various strains of GBS bacteria.
  • FIG. 228: Results of FACS analysis for surface expression of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 on 41 strains of GBS bacteria obtained from the CDC.
  • FIG. 229: Expected immunogenicity coverage of different combinations of GBS 80, GBS 104, GBS 322, GBS 67, and GBS 59 across strains of GBS bacteria.
  • FIG. 230: GBS 59 opsonophagocytic activity is comparable to that of a mixture of GBS 80, GBS 104, GBS 322 and GBS 67.
  • FIG. 231 A-C: Schematic presentation of example hybrid GBS AIs.
  • FIG. 232: Schematic presentation of an example hybrid GBS AI.
  • FIG. 233 A and B: Western blot and FACS analysis detect expression of GBS 80 and GBS 67 on the surface of L. lactis transformed with a hybrid GBS AI.
  • FIG. 234 A-E Hybrid GBS AI cloning strategy.
  • FIG. 235: High magnification of S. pneumoniae strain TIGR4 pili double labeled with α-RrgB (5 nm) and α-RrgC (10 nm). Bar represents 100 nm.
  • FIG. 236: Immuno-gold labeling of the S. pneumoniae TIGR4 rrgA-srtD deletion mutant with no visible pili on the surface detectable by α-RrgB- and α-RrgC. Bar represents 200 nm.
  • FIG. 237: Variability in GBS 67 amino acid sequences between strains 2603 and H36B.
  • FIG. 238: Strain variability in GBS 67 amino acid sequences of allele I (2603).
  • FIG. 239: Stran variability in GBS 67 amino acid sequence of allele II (H36B).
  • BRIEF DESCRIPTION OF THE TABLES
  • TABLE 1: Active Maternal Immunization Assay for fragments of GBS 80
  • TABLE 2: Passive Maternal Immunization Assay for fragments of GBS 80
  • TABLE 3: Lethal dose 50% of AI-1 mutants from GBS strain isolate 2603.
  • TABLE 4: GAS AI-1 sequences from M6 isolate (MGAS10394).
  • TABLE 5: GAS AI-2 sequences from M1 isolate (SF370).
  • TABLE 6: GAS AI-3 sequences from M3 isolate (MGAS315).
  • TABLE 7: GAS AI-3 sequences from M3 isolate (SSI-1).
  • TABLE 8: GAS AI-3 sequences from M18 isolate (MGAS8232).
  • TABLE 9: S. pneumoniae AI sequences from TIGR4 sequence.
  • TABLE 10: GAS AI-3 sequences from M5 isolate (Manfredo).
  • TABLE 11: GAS AI-4 sequences from M12 isolate (A735).
  • TABLE 12: Conservation of GBS 80 and GBS 104 amino acid sequences.
  • TABLE 13: Conservation of GBS 322 and GBS 276 amino acid sequences.
  • TABLE 14: Active maternal immunization assay for a combination of fragments from GBS 322, GBS 80, GBS 104, and GBS 67.
  • TABLE 15: Antigen surface exposure of GBS 80, GBS 322, GBS 104, and GBS 67.
  • TABLE 16: Active maternal immunization assay for each of GBS 80 and GBS 322 antigens.
  • TABLE 17: Active maternal immunization assay for GBS 59.
  • TABLE 18: Summary of FACS values for surface expression of spyM6 0159.
  • TABLE 19: Summary of FACS values for surface expression of spyM6 0160.
  • TABLE 20: Summary of FACS values for surface expression of GAS 15.
  • TABLE 21: Summary of FACS values for surface expression of GAS 16.
  • TABLE 22: Summary of FACS values for surface expression of GAS 16 using a second antisera.
  • TABLE 23: Summary of FACS values for surface expression of GAS 18.
  • TABLE 24: Summary of FACS values for surface expression of GAS 18 using a second antisera.
  • TABLE 25: Summary of FACS values for surface expression of SpyM30098.
  • TABLE 26: Summary of FACS values for surface expression of SpyM3 0100.
  • TABLE 27: Summary of FACS values for surface expression of SpyM3 0102 in M3 serotypes.
  • TABLE 28: Summary of FACS values for surface expression of SpyM3 0102 in M6 serotypes.
  • TABLE 29: Summary of FACS values for surface expression of SpyM30104 in M3 serotypes.
  • TABLE 30: Summary of FACS values for surface expression of SpyM30104 in an M12 serotype.
  • TABLE 31: Summary of FACS values for surface expression of SPs0106 in M3 serotypes.
  • TABLE 32: Summary of FACS values for surface expression of SPs0106 in an M12 serotype.
  • TABLE 33: Summary of FACS values for surface expression of 19224134 in an M12 serotype.
  • TABLE 34: Summary of FACS values for surface expression of 19224134 in M6 serotypes.
  • TABLE 35: Summary of FACS values for surface expression of 19224135 in an M12 serotype.
  • TABLE 36: Summary of FACS values for surface expression of 19224137 in an M12 serotype.
  • TABLE 37: Summary of FACS values for surface expression of 19224141 in an M12 serotype.
  • TABLE 38: S. pneumoniae strain 670 μl sequences.
  • TABLE 39: Pecent identity comparison of S. pneumoniae strains AI sequences.
  • TABLE 40: FACS analysis of L. lactis and GBS bacteria strains expressing GBS AI-1.
  • TABLE 41: Sequences of primers used to amplify AI locus.
  • TABLE 42: Conservation of amino acid sequences encoded by the S. pneumoniae AI locus.
  • TABLE 43: Protection of Mice Immunized with L. lactis expressing GBS AI-1.
  • TABLE 44: GAS AI-3 sequences from M49 isolate (591).
  • TABLE 45: Comparison of Sequences Between the Four GAS AIs.
  • TABLE 46: Antibody Responses against GBS 80 in Serum of Mice Immunized with L. lactis Expressing GBS AI-1
  • TABLE 47: Anti-GBS 80 IgA Antibodies Detected in Mouse Tissues Following Immunization with L. lactis Expressing GBS AI-1
  • TABLE 48: GBS 67 Protects Mice in an Immunization Assay
  • TABLE 49: Exposure Levels of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 on GBS Strains
  • TABLE 50: High Levels of Surface Protein Expression on GBS Serotypes
  • TABLE 51: Further Protection of Mice Immunized with L. lactis expressing GBS AI-1
  • DETAILED DESCRIPTION OF THE INVENTION
  • The practice of the present invention will employ, unless otherwise indicated, conventional methods of chemistry, biochemistry, molecular biology, immunology and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 19th Edition (1995); Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.); and Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell, eds., 1986, Blackwell Scientific Publications); Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Handbook of Surface and Colloidal Chemistry (Birdi, K. S. ed., CRC Press, 1997); Short Protocols in Molecular Biology, 4th ed. (Ausubel et al. eds., 1999, John Wiley & Sons); Molecular Biology Techniques: An Intensive Laboratory Course, (Ream et al., eds., 1998, Academic Press); PCR (Introduction to Biotechniques Series), 2nd ed. (Newton & Graham eds., 1997, Springer Verlag); Peters and Dalrymple, Fields Virology (2d ed), Fields et al. (eds.), B. N. Raven Press, New York, N.Y.
  • All publications, patents and patent applications cited herein, are hereby incorporated by reference in their entireties.
  • As used herein, an “Adhesin Island” or “AI” refers to a series of open reading frames within a bacterial genome, such as the genome for Group A or Group B Streptococcus or other gram positive bacteria, that encodes for a collection of surface proteins and sortases. An Adhesin Island may encode for amino acid sequences comprising at least one surface protein. The Adhesin Island may encode at least one surface protein. Alternatively, an Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, an Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more AI surface proteins may participate in the formation of a pilus structure on the surface of the gram positive bacteria.
  • Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction). The transcriptional regulator may regulate the expression of the AI operon.
  • GBS Adhesin Island 1
  • As discussed above, Applicants have identified a new adhesin island, “Adhesin Island 1”, “AI-1”, or “GBS AI-1”, within the genomes of several Group B Streptococcus serotypes and isolates. AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“AI-1 proteins”). Specifically, AI-1 includes open reading frames encoding for two or more (i.e., 2, 3, 4 or 5) of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648. One or more of the AI-1 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the AI-1 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • A schematic of AI-1 is presented in FIG. 1. AI-1 typically resides on an approximately 16.1 kb transposon-like element frequently inserted into the open reading frame for trmA. One or more of the AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) motif or other sortase substrate motif. The AI surface proteins of the invention may affect the ability of the GBS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The AI-1 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. AI-1 may encode for at least one surface protein. Alternatively, AI-1 may encode for at least two surface exposed proteins and at least one sortase. Preferably, AI-1 encodes for at least three surface exposed proteins and at least two sortases. The AI-1 protein preferably includes GBS 80 or a fragment thereof or a sequence having sequence identity thereto.
  • As used herein, an LPXTG motif represents an amino acid sequence comprising at least five amino acid residues. Preferably, the motif includes a leucine (L) in the first amino acid position, a proline (P) in the second amino acid position, a threonine (T) in the fourth amino acid position and a glycine (G) in the fifth amino acid position. The third position, represented by X, may be occupied by any amino acid residue. Preferably, the X is occupied by lysine (K), Glutamate (E), Asparagine (N), Glutamine (Q) or Alanine (A). Preferably, the X position is occupied by lysine (K). In some embodiments, one of the assigned LPXTG amino acid positions is replaced with another amino acid. Preferably, such replacements comprise conservative amino acid replacements, meaning that the replaced amino acid residue has similar physiological properties to the removed amino acid residue. Genetically encoded amino acids may be divided into four families based on physiological properties: (1) acidic (asparatate and glutamate), (2) basic (lysine, arginine, histitidine), (3) non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophane) and (4) uncharged polar (glycine, asparagines, glutamine, cysteine, serine, threonine, and tyrosine). Phenylalanine, tryptophan and tyrosine are sometimes classified jointly as aromatic amino acids. For example, it is reasonably predictable that an isolated replacement of a leucine with an isoleucine or valine, an asparate with a glutamate, a threonine with a serine, or a similar conservative replacement of an amino acid with a structurally related amino acid will not have a major effect on the biological activity.
  • The first amino acid position of the LPXTG motif may be replaced with another amino acid residue. Preferably, the first amino acid residue (leucine) is replaced with an alanine (A), valine (V), isoleucine (I), proline (P), phenylalanine (F), methionine (M), glutamic acid (E), glutamine (Q), or tryptophan (Y) residue. In one preferred embodiment, the first amino acid residue is replaced with an isoleucine (I).
  • The second amino acid residue of the LPXTG motif may be replaced with another amino acid residue. Preferably, the second amino acid residue praline (P) is replaced with a valine (V) residue.
  • The fourth amino acid residue of the LPXTG motif may be replaced with another amino acid residue. Preferably, the fourth amino acid residue (threonine) is replaced with a serine (S) or an alanine (A).
  • In general, an LPXTG motif may be represented by the amino acid sequence XXXXG, in which X at amino acid position 1 is an L, a V, an E, an I, an F, or a Q; X at amino acid position 2 is a P if X at amino acid position 1 is an L, an I, or an F; X at amino acid position 2 is a V if X at amino acid position 1 is a E or a Q; X at amino acid position 2 is a V or a P if X at amino acid position 1 is a V; X at amino acid position 3 is any amino acid residue; X at amino acid position 4 is a T if X at amino acid position 1 is a V, E, I, F, or Q; and X at amino acid position 4 is a T, S, or A if X at amino acid position 1 is an L.
  • Generally, the LPXTG motif of a GBS AI protein may be represented by the amino acid sequence XPXTG, in which X at amino acid position 1 is L, I, or F, and X at amino acid position 3 is any amino acid residue. Specific examples of LPXTG motifs in GBS AI proteins may include LPXTG (SEQ ID NO: 122) or IPXTG (SEQ ID NO: 133).
  • As discussed further below, the threonine in the fourth amino acid position of the LPXTG motif may be involved in the formation of a bond between the LPXTG containing protein and a cell wall precursor. Accordingly, in preferred LPXTG motifs, the threonine in the fourth amino acid position is not replaced with another amino acid or, if the threonine is replaced, the replacement amino acid is preferably a conservative amino acid replacement, such as serine.
  • Instead of an LPXTG motif, the AI surface proteins of the invention may contain alternative sortase substrate motifs such as NPQTN (SEQ ID NO: 142), NPKTN (SEQ ID NO: 168), NPQTG (SEQ ID NO: 169), NPKTG (SEQ ID NO: 170), XPXTGG (SEQ ID NO: 143), LPXTAX (SEQ ID NO: 144), or LAXTGX (SEQ ID NO: 145). (Similar conservative amino acid substitutions can also be made to these membrane motifs).
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • The AI surface proteins may be polymerized into pili by sortase-catalysed transpeptidation. (See FIG. 44.) Cleavage of AI surface proteins by sortase between the threonine and glycine residues of an LPXTG motif yields a thioester-linked acyl intermediate of sortase. Many AI surface proteins include a pilin motif amino acid sequence which interacts with the sortase and LPXTG amino acid sequence. The first lysine residue in a pilin motif can serve as an amino group acceptor of the cleaved LPXTG motif and thereby provide a covalent linkage between AI subunits to form pili. For example, the pilin motif can make a nucleophilic attack on the acyl enzyme providing a covalent linkage between AI subunits to form pili and regenerate the sortase enzyme. Examples of pilin motifs may include ((YPKN(X10)K; SEQ ID NO: 146), (YPKN(X9)K; SEQ ID NO: 147), (YPK(X7)K; SEQ ID NO: 148), (YPK(X11)K; SEQ ID NO: 149), or (PKN(X9)K; SEQ ID NO: 150)). Preferably, the AI surface proteins of the invention include a pilin motif amino acid sequence.
  • Typically, AI surface proteins of the invention will contain an N-terminal leader or secretion signal to facilitate translocation of the surface protein across the bacterial membrane.
  • Group B Streptococci are known to colonize the urinary tract, the lower gastrointestinal tract and the upper respiratory tract in humans. Electron micrograph images of GBS infection of a cervical epithelial cell line (ME180) are presented in FIG. 25. As shown in these images, the bacteria closely associate with tight junctions between the cells and appear to cross the monolayer by a paracellular route. Similar paracellular invasion of ME180 cells is also shown in the contrast images in FIG. 26. The AI surface proteins of the invention may effect the ability of the GBS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GBS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface.
  • Applicants have discovered that AI-1 surface protein GBS 104 can bind epithelial cells such as ME180 human cervical cells, A549 human lung cells and Caco2 human intestinal cells (See FIGS. 29 and 210). Further, deletion of the GBS 104 sequence in a GBS strain reduces the capacity of GBS to adhere to ME180 cervical epithelial cells. (See FIGS. 30 and 211). Deletion of GBS 104 also reduces the capacity of GBS to invade J774 macrophage-like cells. (See FIGS. 32 and 205). Deletion of GBS 104 also causes GBS to translocate through epithelial monolayers less efficiently. See FIG. 206. GBS 104 protein therefore appears to bind to ME180 epithelial cells and to have a role in adhesion to epithelial cells and macrophage cell lines.
  • Similar to the GBS bacteria that are deletion mutants for GBS 104, GBS 80 knockout mutant strains also partially lose the ability to translocate through an epithelial monolayer. See FIG. 207. Deletion of either GBS 80 or GBS 104 in COH1 cells diminishes adherence to HUVEC endothelial cells. See FIG. 208. Deletion of GBS 80 or GBS 104 in COH1 does not, however, affect growth of COH1 either with ME180 cells or in incubation medium (IM). See FIG. 209. Both GBS 80 and GBS 104, therefore, appear to be involved in translocation of GBS through epithelial cells.
  • GBS 80 does not appear to bind to epithelial cells. Incubation of epithelial cells in the presence of GBS 80 protein followed by FACS analysis using an anti-GBS 80 polyclonal antibody did not detect GBS 80 binding to the epithelial cells. See FIG. 202. Furthermore, deletion of GBS 80 protein does not affect the ability of GBS to adhere and invade ME180 cervical epithelial cells. See FIG. 203
  • Preferably, one or more of the surface proteins may bind to one or more extracellular matrix (ECM) binding proteins, such as fibrinogen, fibronectin, or collagen. As shown in FIGS. 5 and 204, and Example 1, GBS 80, one of the AI-1 surface proteins, can bind to the extracellular matrix binding proteins fibronectin and fibrinogen. While GBS 80 protein apparently does not bind to certain epithelial cells or affect the capacity of a GBS bacteria to adhere to or invade cervical epithelial cells (See FIGS. 27 and 28), removal of GBS 80 from a wild type strain decreases the ability of that strain to translocate through an epithelial cell layer (see FIG. 31).
  • GBS 80 may also be involved in formation of biofilms. COH1 bacteria overexpressing GBS 80 protein have an impaired ability to translocate through an epithelial monolayer. See FIG. 212. These COH1 bacteria overexpressing GBS 80 form microcolonies on epithelial cells. See FIGS. 213 and 214. These microcolonies may be the initiation of biofilm development.
  • AI Surface proteins may also demonstrate functional homology to previously identified adhesion proteins or extracellular matrix (ECM) binding proteins. For example, GBS 80, a surface protein in AI-1, exhibits some functional homology to FimA, a major fimbrial subunit of a Gram positive bacteria A. naeslundii. FimA is thought to be involved in binding salivary proteins and may be a component in a fimbrae on the surface of A. naeslundii. See Yeung et al. (1997) Infection & Immunity 65:2629-2639; Yeunge et al (1998) J. Bacteriol 66:1482-1491; Yeung et al. (1988) J. Bacteriol 170:3803-3809; and Li et al. (2001) Infection & Immunity 69:7224-7233.
  • A similar functional homology has also been identified between GBS 80 and proteins involved in pili formation in the Gram positive bacteria Corynebacterium diphtheriae (SpaA, SpaD, and SpaH). See, Ton-That et al. (2003) Molecular Microbiology 50(4):1429-1438 and Ton-That et al. (2004) Molecular Microbiology 53(1):251-261. The C. diphtheriae proteins all included a pilin motif of WxxxVxVYPK (SEQ ID NO: 151; where x indicates a varying amino acid residue). The lysine (K) residue is particularly conserved in the C. diphtheriae pilus proteins and is thought to be involved in sortase catalized oligomerization of the subunits involved in the C. diphtheriae pilus structure. (The C. diphtheriae pilin subunit SpaA is thought to occur by sortase-catalyzed amide bond cross-linking of adjacent pilin subunits. As the thioester-linked acyl intermediate of sortase requires nucleophilic attack for release, the conserved lysine within the SpaA pilin motif might function as an amino group acceptor of cleaved sorting signals, thereby providing for covalent linkages of the C. diphtheria pilin subunits. See FIG. 6(d) of Ton-That et al., Molecular Microbiology (2003) 50(4): 1429-1438.)
  • In addition, an “E box” comprising a conserved glutamic acid residue has also been identified in the C. diphtheria pilin associated proteins as important in C. diphtheria pilin assembly. The E box motif generally comprises YxLxETxAPxGY (SEQ ID NO: 152; where x indicates a varying amino acid residue). In particular, the conserved glutamic acid residue within the E box is thought necessary for C. diphtheria pilus formation.
  • Preferably, the AI-1 polypeptides of the immunogenic compositions comprise an E box motif. Some examples of E box motifs in the AI-1 polypeptides may include the amino acid sequences YxLxExxxxxGY (SEQ ID NO: 153), YxLxExxxPxGY (SEQ ID NO: 154), or YxLxETxAPxGY (SEQ ID NO: 152). Specifically, the E box motif of the polypeptides may comprise the amino acid sequences YKLKETKAPEGY (SEQ ID NO: 155), YVLKEIETQSGY (SEQ ID NO: 156), or YKLYEISSPDGY (SEQ ID NO: 157).
  • As discussed in more detail below, a pilin motif containing a conserved lysine residue and an E box motif containing a conserved glutamic acid residue have both been identified in GBS 80.
  • While previous publications have speculated that pilus-like structures might be formed on the surface of streptococci, (see, e.g., Ton-That et al., Molecular Microbiology (2003) 50(4): 1429-1438), these structures have not been previously visible in negative stain (non-specific) electron micrographs, throwing such speculations into doubt. For example, FIG. 34 presents electron micrographs of GBS serotype III, strain isolate COH1 with a plasmid insert to facilitate the overexpression of GBS 80. This EM photo was produced with a standard negative stain—no pilus structures are distinguishable. In addition, the use of such AI surface proteins in immunogenic compositions for the treatment or prevention of infection against a Gram positive bacteria has not been previously described.
  • Surprisingly, Applicants have now identified the presence of GBS 80 in surface exposed pilus formations visible in electron micrographs. These structures are only visible when the electron micrographs are specifically stained against an AI surface protein such as GBS 80. Examples of these electron micrographs are shown in FIGS. 11, 16 and 17, which reveal the presence of pilus structures in wild type COH1 Streptococcus agalactiae. Other examples of these electron micrographs are shown in FIG. 49, which reveals that GBS 80 is associated with pili in a wild type clinical isolate of S. agalactiae, JM9030013. (See FIG. 49.)
  • Applicants have also constructed mutant GBS strains containing a plasmid comprising the GBS 80 sequence resulting in the overexpression of GBS 80 within this mutant. The electron micrographs of FIGS. 13-15 are also stained against GBS 80 and reveal long, oligomeric structures containing GBS 80 which appear to cover portions of the surface of the bacteria and stretch far out into the supernatant.
  • In some instances, the formation of pili structures on GBS appears to be correlated to surface expression of GBS 80. FIG. 61 provides FAC analysis of GBS 80 surface levels on bacterial strains COH1 and JM9130013 using an anti-GBS 80 antisera. Immunogold electron microscopy of the COH1 and JM9130013 bacteria using anti-GBS 80 antisera demonstrates that JM9130013 bacteria, which have higher values for GBS 80 surface expression, also form longer pili structures.
  • The surface exposure of GBS 80 on GBS is generally not capsule-dependent. FIG. 62 provides FACS analysis of capsulated and uncapsulated GBS analyzed with anti-GBS 80 and anti-GBS 322 antibodies. Surface exposure of GBS 80, unlike GBS 322, is not capsule dependent.
  • An Adhesin Island surface protein, such as GBS 80 appears to be required for pili formation, as well as an Adhesin Island sortase. Pili are formed in Coh1 bacterial clones that overexpress GBS 80, but lack GBS 104, or one of the AI-1 sortases sag0647 or sag0648. However, pili are not formed in Coh1 bacterial clones that overexpress GBS 80 and lack both sag0647 and sag0648. Thus, for example, it appears that at least GBS 80 and a sortase, sag0647 or sag0648, may be necessary for pili formation. (See FIG. 48.) Overexpression of GBS 80 in GBS strain 515, which lacks an AI-1, also assembles GBS 80 into pili. GBS strain 515 contains an AI-2, and thus AI-2 sortases. The AI-2 sortases in GBS strain 515 apparently polymerize GBS 80 into pili. (See FIG. 42.) Overexpression of GBS 80 in GBS strain 515 cell knocked out for GBS 67 expression also apparently polymerizes GBS 80 into pili. (See FIG. 72.)
  • While GBS 80 appears to be required for GBS AI-1 pili formation, GBS 104 and sortase SAG0648 appears to be important for efficent AI-1 pili assembly. For example, high-molecular structures are not assembled in isogenic COH1 strains which lack expression of GBS 80 due to gene disruption and are less efficiently assembled in isogenic COH1 strains which lack the expression of GBS 104 (see FIG. 41). This GBS strain comprises high molecular weight pili structures composed of covalently linked GBS 80 and GBS 104 subunits. In addition, deleting SAG0648 in COH1 bacteria interferes with assembly of some of the high molecular weight pili structures. Thus, indicating that SAG0648 plays a role in assembly of these pilin species. (See FIG. 41).
  • EM photos confirm the involvement of AI surface protein GBS 104 within the hyperoligomeric structures of a GBS strain adapted for increased GBS 80 expression. (See FIGS. 34-41 and Example 6). In a wild type serotype VIII GBS strain, strain JM9030013, IEM identifies GBS 104 as forming clusters on the bacterial surface. (See FIG. 50.)
  • GBS 52 also appears to be a component of the GBS pili. Immunoblots using an anti-GBS 80 antisera on total cell extracts of Coh1 and a GBS 52 null mutant Coh1 reveal a shift in detected proteins in the Coh1 wild type strain relative to the GBS 52 null mutant Coh1 strain. The shifted proteins were also detected in the wild type Coh1 bacteria with an anti-GBS 52 antisera, indicating that the GBS 52 may be present in the pilus. (See FIG. 45.)
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as GBS 80. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include one or both of a pilin motif comprising a conserved lysine residue and an E box motif comprising a conserved glutamic acid residue.
  • More than one AI surface protein may be present in the oligomeric, pilus-like structures of the invention. For example, GBS 80 and GBS 104 may be incorporated into an oligomeric structure. Alternatively, GBS 80 and GBS 52 may be incorporated into an oligomeric structure, or GBS 80, GBS 104 and GBS 52 may be incorporated into an oligomeric structure.
  • In another embodiment, the invention includes compositions comprising two or more AI surface proteins. The composition may include surface proteins from the same adhesin island. For example, the composition may include two or more GBS AI-1 surface proteins, such as GBS 80, GBS 104 and GBS 52. The surface proteins may be isolated from Gram positve bacteria or they may be produced recombinantly.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a GBS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more GBS Adhesin Island 1 (“AI-1”) proteins and one or more GBS Adhesin Island 2 (“AI-2”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • The oligomeric, pilus-like structures of the invention may be combined with one or more additional GBS proteins. In one embodiment, the oligomeric, pilus-like structures comprise one or more AI surface proteins in combination with a second GBS protein. The second GBS protein may be a known GBS antigen, such as GBS 322 (commonly referred to as “sip”) or GBS 276. Nucleotide and amino acid sequences of GBS 322 sequenced from serotype V isolated strain 2603 V/R are set forth in WO 02/35771 as SEQ ID 8539 and SEQ ID 8540 and in the present specification as SEQ ID NOs: 38 and 39. A particularly preferred GBS 322 polypeptide lacks the N-terminal signal peptide, amino acid residues 1-24. An example of a preferred GBS 322 polypeptide is a 407 amino acid fragment and is shown in SEQ ID NO: 40. Examples of preferred GBS 322 polypeptides are further described in PCTUS04/______, attorney docket number PP20665.002 filed Sep. 15, 2004, hereby incorporated by reference, published as WO 2005/002619.
  • Additional GBS proteins which may be combined with the GBS AI surface proteins of the invention are also described in WO 2005/002619. These GBS proteins include GBS 91, GBS 184, GBS 305, GBS 330, GBS 338, GBS 361, GBS 404, GBS 690, and GBS 691.
  • Additional GBS proteins which may be combined with the GBS AI surface proteins of the invention are described in WO 02/34771.
  • GBS polysaccharides which may be combined with the GBS AI surface proteins of the invention are described in WO 2004/041157. For example, the GBS AI surface proteins of the invention may be combined with a GBS polysaccharides selected from the group consisting of serotype Ia, Ib, Ia/c, II, III, IV, V, VI, VII and VIII.
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an AI surface protein. The invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a GBS bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the GBS bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed AI protein. Preferably, the AI protein is in a hyperoligomeric form. Macromolecular structures associated with oligomeric pili are observed in the supernatant of cultured GBS strain Coh1. (See FIG. 46.) These pili are found in the supernatant at all growth phases of the cultured Coh1 bacteria. (See FIG. 47.)
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein. The invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a GBS bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the GBS bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed Adhesin Island protein. Preferably, the Adhesin Island protein is in a hyperoligomeric form.
  • The GBS bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • GBS bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the GBS bacteria with a plasmid encoding the AI protein. The plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein. Optionally, the sequence encoding the AI protein within the GBS bacterial genome may be deleted. Alternatively, or in addition, the promoter regulating the GBS Adhesin Island may be modified to increase expression.
  • GBS bacteria harbouring a GBS AI-1 may also be adapted to increase AI protein expression by altering the number adenosine nucleotides present at two sites in the intergenic region between AraC and GBS 80. See FIG. 197 A, which is a schematic showing the organization of GBS AI-1 and FIG. 197 B, which provides the sequence of the intergenic region between AraC and GBS 80 in the AI. The adenosine tracts which applicants have identified as influencing GBS 80 surface expression are at nucleotide positions 187 and 233 of the sequence shown in FIG. 197 B (SEQ ID NO: 273). Applicants determined the influence of these adenosine tracts on GBS 80 surface expression in strains of GBS bacteria harboring four adenosines at position 187 and six adenosines at position 233, five adenosines at position 187 and six adenosines position 233, and five adenosines at position 187 and seven adenosines at position 233. FACS analysis of these strains using anti GBS 80 antiserum determined that an intergenic region with five adenosines at position 187 and six adenosines at position 233 had higher expression levels of GBS 80 on their surface than other stains. See FIG. 197 C for results obtained from the FACS analysis. Therefore, manipulating the number of adenosines present at positions 187 and 233 of the AraC and GBS 80 intergenic region may further be used to adapt GBS to increase AI protein expression.
  • The invention further includes GBS bacteria which have been adapted to produce increased levels of AI surface protein. In particular, the invention includes GBS bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein, such as GBS 80. In one embodiment, the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • The invention further includes GBS bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface. The GBS bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide. Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria. Increased expression of a leader peptidase in GBS may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation. The GBS bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • Alternatively, the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes” Infection and Immunity (2004) 72(6):3444-3450). As used herein, non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis. Preferably, the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid. The non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface. Alternatively, the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria. For example, the AI surface protein may be isolated from cell extracts or culture supernatants. Alternatively, the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • The non-pathogenic Gram positive bacteria may be used to express any of the Gram positive bacterial Adhesin Island proteins described herein, including proteins from a GBS Adhesin Island, a GAS Adhesin Island, or a S pneumo Adhesin Island. The non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein. Preferably, the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase. The AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with a pathogenic Gram positive bacteria, such as GBS, GAS or Streptococcus pneumoniae. The non-pathogenic Gram positive bacteria may express the Gram positive bacterial Adheshin Island proteins in oligomeric forms that further comprise adhesin island proteins encoded within the genome of the non-pathogenic Gram positive bacteria.
  • Applicants modified L. lactis to demonstrate that it can express GBS AI polypeptides. L. lactis was transformed with a construct encoding GBS 80 under its own promoter and terminator sequences. The transformed L. lactis appeared to express GBS 80 as shown by Western blot analysis using anti-GBS 80 antiserum. See lanes 6 and 7 of the Western Blots provided in FIGS. 133A and 133B (133A and 133B are two different exposures of the same Western blot). See also Example 13.
  • Applicants also transformed L. lactis with a construct encoding GBS AI-1 polypeptides GBS 80, GBS 52, SAG0647, SAG0648, and GBS 104 under the GBS 80 promoter and terminator sequences. These L. lactis expressed high molecular weight structures that were immunoreactive with anti-GBS 80 in immunoblots. See FIG. 134, lane 2, which shows detection of a GBS 80 monomer and higher molecular weight polymers in total transformed L. lactis extracts. Thus, it appeared that L. lactis is capable of expressing GBS 80 in oligomeric form. The high molecular weight polymers were not only detected in L. lactis extracts, but also in the culture supernatants. See FIG. 135 at lane 4. See also Example 14. Thus, the GBS AI polypeptides in oligomeric form can be isolated and purified from either L. lactis cell extracts or culture supernatants. These oligomeric forms can, for instance, be isolated from cell extracts or culture supernatants by release by sonication. See FIGS. 136A and B. See also FIG. 171, which shows purification of GBS pili from whole extracts of L. lactis expressing the GBS AI-1 following sonication and gel filtration on a Sephacryl HR 400 column.
  • Furthermore, the L. lactis transformed with the construct encoding GBS AI-1 polypeptides GBS 80, GBS 52, SAG0647, SAG0648, and GBS 104 under the GBS 80 promoter and terminator sequences expressed the GBS AI-1 polypeptides on its surface. FACS analysis of these transformed L. lactis detected cell surface expression of both GBS 80 and GBS 104. The surface expression levels of GBS 80 and GBS 104 on the transformed L. lactis were similar to the surface expression levels of GBS 80 and GBS 104 on GBS strains COH1 and JM9130013, which naturally express GBS AI-1.
  • See FIG. 169 for FACS analysis data for L. lactis transformed with GBS AI-1 and wildtype JM9130013 bacteria using anti-GBS 80 and GBS 104 antisera. Table 40 provides the results of FACS analysis of transformed L. lactis, COH1, and JM9130013 bacteria using anti-GBS 80 and anti-GBS 104 antisera. The numbers provided represent the mean fluorescence value difference calculated for immune versus pre-immune sera obtained for each bacterial strain.
    TABLE 40
    FACS analysis of L. lactis and GBS bacteria strains expressing GBS AI-1
    Anti-GBS 80 Anti-GBS 104
    antiserum antiserum
    GBS AI-1 transformed 298 251
    L. lactis
    GBS COH1
    305 305
    GBS JM9130013 461 355

    Immunogold-electronmicroscopy performed with anti-GBS 80 primary antibodies detected the presence of pilus structures on the surface of the L. lactis bacteria expressing GBS AI-1, confirming the results of the FACS analysis. See FIG. 168 B and C. Interestingly, this expression of GBS pili on the surface of the L. lactis induced L. lactis aggregation. See FIG. 170. Thus, GBS AI polypeptides may also be isolated and purified from the surface of L. lactis. The ability of L. lactis to express GBS AI polypeptides on its surface also demonstrates that it may be useful as a host to deliver GBS AI antigens.
  • In fact, immunization of mice with L. lactis transformed with GBS AI-1 was protective in a subsequent challenge with GBS. Female mice were immunized with L. lactis transformed with GBS AI-1. The immunized female mice were bred and their pups were challenged with a dose of GBS sufficient to kill 90% of non-immunized pups. Detailed protocols for intranasal and subcutaneous immunization of mice with transformed L. lactis can be found in Examples 18 and 19, respectively. Table 43 provides data showing that immunization of the female mice with L. lactis expressing GBS AI-1(LL-AI 1) greatly increased survival rate of challenged pups relative to both a negative PBS control (PBS) and a negative L. lactis control (LL 10 E9, which is wild type L. lactis not transformed to express GBS AI-1).
    TABLE 43
    Protection of Mice Immunized with L. lactis expressing GBS AI-1
    Immunization Survival
    Route Antigen Alive/Treated Survival % % Range p value
    Intraperitoneum Recombinant GBS 80 16/18 89 80-100 <0.001
    Subcutaneous LL-AI 1 10 E9 40/49 82 70-90  <0.001
    LL-AI 1 10 E10 50/60 83 60-100 <0.001
    PBS  4/30 13 0-30
    LL 10 E9  3/57 5 0-20
    Intranasal LL-AI 1 10 E9 22/60 37  0-100 0.02
    LL-AI 1 10 E10 31/49 63 30-90  <0.001
    LL 10 E9  2/27 7 0-20
  • Table 51 provides further evidence that immunization of mice with L. lactis transformed with GBS AI-1 is protective against GBS.
    TABLE 51
    Further Protection of Mice Immunized with L. lactis expressing GBS AI-1
    Immunization Alive/ Survival %
    Antigen route Treated (Pval < 0.0000001)
    Recombinant GBS 80 IP 48/50 92
    Recombinant GBS 80 SC 21/30 70
    L. lactis + AI1 106 cfu SC  6/66 9
    L. lactis + AI1 107 cfu SC 47/70 73
    L. lactis + AI1 108 cfu SC 116/153 76
    L. lactis + AI1 109 cfu SC  98/118 83
    L. lactis + AI1 1010 cfu SC 107/129 83
    L. lactis 1010 cfu SC  4/83 5
    PBS SC  6/110 5
    L. lactis + AI1 1010 cfu IN 51/97 52
    L. lactis 1011 cfu IN  1/40 7
    PBS IN  0/37 0
  • Protection of immunized mice with L. lactis expressing the GBS AI-1 is at least partly due to a newly raised antibody response. Table 46 provides anti-GBS 80 antibody titers detected in serum of the mice immunized with L. lactis expressing the GBS AI-1 as described above. Mice immunized with L. lactis expressing the GBS AI-1 have anti-GBS 80 antibody titres, which are not observed in mice immunized with L. lactis not transformed to express the GBS AI-1. Further, as expected from the survival data, mice subcutaneously immunized with L. lactis transformed to express the GBS AI-1 have significantly higher serum anti-GBS 80 antibody titers than mice intranasally immunized with L. lactis transformed to express the GBS AI-1.
    TABLE 46
    Antibody Responses against GBS 80 in Serum of Mice
    Immunized with L. lactis Expressing GBS AI-1
    Ab Titre Obtained Following
    Subcutaneous Intranasal Intraperitoneal
    Antigen Immunization Immunization Immunization
    LL
    10 E9 0 0
    LL-AI 1 10 E9 14000 50
    LL-AI 1 10 E10 25000 406
    Recombinant GBS 80 120000
  • Anti-GBS 80 antibodies of the IgA isotype were specifically detected in various body fluids of the mice subcutaneously or intranasally immunized with L. lactis expressing the GBS AI-1.
    TABLE 47
    Anti-GBS 80 IgA Antibodies Detected in Mouse Tissues Following
    Immunization with L. lactis Expressing GBS AI-1
    Anti-GBS 80 IgA
    Antibodies Detected in
    Antigen Immunization route Serum Vaginal Wash Nasal Wash
    LL
    10 E9 0 0 0
    LL-AI 1 Subcutaneous 0 25 20
    LL-AI 1 Intranasal 140 0 150
    GBS 80 Intraperitoneal 60 0
  • Furthermore, opsonophagocytosis assays also demonstrated that at least some of the antiserum produced against the L. lactis expressing GBS AI 1 is opsonic for GBS. See FIG. 161.
  • To obtain protection of against GBS across a greater number of strains and serotypes, it is possible to transform L. lactis with a recombinant GBS AI encoding both GBS AI-1 and AI-2, i.e., a hybrid GBS AI. By way of example, a hybrid GBS AI may be a GBS AI-1 with a replacement of the GBS 104 gene with a GBS 67 gene. A schematic of such a hybrid GBS AI is depicted in FIG. 231 A. A hybrid GBS AI may alternatively be a GBS AI-1 with a replacement of the GBS 52 gene with a GBS 59 gene. See the schematic at FIG. 231 B. Alternatively, a hybrid GBS AI may be a GBS AI-1 with a substitution of a GBS 59 polypeptide for the GBS 52 gene and a substitution of the GBS 104 gene for genes encoding GBS 59 and the two GBS AI-2 sortases. Another example of a hybrid GBS AI is a GBS AI-1 with the substitution of a GBS 59 gene for the GBS 52 gene and a GBS 67 for the GBS 104 gene. See the schematic at FIG. 232. A further example of a hybrid GBS AI is a GBS AI-1 having a GBS 59 gene and genes encoding the GBS AI-2 sortases in place of the GBS 52 gene. Yet another example of a hybrid GBS AI is a GBS AI-1 with a substitution of either GBS 52 or GBS 104 with a fusion protein comprising GBS 322 and one of GBS 59, GBS 67, or GBS 150. Some of these hybrid GBS AIs may be prepared as briefly outlined in FIG. 234 A-F.
  • Applicants have prepared a hybrid GBS AI having a GBS AI-1 sequence with a substitution of a GBS 67 coding sequence for the GBS 104 gene as depicted in FIG. 231 A. Transformation of L. lactis with the hybrid GBS AI-1 resulted in L. lactis expression of high molecular weight polymers containing the GBS 80 and GBS 67 proteins. See FIG. 233 A, which provides Western blot analysis of L. lactis transformed with the hybrid GBS AI depicted in FIG. 231 A. When L. lactis transformed with the hybrid GBS AI were probed with antibodies to GBS 80 or GBS 67, high molecular weight structures were detected. See lanes labelled LL+a) in both the α-80 and α-67 immunoblots. The GBS 80 and GBS 67 proteins were confirmed to be present on the surface of L. lactis by FACS analysis. See FIG. 233 B, which shows a shift in fluorescence when GBS 80 and GBS 67 antibodies are used to detect GBS 80 and GBS 67 surface expression. The same shifts in fluorescence were not observed in L. lactis control cells, cells not transformed with the hybrid GBS AI.
  • Alternatively, the oligomeric, pilus-like structures may be produced recombinantly. If produced in a recombinant host cell system, the AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention. Such AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • AI Sortases of the invention will typically have a signal peptide sequence within the first 70 amino acid residues. They may also include a transmembrane sequence within 50 amino acid residues of the C terminus. The sortases may also include at least one basic amino acid residue within the last 8 amino acids. Preferably, the sortases have one or more active site residues, such as a catalytic cysteine and histidine.
  • As shown in FIG. 1, AI-1 includes the surface exposed proteins of GBS 80, GBS 52 and GBS 104 and the sortases SAG0647 and SAG0648. AI-1 typically appears as an insertion into the 3′ end of the trmA gene.
  • In addition to the open reading frames encoding the AI-1 proteins, AI-1 may also include a divergently transcribed transcriptional regulator such as araC (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction). It is believed that araC may regulate the expression of the AI operon. (See Korbel et al., Nature Biotechnology (2004) 22(7): 911-917 for a discussion of divergently transcribed regulators in E. coli).
  • AI-1 may also include a sequence encoding a rho independent transcriptional terminator (see hairpin structure in FIG. 1). The presence of this structure within the adhesin island is thought to interrupt transcription after the GBS 80 open reading frame, leading to increased expression of this surface protein.
  • A schematic identifying AI-1 within several GBS serotypes is depicted in FIG. 2. AI-1 sequences were identified in GBS serotype V, strain isolate 2603; GBS serotype III, strain isolate NEM316; GBS serotype II, strain isolate 18RS21; GBS serotype V, strain isolate CJB111; GBS serotype III, strain isolate COH1 and GBS serotype 1a, strain isolate A909. (Percentages shown are amino acid identity to the 2603 sequence). (An AI-1 was not identified in GBS serotype 1b, strain isolate H36B or GBS serotype 1a, strain isolate 515).
  • An alignment of AI-1 polynucleotide sequences from serotype V, strain isolates 2603 and CJB111; serotype II, strain isolate 18RS21; serotype III, strain isolates COH1 and NEM316; and serotype 1a, strain isolate A909 is presented in FIG. 18. An alignment of amino acid sequences of AI-1 surface protein GBS 80 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate A909; serotype III, strain isolates COH1 and NEM316 is presented in FIG. 22. An alignment of amino acid sequences of AI-1 surface protein GBS 104 from serotype V, strain isolates 2603 and CJB111; serotype III, strain isolates COH1 and NEM316; and serotype II, strain isolate 18RS21 is presented in FIG. 23. Preferred AI-1 polynucleotide and amino acid sequences are conserved among two or more GBS serotypes or strain isolates.
  • As shown in this figure, the full length of surface protein GBS 80 is particularly conserved among GBS serotypes V (strain isolates 2603 and CJBIII), III (strain isolates NEM316 and COH1), and Ia (strain isolate A909). The GBS 80 surface protein is missing or fragmented in serotypes II (strain isolate 18RS21), Ib (strain isolate H36B) and Ia (strain isolate 515).
  • Polynucleotide and amino acid sequences for AraC are set forth in FIG. 30.
  • GBS Adhesin Island 2
  • A second adhesin island, “Adhesin Island 2” or “AI-2” or GBS AI-2” has also been identified in numerous GBS serotypes. A schematic depicting the correlation between AI-1 and AI-2 within the GBS serotype V, strain isolate 2603 is shown in FIG. 3. (Homology percentages in FIG. 3 represent amino acid identity of the AI-2 proteins to the AI-1 proteins). Alignments of AI-2 polynucleotide sequences are presented in FIGS. 20 and 21 (FIG. 20 includes sequences from serotype V, strain isolate 2603 and serotype III, strain isolate NEM316. FIG. 21 includes sequences from serotype III, strain isolate COH1 and serotype Ia, strain isolate A909). An alignment of amino acid sequences of AI-2 surface protein GBS 067 from serotype V, strain isolates 2603 and CJB111; serotype 1a, strain isolate 515; serotype II, strain isolate 18RS21; serotype Ib, strain isolate H36B; and serotype III, strain isolate NEM316 is presented in FIG. 24. Preferred AI-2 polynucleotide and amino acid sequences are conserved among two or more GBS serotypes or strain isolates.
  • AI-2 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, AI-2 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5 or more) of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525. In one embodiment, AI-2 includes open reading frames encoding for two or more of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406. Alternatively, AI-2 may include open reading frames encoding for two or more of 01520, 01521, 01522, 01523, 01523, 01524 and 01525.
  • One or more of the surface proteins typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. The GBS AI-2 sortase proteins are thought to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GBS AI-2 may encode for at least one surface protein. Alternatively, AI-2 may encode for at least two surface proteins and at least one sortase. Preferably, GBS AI-2 encodes for at least three surface proteins and at least two sortases. One or more of the AI-2 surface proteins may include an LPXTG or other sortase substrate motif.
  • One or more of the surface proteins may also typically include pilin motif. The pilin motif may be involved in pili formation. Cleavage of AI surface proteins by sortase between the threonine and glycine residue of an LPXTG motif yields a thioester-linked acyl intermediate of sortase. The first lysine residue in a pilin motif can serve as an amino group acceptor of the cleaved LPXTG motif and thereby provide a covalent linkage between AI subunits to form pili. For example, the pilin motif can make a nucleophilic attack on the acyl enzyme providing a covalent linkage between AI subunits to form pili and regenerate the sortase enzyme. Some examples of pilin motifs that may be present in the GBS AI-2 proteins include ((YPKN(X8)K; SEQ ID NO: 158), (PK(X8)K; SEQ ID NO: 159), (YPK(X9)K; SEQ ID NO: 160), (PKN(X8)K; SEQ ID NO: 161), or (PK(X10)K; SEQ ID NO: 162)).
  • One or more of the surface protein may also include an E box motif. The E box motif contains a conserved glutamic acid residue that is believed to be necessary for pilus formation. Some examples of E box motifs may include the amino acid sequences YxLxETxAPxG (SEQ ID NO: 163), YxxxExxAxxGY (SEQ ID NO: 164), YxLxExxxPxDY (SEQ ID NO: 165), or YxLxETxAPxGY (SEQ ID NO: 152).
  • As shown in FIG. 3, GBS AI-2 may include the surface exposed proteins of GBS 67, GBS 59 and GBS 150 and the sortases of SAG1406 and SAG1405. Alternatively, GBS AI-2 may include the proteins 01521, 01524 and 01525 and sortases 01520 and 01522. GBS 067 and 01524 are preferred AI-2 surface proteins.
  • AI-2 may also include a divergently transcribed transcriptional regulator such as a RofA like protein (for example rogB). As in AI-1, rogB is thought to regulate the expression of the AI-2 operon.
  • A schematic depiction of AI-2 within several GBS serotypes is depicted in FIG. 4. (Percentages shown are amino acid identity to the 2603 sequence). While the AI-2 surface proteins GBS 59 and GBS 67 are more variable across GBS serotypes than the corresponding AI-1 surface proteins, AI-2 surface protein GBS 67 appears to be conserved in GBS serotypes where the AI-1 surface proteins are disrupted or missing.
  • For example, as discussed above and in FIG. 2, the AI-1 GBS 80 surface protein is fragmented in GBS serotype II, strain isolate 18RS21. Within AI-2 for this same sequence, as shown in FIG. 4, the GBS 67 surface protein has 99% amino acid sequence homology with the corresponding sequence in strain isolate 2603. Similarly, the AI-1 GBS 80 surface protein appears to be missing in GBS serotype Ib, strain isolate H36B and GBS serotype Ia, strain isolate 515. Within AI-2 for these sequences, however, the GBS 67 surface protein has 97-99% amino acid sequence homology with the corresponding sequence in strain isolate 2603. GBS 67 appears to have two allelic variants, which can be divided according to percent homology with strains 2603 and H36B. See FIGS. 237-239.
  • Unlike for GBS 67, amino acid sequence identity of GBS 59 is variable across different GBS strains. As shown in FIGS. 63 and 224, GBS 59 of GBS strain isolate 2603 shares 100% amino acid residue homology with GBS strain 18RS21, 62% amino acid sequence homology with GBS strain H36B, 48% amino acid residue homology with GBS strain 515 and GBS strain CJB111, and 47% amino acid residue homology with GBS strain NEM316. The amino acid sequence homologies of the different GBS strains suggest that there are two isoforms of GBS 59. The first isoform appears to include the GBS 59 protein of GBS strains CJB111, NEM316, and 515. The second isoform appears to include the GBS 59 protein of GBS strains 18RS21, 2603, and H36B. (See FIGS. 63 and 224.) As expected from the variability in GBS 59 isoforms, antibodies specific for the first GBS 59 isoform detect the first but not the second GBS 59 isoform and antibodies specific for the second GBS 59 isoform detect the second but not the first GBS 59 isoform. See FIG. 226A, which shows FACS analysis of 28 GBS strains having a GBS 59 gene detected using PCR for GBS 59 surface expression. For each of the 28 GBS strains, FACS analysis was performed using either an antibody for GBS 59 isoform 1 (α-Cjb111) or GBS 59 isoform 2 (α-2603). Only one of the two antibodies detected GBS 59 surface expression on each GBS strain. As a negative control, GBS strains in which a GBS 59 gene was not detectable by PCR did not have significant GBS 59 surface expression levels. FIG. 226B.
  • Also, GBS 59 is opsonic only against GBS strains expressing a homologous GBS 59 protein. See FIG. 225.
  • In one embodiment, the immunogenic composition of the invention comprises a first and a second isoform of the GBS 59 protein to provide protection across a wide range of GBS serotypes that express polypeptides from a GBS AI-2. The first isoform may be the GBS 59 protein of GBS strain CJB111, NEM316, or 515. The second isoform may be the GBS 59 protein of GBS strain 18RS21, 2603, or H36B.
  • The gene encoding GBS 59 has been identified in a high number of GBS isolates; the GBS 59 gene was detected in 31 of 40 GBS isolates tested (77.5%). The GBS 59 protein also appears to be present as part of a pilus in whole extracts derived from GBS strains. FIG. 64 shows detection of high molecular weight GBS 59 polymers in whole extracts of GBS strains CJB111, 7357B, COH31, D1363C, 5408, 1999, 5364, 5518, and 515 using antiserum raised against GBS 59 of GBS strain CJB111. FIG. 65 also shows detection of these high molecular weight GBS 59 polymers in whole extracts of GBS strains D136C, 515, and CJB111 with anti-GBS 59 antiserum. (See also FIG. 220 A for detection of GBS 59 high molecular weight polymers in strain 515.) FIG. 65 confirms the presence of different isoforms of GBS 59. Antisera raised against two different GBS 59 isoforms results in different patterns of immunoreactivity depending on the GBS strain origin of the whole extract. FIG. 65 further shows detection of GBS 59 monomers in purified GBS 59 preparations.
  • GBS 59 is also highly expressed on the surface of GBS strains. GBS 59 was detected on the surface of GBS strains CJB111, DK1, DK8, Davis, 515, 2986, 5551, 1169, and 7357B by FACS analysis using mouse antiserum raised against GBS 59 of GBS CJB111. FACS analysis did not detect surface expression of GBS 59 in GBS strains SMU071, JM9130013, and COH1, which do not contain a GBS 59 gene. (See FIG. 66.) Further confirmation that GBS 59 is expressed on the surface of GBS is detection of GBS 59 by immuno-electron microscopy on the surface of GBS strain 515 bacteria. See FIG. 215.
  • GBS 67 and GBS 150 also appear to be included in high molecular weight structures, or pili. FIG. 69 shows that anti-GBS 67 and anti-GBS 150 immunoreact with high molecular weight structures in whole GBS strain 515 extracts. (See also FIG. 220 B and C.) It is also notable in FIG. 69 that the anti-GBS 59 antisera, raised in a mouse following immunization with GBS 59 of GBS strain 2603, does not cross-hybridize with GBS 59 in GBS strain 515. GBS 59 of GBS stain 515 is of a different isotype than GBS 59 of GBS stain 2603. See FIG. 63, which illustrates that the homology of these two GBS 59 polypeptides is 48%, and FIG. 65, which confirms that GBS 59 antisera raised against GBS strain 2603 does not cross-hybridize with GBS 59 of GBS strain 515.
  • Formation of pili containing GBS 150 does not appear to require GBS 67 expression. FIG. 70 provides Western blots showing that higher molecular weight structures in GBS strain 515 total extracts immunoreact with anti-GBS 67 and anti-GBS 150 antiserum. In a GBS strain 515 lacking GBS 67 expression, anti-GBS 67 antiserum no longer immunoreacts with polypeptides in total extracts, while anti-GBS 150 antiserum is still able to cross-hybridze with high molecular weight structures.
  • Likewise, formation of pili containing GBS 59 does not appear to require GBS 67 expression. As expected, FACS detects GBS 67 cell surface expression on wildtype GBS strain 515, but not GBS strain 515 cells knocked out for GBS 67. FACS analysis using anti-GBS 59 antisera, however, detects GBS 59 expression on both the wildtype GBS strain 515 cells and the GBS strain 515 cells knocked out for GBS 67. Thus, GBS 59 cell surface expression is detected on GBS stain 515 cells regardless of GBS 67 expression.
  • GBS 67, while present in pili, appears to be localized around the surface of GBS strain 515 cells. See the immuno-electron micrographs presented in FIG. 216. GBS 67 binds to fibronectin. See FIG. 217.
  • Formation of pili encoded by GBS AI-2 does require expression of GBS 59. Deletion of GBS 59 from strain 515 bacteria eliminates detection of high molecular weight structures by antibodies that bind to GBS 59 (FIG. 221 A, lane 3), GBS 67 (FIG. 221 B, lane 3), and GBS 150 (FIG. 221 C, lane 3). By contrast, Western blot analysis of 515 bacteria with a deletion of the GBS 67 gene detects high molecular weight structures using GBS 59 (FIG. 221 A, lane 2) and GBS 150 (FIG. 221 C, lane 2) antisera. Similarly, Western blot analysis of 515 bacteria with a deletion of the GBS 150 gene detects high molecular weight structures using GBS 59 (FIG. 221 A, lane 4) and GBS 67 (FIG. 221 B, lane 4). See also FIG. 223, which provides Western blots of each of the 515 strains interrogated with antibodies for GBS 59, GBS 67, and GBS 150. FACS analysis of strain 515 bacteria deleted for either GBS 59 or GBS 67 confirms these results. See FIG. 222, which shows that only deletion of GBS 59 abolishes surface expression of both GBS 59 and GBS 67.
  • Formation of pili encoded by GBS AI-2 also requires expression of both GBS adhesin island-2 encoded sortases. See FIG. 218, which provides Western blot analysis of strain 515 bacteria lacking Srt1, Srt2, or both Srt1 and Srt2. Only deletion of both Srt1 and Srt2 abolishes pilus assembly as detected by antibodies that cross-hybridize with each of GBS 59, GBS 67 and GBS 150. The results of the Western blot analysis were verified by FACS, which provided similar results. See FIG. 219.
  • As shown in FIG. 4, two of the GBS strain isolates (COH1 and A909) do not appear to contain homologues to the surface proteins GBS 59 and GBS 67. For these two strains, the percentages shown in FIG. 4 are amino acid identity to the COH1 protein). Notwithstanding the difference in the surface protein lengths for these two strains, AI-2 within these sequences still contains two sortase proteins and three LPXTG containing surface proteins, as well as a signal peptidase sequence leading into the first surface protein. One of the surface proteins in this variant of AI-2, spb1, has previously been identified as a potential adhesion protein. (See Adderson et al., Infection and Immunity (2003) 71(12):6857-6863). Alternatively, because of the lack of GBS 59 and GBS 67 sequences, this variant of AI-2 may be a third type of AI (Adhesin Island-3, AI-3, or GBS AI-3).
  • More than one AI surface protein may be present in the oligomeric, pilus-like structures of the invention. For example, GBS 59 and GBS 67 may be incorporated into an oligomeric structure. Alternatively, GBS 59 and GBS 150 may be incorporated into an oligomeric structure, or GBS 59, GBS 150 and GBS 67 may be incorporated into an oligomeric structure.
  • In another embodiment, the invention includes compositions comprising two or more AI surface proteins. The composition may include surface proteins from the same adhesin island. For example, the composition may include two or more GBS AI-2 surface proteins, such as GBS 59, GBS 67 and GBS 150. The surface proteins may be isolated from Gram positve bacteria or they may be produced recombinantly.
  • GAS Adhesin Islands
  • As discussed above, Applicants have identified at least four different GAS Adhesin Islands. These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fascilitis. In addition, post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases. The first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused. In the second stage of infection, the bacteria secretes a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection. The final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart. A general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • In order to prevent the pathogenic effects associated with the later stages of GAS infection, an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above. The M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation. The carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci. The amino terminus, which extend through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen. Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens. Antisera to define T types is commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • The gene coding for one form of T-antigen, T-type 6, from an M6 strain of GAS (D741) has been cloned and characterized and maps to an approximately 11 kb highly variable pathogenicity island. Schneewind et al., J. Bacteriol. (1990) 172(6):3310-3317. This island is known as the Fibronectin-binding, Collagen-binding T-antigen (FCT) region because it contains, in addition to the T6 coding gene (tee6), members of a family of genes coding for Extra Cellular Matrix (ECM) binding proteins. Bessen et al., Infection & Immunity (2002) 70(3):1159-1167. Several of the protein products of this gene family have been shown to directly bind either fibronectin and/or collagen. See Hanski et al., Infection & Immunity (1992) 60(12):5119-5125; Talay et al., Infection & Immunity (1992(60(9):3837-3844; Jaffe et al. (1996) 21(2):373-384; Rocha et al., Adv Exp Med Biol. (1997) 418:737-739; Kreikemeyer et al., J Biol Chem (2004) 279(16):15850-15859; Podbielski et al., Mol. Microbiol. (1999) 31(4):1051-64; and Kreikemeyer et al., Int. J. Med Microbiol (2004) 294(2-3):177-88. In some cases direct evidence for a role of these proteins in adhesion and invasion has been obtained.
  • Applicants raised antiserum against a recombinant product of the tee6 gene and used it to explore the expression of T6 in M6 strain 2724. In immunoblot of mutanolysin extracts of this strain, the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used.
  • This pattern of high molecular weight products is similar to that observed in immunoblots of the protein components of the pili identified in Streptococcus agalactiae (described above) and previously in Corynebacterium diphtheriae. Electron microscropy of strain M6 2724 with antisera specific for the product of tee6 revealed abundant surface staining and long pilus like structures extending up to 700 nanometers from the bacterial surface, revealing that the T6 protein, one of the antigens recognized in the original Lancefiled serotyping system, is located within a GAS Adhesin Island (GAS AI-1) and forms long covalently linked pilus structures.
  • Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms. Immunological recognition of these proteins may allow the host immune response to slow or prevent the bacteria's transition into the more pathogenic later stages of infection.
  • In addition, Applicants have discovered that the GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix). Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm. Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment (i.e., before complete biofilm formation) is preferable.
  • The invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes. The immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form. The invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • While there is surprising variability in the number and sequence of the GAS AI components across isolates, GAS AI sequences may be generally characterized as Type 1, Type 2, Type 3, and Type 4, depending on the number and type of sortase sequence within the island and the percentage identity of other proteins within the island. Schematics of the GAS adhesin islands are set forth in FIG. 51A and FIG. 162. In all strains identified so far, the adhesin island region is flanked by highly conserved open reading frames M1 123 and M1 136. Between three and five genes in each GAS adhesin island code for ECM binding adhesin proteins containing LPXTG motifs.
  • GAS Adhesin Island 1
  • As discussed above, Applicants have identified adhesin islands, “GAS Adhesin Island 1” or “GAS AI-1”, within the genome Group A Streptococcus serotypes and isolates. GAS AI-1 comprises a series of approximately five open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-1 proteins”). GAS AI-1 preferably comprises surface proteins, a srtB sortase, and a rofA divergently transcribed transcriptional regulator. GAS AI-1 surface proteins may include a fibronectin binding protein, a collagen adhesion protein and a fimbrial structural subunit. Preferably, each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine). Specifically, GAS AI-1 includes open reading frames encoding for two or more (i.e., 2, 3, 4 or 5) of M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161.
  • Applicants have also identified open reading frames encoding fimbrial structural subunits in other GAS bacteria harbouring an AI-1. These open reading frames encode fimbrial structural subunits CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial. A GAS AI-1 may comprise a polynucleotide encoding any one of CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • As discussed above, the hyper-oligomeric pilus structure of GAS AI-1 appears to be responsible for the T-antigen type 6 classification, and GAS AI-1 corresponds to the FCT region previously identified for tee6. As in GAS AI-1, the tee6 FCT region includes open reading frames encoding for a collagen adhesion protein (cpa, capsular polysaccharide adhesion) and a fibronectin binding protein (prtF1). Immunoblots of tee6, a GAS AI-1 fimbrial structural subunit corresponding to M6_Spy160, reveal high molecular weight structures indicative of the hyper-oligomeric pilus structures. Immunoblots with antiserum specific for Cpa also recognize a high molecular weight ladder structure, indicating Cpa involvement in the GAS AI-1 pilus structure or formation. In EM photos of GAS bacteria, Cpa antiserum reveals abundant staining on the surface of the bacteria and occasional gold particles extended from the surface of the bacteria. In contrast, immunoblots with antiserum specific for PrtF1 recognize only a single molecular species with electrophoretic mobility corresponding to its predicted molecular mass, indicating that PrtF1 may not be associated with the oligomeric pilus structure. A preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which may be formulated or purified in an oligomeric (pilis) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Another preferred immunogenic composition of the invention comprises a GAS AI-1 surface protein which has been isolated in an oligomeric (pilis) form. The oligomer or hyperoligomeric pilus structures comprising the GAS AI-1 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-1 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-1 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the GAS AI-1 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The LPXTG sortase substrate motif of a GAS AI surface protein may be generally represented by the formula XXXXG, wherein X at amino acid position 1 is an L, a V, an E, or a Q, wherein X at amino acid position 2 is a P if X at amino acid position 1 is an L, wherein X at amino acid position 2 is a V if X at amino acid position 1 is a E or a Q, wherein X at amino acid position 2 is a V or a P if X at amino acid position 1 is a V, wherein X at amino acid position 3 is any amino acid residue, wherein X at amino acid position 4 is a T if X at amino acid position 1 is a V, E, or Q, and wherein X at amino acid position 4 is a T, S, or A if X at amino acid position 1 is an L. Some examples of LPXTG motifs present in GAS AI surface proteins include LPSXG (SEQ ID NO: 134), VVXTG (SEQ ID NO: 135), EVXTG (SEQ ID NO: 136), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138), LPXAG (SEQ ID NO: 139), QVPTG (SEQ ID NO: 140), and FPXTG (SEQ ID NO: 141).
  • The GAS AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer. Preferably, one or more GAS AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. GAS AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The GAS AI-1 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-1 may encode for at least one surface protein. Alternatively, GAS AI-1 may encode for at least two surface exposed proteins and at least one sortase. Preferably, GAS AI-1 encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • GAS AI-1 preferably includes a srtB sortase. GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a GAS AI-1 surface protein such as M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, or DSM2071_fimbrial. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively:
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more GAS Adhesin Island 1 (“GAS AI-1”) proteins and one or more GAS Adhesin Island 2 (“GAS AI-2”), GAS Adhesin Island 3 (“GAS AI-3”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the GAS Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the GAS AI-1 proteins, GAS AI-1 may also include a divergently transcribed transcriptional regulator such as RofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • GAS Adhesin Island 2
  • A second adhesin island, “GAS Adhesin Island 2” or “GAS AI-2” has also been identified in Group A Streptococcus serotypes and isolates. GAS AI-2 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-2 proteins”). Specifically, GAS AI-2 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, 7, or 8) of GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20.
  • A preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which may be formulated or purified in an oligomeric (pilis) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Another preferred immunogenic composition of the invention comprises a GAS AI-2 surface protein which has been isolated in an oligomeric (pilis) form. The oligomer or hyperoligomeric pilus structures comprising the GAS AI-2 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-2 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-2 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the GAS AI-2 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. The AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The GAS AI-2 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-2 may encode for at least one surface protein. Alternatively, GAS AI-2 may encode for at least two surface exposed proteins and at least one sortase. Preferably, GAS AI-2 encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as GAS 15, GAS 16, or GAS 18. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more GAS Adhesin Island 2 (“GAS AI-2”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 3 (“GAS AI-3”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the GAS AI-2 proteins, GAS AI-2 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • GAS Adhesin Island 3
  • A third adhesin island, “GAS Adhesin Island 3” or “GAS AI-3” has also been identified in several Group A Streptococcus serotypes and isolates. GAS AI-3 comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-3 proteins”). Specifically, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, SpyM30104, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, SPs0106, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149. In one embodiment, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, and SpyM30104. In another embodiment, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, and SPs0106. In a further embodiment, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of orf78, orf79, orf80, orf81, orf82, orf83, and orf84. In yet another embodiment, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, and spyM180132. In yet another embodiment, GAS AI-3 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149.
  • Applicants have also identified open reading frames encoding fimbrial structural subunits in other GAS bacteria harbouring an AI-3. These open reading frames encode fimbrial structural subunits ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. A GAS AI-3 may comprise a polynucleotide encoding any one of ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial.
  • One or more of the GAS AI-3 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-3 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • A preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which may be formulated or purified in an oligomeric (pilis) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Another preferred immunogenic composition of the invention comprises a GAS AI-3 surface protein which has been isolated in an oligomeric (pilis) form. The oligomer or hyperoligomeric pilus structures comprising the GAS AI-3 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-3 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. The AI surface proteins of the invention may affect the ability of the GAS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The GAS AI-3 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-3 may encode for at least one surface protein. Alternatively, GAS AI-3 may encode for at least two surface exposed proteins and at least one sortase. Preferably, GAS AI-3 encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine or alanine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • The invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyM30098, SpyM3 0100, SpyM3 0102, SpyM30104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM180126, spyM18 0128, spyM180130, spyM180132, SpyoM01000155, SpyoM01000153, SpyoM01000151, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyM30098, SpyM3 0100, SpyM3 0102, and SpyM30104. In another embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SPs0100, SPs0102, SPs0104, and SPs0106. In another embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as orf78, orf80, orf82, and orf84. In yet another embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as spyM180126, spyM18 0128, spyM180130, and spyM180132. In a further embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as SpyoM01000155, SpyoM01000153, SpyoM0000151, and SpyoM1000149. In yet a further embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more GAS Adhesin Island 3 (“GAS AI-3”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 2 (“GAS AI-2”), or GAS Adhesin Island 4 (“GAS AI-4”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the GAS AI-3 proteins, GAS AI-3 may also include a transcriptional regulator such as Nra.
  • GAS Adhesin Island 4
  • A fourth adhesin island, “GAS Adhesin Island 4” or “GAS AI-4” has also been identified in Group A Streptococcus serotypes and isolates. GAS AI-4 comprises a series of approximately eight open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases (“GAS AI-4 proteins”). Specifically, GAS AI-4 includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, 7, or 8) of 19224134, 19224135, 19223136, 19223137, 19224138, 19224139, 19224140, and 19224141.
  • Applicants have also identified open reading frames encoding fimbrial structural subunits in other GAS bacteria harbouring an AI-4. These open reading frames encode fimbrial structural subunits 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial. A GAS AI-4 may comprise a polynucleotide encoding any one of 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • One or more of the GAS AI-4 open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the GAS AI-4 open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • A preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which may be formulated or purified in an oligomeric (pilis) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Another preferred immunogenic composition of the invention comprises a GAS AI-4 surface protein which has been isolated in an oligomeric (pilis) form. The oligomer or hyperoligomeric pilus structures comprising the GAS AI-4 surface proteins may be purified or otherwise formulate for use in immunogenic compositions.
  • One or more of the GAS AI-4 surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. The AI surface proteins of the invention may effect the ability of the GAS bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of GAS to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The GAS AI-4 sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. GAS AI-4 may encode for at least one surface protein. Alternatively, GAS AI-4 may encode for at least two surface exposed proteins and at least one sortase. Preferably, GAS AI-4 encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising an AI surface protein such as 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a GAS Adhesin Island protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more GAS Adhesin Island 4 (“GAS AI-4”) proteins and one or more GAS Adhesin Island 1 (“GAS AI-1”), GAS Adhesin Island 2 (“GAS AI-2”), or GAS Adhesin Island 3 (“GAS AI-3”) proteins, wherein one or more of the Adhesin Island proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the GAS AI-4 proteins, GAS AI-4 may also include a divergently transcribed transcriptional regulator such as rofA (i.e., the transcriptional regulator is located near or adjacent to the AI protein open reading frames, but it transcribed in the opposite direction).
  • The oligomeric, pilus-like structures of the invention may be combined with one or more additional GAS proteins. In one embodiment, the oligomeric, pilus-like structures comprise one or more AI surface proteins in combination with a second GAS protein.
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an AI surface protein. The invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a GAS bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the GAS bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed AI protein. Preferably, the AI protein is in a hyperoligomeric form.
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein. The invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a GAS bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the GAS bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed Adhesin Island protein. Preferably, the Adhesin Island protein is in a hyperoligomeric form.
  • The GAS bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • GAS bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the GAS bacteria with a plasmid encoding the AI protein. The plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein. Optionally, the sequence encoding the AI protein within the GAS bacterial genome may be deleted. Alternatively, or in addition, the promoter regulating the GAS Adhesin Island may be modified to increase expression.
  • The invention further includes GAS bacteria which have been adapted to produce increased levels of AI surface protein. In particular, the invention includes GAS bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein. In one embodiment, the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • The invention further includes GAS bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface. The GAS bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of LepA polypeptide, or an equivalent signal peptidase, in the GAS bacteria. Applicants have shown that deletion of LepA in strain SF370 bacteria, which harbour a GAS AI-2, abolishes surface exposure of M and pili proteins on the GAS. Increased levels of LepA expression in GAS are expected to result in increased exposure of M and pili proteins on the surface of GAS. Increased expression of LepA in GAS may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation. The GAS bacteria adapted to have increased levels of LepA expression may additionally be adapted to express increased levels of at least one pili protein.
  • Alternatively, the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes” Infection and Immunity (2004) 72(6):3444-3450). As used herein, non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis. Preferably, the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid. The non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface. Alternatively, the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria. For example, the AI surface protein may be isolated from cell extracts or culture supernatants. Alternatively, the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • The non-pathogenic Gram positive bacteria may be used to express any of the GAS Adhesin Island proteins described herein. The non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein. Preferably, the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase. The AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with pathogenic GAS.
  • Applicants modified L. lactis to demonstrate that, like GBS polypeptides, it can express GAS AI polypeptides. L. lactis was transformed with pAM401 constructs encoding entire pili gene clusters of AI-1, AI-2, and AI-4 adhesin islands. Briefly, the pAM401 is a promoterless high-copy plasmid. The entire pili gene clusters of an M6 (AI-1), M1 (AI-2), and M12 (AI-4) bacteria were inserted into the pAM401 construct. The gene clusters were transcribed under the control their own (M6, M1, or M12) promoter or the GBS promoter that successfully initiated expression of the GBS AI-1 adhesin islands in L. lactis, described above. FIG. 172 provides a schematic depiction of GAS M6 (AI-1), M1 (AI-2), and M12 (AI-4) adhesin islands and indicates the portions of the adhesin island sequences inserted in the pAM401 construct.
  • Each of the L. lactis transformed with one of the M6, M1, or M12 adhesin island gene clusters expressed high molecular weight structures that were immunoreactive with antibodies that bind to polypeptides present in their respective pili. FIGS. 173 A-C provide results of Western blot analysis of surface protein-enriched extracts of L. lactis transformed with M6 (FIG. 173 A), M1 (FIG. 173 B), or M12 (FIG. 173 C) adhesin island gene clusters using antibodies that bind to the fimbrial structural subunit encoded by each cluster. FIG. 173A at lanes 3 and 4 shows detection of high molecular structures in L. lactis transformed with an adhesin island pilus gene cluster from an M1 AI-2 using an antibody that binds to fimbrial structural subunit Spy0128. FIG. 173B at lanes 3 and 4 shows detection of high molecular weight structures in L. lactis transformed with an adhesin island pilus gene cluster from an M12 AI-4 using an antibody that binds to fimbrial structural subunit EftLSL.A. FIG. 173C at lane 3 shows detection of high molecular weight structures in L. lactis transformed with an adhesin island pilus gene cluster from an M6 AI-1 using an antibody that binds to fimbrial structural subunit M6_Spy0160. In FIGS. 173 A-C, “p1” immediately following the notation of AI subtype indicates that the promoter present in the Adhesin Island is used to drive transcription of the adhesin island gene cluster and “p2” indicates that the promoter was the GBS promoter described above. Thus, it appears that L. lactis is capable of expressing the fimbrial structural subunits encoded by GAS adhesin islands in an oligomeric form.
  • Alternatively, the oligomeric, pilus-like structures may be produced recombinantly. If produced in a recombinant host cell system, the AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention. Such AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • S. pneumoniae from TIGR4 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae from TIGR4. The S. pneumoniae from TIGR4 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae from TIGR4 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, and SP0468.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae from TIGR4 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. In a preferred embodiment, the oligomeric form is a hyperoligomer. Another preferred immunogenic composition of the invention comprises a S. pneumoniae from TIGR4 AI surface protein which has been isolated in an oligomeric (pilis) form. The oligomer or hyperoligomer pilus structures comprising S. pneumoniae surface proteins may be purified or otherwise formulated for use in immunogenic compositions.
  • One or more of the S. pneumoniae from TIGR4 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae from TIGR4 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae from TIGR4 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae from TIGR4 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae from TIGR4 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae from TIGR4 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae from TIGR4 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae from TIGR4 AI may encode for at least one surface protein. Alternatively, S. pneumoniae from TIGR4 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae from TIGR4 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae from TIGR4 AI surface protein such as SP0462, SP0463, SP0464, or SP0465. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae from TIGR4 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae from TIGR4 AI proteins and one or more S. pneumoniae strain 670 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae from TIGR4 AI proteins, S. pneumoniae from TIGR4 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 670 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 670. The S. pneumoniae strain 670 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 670 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of orf1 670, orf3 670, orf4 670, orf5 670, orf6 670, orf7 670, orf8 670.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 670 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 670 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 670 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 670 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 670 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 670 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 670 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 670 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 670 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 670 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 670 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 670 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 670 AI surface protein such as orf3 670, orf4 670, or orf5 670. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 670 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 670 AI proteins and one or more S. pneumoniae from TIGR4 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 670 AI proteins, S. pneumoniae strain 670 AI may also include a transcriptional regulator.
  • S. pneumoniae strain 14 CSR 10 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 14 CSR 10. The S. pneumoniae strain 14 CSR 10 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 14 CSR 10 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF214CSR, ORF314CSR, ORF414CSR, ORF514CSR, ORF614CSR, ORF714CSR, ORF814CSR.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 14 CSR 10 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 14 CSR 10 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 14 CSR 10 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 14 CSR 10 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 14 CSR 10 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 14 CSR 10 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 14 CSR 10 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 14 CSR 10 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 14 CSR 10 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 14 CSR 10 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 14 CSR 10 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 14 CSR 10 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 14 CSR 10 AI surface protein such as orf3_CSR, orf4_CSR, or orf5_CSR. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 14 CSR 10 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 14 CSR 10 AI proteins, and one or more AI proteins of any of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 14 CSR 10 AI proteins, S. pneumoniae strain 14 CSR 10 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 19A Hungary 6 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 19A Hungary 6. The S. pneumoniae strain 19A Hungary 6 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 19A Hungary 6 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF219AH, ORF319AH, ORF419AH, ORF5 1 gAH, ORF6 1 gAH, ORF719AH, ORF819AH.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19A Hungary 6 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19A Hungary 6 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 19A Hungary 6 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 19A Hungary 6 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 19A Hungary 6 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 19A Hungary 6 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 19A Hungary 6 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 19A Hungary 6 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 19A Hungary 6 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 19A Hungary 6 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 19A Hungary 6 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 19A Hungary 6 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 19A Hungary 6 AI surface protein such as orf319AH, orf419AH, or orf519AH. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 19A Hungary 6 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 19A Hungary 6 AI proteins and one or more AI proteins from one of any one of S. pneumoniae from TIGR4, 670, 14 CSR 10, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI GR4 AI proteins, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 19A Hungary 6 AI proteins, S. pneumoniae strain 19A Hungary 6 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 19F Taiwan 14 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 19F Taiwan 14. The S. pneumoniae strain 19F Taiwan 14 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 19F Taiwan 14 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF219FTW, ORF319FTW, ORF419FTW, ORF519FTW, ORF619FTW, ORF719FTW, ORF819FTW.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 19F Taiwan 14 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 19F Taiwan 14 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 19F Taiwan 14 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 19F Taiwan 14 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 19F Taiwan 14 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 19F Taiwan 14 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 19F Taiwan 14 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 19F Taiwan 14 μl may encode for at least one surface protein. Alternatively, S. pneumoniae strain 19F Taiwan 14 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 19F Taiwan 14 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 19F Taiwan 14 AI surface protein such as orf319FTW, orf419FTW, or orf519FTW. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 19F Taiwan 14 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 19F Taiwan 14 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 14 CSR 10, 23F Taiwan 15, or 23F Poland 16, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 19F Taiwan 14 AI proteins, S. pneumoniae strain 19F Taiwan 14 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 23F Poland 16 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 23F Poland 16. The S. pneumoniae strain 23F Poland 16 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 23F Poland 16 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF223FP, ORF323FP, ORF423FP, ORF523FP, ORF623FP, ORF723FP, and ORF823FP.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Poland 16 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Poland 16 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 23F Poland 16 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 23F Poland 16 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 23F Poland 16 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 23F Poland 16 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 23F Poland 16 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 23F Poland 16 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 23F Poland 16 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 23F Poland 16 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 23F Poland 16 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 23F Poland 16 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 23F Poland 16 AI surface protein such as orf323FP, orf423FP, or orf523FP. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 23F Poland 16 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 23F Poland 16 AI proteins and one or more AI proteins from any one or more S. pneumoniae strains of TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 14 CSR 10, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 23F Poland 16 AI proteins, S. pneumoniae strain 23F Poland 16 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 23F Taiwan 15 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 23F Taiwan 15. The S. pneumoniae strain 23F Taiwan 15 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 23F Taiwan 15 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF223FTW, ORF323FTW, ORF423FTW, ORF523FTW, ORF623FTW, ORF723F[W, ORF823FTW.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 23F Taiwan 15 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 23F Taiwan 15 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 23F Taiwan 15 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 23F Taiwan 15 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 23F Taiwan 15 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 23F Taiwan 15 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 23F Taiwan 15 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 23F Taiwan 15 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 23F Taiwan 15 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 23F Taiwan 15 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 23F Taiwan 15 AI surface protein such as orf323FTW, orf423FTW, or orf523FTW. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 23F Taiwan 15 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 23F Taiwan 15 AI proteins and one or more AI proteins from any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 14 CSR 10, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 23F Taiwan 15 AI proteins, S. pneumoniae strain 23F Taiwan 15 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 6B Finland 12 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 6B Finland 12. The S. pneumoniae strain 6B Finland 12 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 6B Finland 12 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF26BF, ORF36BF, ORF46BF, ORF56BF, ORF66BF, ORF76BF, ORF86BF.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Finland 12 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Finland 12 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 6B Finland 12 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 6B Finland 12 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 6B Finland 12 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 6B Finland 12 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 6B Finland 12 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 6B Finland 12 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 6B Finland 12 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 6B Finland 12 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 6B Finland 12 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 6B Finland 12 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 6B Finland 12 AI surface protein such as orf36BF, orf46BF, or orf56BF. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 6B Finland 12 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 6B Finland 12 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 6B Finland 12 AI proteins, S. pneumoniae strain 6B Finland 12 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 6B Spain 2 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 6B Spain 2. The S. pneumoniae strain 6B Spain 2 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 6B Spain 2 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF26BSP, ORF36BSP, ORF46BSP, ORF56BSP, ORF66BSP, ORF76BSP, and ORF86BSP.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Spain 2 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 6B Spain 2 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 6B Spain 2 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 6B Spain 2 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 6B Spain 2 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 6B Spain 2 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 6B Spain 2 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 6B Spain 2 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 6B Spain 2 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 6B Spain 2 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 6B Spain 2 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 6B Spain 2 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 6B Spain 2 AI surface protein such as orf36BSP, orf46BSP, or orf56BSP. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 6B Spain 2 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 6B Spain 2 AI proteins and one or more AI proteins of any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 14 CSR 10, 9V Spain 3, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 6B Spain 2 AI proteins, S. pneumoniae strain 6B Spain 2 AI may also include a transcriptional regulator.
  • S. pneumoniae Strain 9V Spain 3 Adhesin Island
  • As discussed above, Applicants have identified adhesin islands within the genome of S. pneumoniae strain 9V Spain 3. The S. pneumoniae strain 9V Spain 3 Adhesin Island comprises a series of approximately seven open reading frames encoding for a collection of amino acid sequences comprising surface proteins and sortases. Specifically, the S. pneumoniae strain 9V Spain 3 AI proteins includes open reading frames encoding for two or more (i.e., 2, 3, 4, 5, 6, or 7) of ORF29VSP, ORF39VSP, ORF49VSP, ORF59VSP, ORF69VSP, ORF79VSP, and ORF89VSP.
  • A preferred immunogenic composition of the invention comprises a S. pneumoniae strain 9V Spain 3 AI surface protein which may be formulated or purified in an oligomeric (pilis) form. Another preferred immunogenic composition of the invention comprises a S. pneumoniae strain 9V Spain 3 AI surface protein which has been isolated in an oligomeric (pilis) form.
  • One or more of the S. pneumoniae strain 9V Spain 3 AI open reading frame polynucleotide sequences may be replaced by a polynucleotide sequence coding for a fragment of the replaced ORF. Alternatively, one or more of the S. pneumoniae strain 9V Spain 3 AI open reading frames may be replaced by a sequence having sequence homology to the replaced ORF.
  • One or more of the S. pneumoniae strain 9V Spain 3 AI surface protein sequences typically include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif.
  • The S. pneumoniae strain 9V Spain 3 AI surface proteins of the invention may affect the ability of the S. pneumoniae bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of S. pneumoniae to translocate through an epithelial cell layer. Preferably, one or more S. pneumoniae strain 9V Spain 3 AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. S. pneumoniae strain 9V Spain 3 AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • The S. pneumoniae strain 9V Spain 3 AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. S. pneumoniae strain 9V Spain 3 AI may encode for at least one surface protein. Alternatively, S. pneumoniae strain 9V Spain 3 AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, S. pneumoniae strain 9V Spain 3 AI encodes for at least three surface exposed proteins and at least two sortases.
  • The AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a S. pneumoniae strain 9V Spain 3 AI surface protein such as orf39VSP, orf49VSP, or orf59VSP. The oligomeric, pilus-like structure may comprise numerous units of AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine or serine amino acid residue, respectively.
  • AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include a pilin motif.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a S. pneumoniae strain 9V Spain 3 AI protein in oligomeric form, preferably in a hyperoligomeric form. In one embodiment, the invention comprises a composition comprising one or more S. pneumoniae strain 9V Spain 3 AI proteins and one or more AI proteins from any one or more of S. pneumoniae from TIGR4, 670, 19A Hungary 6, 6B Finland 12, 6B Spain 2, 14 CSR 10, 19F Taiwan 14, 23F Taiwan 15, or 23F Poland 16 AI, wherein one or more of the S. pneumoniae AI proteins is in the form of an oligomer, preferably in a hyperoligomeric form.
  • In addition to the open reading frames encoding the S. pneumoniae strain 9V Spain 3 AI proteins, S. pneumoniae strain 9V Spain 3 AI may also include a transcriptional regulator.
  • The S. pneumoniae oligomeric, pilus-like structures may be isolated or purified from bacterial cultures in which the bacteria express an S. pneumoniae AI surface protein. The invention therefore includes a method for manufacturing an oligomeric AI surface antigen comprising culturing a S. pneumoniae bacterium that expresses the oligomeric AI protein and isolating the expressed oligomeric AI protein from the S. pneumoniae bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed AI protein. Preferably, the AI protein is in a hyperoligomeric form.
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing an AI surface protein. The invention therefore includes a method for manufacturing an S. pneumoniae oligomeric Adhesin Island surface antigen comprising culturing a S. pneumoniae bacterium adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the S. pneumoniae bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed Adhesin Island protein. Preferably, the Adhesin Island protein is in a hyperoligomeric form.
  • The S. pneumoniae bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • S. pneumoniae bacteria may be adapted to increase AI protein expression by any means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the S. pneumoniae bacteria with a plasmid encoding the AI protein. The plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein. Optionally, the sequence encoding the AI protein within the S. pneumoniae bacterial genome may be deleted. Alternatively, or in addition, the promoter regulating the S. pneumoniae Adhesin Island may be modified to increase expression.
  • The invention further includes S. pneumoniae bacteria which have been adapted to produce increased levels of AI surface protein. In particular, the invention includes S. pneumoniae bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein. In one embodiment, the S. pneumoniae of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • The invention further includes S. pneumoniae bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface. The S. pneumoniae bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide. Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria. Increased expression of a leader peptidase in S. pneumoniae may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation. The S. pneumoniae bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • Alternatively, the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal Vaccine Made from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes” Infection and Immunity (2004) 72(6):3444-3450). As used herein, non-pathogenic Gram positive bacteria refer to Gram positive bacteria which are compatible with a human host subject and are not associated with human pathogenisis. Preferably, the non-pathogenic bacteria are modified to express the AI surface protein in oligomeric, or hyper-oligomeric form. Sequences encoding for an AI surface protein and, optionally, an AI sortase, may be integrated into the non-pathogenic Gram positive bacterial genome or inserted into a plasmid. The non-pathogenic Gram positive bacteria may be inactivated or attenuated to facilitate in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface. Alternatively, the AI surface protein may be isolated or purified from a bacterial culture of the non-pathogenic Gram positive bacteria. For example, the AI surface protein may be isolated from cell extracts or culture supernatants. Alternatively, the AI surface protein may be isolated or purified from the surface of the non-pathogenic Gram positive bacteria.
  • The non-pathogenic Gram positive bacteria may be used to express any of the S. pneumoniae Adhesin Island proteins described herein. The non-pathogenic Gram positive bacteria are transformed to express an Adhesin Island surface protein. Preferably, the non-pathogenic Gram positive bacteria also express at least one Adhesin Island sortase. The AI transformed non-pathogenic Gram positive bacteria of the invention may be used to prevent or treat infection with pathogenic S. pneumoniae.
  • FIGS. 190 A and B, and 193-195 provide examples of three methods successfully practiced by applicants to purify pili from S. pneumoniae TIGR4.
  • Immunogenic Compositions
  • The Gram positive bacteria AI proteins described herein are useful in immunogenic compositions for the prevention or treatment of Gram positive bacterial infection. For example, the GBS AI surface proteins described herein are useful in immunogenic compositions for the prevention or treatment of GBS infection. As another example, the GAS AI surface proteins described herein may be useful in immunogenic compositions for the prevention or treatment of GAS infection. As another example, the S. pneumoniae AI surface proteins may be useful in immunogenic cojmpositions for the prevention or treatment of S. pneumoniae infection.
  • Gram positive bacteria AI surface proteins that can provide protection across more than one serotype or strain isolate may be used to increase immunogenic effectiveness. For example, a particular GBS AI surface protein having an amino acid sequence that is at least 50% (i.e., at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%) homologous to the particular GBS AI surface protein of at least 2 (i.e., at least 3, 4, 5, 6, 7, 8, 9, 10, or more) other GBS serotypes or strain isolates may be used to increase the effectiveness of such compositions.
  • As another example, fragments of Gram positive bacteria AI surface proteins that can provide protection across more than one serotype or strain isolate may be used to increase immunogenic effectiveness. Such a fragment may be identified within a consensus sequence of a full length amino acid sequence of a Gram positive bacteria AI surface protein. Such a fragment can be identified in the consensus sequence by its high degree of homology or identity across multiple (i.e, at least 3, 4, 5, 6, 7, 8, 9, 10, or more) Gram positive bacteria serotypes or strain isolates. Preferably, a high degree of homology is a degree of homology of at least 90% (i.e., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) across Gram positive bacteria serotypes or strain isolates. Preferably, a high degree of identity is a degree of identity of at least 90% (i.e., at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) across Gram positive bacteria serotypes or strain isolates. In one embodiment of the invention, such a fragment of a Gram positive bacteria AI surface protein may be used in the immunogenic compositions.
  • In addition, the AI surface protein oligomeric pilus structures may be formulated or purified for use in immunization. Isolated AI surface protein oligomeric pilus structures may also be used for immunization.
  • The invention includes an immunogenic composition comprising a first Gram positive bacteria AI protein and a second Gram positive bacterial AI protein. One or more of the AI proteins may be a surface protein. Such surface proteins may contain an LPXTG motif or other sortase substrate motif.
  • The first and second AI proteins may be from the same or different genus or species of Gram positive bacteria. If within the same species, the first and second AI proteins may be from the same or different AI subtypes. If two AIs are of the same subtype, the AIs have the same numerical designation. For example, all AIs designated as AI-1 are of the same AI subtype. If two AIs are of a different subtype, the AIs have different numerical designations. For example, AI-1 is of a different AI subtype from AI-2, AI-3, AI-4, etc. Likewise, AI-2 is of a different AI subtype from AI-1, AI-3, and AI-4, etc.
  • For example, the invention includes an immunogenic composition comprising one or more GBS AI-1 proteins and one or more GBS AI-2 proteins. One or more of the AI proteins may be a surface protein. Such surface proteins may contain an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) and may bind fibrinogen, fibronectin, or collagen. One or more of the AI proteins may be a sortase. The GBS AI-1 proteins may be selected from the group consisting of GBS 80, GBS 104, GBS 52, SAG0647 and SAG0648. Preferably, the GBS AI-1 proteins include GBS 80 or GBS 104.
  • The GBS AI-2 proteins may be selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, SAG1406, 01520, 01521, 01522, 01523, 01523, 01524 and 01525. In one embodiment, the GBS AI-2 proteins are selected from the group consisting of GBS 67, GBS 59, GBS 150, SAG1405, and SAG1406. In another embodiment, the GBS AI-2 proteins may be selected from the group consisting of 01520, 01521, 01522, 01523, 01523, 01524 and 01525. Preferably, the GBS AI-2 protein includes GBS 59 or GBS 67.
  • As another example, the invention includes an immunogenic composition comprising one or more of any combination of GAS AI-1, GAS AI-2, GAS AI-3, or GAS AI-4 proteins. One or more of the GAS AI proteins may be a sortase. The GAS AI-1 proteins may be selected from the group consisting of M6_Spy0156, M6_Spy0157, M6_Spy0158, M6_Spy0159, M6_Spy0160, M6_Spy0161, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial. Preferably, the GAS AI-1 proteins are selected from the group consisting of M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, and DSM2071_fimbrial.
  • The GAS AI-2 proteins may be selected from the group consisting of Spy0124, GAS15, Spy0127, GAS16, GAS17, GAS18, Spy0131, Spy0133, and GAS20. Preferably, the GAS AI-2 proteins are selected from the group consisting of GAS 15, GAS 16, and GAS 18.
  • The GAS AI-3 proteins may be selected from the group consisting of SpyM30097, SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, SpyM30104, SPs0099, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, SPs0106, orf77, orf78, orf79, orf80, orf81, orf82, orf83, orf84, spyM180125, spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, and ISS4959_fimbrial. In one embodiment the GAS AI-3 proteins are selected from the group consisting of SpyM30097, SpyM30098, SpyM30099, SpyM3 0100, SpyM30101, SpyM3 0102, SpyM30103, and SpyM30104. In another embodiment, the GAS AI-3 proteins are selected from the group consisting of SPs0099, SPs0100, SPs0101, SPs0102, SPs0103, SPs0104, SPs0105, and SPs0106. In yet another embodiment, the GAS AI-3 proteins are selected from the group consisting of orf77, orf78, orf79, orf80, orf81, orf82, orf83, and orf84. In a further embodiment, the GAS AI-3 proteins are selected from the group consisting of spyM180125, spyM180126, spyM180127, spyM18 0128, spyM180129, spyM180130, spyM180131, and spyM180132. In yet another embodiment the GAS AI-3 proteins are selected from the group consisting of SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, and SpyoM01000149.
  • The GAS AI-4 proteins may be selected from the group consisting of 19224133, 19224134, 19224135, 19224136, 19224137, 19224138, 19224139, 19224140, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial. Preferably, the GAS-AI-4 proteins are selected from the group consisting of 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, and ISS4538_fimbrial.
  • As yet another example, the invention includes an immunogenic composition comprising one or more of any combination of S. pneumonaie from TIGR4, S. pneumonaie strain 670, S. pneumonaie from 19A Hungary 6, S. pneumonaie from 6B Finland 12, S. pneumonaie from 6B Spain 2, S. pneumonaie from 9V Spain 3, S. pneumonaie from 14 CSR 10, S. pneumonaie from 19F Taiwan 14, S. pneumonaie from 23F Taiwan 15, or S. pneumonaie from 23F Poland 16 AI proteins. One or more of the AI proteins may be a surface protein. Such surface proteins may contain an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) and may bind fibrinogen, fibronectin, or collagen. One or more of the AI proteins may be a sortase.
  • The S. pneumonaie from TIGR4 AI proteins may be selected from the group consisting of SP0462, SP0463, SP0464, SP0465, SP0466, SP0467, SP0468. Preferably, the S. pneumonaie from TIGR4 AI proteins include SP0462, SP0463, or SP0464.
  • The S. pneumonaie strain 670 AI proteins may be selected from the group consisting of Orf1 670, Orf3 670, Orf4 670, Orf5 670, Orf6 670, Orf7 670, and Orf8 670. Preferably, the S. pneumonaie strain 670 AI proteins include Orf3 670, Orf4 670, or Orf5 670.
  • The S. pneumonaie from 19A Hungary 6 AI proteins may be selected from the group consisting of ORF2 1 gAH, ORF3 1 gAH, ORF419AH, ORF519AH, ORF6 1 gAH, ORF719AH, or ORF819AH.
  • The S. pneumonaie from 6B Finland 12 AI proteins may be selected from the group consisting of ORF26BF, ORF36BF, ORF46BF, ORF56BF, ORF66BF, ORF76BF, or ORF86BF.
  • The S. pneumonaie from 6B Spain 2 AI proteins may be selected from the group consisting of ORF26BSP, ORF36BSP, ORF46BSP, ORF56BSP, ORF66BSP, ORF76BSP, or ORF86BSP.
  • The S. pneumonaie from 9V Spain 3 AI proteins may be selected from the group consisting of ORF29VSP, ORF39VSP, ORF49VSP, ORF59VSP, ORF69VSP, ORF79VSP, or ORF89VSP.
  • The S. pneumonaie from 14 CSR 10 AI proteins may be selected from the group consisting of ORF214CSR, ORF314CSR, ORF414CSR, ORF514CSR, ORF614CSR, ORF714CSR, or ORF814CSR.
  • The S. pneumonaie from 19F Taiwan 14 AI proteins may be selected from the group consisting of ORF2 1 gFTW, ORF319FTW, ORF419FTW, ORF519FTW, ORF619FTW, ORF719FTW, or ORF819FTW.
  • The S. pneumonaie from 23F Taiwan 15 AI proteins may be selected from the group consisting of ORF223FTW, ORF323FTW, ORF423FTW, ORF523FTW, ORF623FTW, ORF723FTW, or ORF823FTW.
  • The S. pneumonaie from 23F Poland 16 AI proteins may be selected from the group consisting of ORF223FP, ORF323FP, ORF423FP, ORF523FP, ORF623FP, ORF723FP or ORF823FP.
  • Preferably, the Gram positive bacteria AI proteins included in the immunogenic compositions of the invention can provide protection across more than one serotype or strain isolate. For example, the immunogenic composition may comprise a first AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different serotypes of a Gram positive bacteria. The first AI protein may also be homologous to the amino acid sequence of a third AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different serotypes of a Gram positive bacteria. The first AI protein may also be homologous to the amino acid sequence of a fourth AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different serotypes of a Gram positive bacteria.
  • For example, preferably, the GBS AI proteins included in the immunogenic compositions of the invention can provide protection across more than one GBS serotype or strain isolate. For example, the immunogenic composition may comprise a first GBS AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second GBS AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different GBS serotypes. The first GBS AI protein may also be homologous to the amino acid sequence of a third GBS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GBS serotypes. The first AI protein may also be homologous to the amino acid sequence of a fourth GBS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GBS serotypes.
  • The first AI protein may be selected from an AI-1 protein or an AI-2 protein. For example, the first AI protein may be a GBS AI-1 surface protein such as GBS 80. The amino acid sequence of GBS 80 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 80 amino acid sequence from GBS serotype III, strain isolates NEM316 and COH1 and the GBS 80 amino acid sequence from GBS serotype 1a, strain isolate A909.
  • As another example, the first AI protein may be GBS 104. The amino acid sequence of GBS 104 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 104 amino acid sequence from GBS serotype III, strain isolates NEM316 and COH1, the GBS 104 amino acid sequence from GBS serotype 1a, strain isolate A909, and the GBS 104 amino acid sequence serotype II, strain isolate 18RS21.
  • Table 12 provides the amino acid sequence identity of GBS 80 and GBS 104 across GBS serotypes Ia, Ib, II, III, V, and VIII. The GBS strains in which genes encoding GBS 80 and GBS 104 were identified share, on average, 99.88 and 99.96 amino acid sequence identity, respectively. This high degree of amino acid identity indicates that an immunogenic composition comprising a first protein of GBS 80 or GBS 104 may provide protection across more than one GBS serotype or strain isolate.
    TABLE 12
    Conservation of GBS 80 and GBS 104 amino acid sequences
    GBS
    80 GBS 104
    Serotype Strains cGH % AA identity cGH % AA identity
    Ia
    090 + 99.79 + 100.00
    A909 + 100.00 + 100.00
    515
    DK1
    DK8
    Davis
    Ib 7357b + 100.00 +
    H36B
    II 18RS21 + 100.00
    DK21
    III NEM316 + 100.00 + 100.00
    COH31 + 100.00 +
    D136 + 100.00 +
    M732 + 100.00 + 99.88
    COH1 + 99.79 + 99.88
    M781 + 99.79 + 99.88
    No type CJB110 + 99.37 + 100.00
    1169NT
    V CJB111 + 100.00 + 100.00
    2603 + 100.00 + 100.00
    VIII JM130013 + 99.79 + 100.00
    SMU014 + 100.00 +
    total 14/22 99.88 +/− 0.19 15/22 99.96 +/− 0.056
  • As another example, the first AI protein may be an AI-2 protein such as GBS 67. The amino acid sequence of GBS 67 from GBS serotype V, strain isolate 2603 is greater than 90% homologous to the GBS 67 amino acid sequence from GBS serotype III, strain isolate NEM316, the GBS 67 amino acid sequence from GBS serotype 1b, strain isolate H36B, and the GBS 67 amino acid sequence from GBS serotype II, strain isolate 17RS21.
  • As another example, the first AI protein may be an AI-2 protein such as spb1. The amino acid sequence of spb1 from GBS serotype III, strain isolate COH1 is greater than 90% homologous to the spb1 amino acid sequence from GBS serotype Ia, strain isolate A909.
  • As yet another example, the first AI protein may be an AI-2 protein such as GBS 59. The amino acid sequence of GBS 59 from GBS serotype II, strain isolate 18RS21 is 100% homologous to the GBS 59 amino acid sequence from GBS serotype V, strain isolate 2603. The amino acid sequence of GBS 59 from GBS serotype V, strain isolate CJB111 is 98% homologous to the GBS 59 amino acid sequence from GBS serotype III, strain isolate NEM316.
  • The compositions of the invention may also be designed to include Gram positive AI proteins from divergent serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of serotypes or strain isolates of a Gram positive bacteria and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • For example, the invention may include an immunogenic composition comprising a first and second Gram positive bacteria AI protein, wherein a polynucleotide sequence encoding for the full length sequence of the first AI protein is not present in a similar Gram positive bacterial genome comprising a polynucleotide sequence encoding for the second AI protein.
  • The compositions of the invention may also be designed to include AI proteins from divergent GBS serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of GBS serotypes or strain isolates and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • For example, the invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein a polynucleotide sequence encoding for the full length sequence of the first GBS AI protein is not present in a genome comprising a polynucleotide sequence encoding for the second GBS AI protein. For example, the first AI protein could be GBS 80 (such as the GBS 80 sequence from GBS serotype V, strain isolate 2603). As previously discussed (and depicted in FIG. 2), the sequence for GBS 80 in GBS sertoype II, strain isolate 18RS21 is disrupted. In this instance, the second AI protein could be GBS 104 or GBS 67 (sequences selected from the GBS serotype II, strain isolate 18RS21).
  • Further, the the invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein the first GBS AI protein has detectable surface exposure on a first GBS strain or serotype but not a second GBS strain or serotype and the second GBS AI protein has detectable surface exposure on a second GBS strain or serotype but not a first GBS strain or serotype. For example, the first AI protein could be GBS 80 and the second AI protein could be GBS 67. As seen in Table 15, there are some GBS serotypes and strains that have surface exposed GBS 80 but that do not have surface exposed GBS 67 and vice versa. An immunogenic composition comprising a GBS 80 and a GBS 67 protein may provide protection across a wider group of GBS strains and serotypes.
    TABLE 15
    Antigen surface exposure of GBS 80, GBS 322, GBS 104, and GBS 67
    GBS strains Type GBS 80 GBS 322 GBS 104 GBS 67
    DK1* Ia 0 nd 237 478
    DK8* 0 213 151 475
    Davis* 0 86 271 430
    515* 0 227 262 409
    090 0 0 0 0
    A909 0 0 0 0
    2986 0 0 157 397
    5551 0 36 384 485
    2177 Ib 477 323 328 66
    H36B* 0 105 518 444
    7357b- 91 102 309 316
    2129 57 71 132 0
    5518 31 nd 60 28
    COH1 III 305 130 305 0
    D136C 16 460 226 406
    COH31 0 479 71 273
    M732 105 292 101 0
    M781 65 224 136 0
    1998 95 288 205 350
    5376 165 76 156 0
    5435 93 88 100 0
    18RS21 II 0 471 50 103
    DK21* 0 342 419 331
    3050 43 188 289 460
    5401 170 135 494 618
    2141 0 76 0 69
    CJB111 V 365 58 355 481
    2603 62 293 100 105
    5364 454 463 379 394
    2110 0 11 345 589
    2274 IV 113 161 465 484
    1999 0 55 492 453
    2210 0 0 363 574
    2928 VII 0 0 0 0
    SMU071 VIII 556 170 393 79
    JM9130013 587 133 436 83
    2189 0 0 0 0
    5408 0 0 159 433
    CJB110 NT 71 587 169 245
    1169* 0 213 371 443
    Δ Mean >100 9/40 22/38 32/40 25/40
    22% 58% 80% 62%
  • Alternatively, the invention may include an immunogenic composition comprising a first and second Gram positive bacteria AI protein, wherein the polynucleotide sequence encoding the sequence of the first AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second AI protein.
  • The invention may include an immunogenic composition comprising a first and second GBS AI protein, wherein the polynucleotide sequence encoding the sequence of the first GBS AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second GBS AI protein. For example, the first GBS AI protein could be GBS 67 (such as the GBS 67 sequence from GBS serotype 1b, strain isolate H36B). As shown in FIGS. 2 and 4, the GBS 67 sequence for this strain is less than 90% homologous (87%) to the corresponding GBS 67 sequence in GBS serotype V, strain isolate 2603. In this instance, the second GBS AI protein could then be the GBS 80 sequence from GBS serotype V, strain isolate 2603.
  • An example immunogenic composition of the invention may comprise adhesin island proteins GBS 80, GBS 104, GBS 67, and GBS 59, and non-AI protein GBS 322. FACS analysis of different GBS strains demonstrates that at least one of these five proteins is always found to be expressed on the surface of GBS bacteria. An initial FACS analysis of 70 strains of GBS bacteria, obtained from the CDC in the United States (33 strains), ISS in Italy (17 strains), and Houston/Harvard (20 strains), detected surface exposure of at least one of GBS 80, GBS 104, GBS 322, GBS 67, or GBS 59 on the surface of the GBS bacteria. FIG. 227 provides the FACS data obtained for surface exposure of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 on each of 37 GBS strains. FIG. 228 provides the FACS data obtained for surface exposure of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 on each of 41 GBS strains obtained from the CDC. As can be seen from FIGS. 227 and 228, each GBS strain had surface expression of at least one of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59. The surface exposure of at least one of these proteins on each bacterial strain indicates that an immunogenic composition comprising these proteins will provide wide protection across GBS strains and serotypes.
  • The surface exposed GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 proteins are also present at high levels as determined by FACS. Table 49 summarizes the FACS results for the initial 70 GBS strains examined for GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59 surface expression. A protein was designated as having high levels of surface expression of a protein if a five-fold shift in fluorescence was observed when using antibodies for the protein relative to preimmune control serum.
    TABLE 49
    Exposure Levels of GBS 80, GBS 104, GBS 67, GBS 322, and GBS 59
    on GBS Strains
    GBS
    80 GBS 104 GBS 67 GBS 59 GBS 322
    5-fold shift in 17/70 14/70 49/70 46/70 33/70
    fluorescence 24% 20% 70% 66% 47%
    by FACS
  • Table 50 details which of the surface proteins is highly expressed on the different GBS serotype.
    TABLE 50
    High Levels of Surface Protein Expression on GBS Serotypes
    5-fold shift in
    fluorescence
    by FACS GBS 80 GBS 104 GBS 67 GBS 59 GBS 322
    Ia + Ib + III  4/36 2/36 22/36 20/36 18/36
    II + V 11/25 9/25 21/25 21/25 13/25
    Others 2/9 3/9  6/9  5/9 2/9
  • Alternatively, the immunogenic composition of the invention may include GBS 80, GBS 104, GBS 67, and GBS 322. Assuming that protein antigens that are highly accessible to antibodies confer 100% protection with suitable adjuvants, an immunogenic composition containing GBS 80, GBS 104, GBS 67, GBS 59 and GBS 322 will provide protection for 89% of GBS strains and serotypes, the same percentage as an immunogenic composition containing GBS 80, GBS 104, GBS 67, and GBS 322 proteins. See FIG. 229. However, it may be preferable to include GBS 59 in the composition to increase its immunogenic strength. As seen from Table 50, GBS 59 is highly expressed on the surface two-thirds of GBS bacteria examined by FACS analysis, unlike GBS 80, GBS 104, and GBS 322, which are highly expressed in less than half of GBS bacteria examined. GBS 59 opsonophagocytic activity is also comparable to that of a mix of GBS 322, GBS 104, GBS 67, and GBS 80 proteins. See FIG. 230.
  • By way of another example, preferably, the GAS AI proteins included in the immunogenic compositions of the invention can provide protection across more than one GAS serotype or strain isolate. For example, the immunogenic composition may comprise a first GAS AI protein, wherein the amino acid sequence of said AI protein is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100%) homologous to the amino acid sequence of a second GAS AI protein, and wherein said first AI protein and said second AI protein are derived from the genomes of different GAS serotypes. The first GAS AI protein may also be homologous to the amino acid sequence of a third GAS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GAS serotypes. The first AI protein may also be homologous to the amino acid sequence of a fourth GAS AI protein, such that the first AI protein, the second AI protein and the third AI protein are derived from the genomes of different GAS serotypes.
  • The compositions of the invention may also be designed to include GAS AI proteins from divergent serotypes or strain isolates, i.e., to include a first AI protein which is present in one collection of serotypes or strain isolates of a GAS bacteria and a second AI protein which is present in those serotypes or strain isolates not represented by the first AI protein.
  • For example, the first AI protein could be a prtF2 protein (such as the 19224141 protein from GAS serotype M12, strain isolate A735). As previously discussed (and depicted in FIG. 164), the sequence for a prtF2 protein is not present in GAS AI types 1 or 2. In this instance, the second AI protein could be collagen binding protein M6_Spy0159 (from M6 isolate (MGAS10394), which comprises an AI-1) or GAS15 (from M1 isolate (SF370), which comprises an AI-2).
  • Further, the invention may include an immunogenic composition comprising a first and second GAS AI protein, wherein the first GAS AI protein has detectable surface exposure on a first GAS strain or serotype but not a second GAS strain or serotype and the second GAS AI protein has detectable surface exposure on a second GAS strain or serotype but not a first GAS strain or serotype.
  • The invention may include an immunogenic composition comprising a first and second GAS AI protein, wherein the polynucleotide sequence encoding the sequence of the first GAS AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 80, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35 or 30 percent) homologous than the corresponding sequence in the genome of the second GAS AI protein. Preferably the first and second GAS AI proteins are subunits of the pilus. More preferably the first and second GAS AI proteins are selected from the major pilus forming proteins (i.e., M6_Spy0160 from M6 strain 10394, SPy0128 from M1 strain SF370, SpyM3 0100 from M3 strain 315, SPs0102 from M3 strain SSI, orf80 from M5 isolate Manfredo, spyM18 0128 from M18 strain 8232, SpyoM01000153 from M49 strain 591, 19224137 from M12 strain A735, fimbrial structural subunit from M77 strain ISS4959, fimbrial structural subunit from M44 strain ISS3776, fimbrial structural subunit from M50 strain ISS3776 ISS 4538, fimbrial structural subunit from M12 strain CDC SS635, fimbrial structural subunit from M23 strain DSM2071, fimbrial structural subunit from M6 strain CDC SS410). Table 45 provides the percent identity between the amino acidic sequences of each of the main pilus forming subunits from GAS AI-1, AI-2, AI-3, and AI-4 representative strains (i.e., M6_Spy0160 from M6 strain 10394, SPy0128 from M1 strain SF370, SpyM3 0100 from M3 strain 315, SPs0102 from M3 strain SSI, orf80 from M5 isolate Manfredo, spyM18 0128 from M18 strain 8232, SpyoM01000153 from M49 strain 591, 19224137 from M12 strain A735, Fimbrial structural subunit from M77 strain ISS4959, fimbrial structural subunit from M44 strain ISS3776, fimbrial structural subunit from M50 strain ISS3776 ISS 4538, fimbrial structural subunit from M12 strain CDC SS635, fimbrial structural subunit from M23 strain DSM2071, fimbrial structural subunit from M6 strain CDC SS410).
    TABLE 45
    Comparison of Amino Acid Sequences of Major Pilus Proteins in the Four GAS
    AIs
    Figure US20060165716A1-20060727-C00001
  • For example, the first main pilus subunit may be selected from bacteria of GAS serotype M6 strain 10394 and the second main pilus subunit may be selected from bacteria of GAS serotype M1 strain 370. As can be seen from Table 45, the main pilus subunits encoded by these strains of bacteria share only 23% nucleotide identity. An immunogenic composition comprising pilus main subunits from each of these strains of bacteria is expected to provide protection across a wider group of GAS strains and serotypes. Other examples of main pilus subunits that can be used in combination to provide increased protection across a wider range of GAS strains and serotypes include proteins encoded by GAS serotype M5 Manfredo isolate and serotype M6 strain 10394, which share 23% sequence identity, GAS serotype M18 strain 8232 and serotype M1 strain 370, which share 38% sequence identity, GAS serotype M3 strain 315 and serotype M12 strain A735, which share 61% sequence identity, and GAS serotype M3 strain 315 and serotype M6 strain 10394 which share 25% sequence identity.
  • As also can be seen from Table 45, the amino acid sequences of the four types of main pilus subunits present in GAS are relatively divergent. FIGS. 198-201 provide further tables comparing the percent identity of adhesin island-encoded surface exposed proteins for different GAS serotypes relative to other GAS serotypes harbouring an adhesin island of the same or a different subtype (GAS AI-1, GAS AI-2, GAS AI-3, and GAS AI-4). See also further discussion below.
  • Immunizations with the Adhesin Island proteins of the invention are discussed further in the Examples.
  • Co-Expression of GBS Adhesin Island Proteins and Role of GBS AI Proteins in Surface Presentation
  • In addition to the use of the GBS adhesin island proteins for cross strain and cross serotype protection, Applicants have identified interactions between adhesin island proteins which appear to affect the delivery or presentation of the surface proteins on the surface of the bacteria.
  • In particular, Applicants have discovered that surface exposure of GBS 104 is dependent on the concurrent expression of GBS 80. As discussed further in Example 2, reverse transcriptase PCR analysis of AI-1 shows that all of the AI genes are co-transcribed as an operon. Applicants constructed a series of mutant GBS containing in frame deletions of various AI-1 genes. (A schematic of the GBS mutants is presented in FIG. 7). FACS analysis of the various mutants comparing mean shift values using anti-GBS 80 versus anti-GBS 104 antibodies is presented in FIG. 8. Removal of the GBS 80 operon prevented surface exposure of GBS 104; removal of the GBS 104 operon did not affect surface exposure of GBS 80. While not being limited to a specific theory, it is thought that GBS 80 is involved in the transport or localization of GBS 104 to the surface of the bacteria. The two proteins may be oligomerized or otherwise associated. It is possible that this association involves a conformational change in GBS 104 that facilitates its transition to the surface of the GBS bacteria.
  • Pili structures that comprise GBS 104 appear to be of a lower molecular weight than pili structures lacking GBS 104. FIG. 68 shows that polyclonal anti-GBS 104 antibodies (see lane marked α-104 POLIC.) cross-hybridize with smaller structures than do polyclonal anti-GBS 80 antibodies (see lane marked α-GBS 80 POLIC.).
  • In addition, Applicants have shown that removal of GBS 80 can cause attenuation, further suggesting the protein contributes to virulence. As described in more detail in Example 3, the LD50's for the Δ80 mutant and the Δ80, Δ104 double mutant were reduced by an order of magnitude compared to wildtype and Δ104 mutant.
  • The sortases within the adhesin island also appear to play a role in localization and presentation of the surface proteins. As discussed further in Example 4, FACS analysis of various sortase deletion mutants showed that removal of sortase SAG0648 prevented GBS 104 from reaching the surface and slightly reduced the surface exposure of GBS 80. When sortase SAG0647 and sortase SAG0648 were both knocked out, neither GBS 80 nor GBS 104 were surface exposed. Expression of either sortase alone was sufficient for GBS 80 to arrive at the bacterial surface. Expression of SAG0648, however, was required for GBS 104 surface localization.
  • Accordingly, the compositions of the invention may include two or more AI proteins, wherein the AI proteins are physically or chemically associated. For example, the two AI proteins may form an oligomer. In one embodiment, the associated proteins are two AI surface proteins, such as GBS 80 and GBS 104. The associated proteins may be AI surface proteins from different adhesin islands, including host cell adhesin island proteins if the AI surface proteins are expressed in a recombinant system. For example, the associated proteins may be GBS 80 and GBS 67.
  • Adhesin Island Proteins from Other Gram Positive Bacteria
  • Applicants' identification and analysis of the GBS adhesin islands and the immunological and biological functions of these AI proteins and their pilus structures provides insight into similar structures in other Gram positive bacteria.
  • As discussed above, “Adhesin Island” or “AI” refers to a series of open reading frames within a bacterial genome that encode for a collection of surface proteins and sortases. An Adhesin Island may encode for amino acid sequences comprising at least one surface protein. The Adhesin Island may encode at least one surface protein. Alternatively, an Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, an Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • Gram positive adhesin islands of the invention preferably include a divergently transcribed transcriptional regulator. The transcriptional regulator may regulate the expression of the AI operon.
  • The invention includes a composition comprising one or more Gram positive bacteria AI surface proteins. Such AI surface proteins may be associated in an oligomeric or hyperoligomeric structure.
  • Preferred Gram positive adhesin island proteins for use in the invention may be derived from Staphylococcus (such as S. aureus), Streptococcus (such as S. agalactiae (GBS), S. pyogenes (GAS), S. pneumonaie, S. mutans), Enterococcus (such as E. faecalis and E. faecium), Clostridium (such as C. difficile), Listeria (such as L. monocytogenes) and Corynebacterium (such as C. diphtheria).
  • One or more of the Gram positive AI surface protein sequences typically include an LPXTG motif or other sortase substrate motif. Gram positive AI surface proteins of the invention may affect the ability of the Gram positive bacteria to adhere to and invade epithelial cells. AI surface proteins may also affect the ability of Gram positive bacteria to translocate through an epithelial cell layer. Preferably, one or more AI surface proteins are capable of binding to or otherwise associating with an epithelial cell surface. Gram positive AI surface proteins may also be able to bind to or associate with fibrinogen, fibronectin, or collagen.
  • Gram positive AI sortase proteins are predicted to be involved in the secretion and anchoring of the LPXTG containing surface proteins. A Gram positive bacteria AI may encode for at least one surface exposed protein. The Adhesin Island may encode at least one surface protein. Alternatively, a Gram positive bacteria AI may encode for at least two surface exposed proteins and at least one sortase. Preferably, a Gram positive AI encodes for at least three surface exposed proteins and at least two sortases.
  • Gram positive AI surface proteins may be covalently attached to the bacterial cell wall by membrane-associated transpeptidases, such as an AI sortase. The sortase may function to cleave the surface protein, preferably between the threonine and glycine residues of an LPXTG motif. The sortase may then assist in the formation of an amide link between the threonine carboxyl group and a cell wall precursor such as lipid II. The precursor can then be incorporated into the peptidoglycan via the transglycoslylation and transpeptidation reactions of bacterial wall synthesis. See Comfort et al., Infection & Immunity (2004) 72(5): 2710-2722. Typically, Gram positive bacteria AI surface proteins of the invention will contain an N-terminal leader or secretion signal to facilitate translocation of the surface protein across the bacterial membrane.
  • Gram positive bacteria AI surface proteins of the invention may affect the ability of the Gram positive bacteria to adhere to and invade target host cells, such as epithelial cells. Gram positive bacteria AI surface proteins may also affect the ability of the gram positive bacteria to translocate through an epithelial cell layer. Preferably, one or more of the Gram positive AI surface proteins are capable of binding to or other associating with an epithelial cell surface. Further, one or more Gram positive AI surface proteins may bind to fibrinogen, fibronectin, or collagen protein.
  • In one embodiment, the invention includes a composition comprising oligomeric, pilus-like structures comprising a Gram positive bacteria AI surface protein. The oligomeric, pilus-like structure may comprise numerous units of the AI surface protein. Preferably, the oligomeric, pilus-like structures comprise two or more AI surface proteins. Still more preferably, the oligomeric, pilus-like structure comprises a hyper-oligomeric pilus-like structure comprising at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 150, 200 or more) oligomeric subunits, wherein each subunit comprises an AI surface protein or a fragment thereof. The oligomeric subunits may be covalently associated via a conserved lysine within a pilin motif. The oligomeric subunits may be covalently associated via an LPXTG motif, preferably, via the threonine amino acid residue.
  • Gram positive bacteria AI surface proteins or fragments thereof to be incorporated into the oligomeric, pilus-like structures of the invention will preferably include one or both of a pilin motif comprising a conserved lysine residue and an E box motif comprising a conserved glutamic acid residue.
  • The oligomeric, pilus like structures may be used alone or in the combinations of the invention. In one embodiment, the invention comprises a Gram positive bacteria Adhesin Island in oligomeric form, preferably in a hyperoligomeric form.
  • The oligomeric, pilus-like structures of the invention may be combined with one or more additional Gram positive AI proteins (from the same or a different Gram positive species or genus). In one embodiment, the oligomeric, pilus-like structures comprise one or more Gram positive bacteria AI surface proteins in combination with a second Gram positive bacteria protein. The second Gram positive bacteria protein may be a known antigen, and need not normally be associated with an AI protein.
  • The oligomeric, pilus-like structures may be isolated or purified from bacterial cultures overexpressing a Gram positive bacteria AI surface protein. The invention therefore includes a method for manufacturing an oligomeric Adhesin Island surface antigen comprising culturing a Gram positive bacteria adapted for increased AI protein expression and isolation of the expressed oligomeric Adhesin Island protein from the Gram positive bacteria. The AI protein may be collected from secretions into the supernatant or it may be purified from the bacterial surface. The method may further comprise purification of the expressed Adhesin Island protein. Preferably, the Adhesin Island protein is in a hyperoligomeric form.
  • Gram positive bacteria are preferably adapted to increase AI protein expression by at least two (e.g., 2, 3, 4, 5, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 125, 150 or 200) times wild type expression levels.
  • Gram positive bacteria may be adapted to increase AI protein expression by means known in the art, including methods of increasing gene dosage and methods of gene upregulation. Such means include, for example, transformation of the Gram positive bacteria with a plasmid encoding the AI protein. The plasmid may include a strong promoter or it may include multiple copies of the sequence encoding the AI protein. Optionally, the sequence encoding the AI protein within the Gram positive bacterial genome may be deleted. Alternatively, or in addition, the promoter regulating the Gram positive Adhesin Island may be modified to increase expression.
  • The invention further includes Gram positive bacteria which have been adapted to produce increased levels of AI surface protein. In particular, the invention includes Gram positive bacteria which have been adapted to produce oligomeric or hyperoligomeric AI surface protein. In one embodiment, the Gram positive bacteria of the invention are inactivated or attenuated to permit in vivo delivery of the whole bacteria, with the AI surface protein exposed on its surface.
  • The invention further includes Gram positive bacteria which have been adapted to have increased levels of expressed AI protein incorporated in pili on their surface. The Gram positive bacteria may be adapted to have increased exposure of oligomeric or hyperoligomeric AI proteins on its surface by increasing expression levels of a signal peptidase polypeptide. Increased levels of a local signal peptidase expression in Gram positive bacteria (such us LepA in GAS) are expected to result in increased exposure of pili proteins on the surface of Gram positive bacteria. Increased expression of a leader peptidase in Gram positive may be achieved by any means known in the art, such as increasing gene dosage and methods of gene upregulation. The Gram positive bacteria adapted to have increased levels of leader peptidase may additionally be adapted to express increased levels of at least one pili protein.
  • Alternatively, the AI proteins of the invention may be expressed on the surface of a non-pathogenic Gram positive bacteria, such as Streptococus gordonii (See, e.g., Byrd et al., “Biological consequences of antigen and cytokine co-expression by recombinant Streptococcus gordonii vaccine vectors”, Vaccine (2002) 20:2197-2205) or Lactococcus lactis (See, e.g., Mannam et al., “Mucosal VaccineMade from Live, Recombinant Lactococcus lactis Protects Mice against Pharangeal Infection with Streptococcus pyogenes” Infection and Immunity (2004) 72(6):3444-3450). It has already been demonstrated, above, that L. lactis expresses GBS and GAS AI polypeptides in oligomeric form and on its surface.
  • Alternatively, the oligomeric, pilus-like structures may be produced recombinantly. If produced in a recombinant host cell system, the Gram positive bacteria AI surface protein will preferably be expressed in coordination with the expression of one or more of the AI sortases of the invention. Such AI sortases will facilitate oligomeric or hyperoligomeric formation of the AI surface protein subunits.
  • Gram positive AI Sortases of the invention will typically have a signal peptide sequence within the first 70 amino acid residues. They may also include a transmembrane sequence within 50 amino acid residues of the C terminus. The sortases may also include at least one basic amino acid residue within the last 8 amino acids. Preferably, the sortases have one or more active site residues, such as a catalytic cysteine and histidine.
  • Adhesin island surface proteins from two or more Gram positive bacterial genus or species may be combined to provide an immunogenic composition for prophylactic or therapeutic treatment of disease or infection of two more Gram positive bacterial genus or species. Optionally, the adhesin island surface proteins may be associated together in an oligomeric or hyperoligomeric structure.
  • In one embodiment, the invention comprises an adhesin island surface proteins from two or more Streptococcus species. For example, the invention includes a composition comprising a GBS AI surface protein and a GAS adhesin island surface protein. As another example, the invention includes a composition comprising a GAS adhesin island surface protein and a S. pneumoniae adhesin island surface protein.
  • In one embodiment, the invention comprises an adhesin island surface protein from two or more Gram positive bacterial genus. For example, the invention includes a composition comprising a Streptococcus adhesin island protein and a Corynebacterium adhesin island protein.
  • Examples of AI sequences in several Gram positive bacteria are discussed further below.
  • Streptococcus pyogenes (GAS)
  • As discussed above, Applicants have identified at least four different GAS Adhesin Islands. These adhesion islands are thought to encode surface proteins which are important in the bacteria's virulence, and Applicants have obtained the first electron micrographs revealing the presence of these adhesin island proteins in hyperoligomeric pilus structures on the surface of Group A Streptococcus.
  • Group A Streptococcus is a human specific pathogen which causes a wide variety of diseases ranging from pharyngitis and impetigo through life threatening invasive disease and necrotizing fasciitis. In addition, post-streptococcal autoimmune responses are still a major cause of cardiac pathology in children.
  • Group A Streptococcal infection of its human host can generally occur in three phases. The first phase involves attachment and/or invasion of the bacteria into host tissue and multiplication of the bacteria within the extracellular spaces. Generally this attachment phase begins in the throat or the skin. The deeper the tissue level infected, the more severe the damage that can be caused. In the second stage of infection, the bacteria secrete a soluble toxin that diffuses into the surrounding tissue or even systemically through the vasculature. This toxin binds to susceptible host cell receptors and triggers innappropropriate immune responses by these host cells, resulting in pathology. Because the toxin can diffuse throughout the host, the necrosis directly caused by the GAS toxins may be physically located in sites distant from the bacterial infection. The final phase of GAS infection can occur long after the original bacteria have been cleared from the host system. At this stage, the host's previous immune response to the GAS bacteria due to cross reactivity between epitopes of a GAS surface protein, M, and host tissues, such as the heart. A general review of GAS infection can be found in Principles of Bacterial Pathogeneis, Groisman ed., Chapter 15 (2001).
  • In order to prevent the pathogenic effects associated with the later stages of GAS infection, an effective vaccine against GAS will preferably facilitate host elimination of the bacteria during the initial attachment and invasion stage.
  • Isolates of Group A Streptococcus are historically classified according to the M surface protein described above. The M protein is surface exposed trypsin-sensitive protein generally comprising two polypeptide chains complexed in an alpha helical formation. The carboxyl terminus is anchored in the cytoplasmic membrane and is highly conserved among all group A streptococci. The amino terminus, which extends through the cell wall to the cell surface, is responsible for the antigenic variability observed among the 80 or more serotypes of M proteins.
  • A second layer of classification is based on a variable, trypsin-resistant surface antigen, commonly referred to as the T-antigen. Decades of epidemiology based on M and T serological typing have been central to studies on the biological diversity and disease causing potential of Group A Streptococci. While the M-protein component and its inherent variability have been extensively characterized, even after five decades of study, there is still very little known about the structure and variability of T-antigens. Antisera to define T types are commercially available from several sources, including Sevapharma (http://www.sevapharma.cz/en).
  • The gene coding for one form of T-antigen, T-type 6, from an M6 strain of GAS (D741) has been cloned and characterized and maps to an approximately 11 kb highly variable pathogenicity island. Schneewind et al., J. Bacteriol. (1990) 172(6):3310-3317. This island is known as the Fibronectin-binding, Collagen-binding T-antigen (FCT) region because it contains, in addition to the T6 coding gene (tee6), members of a family of genes coding for Extra Cellular Matrix (ECM) binding proteins. Bessen et al., Infection & Immunity (2002) 70(3):1159-1167. Several of the protein products of this gene family have been shown to directly bind either fibronectin and/or collagen. See Hanski et al., Infection & Immunity (1992) 60(12):5119-5125; Talay et al., Infection & Immunity (1992(60(9):3837-3844; Jaffe et al. (1996) 21(2):373-384; Rocha et al., Adv Exp Med Biol. (1997) 418:737-739; Kreikemeyer et al., J Biol Chem (2004) 279(16):15850-15859; Podbielski et al., Mol. Microbiol. (1999) 31(4):1051-64; and Kreikemeyer et al., Int. J. Med Microbiol (2004) 294(2-3):177-88. In some cases direct evidence for a role of these proteins in adhesion and invasion has been obtained.
  • Applicants raised antiserum against a recombinant product of the tee6 gene and used it to explore the expression of T6 in M6 strain ISS3650. In immunoblot of mutanolysin extracts of this strain, the antiserum recognized, in addition to a band corresponding to the predicted molecular mass of the tee6 gene product, very high molecular weight ladders ranging in mobility from about 100 kDa to beyond the resolution of the 3-8% gradient gels used. See FIG. 163A, last lane labeled “M6 Tee6.”
  • This pattern of high molecular weight products is similar to that observed in immunoblots of the protein components of the pili identified in Streptococcus agalactiae (described above) and previously in Corynebacterium diphtheriae. Electron microscropy of strain M6 ISS3650 with antisera specific for the product of tee6 revealed abundant surface staining and long pilus like structures extending up to 700 nanometers from the bacterial surface, revealing that the T6 protein, one of the antigens recognized in the original Lancefield serotyping system, is located within a GAS Adhesin Island (GAS AI-1) and forms long covalently linked pilus structures. See FIG. 163I.
  • In addition to the tee6 gene, the FCT region in M6_ISS3650 (GAS AI-1) contains two other genes (prtF1 and cpa) predicted to code for surface exposed proteins; these proteins are characterized as containing the cell wall attachment motif LPXTG. Western blot analysis using antiserum specific for PrtF1 detected a single molecular species with electrophoretic mobility corresponding to the predicted molecular mass of the protein and one smaller band of unknown origin. Western blot analysis using antisera specific for Cpa recognized a high molecular weight covalently linked ladder (FIG. 163A, second lane). Immunogold labelling of Cpa with specific antiserum followed by transmission electron microscopy detected an abundance of Cpa at the cell surface and only occasional structures extending from the cell surface (FIG. 163J).
  • Four classes of FCT region can be discerned by the types and order of the genes contained within the region. The FCT region of strains of types M3, M5, M18 and M49 have a similar organization whereas those of M6, M1 and M12 differ. See FIG. 164. As discussed below, these four FCT regions correlate to four GAS Adhesin Island types (AI-1, AI-2, AI-3 and AI-4).
  • Applicants discovery of genes coding for pili in the FCT region of strain M6_ISS3650 prompted them to examine the predicted surface exposed proteins in the variant FCT regions of three other GAS strains of having different M-type (M1_SF370, M5_ISS4883 and M1220010296) representing the other three FCT variants. Each gene present in the FCT region of each bacteria was cloned and expressed. Antisera specific for each recombinant protein was then used to probe mutanolysin extracts of the respective strains (6). In M1 strain SF370, there are three predicted surface proteins (Cpa (also referred to as M1 126 and GAS 15), M1128 (a fimbrial protein also referred to as Spy0128 and GAS 16), and M1130 (also referred to as Spy0130 and GAS 18)) (GAS AI-2). Antisera specific for each surface protein reacted with a ladder of high molecular weight material (FIG. 163B). Immunogold staining of M1 strain SF370 with antiserum specific for M1 128 revealed pili structures similar to those seen when M6 strain ISS3650 was immunogold stained with antiserum specific for tee6 (See FIG. 1163K). Antisera specific for surface proteins Cpa and M1 130 revealed abundant surface staining and occasional structures extending from the surface of M1 strain SF370 bacteria (FIG. 163S).
  • The M1 128 protein appears to be necessary for polymerization of Cpa and M1 130 proteins. If the M1 128 gene in M1_SF370 was deleted, Western blot analysis using antibodies that hybridize to Cpa and M1 130 no longer detected high molecular weight ladders comprising the Cpa and M1 130 proteins (FIG. 163 E). See also FIGS. 177 A-C which provide the results of Western blot analysis of the M1128 (Δ128) deleted bacteria using anti-M1 130 antiserum (FIG. 177 A), anti-M1 128 antiserum (FIG. 177 B), and anti-M1 126 antiserum (FIG. 177 C). High molecular weight ladders, indicative of pilus formation on the surface of M1 strain SF370, could not be detected by any of the three antisera in Δ128 bacteria. If the Δ128 bacteria were transformed with a plasmid containing the gene for M1 128, Western blot analysis using antisera specific for Cpa and M1 130 again detected high molecular weight ladders (FIG. 163 H).
  • In agreement with the Western blot analysis, immunoelectron microscopy failed to detect pilus assembly on the Δ128 strain SF370 bacteria using M1 128 antisera (FIG. 178 B). Although Δ128 SF370 bacteria were unable to form pili, M1126 (cpa) and M1 130, which contain sortase substrate motifs, were present on the bacteria's surface. FACS analysis of the M1 128 deleted (Δ128) strain SF370 bacteria also detected both M1 126 and M1 130 on the surface of the Δ128 strain SF370 bacteria. See FIG. 179 D and F, which show a shift in fluorescence when antibodies immunoreactive to M1 126 and M1 130 are used on Δ128 bacteria. As expected, virtually no shift in fluorescence is observed when antibodies immunoreactive to M1 128 are used with the Δ128 bacteria (FIG. 179 E).
  • By contrast, deletion of the M1 130 gene did not effect polymerization of M1128 (FIG. 163 F). See also FIGS. 177 A-C, which provide Western blot analysis results of the M1 130 deleted (Δ130) strain SF370 bacteria using anti-M1130 (FIG. 177 A), anti-M1128 (FIG. 177 B), and anti-M1 126 antiserum (FIG. 177 C). The anti-M1128 and anti-M1126 antiserum both detected the presence of high molecular weight ladders in the A130 strain SF370 bacteria, indicating that the A130 bacteria form pili that comprise M1 126 and M1 128 polypeptides in the absence of M1 130. As expected, the Western blot probed with antiserum immunoreactive with M1 130 did not detect any proteins for the Δ130 bacteria (FIG. 177A).
  • Hence, the composition of the pili in GAS resembles that previously described for both C. diphtheria (7, 8) and S. agalactiae (described above) (9) in that each pilus is formed by a backbone component which abundantly stains the pili in EM and is essential for the incorporation of the other components.
  • Also similar to C. diphtheria, elimination of the srtC1 gene from the FCT region of M1_SF370 abolished polymerization of all three proteins and assembly of pili (FIG. 163 G). See also FIGS. 177 A-C, which provide Western blot analysis of the SrtC1 deleted (ΔsrtC1) strain SF370 bacteria using anti-M1130 (FIG. 177 A), anti-M1128 (FIG. 177 B), and anti-M1 126 antiserum (FIG. 177 C). None of the three antisera immunoreacted with high molecular weight structures (pili) in the ΔSrtC1 bacteria. Confirming that deletion of the SrtC1 gene abrogates pilus assembly in strain SF370, immunoelectron microscopy using antisera against M1 128 failed to detect pilus formation on the bacteria surface. See FIG. 178 C. Although no assembled pili were detected on ΔSrtC1 SF370, M1 128 proteins could be detected on the surface of SF370. Thus, it appeared that SrtC1 deletion prevented pilus assembly on the surface of the SF370 bacteria, but not anchoring of the proteins that comprise pili to the bacterial cell wall. FACS analysis of the ΔSrtC1 strain SF370 confirmed that deletion of SrtC1 does not eliminate cell surface expression of M1 126, M1 128 or M1 130. See FIG. 179 G-I, which show a shift in fluorescence when antibodies immunoreactive to M1126 (FIG. 179 G), M1128 (FIG. 179 H), and M1130 (FIG. 179 I) are used to detect cell surface protein expression on ΔSrtC1 bacteria. Thus, SrtC1 deletion prevents pilus formation, but not surface anchoring of proteins involved in pilus formation on the surface of bacteria. Another sortase is possibly involved in anchoring of the proteins to the bacteria surface. Pilus polymerization in C. diphtheriae is also dependent on particular sortase enzyme whose gene resides at the same genetic locus as the pilus components (7, 8).
  • The LepA signal peptidase, Spy0127, also appears to be essential for pilus assembly in strain SF370. LepA deletion mutants (ΔLepA) of strain SF370 fail to assemble pili on the cell surface. Not only are the ΔLepA mutants unable to assemble pili, they are also deficient at cell surface M1 expression. See FIG. 180, which provides a FACS analysis of the wildtype (A) and ΔLepA mutant (B) SF370 bacteria using M1 antisera. No shift in fluorescence is observed for the ΔLepA mutant bacteria in the presence of M1 immune serum. It is possible that these deletion mutants of LepA will be useful for detecting non-M, non-pili, surface exposed antigens on the surface of GAS, or any Gram positive bacteria. These antigens may also be useful in immunogenic compositions.
  • Pili were also observed in M5 strain ISS4882 and M12 strain 20010296. The M5 strain ISS4882 contains genes for four predicted surface exposed proteins (GAS AI-3). Antisera against three of the four products of the FCT region (GAS AI-3) of M5_ISS4883 (Cpa, M5_orf80, M5_orf82) stained high molecular weight ladders in Western blot analysis (FIG. 163 C). Long pili were visible when antisera against M5_orf80 was used in immunogold staining followed by electron microscopy (FIG. 163L).
  • The M12 strain 20010296 contains genes for five predicted surface exposed proteins. (GAS AI-4) Antisera against three of the five products of the FCT region (GAS AI-4) of M1220010296 (Cpa, EftLSL.A, Orf2) stained high molecular weight ladders in Westen blot analysis (FIG. 163 D). Long pili were visible when antisera against EftLSL.A were used (FIG. 163M).
  • The major pilus forming proteins identified in the four strains studied by applicants (T6, M1 128, M5_orf80 and EftLSL.A) share between 23% and 65% amino acid identity in any pairwise comparison, indicating that each pilus may represent a different Lancefield T-antigen. Each pilus is part of a trypsin resistant structure on the GAS bacteria surface, as is the case for the Lancefield T antigens. See FIG. 165, which provides a FACS analysis of bacteria harboring each of the FCT types that had or had not been treated with trypsin (6). Following treatment, surface expression of the pilus proteins was assayed by indirect immunofluorescence and flow cytometry using antibodies specific for the pilus proteins, the bacteria's respective M proteins, or surface proteins not associated with the pili (FIG. 165). Staining the cells with sera specific for proteins associated with the pili was not effected by trypsin treatment, whereas trypsin treatment substantially reduced detection of M-proteins or surface proteins not associated with pili.
  • The pili structures identified on the surface of the GAS bacteria were confirmed to be Lancefield T antigens when commercially available T-serotyping sera detected the pili on the surface of bacteria. Western blot analysis was initially performed to determine if polyvalent serum pools (designated T, U, W, X, and Y) could detect recombinant proteins for each of the major pilis components (T6, M1 128, M5_orf80 and EftLSL.A) identified in the strains of bacteria discussed above. Pool U, which contains the T6 serum, recognized the T6 protein specifically (a surface exposed pilus protein from GAS AI-1) (FIG. 166 B). Pool T specifically recognized M1128 (a surface exposed pilus protein from GAS AI-2) (FIG. 166 A). Pool W recognized both M5_orf80 and EftLSL.A (FIG. 166 C). Using monovalent sera representative of each of the components of each polyvalent pool, applicants confirmed the specificity of the T6 antigen (corresponding to a surface exposed pilus protein from GAS AI-1) (FIG. 166 E) and identified M1 28 as antigen T1 (corresponding to a surface exposed pilus protein from GAS AI-2) (FIG. 166 D), EftLSL.A as antigen T12 (corresponding to a surface exposed pilus protein from GAS AI-4) (FIG. 166 G) and M5_orf80 as a common antigen recognized by the related sera T5, T27 and T44 (corresponding to a surface exposed pilus protein from GAS AI-3).
  • Confirming applicants observations, discussed above, that deleting the M1 128 gene from M1_SF370 abolishes pilus formation, the pool T sera stained whole M1_SF370 bacteria (FIG. 166 H) but failed to stain M1_SF370 bacteria lacking the M1 128 gene (FIG. 166 I).
  • As discussed above, Applicants have identified at least four different Group A Streptococcus Adhesin Islands. While these GAS AI sequences can be identified in numerous M types, Applicants have surprisingly discovered a correlation between the four main pilus subunits from the four different GAS AI types and specific T classifications. While other trypsin-resistant surface exposed proteins are likely also implicated in the T classification designations, the discovery of the role of the GAS adhesin islands (and the associated hyper-oligomeric pilus like structures) in T classification and GAS serotype variance has important implications for prevention and treatment of GAS infections. Applicants have identified protein components within each of the GAS adhesin islands which are associated with the pilus formation. These proteins are believed to be involved in the bacteria's initial adherence mechanisms. Immunological recognition of these proteins may allow the host immune response to slow or prevent the bacteria's transition into the more pathogenic later stages of infection. In addition, the GAS pili may be involved in formation of biofilms. Applicants have discovered that the GBS pili structures appear to be implicated in the formation of biofilms (populations of bacteria growing on a surface, often enclosed in an exopolysaccharide matrix). Biofilms are generally associated with bacterial resistance, as antibiotic treatments and host immune response are frequently unable to erradicate all of the bacteria components of the biofilm. Direction of a host immune response against surface proteins exposed during the first steps of bacterial attachment (i.e., before complete biofilm formation) is preferable.
  • The invention therefore provides for improved immunogenic compositions against GAS infection which may target GAS bacteria during their initial attachment efforts to the host epithelial cells and may provide protection against a wide range of GAS serotypes. The immunogenic compositions of the invention include GAS AI surface proteins which may be formulated in an oligomeric, or hyperoligomeric (pilus) form. The invention also includes combinations of GAS AI surface proteins. Combinations of GAS AI surface proteins may be selected from the same adhesin island or they may be selected from different GAS adhesin islands.
  • The invention comprises compositions comprising a first GAS AI protein and a second GAS AI protein wherein the first and second GAS AI proteins are derived from different GAS adhesin islands. For example, the invention includes a composition comprising at least two GAS AI proteins wherein the GAS AI proteins are encoded by the adhesin islands selected from the group consisting of GAS AI-1 and AI-2; GAS AI-1 and GAS AI-3; GAS AI-1 and GAS AI-4; GAS AI-2 and GAS AI-3; GAS AI-2 and GAS AI-4; and GAS AI-3 and GAS AI-4. Preferably the two GAS AI proteins are derived from different T-types.
  • A schematic arrangement of GAS Adhesin Island sequences is set forth in FIG. 162. In all strains, the AI region is flanked by the highly conserved open reading frames M1 123 and M1-136. Between three and five genes in each locus code for surface proteins containing LPXTG motifs. These surface proteins also all belong to the family of genes coding for ECM binding adhesins.
  • Adhesin island sequences can be identified in numerous M types of Group A Streptococcus. Examples of AI sequences within M1, M6, M3, M5, M12, M18, and M49 serotypes are discussed below.
  • GAS Adhesin Islands generally include a series of open reading frames within a GAS genome that encode for a collection of surface proteins and sortases. A GAS Adhesin Island may encode for amino acid sequences comprising at least one surface protein. Alternatively, a GAS Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, a GAS Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more GAS AI surface proteins may participate in the formation of a pilus structure on the surface of the Gram positive bacteria.
  • GAS Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator. The transcriptional regulator may regulate the expression of the GAS AI operon. Examples of transcriptional regulators found in GAS AI sequences include RofA and Nra.
  • The GAS AI surface proteins may bind or otherwise adhere to fibrinogen, fibronectin, or collagen. One or more of the GAS AI surface proteins may comprise a fimbrial structural subunit.
  • One or more of the GAS AI surface proteins may include an LPXTG motif or other sortase substrate motif. The LPXTG motif may be followed by a hydrophobic region and a charged C terminus, which are thought to retard the protein in the cell membrane to facilitate recognition by the membrane-localized sortase. See Barnett, et al., J. Bacteriology (2004) 186 (17): 5865-5875.
  • GAS AI sequences may be generally categorized as Type 1, Type 2, Type 3, or Type 4, depending on the number and type of sortase sequences within the island and the percentage identity of other proteins (with the exception of RofA and cpa) within the island. FIG. 167 provides a chart indicating the number and type of sortase sequences identified within the adhesin islands of various strains and serotypes of GAS. As can be seen in this figure, all GAS strains and serotypes thus far characterized as an AI-1 have a SrtB type sortase, all GAS strains and serotypes thus far characterized as an AI-2 have SrtB and SrtC1 type sortases, all GAS strains and serotypes thus far characterized as an AI-3 have a SrtC2 type sortase, and all GAS strains and serotypes thus far characterized as an AI-4 have SrtB and SrtC2 type sortases. A comparison of the percentage identity of sequences within the adhesin islands was presented in Table 45, see above.
  • (1) Adhesin Island Sequence within M6: GAS Adhesin Island 1 (“GAS AI-1”)
  • A GAS Adhesin Island within M6 serotype (MGAS10394) is outlined in Table 4 below. This GAS adhesin island 1 (“GAS AI-1”) comprises surface proteins, a srtB sortase and a rofA divergently transcribed transcriptional regulator.
  • GAS AI-1 surface proteins include Spy0157 (a fibronectin binding protein), Spy0159 (a collagen adhesion protein) and Spy0160 (a fimbrial structural subunit). Preferably, each of these GAS AI-1 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122) or LPXSG (SEQ ID NO: 134) (conservative replacement of threonine with serine).
  • GAS AI-1 includes a srtB type sortase. GAS srtB sortases may preferably anchor surface proteins with an LPSTG motif (SEQ ID NO: 166), particularly where the motif is followed by a serine.
    TABLE 4
    GAS AI-1 sequences from M6 isolate (MGAS10394)
    Sortase substrate
    AI-1 sequence sequence or sortase
    identifier type functional description
    M6_Spy0156 Transcriptional regulator (rofA)
    M6_Spy0157 LPXTG Fibronectin-binding protein
    M6_Spy0158 Reverse transcriptase
    M6_Spy0159 LPXSG Collagen adhesion protein
    M6_Spy0160 LPXTG Fimbrial structural subunit
    M6_Spy0161 srtB Sortase
  • M6_Spy0160 appears to be present on the surface of GAS as part of oligomeric (pilus) structures. FIGS. 127-132 present electron micrographs of GAS serotype M6 strain 3650 immunogold stained for M6 Spy0160 using anti-M6 Spy0160 antiserum. Oligomeric or hyperoligmeric structures labelled with gold particles can be seen extending from the surface of the GAS in each of these figures, indicating the presence of multiple M6_Spy0160 polypeptides in the oligomeric or hyperoligomeric structures. FIG. 176 A-F present electron micrographs of GAS M6 strain 2724 immunogold stained for M6_Spy0160 using anti-M6_Spy0160 antiserum (FIGS. 176 A-E) or immunogold stained for M6_Spy0159 using anti-M6_Spy0159 antiserum (FIG. 176 F). Oligomeric or hyperoligomeric structures labelled with gold particles can again be seen extending from the surface of the M6 strain 2724 GAS bacteria immunogold stained for M6_Spy0160. M6_Spy0159 is also detected on the surface of the M6 strain 2724 GAS.
  • FACS analysis has confirmed that the GAS AI-1 surface proteins spyM6 0159 and spyM6 0160 are indeed expressed on the surface of GAS. FIG. 73 provides the results of FACS analysis for surface expression of spyM6 0159 on each of GAS serotypes M6 2724, M6 3650, and M6 2894. A shift in fluorescence is observed for each GAS serotype when anti-spyM60159 antiserum is present, demonstrating cell surface expression. Table 18, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-spyM60159 antiserum, and the difference in fluorescence value between the pre-immune and anti-spyM60159 antiserum.
    TABLE 18
    Summary of FACS values for surface expression of spyM6_0159
    2724 3650 2894
    Pre- Anti- Pre- Anti- Pre- Anti-
    immune spyM6_0159 Change immune spyM6_0159 Change immune spyM6_0159 Change
    134.84 427.48 293 149.68 712.62 563 193.86 597.8 404
  • FIG. 74 provides the results of FACS analysis for surface expression of spyM6 0160 on each of GAS serotypes M6 2724, M6 3650, and M6 2894. In the presence of of anti-spyM60160 antiserum, a shift in fluorescence is observed for each GAS serotype, which demonstrates its cell surface expression. Table 19, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-spyM60160 antiserum, and the change in fluorescence value between the pre-immune and anti-spyM60160 antiserum.
    TABLE 19
    Summary of FACS values for surface expression of spyM6_0160
    2724 3650 2894
    Pre- Anti- Pre- Anti- Pre- Anti-
    immune spyM6_0160 change immune spyM6_0160 change immune spyM6_0160 change
    117.12 443.24 326 128.57 776.39 648 125.87 621.17 495
  • Surface expression of M6_Spy0159 and M6_Spy0160 on M6 serotype GAS has also been confirmed by Western blot analysis. FIG. 98 shows that while pre-immune sera (P α-0159) does not detect expression of M6_Spy0159 in GAS serotype M6, anti-M6_Spy0159 immune sera (I α-0159) is able to detect M6_Spy0159 protein in both total GAS M6 extracts (M6 tot) and GAS M6 fractions enriched for cell surface proteins (M6 surf prot). The M6_Spy0159 proteins detected in the total GAS M6 extracts or the GAS M6 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that M6_Spy0159 may be in an oligomeric (pilus) form.
  • FIG. 112 shows that while preimmune sera (Preimmune Anti 106) does not detect expression of M6_Spy0160 in GAS serotype M6 strain 2724, anti-M6_Spy0160 immune sera (Anti 160) does in both total GAS M6 strain 2724 extracts (M6 2724 tot) and GAS M6 strain 2724 fractions enriched for surface proteins. The M6_Spy0160 proteins detected in the total GAS M6 strain 2724 extracts or the GAS M6 strain 2724 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that M6_Spy0160 may be in an oligomeric (pilus) form.
  • FIGS. 110 and 111 both further verify the presence of M6_Spy0159 and M6_Spy0160 in higher molecular weight structures on the surface of GAS. FIG. 110 provides a Western blot performed to detect M6_Spy0159 and M6_Spy0160 in GAS M6 strain 2724 extracts enriched for surface proteins. Antiserum raised against either M6_Spy0159 (Anti-159) or M6_Spy0160 (Anti-160) cross-hybridizes with high molecular weight structures (pili) in these extracts. FIG. 111 provides a similar Western blot that verifies the presence of M6_Spy0159 and M6_Spy0160 in high molecular weight structures in GAS M6 strain 3650 extracts enriched for surface proteins.
  • SpyM60157 (a fibronectin-binding protein) may also be expressed on the surface of GAS serotype M6 bacteria. FIG. 174 shows the results of FACS analysis for surface expression of spyM60157 on M6 strain 3650. A slight shift in fluorescence is observed, which demonstrates that some spyM60157 may be expressed on the GAS cell surface.
  • Adhesin Island Sequence within M6: GAS Adhesin Island 2 (“GAS AI-2”)
  • A GAS Adhesin Island within M1 serotype (SF370) is outlined in Table 5 below. This GAS adhesin island 2 (“GAS AI-2”) comprises surface proteins, a SrtB sortase, a SrtC1 sortase and a RofA divergently transcribed transcriptional regulator.
  • GAS AI-2 surface proteins include GAS 15 (Cpa), Spy0128 (thought to be a fimbrial protein) and Spy0130 (a hypothetical protein). Preferably, each of these GAS AI-2 surface proteins includes an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VVXTG (SEQ ID NO: 135), or EVXTG (SEQ ID NO: 136).
  • GAS AI-2 includes a srtB type sortase and a srtC1 sortase. As discussed above, GAS SrtB sortases may preferably anchor surface proteins with an LPSTG (SEQ ID NO: 166) motif, particularly where the motif is followed by a serine. GAS SrtC1 sortase may preferentially anchor surface proteins with a V(P/V)PTG (SEQ ID NO: 167) motif. GAS SrtC1 may be differentially regulated by RofA.
  • GAS AI-2 may also include a LepA putative signal peptidase I protein.
    TABLE 5
    GAS AI-2 sequence from M1 isolate (SF370)
    Sortase substrate
    AI-2 sequence sequence or
    identifier sortase type functional description
    SPy0124 rofA regulatory protein
    GAS15(not VVXTG cpa
    annotated
    in SF370)
    SPy0127 LepA putative signal peptidase I
    SPy0128 (GAS16) EVXTG hypothetical protein (fimbrial)
    SPy0129 (GAS17) srtC1 sortase
    SPy0130 (GAS18) LPXTG hypothetical protein
    SPy0131 conserved hypothetical protein
    SPy0133 conserved hypothetical protein
    SPy0135 (GAS20) srtB sortase (putative fimbrial-
    associated protein)
  • GAS 15, GAS 16, and GAS 18 appear to be present on the surface of GAS as part of oligomeric (pilus) structures. FIGS. 113-115 present electron micrographs of GAS serotype M1 strain SF370 immunogold stained for GAS 15 using anti-GAS 15 antiserum. FIGS. 116-121 provide electron micrographs of GAS serotype M1 strain SF370 immunogold stained for GAS 16 using anti-GAS 16 antiserum. FIGS. 122-125 present electron micrograph of GAS serotype M1 strain SF370 immunogold stained for GAS 18 using anti-GAS 18 antiserum. Oligomers of these proteins can be seen on the surface of SF370 bacteria in the immuno-gold stained micrographs.
  • FIG. 126 reveals a hyperoligomer on the surface of a GAS serotype M1 strain SF370 bacterium immunogold stained for GAS 18. This long hyperoliogmeric structure comprising GAS 18 stretches far out into the supernatant from the surface of the bacteria.
  • FACS analysis has confirmed that the GAS AI-2 surface proteins GAS 15, GAS 16, and GAS 18 are expressed on the surface of GAS. FIG. 75 provides the results of FACS analysis for surface expression of GAS 15 on each of GAS serotypes M1 2719, M1 2580, M1 3280, M1 SF370, M1 2913, and M1 3348. A shift in fluorescence is observed for each GAS serotype when anti-GAS 15 antiserum is present, demonstrating cell surface expression. Table 20, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-GAS 15 antiserum, and the difference in fluorescence value between the pre-immune and anti-GAS 15 antiserum.
    TABLE 20
    Summary of FACS values for surface expression of GAS 15
    Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS
    immune 15 Change immune 15 Change immune 15 Change
    2719 2580 3280
    159.46 712.71 553 123.9  682.84 559 217.02 639.69 423
    SF370 2913 3348
    201.93 722.68 521 121.41 600.45 479 152.09 446.41 294
  • FIGS. 76 and 79 provide the results of FACS analysis for surface expression of GAS 16 on each of GAS serotypes M1 2719, M1 2580, M1 3280, M1 SF370, M1 2913, and M1 3348. The FACS data in FIG. 76 was obtained using antisera was raised against full length GAS 16. In the presence of this anti-GAS 16 antiserum, a shift in fluorescence is observed for each GAS serotype, demonstrating its cell surface expression. Table 21, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-GAS 16 antiserum, and the change in fluorescence value between the pre-immune and anti-GAS 16 antiserum.
    TABLE 21
    Summary of FACS values for surface expression of GAS 16
    Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS
    immune 16 Change immune 16 Change immune 16 Change
    2719 2580 3280
    233.27 690.09 457 133.82 732.29 598 264.47 649.43 385
    SF370 2913 3348
    237.2 727.46 490 138.52 588.04 450 180.56 420.93 240
  • The FACS data in FIG. 79 was obtained using antisera was raised against a truncated GAS 16, which is encoded by SEQ ID NO: 179, shown below.
    SEQ ID NO: 179:
    GCTACAACAGTTCACGGGGAGACTGTTGTAAACGGAGCCAAACTAACAGT
    TACAAAAAACCTTGATTTAGTTAATAGCAATGCATTAATTCCAAATACAG
    ATTTTACATTTAAAATCGAACCTGATACTACTGTCAACGAAGACGGAAAT
    AAGTTTAAAGGTGTAGCTTTGAACACACCGATGACTAAAGTCACTTACAC
    CAATTCAGATAAAGGTGGATCAAATACGAAAACTGCAGAATTTGATTTTT
    CAGAAGTTACTTTTGAAAAACCAGGTGTTTATTATTACAAAGTAACTGAG
    GAGAAGATAGATAAAGTTCCTGGTGTTTCTTATGATACAACATCTTACAC
    TGTTCAAGTTCATGTCTTGTGGAATGAAGAGCAACAAAAACCAGTAGCTA
    CTTATATTGTTGGTTATAAAGAAGGTAGTAAGGTGCCAATTCAGTTCAAA
    AATAGCTTAGATTCTACTACATTAACGGTGAAGAAAAAAGTTTCAGGTAC
    CGGTGGAGATCGCTCTAAAGATTTTAATTTTGGTCTGACTTTAAAAGCAA
    ATCAGTATTATAAGGCGTCAGAAAAAGTCATGATTGAGAAGACAACTAAA
    GGTGGTCAAGCTCCTGTTCAAACAGAGGCTAGTATAGATCAACTCTATCA
    TTTTACCTTGAAAGATGGTGAATCAATCAAAGTCACAAATCTTCCAGTAG
    GTGTGGATTATGTTGTCACTGAAGACGATTACAAATCAGAAAAATATACA
    ACCAACGTGGAAGTTAGTCCTCAAGATGGAGCTGTAAAAAATATCGCAGG
    TAATTCAACTGAACAAGAGACATCTACTGATAAAGATATGACCATTACTT
    TTACAAATAAAAAAGATTT
  • In the presence of this anti-GAS 16 antiserum, a shift in fluorescence is observed for each GAS serotype, demonstrating its cell surface expression. Table 22, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-GAS 16 antiserum, and the change in fluorescence value between the pre-immune and anti-GAS 16 antiserum.
    TABLE 22
    Summary of FACS values for surface expression of GAS 16 using a second antisera
    Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS
    immune 16 Change immune 16 Change immune 16 Change
    2719 2580 3280
    141.55 650.22 509 119.57 672.35 553 209.18 666.71 458
    SF370 2913 3348
    159.92 719.32 559 115.97 585.9 470 146.1 414.01 268
  • FIGS. 77 and 78 provide the results of FACS analysis for surface expression of GAS 18 on each of GAS serotypes M12719, M12580, M1 3280, M1 SF370, M12913, and M13348. The antiserum used to obtain the FACS data in each of FIGS. 77 and 78 was different, although each was raised against full length GAS 18. In the presence of each of the anti-GAS 18 antisera, a shift in fluorescence is observed for each GAS serotype, demonstrating its cell surface expression. Tables 23 and 24, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, first or second anti-GAS 18 antiserum, and the change in fluorescence value between the pre-immune and first or second anti-GAS 18 antiserum.
    TABLE 23
    Summary of FACS values for surface expression of GAS 18
    Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS
    immune 18 Change immune 18 Change immune 18 Change
    2719 2580 3280
    135.68 327.98 192 116.32 379.41 263 208.12 380.84 173
    SF370 2913 3348
    185.39 438.23 253 119.95 373.32 253 147.12 266.51 119
  • TABLE 24
    Summary of FACS values for surface expression of GAS 18 using a second antisera
    Pre- Anti-GAS Pre- Anti-GAS Pre- Anti-GAS
    immune 18 Change immune 18 Change immune 18 Change
    2719 2580 3280
    150.4 250.39 100 139.18 386.38 247 253.38 347.72 94
    SF370 2913 3348
    188.64 373.11 184 124.94 384.82 260 168.8 213.65 45
  • Surface expression of GAS 15, GAS 16, and GAS 18 on M1 serotype GAS has also been confirmed by Western blot analysis. FIG. 89 shows that while pre-immune sera does not detect GAS M1 expression of GAS 15, anti-GAS 15 immune sera is able to detect GAS 15 protein in both total GAS M1 extracts and GAS M1 proteins enriched for cell surface proteins. The GAS 15 proteins detected in the M1 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that GAS 15 may be in an oligomeric (pilus) form. FIG. 90 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 15 antisera. Again, the lanes that contain GAS M1 extracts enriched for surface proteins (M1 prot sup) show the presence of high molecular weight structures that may be oligomers of GAS 15. FIG. 91 provides an additional Western blot identical to that of FIG. 90, but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIG. 92 provides a Western blot that was probed for GAS 16 protein. While pre-immune sera does not detect GAS M1 expression of GAS 16, anti-GAS 16 immune sera is able to detect GAS 16 protein in GAS M1 extracts enriched for cell surface proteins. The GAS 16 proteins detected in the M1 extracts enriched for surface proteins are present as high molecular weight structures, indicating that GAS 16 may be in an oligomeric (pilus) form. FIG. 93 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 16 antisera. The lanes that contain total GAS M1 protein (M1 tot new and M1 tot old) and the lane that contains GAS M1 extracts enriched for surface proteins (M1 prot sup) show the presence of high molecular weight structures that may be oligomers of GAS 16. FIG. 94 provides an additional Western blot identical to that of FIG. 93, but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIG. 95 provides a Western blot that was probed for GAS 18 protein. While pre-immune sera does not detect GAS M1 expression of GAS 18, anti-GAS 18 immune sera is able to detect GAS 18 protein in GAS M1 extracts enriched for cell surface proteins. The GAS 18 proteins detected in the M1 extracts enriched for surface proteins are present as high molecular weight structures, indicating that GAS 18 may be in an oligomeric (pilus) form. FIG. 96 also shows the results of Western blot analysis of M1 serotype GAS using anti-GAS 18 antisera. The lane that contains GAS M1 extracts enriched for surface proteins (M1 prot sup) show the presence of high molecular weight structures that may be oligomers of GAS 18. FIG. 97 provides an additional Western blot identical to that of FIG. 96, but that was probed with pre-immune sera. As expected, no proteins were detected on this membrane.
  • FIGS. 102-106 provide additional Western blots to verify the presence of GAS 15, GAS 16, and GAS 18 in high molecular weight structures in GAS. Each Western blot was performed using proteins from a different GAS M1 strain, 2580, 2913, 3280, 3348, and 2719. Each Western blot was probed with antisera raised against each of GAS 15, GAS 16, and GAS 18. As can be seen in FIGS. 102-106, none of the Western blots shows detection of proteins using pre-immune serum (Pα-158, Pα-15, Pα-16, or Pα-18), while each Western blot shows cross-hybridization of the GAS 15 (1α-15), GAS 16 (1α-16), and GAS 18 (1α-18) antisera to high molecular weight structures. Thus, these Western blots confirm that GAS 15, GAS 16, and GAS 18 can be present in pili in GAS M1.
  • FIG. 107 provides a similar Western blot performed to detect GAS 15, GAS 16, and GAS 18 proteins in a GAS serotype M1 strain SF370 protein fraction enriched for surface proteins. This Western blot also shows detection of GAS 15 (Anti-15), GAS 16 (Anti-16), and GAS 18 (Anti-18) as high molecular weight structures.
  • (3) Adhesin Island Sequence within M3, M5 and M18: GAS Adhesin Island 3 (“GAS AI-3”)
  • GAS Adhesin Island sequences within M3, M5, and M18 serotypes are outlined in Tables 6-8 and 10 below. This GAS adhesin island 3 (“GAS AI-3”) comprises surface proteins, a SrtC2 sortase, and a Negative transcriptional regulator (Nra) divergently transcribed transcriptional regulator.
  • GAS AI-3 surface proteins within include a collagen binding protein, a fimbrial protein, a F2 like fibronectin-binding protein. GAS AI-3 surface proteins may also include a hypothetical surface protein. Preferably, each of these GAS AI-3 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-3 includes a SrtC2 type sortase. GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail. GAS SrtC2 may be differentially regulated by Nra.
  • GAS AI-3 may also include a LepA putative signal peptidase I protein.
  • GAS AI-3 may also include a putative multiple sugar metabolism regulator.
    TABLE 6
    GAS AI-3 sequences from M3 isolate (MGAS315)
    Sortase
    substrate
    AI-3 sequence sequence or
    identifier sortase type Functional description
    SpyM3_0097 Negative transcriptional regulator (Nra)
    SpyM3_0098 VPXTG putative collagen binding protein (Cpb)
    SpyM3_0099 LepA putative signal peptidase I
    SpyM3_0100 QVXTG conserved hypothetical protein (fimbrial)
    SpyM3_0101 SrtC2 sortase
    SpyM3_0102 LPXAG hypothetical protein
    SpyM3_0103 putative multiple sugar metabolism
    regulator
    SpyM3_0104 LPXTG protein F2 like fibronectin-binding protein
  • TABLE 7
    GAS AI-3 sequence from M3 isolate (SSI-1)
    Sortase
    Substrate
    AI-3 sequence sequence or
    identifier sortase type Functional description
    SPs0099 Negative transcriptional regulator (Nra)
    SPs0100 VPXTG putative collagen binding protein (Cpb)
    SPs0101 LepA putative signal peptidase I
    SPs0102 QVXTG conserved hypothetical protein (fimbrial)
    SPs0103 SrtC2 sortase
    SPs0104 LPXAG hypothetical protein
    SPs0105 putative multiple sugar metabolism
    regulator
    SPs0106 LPXTG protein F2 like fibronectin-binding protein
  • TABLE 10
    GAS AI-3 sequences from M5 isolate (Manfredo)
    AI-3 Sortase substrate
    sequence sequence or
    identifier sortase type Functional description
    orf77 Negative transcriptional regulator (Nra)
    orf78 VPXTG putative collagen binding protein (Cpb)
    orf79 LepA putative signal peptidase I
    orf80 QVXTG conserved hypothetical protein (fimbrial)
    orf81 SrtC2 sortase
    orf82 LPXAG hypothetical protein
    orf83 putative multiple sugar metabolism
    regulator
    orf84 LPXTG protein F2 like fibronectin-binding protein
  • TABLE 8
    GAS AI-3 sequences from M18 isolate (MGAS8232)
    Sortase
    substrate
    AI-3 sequence sequence or
    identifier sortase type Functional description
    spyM18_0125 Negative transcriptional regulator (Nra)
    (N-terminal fragment)
    spyM18_0126 VPXTG putative collagen binding protein (Cpb)
    spyM18_0127 LepA putative signal peptidase I
    spyM18_0128 QVXTG conserved hypothetical protein (fimbrial)
    spyM18_0129 SrtC2 sortase
    spyM18_0130 LPXAG hypothetical protein
    spyM18_0131 putative multiple sugar metabolism
    regulator
    spyM18_0132 LPXTG protein F2 like fibronectin-binding protein
  • TABLE 44
    GAS AI-3 sequences from M49 isolate (591)
    Sortase substrate
    AI-3 sequence sequence or
    identifier sortase type Functional description
    SpyoM01000156 Negative transcriptional regulator
    (Nra)
    SpyoM01000155 VPXTG collagen binding protein (Cpa)
    SpyoM01000154 LepA putative signal peptidase I
    SpyoM01000153 QVXTG conserved hypothetical protein
    (fimbrial)
    SpyoM01000152 SrtC2 sortase
    SpyoM01000151 LPXAG hypothetical protein
    SpyoM01000150 MsmRL
    SpyoM01000149 LPXTG protein F2 like fibronectin-
    binding protein
  • A schematic of AI-3 serotypes M3, M5, M 18, and M49 is shown in FIG. 51A. Each contains an open reading frame encoding a SrtC2-type sortase of nearly identical amino acid sequence. See FIG. 52B for an amino acid sequence alignment for each of the SrtC2 amino acid sequences.
  • The protein F2-like fibronectin-binding protein of each these type 3 adhesin islands contains a pilin motif and an E-box. FIG. 60 indicates the amino acid sequence of the pilin motif and E-box of each of GAS AI-3 serotype M3 MGAS315 (SpyM30104/21909640), GAS AI-3 serotype M3 SSI (Sps0106/28895018), GAS AI-3 serotype M18 (SpyM180132/19745307), and GASAI-3 serotype M5 (orf84).
  • FACS analysis has confirmed that the GAS AI-3 surface proteins SpyM30098, SpyM3 0100, SpyM3 0102, and SpyM30104 are expressed on the surface of GAS. FIG. 80 provides the results of FACS analysis for surface expression of SpyM30098 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM30098 antiserum is present, demonstrating cell surface expression. Table 25, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM30098 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30098 antiserum.
    TABLE 25
    Summary of FACS values for surface expression of SpyM3_0098
    2721 3135
    Pre- Anti- Pre- Anti-
    immune spyM3_0098 Change immune spyM3_0098 Change
    117.85 249.51 132 99.17 277.21 178
  • FIG. 81 provides the results of FACS analysis for surface expression of SpyM3 0100 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM30100 antiserum is present, demonstrating cell surface expression. Table 26, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM30100 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30100 antiserum.
    TABLE 26
    Summary of FACS values for surface expression of SpyM3_0100
    2721 3135
    Pre- Anti- Pre- Anti-
    immune spyM3_0100 Change immune spyM3_0100 Change
    110.31 181.91 72 97.87 250.01 152
  • FIG. 82 provides the results of FACS analysis for surface expression of SpyM3 0102 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM30102 antiserum is present, demonstrating cell surface expression. Table 27, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM30102 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30102 antiserum.
    TABLE 27
    Summary of FACS values for surface expression
    of SpyM3_0102 in M3 serotypes
    2721 3135
    Pre- Anti- Pre- Anti-
    immune spyM3_0102 Change immune spyM3_0102 Change
    109.86 155.26 45 100.02 112.58 13
  • FIG. 82 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SpyM3 0102 identified in a different GAS serotype, M6. FACS analysis conducted with the SpyM3 0102 antisera was able to detect surface expression of the homologous SpyM3 0102 antigen on each of GAS serotypes M6 2724, M6 3650, and M6 2894. Table 28, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM30102 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30102 antiserum.
    TABLE 28
    Summary of FACS values for surface expression of SpyM3_0102 in M6 serotypes
    2724 3650 2894
    Pre- Anti- Pre- Anti- Pre- Anti-
    immune spyM3_0102 Change immune spyM3_0102 Change immune spyM3_0102 Change
    146.59 254.03 107 162.56 294.03 131 175.49 313.69 138
  • SpyM3 0102 is also homologous to pilin antigen 19224139 of GAS serotype M12. Antisera raised against SpyM3 0102 is able to detect high molecular weight structures in GAS serotype M12 strain 2728 protein fractions enriched for surface proteins, which would contain the 19224139 antigen. See FIG. 109 at the lane labelled M112 2728 surf prot.
  • FIG. 83 provides the results of FACS analysis for surface expression of SpyM30104 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SpyM30104 antiserum is present, demonstrating cell surface expression. Table 29, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SpyM30104 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30104 antiserum.
    TABLE 29
    Summary of FACS values for surface expression of
    SpyM3_0104 in M3 serotypes
    2721 3135
    Pre- Anti- Pre- Anti-
    immune spyM3_0104 Change immune spyM3_0104 Change
    128.45 351.65 223 105.1 339.88 235
  • FIG. 83 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SpyM30104 identified in a different GAS serotype, M12. FACS analysis conducted with the SpyM30104 antisera was able to detect surface expression of the homologous SpyM30104 antigen on GAS serotype M12 2728. Table 30, below, quantitatively summarizes the FACS fluorescence values obtained for this GAS serotype in the presence of pre-immune antiserum, anti-SpyM30104 antiserum, and the difference in fluorescence value between the pre-immune and anti-SpyM30104 antiserum.
    TABLE 30
    Summary of FACS values for surface expression of SpyM3_0104
    in an M12 serotype
    2728
    Pre-immune Anti-spyM3_0104 Change
    198.57 288.75 90
  • FIG. 84 provides the results of FACS analysis for surface expression of SPs0106 on each of GAS serotypes M3 2721 and M3 3135. A shift in fluorescence is observed for each GAS serotype when anti-SPs0106 antiserum is present, demonstrating cell surface expression. Table 31, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SPs0106 antiserum, and the difference in fluorescence value between the pre-immune and anti-SPs0106 antiserum.
    TABLE 31
    Summary of FACS values for surface expression
    of SPs_0106 in M3 serotypes
    2721 3135
    Anti- Anti-
    Pre-immune SPs_0106 Change Pre-immune SPs_0106 Change
    116 463.28 347 103.02 494.27 391
  • FIG. 84 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to SPs0106 identified in a different GAS serotype, M12. FACS analysis conducted with the SPs0106 antisera was able to detect surface expression of the homologous SPs0106 antigen on GAS serotype M12 2728. Table 32, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-SPs0106 antiserum, and the difference in fluorescence value between the pre-immune and anti-SPs0106 antiserum.
    TABLE 32
    Summary of FACS values for surface expression of SPs_0106
    in an M12 serotype
    2728
    Pre-immune Anti-SPs_0106 Change
    304.01 254.64 −49
  • (4) Adhesin Island Sequence within M12: GAS Adhesin Island 4 (“GAS AI-4”)
  • GAS Adhesin Island sequences within M12 serotype are outlined in Table 11 below. This GAS adhesin island 4 (“GAS AI-4”) comprises surface proteins, a SrtC2 sortase, and a RofA regulatory protein.
  • GAS AI-4 surface proteins within may include a fimbrial protein, an F or F2 like fibronectin-binding protein, and a capsular polysaccharide adhesion protein (Cpa). GAS AI-4 surface proteins may also include a hypothetical surface protein in an open reading frame (orf). Preferably, each of the GAS AI-4 surface proteins include an LPXTG sortase substrate motif, such as LPXTG (SEQ ID NO: 122), VPXTG (SEQ ID NO: 137), QVXTG (SEQ ID NO: 138) or LPXAG (SEQ ID NO: 139).
  • GAS AI-4 includes a SrtC2 type sortase. GAS SrtC2 type sortases may preferably anchor surface proteins with a QVPTG (SEQ ID NO: 140) motif, particularly when the motif is followed by a hydrophobic region and a charged C terminus tail.
  • GAS AI-4 may also include a LepA putative signal peptidase I protein and a MsmRL protein.
    TABLE 11
    GAS AI-4 sequences from M12 isolate (A735)
    Sortase substrate
    AI-4 sequence sequence or sortase
    identifier type Functional description
    19224133 RofA regulatory protein
    19224134 LPXTG protein F
    SrtB SrtB (stop codon*)
    19224135 VPXTG Cpa
    19224136 LepA
    19224137 QVXTG EftLSL.A (fimbrial)
    19224138 SrtC2 EftLSL.B
    19224139 LPXAG Orf2
    19224140 MsmRL
    19224141 LPXTG protein F2
  • A schematic of AI-4 serotype M12 is shown in FIG. 51A.
  • One of the open reading frames encodes a SrtC2-type sortase having an amino acid sequence nearly identical to the amino acid sequence of the SrtC2-type sortase of the AI-3 serotypes described above. See FIG. 52B for an amino acid sequence alignment for each of the SrtC2 amino acid sequences.
  • Other proteins encoded by the open reading frames of the AI-4 serotype M12 are homologous to proteins encoded by other known adhesin islands in S. pyogenes, as well as the GAS AI-3 serotype M5 (Manfredo). FIG. 52 is an amino acid alignment of the capsular polysaccharide adhesion protein (cpa) of AI-4 serotype M112 (19224135), GAS AI-3 serotype M5 (ORF78), S. pyogenes strain MGAS315 serotype M3 (21909634), S. pyogenes SSI-1 serotype M3 (28810257), S. pyogenes MGAS8232 serotype M3 (19745301), and GAS AI-2 serotype M1 (GAS15). The amino acid sequence of the AI-4 serotype M12 cpa shares a high degree of homology with other cpa proteins.
  • FIG. 53 shows that the F-like fibronectin-binding protein encoded by the AI-4 serotype M12 open frame (19224134) shares homology with a F-like fibronectin-binding protein found in S. pyogenes strain MGAS10394 serotype M6 (50913503).
  • FIG. 54 is an amino acid sequence alignment that illustrates that the F2-like fibronectin-binding protein of AI-4 serotype M12 (19224141) shares homology with the F2-like fibronectin-binding protein of S. pyogenes strain MGAS8232 serotype M3 (19745307), GAS AI-3 serotype M5 (ORF84), S. pyogenes strain SSI serotype M3 (28810263), and S. pyogenes strain MGAS315 serotype M3 (21909640).
  • FIG. 55 is an amino acid sequence alignment that illustrates that the fimbrial protein of AI-4 serotype M12 (19224137) shares homology with the fimbrial protein of GAS AI-3 serotype M5 (ORF80), and the hypothetical protein of S. pyogenes strain MGAS315 serotype M3 (21909636), S. pyogenes strain SSI serotype M3 (28810259), S. pyogenes strain MGAS8732 serotype M3 (19745303), and S. pyogenes strain M1 GAS serotype M1 (13621428).
  • FIG. 56 is an amino acid sequence alignment that illustrates that the hypothetical protein of GAS AI-4 serotype M12 (19224139) shares homology with the hypothetical protein of S. pyogenes strain MGAS315 serotype M3 (21909638), S. pyogenes strain SSI-1 serotype M3 (28810261), GAS AI-3 serotype M5 (ORF82), and S. pyogenes strain MGAS8232 serotype M3 (19745305).
  • The protein F2-like fibronectin-binding protein of the type 4 adhesin island also contains a highly conserved pilin motif and an E-box. FIG. 60 indicates the amino acid sequence of the pilin motif and E-box in AI-4 serotype M12.
  • FACS analysis has confirmed that the GAS AI-4 surface proteins 19224134, 19224135, 19224137, and 19224141 are expressed on the surface of GAS. FIG. 85 provides the results of FACS analysis for surface expression of 19224134 on GAS serotype M12 2728. A shift in fluorescence is observed when anti-19224134 antiserum is present, demonstrating cell surface expression. Table 33, below, quantitatively summarizes the FACS fluorescence values obtained for GAS serotype M12 2728 in the presence of pre-immune antiserum, anti-19224134 antiserum, and the difference in fluorescence value between the pre-immune and anti-19224134 antiserum.
    TABLE 33
    Summary of FACS values for surface expression of 19224134 in an
    M12 serotype
    2728
    Pre-immune Anti-19224134 Change
    137.8 485.32 348
  • FIG. 85 also provides the results of FACS analysis for surface expression of a pilin antigen that has homology to 19224134 identified in a different GAS serotype, M6. FACS analysis conducted with the 19224134 antisera was able to detect surface expression of the homologous 19224134 antigen on each of GAS serotypes M6 2724, M6 3650, and M6 2894. Table 34, below, quantitatively summarizes the FACS fluorescence values obtained for each GAS serotype in the presence of pre-immune antiserum, anti-19224134 antiserum, and the difference in fluorescence value between the pre-immune and anti-19224134 antiserum.
    TABLE 34
    Summary of FACS values for surface expression of 19224134 in M6 serotypes
    2724 3650 2894
    Pre- Anti- Pre- Anti- Pre- Anti-
    immune 19224134 Change immune 19224134 Change immune 19224134 Change
    123.58 264.59 141 140.82 262.64 122 135.4 307.25 172
  • FIG. 86 provides the results of FACS analysis for surface expression of 19224135 on GAS serotype M12 2728. A shift in fluorescence is observed when anti-19224135 antiserum is present, demonstrating cell surface expression. Table 35, below, quantitatively summarizes the FACS fluorescence values obtained for GAS serotype M12 2728 in the presence of pre-immune antiserum, anti-19224135 antiserum, and the difference in fluorescence value between the pre-immune and anti-19224135 antiserum.
    TABLE 35
    Summary of FACS values for surface expression of 19224135
    in an M12 serotype
    2728
    Pre-immune Anti-19224135 Change
    151.38 471.95 321
  • FIG. 87 provides the results of FACS analysis for surface expression of 19224137 on GAS serotype M12 2728. A shift in fluorescence is observed when anti-19224137 antiserum is present, demonstrating cell surface expression. Table 36, below, quantitatively summarizes the FACS fluorescence values obtained for GAS serotype M12 2728 in the presence of pre-immune antiserum, anti-19224137 antiserum, and the difference in fluorescence value between the pre-immune and anti-19224137 antiserum.
    TABLE 36
    Summary of FACS values for surface expression of 19224137
    in an M12 serotype
    2728
    Pre-immune Anti-19224137 Change
    140.44 433.25 293
  • FIG. 88 provides the results of FACS analysis for surface expression of 19224141 on GAS serotype M12 2728. A shift in fluorescence is observed when anti-19224141 antiserum is present, demonstrating cell surface expression. Table 37, below, quantitatively summarizes the FACS fluorescence values obtained for GAS serotype M12 2728 in the presence of pre-immune antiserum, anti-19224141 antiserum, and the difference in fluorescence value between the pre-immune and anti-19224141 antiserum.
    TABLE 37
    Summary of FACS values for surface expression of 19224141
    in an M12 serotype
    2728
    Pre-immune Anti-19224141 Change
    147.02 498 351
  • 19224139 (designated as orf2) may also be expressed on the surface of GAS serotype M12 bacteria. FIG. 175 shows the results of FACS analysis for surface expression of 19224139 on M12 strain 2728. A slight shift in fluorescence is observed, which demonstrates that some 19224139 may be expressed on the GAS cell surface.
  • Surface expression of 19224135 on M12 serotype GAS has also been confirmed by Western blot analysis. FIG. 99 shows that while pre-immune sera (P α-4135) does not detect GAS M12 expression of 19224135, anti-19224135 immune sera (I α-4135) is able to detect 19224135 protein in both total GAS M12 extracts (M12 tot) and GAS M12 fractions enriched for cell surface proteins (M12 surf prot). The 19224135 proteins detected in the total GAS M12 extracts or the GAS M12 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that 19224135 may be in an oligomeric (pilus) form. See also FIG. 108, which provides a further Western blot showing that anti-19224135 antiserum (Anti-19224135) immunoreacts with high molecular weight structures in GAS M12 strain 2728 protein extracts enriched for surface proteins.
  • Surface expression of 19224137 on M12 serotype GAS has also been confirmed by Western blot analysis. FIG. 100 shows that while pre-immune sera (P α-4137) does not detect GAS M12 expression of 19224137, anti-19224137 immune sera (I α-4137) is able to detect 19224137 protein in both total GAS M12 extracts (M12 tot) and GAS M12 fractions enriched for cell surface proteins (M12 surf prot). The 19224137 proteins detected in the total GAS M12 extracts or the GAS M12 extracts enriched for surface proteins are also present as high molecular weight structures, indicating that 19224137 may be in an oligomeric (pilus) form. See also FIG. 108, which provides a further Western blot showing that anti-19224137 antiserum (Anti-19224137) immunoreacts with high molecular weight structures in GAS M12 strain 2728 protein extracts enriched for surface proteins.
  • Streptococcus pneumoniae
  • Adhesin island sequences can be identified in Streptococcus pneumoniae genomes. Several of these genomes include the publicly available Streptococcus pneumoniae TIGR4 genome or Streptococcus pneumoniae strain 670 genome. Examples of these S. pneumoniae AI sequence are discussed below.
  • S. pneumoniae Adhesin Islands generally include a series of open reading frames within a S. pneumoniae genome that encode for a collection of surface proteins and sortases. A S. pneumoniae Adhesin Island may encode for amino acid sequences comprising at least one surface protein. Alternatively, an S. pneumoniae Adhesin Island may encode for at least two surface proteins and at least one sortase. Preferably, a S. pneumoniae Adhesin Island encodes for at least three surface proteins and at least two sortases. One or more of the surface proteins may include an LPXTG motif (such as LPXTG (SEQ ID NO: 122)) or other sortase substrate motif. One or more S. pneumoniae AI surface proteins may participate in the formation of a pilus structure on the surface of the S. pneumoniae bacteria.
  • S. pneumoniae Adhesin Islands of the invention preferably include a divergently transcribed transcriptional regulator. The transcriptional regulator may regulate the expression of the S. pneumoniae AI operon.
  • The S. pneumoniae AI surface proteins may bind or otherwise adhere to fibrinogen, fibronectin, or collagen.
  • A schematic of the organization of a S. pneumoniae AI locus is provided in FIG. 137. The locus comprises open reading frames encoding a transcriptional regulator (rlrA), cell wall surface proteins (rrgA, rrgB, rrgc), and sortases (srtB, srtC, srtD). FIG. 137 also indicates the S. pneumoniae strain TIGR4 gene name corresponding to each of these open reading reading frames.
  • Tables 9 and 38 identify the genomic location of each of these open reading frames in S. pneumoniae strains TIGR4 and 670, respectively.
    TABLE 9
    S. pneumoniae AI sequences from TIGR4
    Synonym (AI
    Genomic Location Strand Length PID Sequence Identifier) Functional description
    436302 . . . 437831 509 15900377 SP0461 transcriptional regulator
    438326 . . . 441007 + 893 15900378 SP0462 cell wall surface anchor family protein
    441231 . . . 443228 + 665 15900379 SP0463 cell wall surface anchor family protein
    443275 . . . 444456 + 393 15900380 SP0464 cell wall surface anchor family protein
    444675 . . . 444806 43 15900381 SP0465 hypothetical protein
    444857 . . . 445696 + 279 15900382 SP0466 sortase
    445791 . . . 446576 + 261 15900383 SP0467 sortase
    446563 . . . 447414 + 283 15900384 SP0468 sortase
  • TABLE 38
    S. pneumoniae strain 670 AI sequences
    AI Sequence
    Genomic Location Strand Identifier\ Functional description
     4383-5645 Orf1_670 IS1167, transposase
     5910-7439 Orf2_670 transcriptional regulator,
    putative
     7934-10606 + Orf3_670 cell wall surface anchor
    family protein
    10839-12773 + Orf4_670 cell wall surface anchor
    family protein
    12796-14001 + Orf5_670 cell wall surface anchor
    family protein
    14327-15241 + Orf6_670 sortase, putative
    15336-16121 + Orf7_670 sortase, putative
    16108-16959 + Orf8_670 sortase, putative
  • The full-length nucleotide sequence of the S. pneumoniae strain 670 μl is also shown in FIG. 101, as is its translated amino acid sequence.
  • At least eight other S. pneumoniae strains contain an adhesin island locus described by the locus depicted in FIG. 137. These strains were identified by an amplification analysis. The genomes of different S. pneumoniae strains were amplified with eleven separate sets of primers. The sequence of each of these primers is provided below in Table 41.
    TABLE 41
    Sequences of primers used to amplify AI locus
    Primer
    Pair Forward Primer Sequence Reverse Primer Sequence
    1 ACTTTCTAATGAGTTGTTTAGGCG AGCGACAAGCCACTGTATCATATT
    2 CTGGTCGATAACTCCTTCAATCTT GTACGACAAAAGTGTGGCTTGTT
    3 GAATGCGATATTCAGGACCAACTA ATCTCACTGAGTTAATCCGTTCAC
    4 TGTATACAAGTGTGTCATTGCCAG CATCTTCACCTGTTCTCACATTTT
    5 GCGGTCTTTAGTCTTCAAAAACA CAAGAGAAAAACACAGAGCCATAA
    6 TTGCTTAAGTAAGAGAGAAAGGAGC CAGGAGTATAGTGTCCGCTTTCTT
    7 GGCAATGTTGACTTTATGAAGGTG TATCAGCATCCCTTTATCTTCAAAC
    8 TGAGATTTTCTCGTTTCTCTTAGC AATAGACGATGGGTATTGATCATGT
    9 CCGACGAACTTTGATGATTTATTG ACCAACAGACGATGACTGTTAATC
    10 AATGACTTTGAGCCTGTCTTGAT TTCTACAATTTCCTGGCCATTATC
    11 GCCATTTGGATCAGCTAAAAGTT TTTTTCAACCCACTACAGTTGACA

    These primers hybridized along the entire length of the AI locus to generate amplification products representative of sequences throughout the locus. See FIG. 138, which is a schematic of the location where each of these primers hybridizes to the S. pneumoniae AI locus. FIG. 139A provides the set of amplicons obtained from amplification of the AI locus in S. pneumoniae strain TIGR4. FIG. 139B provides the length, in base pairs, of each amplicon in S. pneumoniae strain TIGR4. Amplification of the genome of S. pneumoniae strains 19A Hungary 6, 6B Finland 12, 6B Spain 2, 9V Spain 3, 14 CSR 10, 19F Taiwan 14, 23F Taiwan 15, and 23F Poland 16 produced a set of eleven amplicons for the eleven primer pairs, indicating that each of these strains also contained the S. pneumoniae AI locus.
  • The S. pneumoniae strains were also identified as containing the AI locus by comparative genome hybridization (CGH) analysis. The genomes of sixteen S. pneumoniae strains were interrogated for the presence of the AI locus by comparison to unique open reading frames of strain TIGR4. The AI locus was detected by this method in strains 19A Hungary 6 (19AHUN), 6B Finland 12 (6BFIN12), 6B Spain 2 (6BSP2), 14CSR10 (14 CSR10), 9V Spain 3 (9VSP3), 19F Taiwan 14 (19FTW14), 23F Taiwan 15 (19FTW15), and 23F Poland 16 (23FP16). See FIG. 140.
  • The AI locus has been sequenced for each of these strains and the nucleotide and encoded amino acid seqeunce for each orf has been determined. An alignment of the complete nucleotide sequence of the adhesin island present in each of the ten strains is provided in FIG. 196. Aligning the amino acid sequences encoded by the orfs reveals conservation of many of the AI polypeptide amino acid sequences. For example, Table 39 provides a comparison of the percent identities of the polypeptides encoded within the S. pneumoniae strain 670 and TIGR4 adhesin islands.
    TABLE 39
    Pecent identity comparison of S. pneumoniae strains AI sequences
    S. pneumoniae S. pneumoniae
    strain
    670 from TIGR4
    polypeptide polypeptide Shared identity of polypeptides
    Orf1_670 SP0460  99.3% identity in 422 aa overlap
    Orf2_670 SP0461 100.0% identity in 509 aa overlap
    Orf3_670 SP0462  83.2% identity in 895 aa overlap
    Orf4_670 SP0463  47.9% identity in 678 aa overlap
    Orf5_670 SP0464  99.7% identity in 393 aa overlap
    Orf6_670 SP0466 100.0% identity in 279 aa overlap
    Orf7_670 SP0467  94.2% identity in 260 aa overlap
    Orf8_670 SP0468  91.5% identity in 283 aa overlap

    FIGS. 141-147 each provide a multiple sequence alignment for the polypeptides encoded by one of the open reading frames in all ten AI-positive S. pneumoniae strains. In each of the sequence alignments, light shading indicates an LPXTG motif and dark shading indicates the presence of an E-box motif with the conserved glutamic acid residue of the E-box motif in bold.
  • The sequence alignments also revealed that the polypeptides encoded by most of the open reading frames may be divided into two groups of homology, S. pneumoniae AI-a and AI-b. S. pneumoniae strains that comprise AI-a include 14 CSR 10, 19A Hungary 6, 23F Poland 15, 670, 6B Finland 12, and 6B Spain 2. S. pneumoniae strains that comprise AI-b include 19F Taiwan 14, 9V Spain 3, 23F Taiwan 15, and TIGR4. An immunogenic composition of the invention may comprise one or more polypeptides from within each of S. pneumoniae AI-a and AI-b. For example, polypeptide RrgB, encoded by open reading frame 4, may be divided within two such groups of homology. One group contains the RrgB sequences of six S. pneumoniae strains and a second group contains the RrgB sequences of four S. pneumoniae strains. While the amino acid sequence of the strains within each individual group is 99-100 percent identical, the amino acid sequence identity of the strains in the first relative to the second group is only 48%. Table 41 provides the identity comparisons of the amino acid sequences encoded by each open reading frame for the ten S. pneumoniae strains.
    TABLE 42
    Conservation of amino acid sequences encoded
    by the S. pneumoniae AI locus
    % Identity % Identity
    Putative Role Encoded Groups of in Between
    of Polypeptide by Orf Homology Group Groups
    RlrA, 2 1 group (10 strains) 100
    transcriptional
    regulator
    RrgA, cell 3 2 groups (6 + 4) 98-100 83
    wall surface
    protein
    RrgB, cell 4 2 groups (6 + 4) 99-100 48
    wall surface
    protein
    RrgC, cell 5 2 groups (6 + 4) 99-100 97
    wall surface
    protein
    SrtB, putative 6 2 groups (7 + 3) 99-100 97
    sortase
    SrtC, putative 7 2 groups (6 + 4) 95-100 93
    sortase
    SrtD, putative 8 2 groups (6 + 4) 99-100 92
    sortase
  • The division of homology between the RrgB polypeptide in the S. pneumoniae strains is due a lack of amino acid sequence identity in the central amino acid residues. Amino acid residues 1-30 and 617-665 are identical for each of the ten S. pneumoniae strains. However, amino acid residues 31-616 share between 42 and 100 percent identity between strains. See FIG. 149. The shared N- and C-terminal regions of identity in the RrgB polypeptides may be preferred portions of the RrgB polypeptide for use in an immunogenic composition. Similarly, shared regions of identity in any of the polypeptides encoded by the S. pneumoniae AI locus may be preferable for use in immunogenic compositions. One of skill in the art, using the amino acid alignments provided in FIGS. 141-147, would readily be able to determine these regions of identity.
  • The S. pneumoniae comprising these AI loci do, in fact, express high molecular weight polymers on their surface, indicating the presence of pili. See FIG. 182, which shows detection of high molecular weight structures expressed by S. pneumoniae strains that comprise the adhesin island locus depicted in FIG. 137, these strains are indicated as rlrA+. Confirming these findings, electron microscopy and negative staining detects the presence of pili extending from the surface of S. pneumoniae. See FIG. 185. To demonstrate that the adhesin island locus was responsible for the pili, the rrgA-srtD region of TIGR 4 were deleted. Deletion of this region of the adhesin island resulted in a loss of pili expression. See FIG. 186. See also FIG. 235, which provides an electron micrograph of S. pneumoniae lacking the rrgA-srtD region immunogold stained using anti-RrgB and anti-RrgC antibodies. No pili can be seen. Similarly to that described above, a S. pneumoniae bacteria that lacks a transcriptional repressor, mgrA, of genes in the adhesin island expresses pili. See FIG. 187. However, and as expected, a S. pneumoniae bacteria that lacks both the mgrA and adhesin island genes in the rrgA-srtD region does not express pili. See FIG. 188.
  • These high molecular weight pili structures appear to play a role in adherence of S. pneumoniae to cells. S. pneumoniae TIGR4 that lack the pilus operon have significantly diminished ability to adhere to A549 Alveolar cells in vitro. See FIG. 184.
  • The Sp0463 (S. pneumoniae TIGR4 rrgB) adhesion island polypeptide is expressed in oligomeric form. Whole cell extracts were analyzed by Western blot using a Sp0463 antiserum. The antiserum cross-hybridized with high molecular weight Sp0463 polymers. See FIG. 156. The antiserum did not cross-hybridize with polypeptides from D39 or R6 strains of S. pneumoniae, which do not contain the AI locus depicted in FIG. 137. Immunogold labelling of S. pneumoniae TIGR 4 using RrgB antiserum confirms the presence of RrgB in pili. FIG. 189 shows double-labeling of S. pneumoniae TIGR 4 bacteria with immunolabeling for RrgB (5 nm gold particles) and RrgC (10 nm gold particles) protein. The RrgB protein is detected as present at intervals along the pilus structure. The RrgC protein is detected at the tips of the pili. See FIG. 234 at arrows; FIG. 234 is a close up of a pilus in FIG. 189 at the location indicated by *.
  • The RrgA protein appears to be present in and necessary for formation of high molecular weight structures on the surface of S. pneumoniae TIGR4. See FIG. 181 which provides the results of Western blot analysis of TIGR4 S. pneumoniae lacking the gene encoding RrgA. No high molecular weight structures are detected in S. pneumoniae that do not express RrgA using antiserum raised against RrgB. See also FIG. 183.
  • A detailed diagram of the amino acid sequence comparions of the RrgA protein in the ten S. pneumoniae strains is shown in FIG. 148. The diagram reveals the division of the individual S. pneumoniae strains into the two different homology groups.
  • The cell surface polypeptides encoded by the S. pneumoniae TIGR4 AI, Sp0462 (rrgA), Sp0463 (rrgB), and Sp0464 (rrgC), have been cloned and expressed. See examples 15-17. A polyacrylamide gel showing successful recombinant expression of RrgA is provided in FIG. 190A. Detection of the RrgA protein, which is expressed in pET21b with a histidine tag, is also shown by Western blot analysis in FIG. 190B, using an anti-histidine tag antibody.
  • Antibodies that detect RrgB and RrgC antibodies have been produced in mice. See FIGS. 191 and 192, which show detection of RrgB and RrgC, respectively, using the raised antibodies.
  • In addition to the identification of these S. pneumoniae adhesion islands, coding sequences for SrtB type sortases have been identified in several S. pneumoniae clinical isolates, demonstrating conservation of a SrtB type sortase across these isolates.
  • Recombinantly Produced AI polypeptides
  • It is also an aspect of the invention to alter a non-AI polypeptide to be expressed as an AI polypeptide. The non-AI polypeptide may be genetically manipulated to additionally contain AI polypeptide sequences, e.g., a sortase substrate, pilin, or E-box motif, which may cause expression of the non-AI polypeptide as an AI polypeptide. Alternatively the non-AI polypeptide may be genetically manipulated to replace an amino acid sequence within the non-AI polypeptide for AI polypeptide sequences, e.g., a sortase substrate, pilin, or E-box motif, which may cause expression of the non-AI polypeptide as an AI polypeptide. Any number of amino acid residues may be added to the non-AI polypeptide or may be replaced within the non-AI polypeptide to cause its expression as an AI polypeptide. At least 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 50, 75, 100, 150, 200, or 250 amino acid residues may be replaced or added to the non-AI polypeptide amino acid sequence. GBS 322 may be one such non-AI polypeptide that may be expressed as an AI polypeptide.
  • GBS Adhesin Island Sequences
  • The GBS AI polypeptides of the invention can, of course, be prepared by various means (e.g. recombinant expression, purification from GBS, chemical synthesis etc.) and in various forms (e.g. native, fusions, glycosylated, non-glycosylated etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other streptococcal or host cell proteins) or substantially isolated form.
  • The GBS AI proteins of the invention may include polypeptide sequences having sequence identity to the identified GBS proteins. The degree of sequence identity may vary depending on the amino acid sequence (a) in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more). Polypeptides having sequence identity include homologs, orthologs, allelic variants and functional mutants of the identified GBS proteins. Typically, 50% identity or more between two proteins is considered to be an indication of functional equivalence. Identity between proteins is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affinity gap search with parameters gap open penalty=]12 and gap extension penalty=1.
  • The GBS adhesin island polynucleotide sequences may include polynucleotide sequences having sequence identity to the identified GBS adhesin island polynucleotide sequences. The degree of sequence identity may vary depending on the polynucleotide sequence in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more).
  • The GBS adhesin island polynucleotide sequences of the invention may include polynucleotide fragments of the identified adhesin island sequences. The length of the fragment may vary depending on the polynucleotide sequence of the specific adhesin island sequence, but the fragment is preferably at least 10 consecutive polynucleotides, (e.g. at least 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more).
  • The GBS adhesin island amino acid sequences of the invention may include polypeptide fragments of the identified GBS proteins. The length of the fragment may vary depending on the amino acid sequence of the specific GBS antigen, but the fragment is preferably at least 7 consecutive amino acids, (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more). Preferably the fragment comprises one or more epitopes from the sequence. Other preferred fragments include (1) the N-terminal signal peptides of each identified GBS protein, (2) the identified GBS protein without their N-terminal signal peptides, and (3) each identified GBS protein wherein up to 10 amino acid residues (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) are deleted from the N-terminus and/or the C-terminus e.g. the N-terminal amino acid residue may be deleted. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • GBS 80
  • Examples of preferred GBS 80 fragments are discussed below. Polynucleotide and polypeptide sequences of GBS 80 from a variety of GBS serotypes and strain isolates are set forth in FIGS. 18 and 22. The polynucleotide and polypeptide sequences for GBS 80 from GBS serotype V, strain isolate 2603 are also included below as SEQ ID NOS 1 and 2:
    SEQ ID NO.1
    ATGAAATTATCGAAGAAGTTATTGTTTTCGGCTGCTGTTTTAACAATGGT
    GGCGGGGTCAACTGTTGAACCAGTAGCTCAGTTTGCGACTGGAATGAGTA
    TTGTAAGAGCTGCAGAAGTGTCACAAGAACGCCCAGCGAAAACAACAGTA
    AATATCTATAAATTACAAGCTGATAGTTATAAATCGGAAATTACTTCTAA
    TGGTGGTATCGAGAATAAAGACGGCGAAGTAATATCTAACTATGCTAAAC
    TTGGTGACAATGTAAAAGGTTTGCAAGGTGTACAGTTTAAACGTTATAAA
    GTCAAGACGGATATTTCTGTTGATGAATTGAAAAAATTGACAACAGTTGA
    AGCAGCAGATGCAAAAGTTGGAACGATTCTTGAAGAAGGTGTCAGTCTAC
    CTCAAAAAACTAATGCTCAAGGTTTGGTCGTCGATGCTCTGGATTCAAAA
    AGTAATGTGAGATACTTGTATGTAGAAGATTTAAAGAATTCACCTTCAAA
    CATTACCAAAGCTTATGCTGTACCGTTTGTGTTGGAATTACCAGTTGCTA
    ACTCTACAGGTACAGGTTTCCTTTCTGAAATTAATATTTACCCTAAAAAC
    GTTGTAACTGATGAACCAAAAACAGATAAAGATGTTAAAAAATTAGGTCA
    GGACGATGCAGGTTATACGATTGGTGAAGAATTCAAATGGTTCTTGAAAT
    CTACAATCCCTGCCAATTTAGGTGACTATGAAAAATTTGAAATTACTGAT
    AAATTTGCAGATGGCTTGACTTATAAATCTGTTGGAAAAATCAAGATTGG
    TTCGAAAACACTGAATAGAGATGAGCACTACACTATTGATGAACCAACAG
    TTGATAACCAAAATACATTAAAAATTACGTTTAAACCAGAGAAATTTAAA
    GAAATTGCTGAGCTACTTAAAGGAATGACCCTTGTTAAAAATCAAGATGC
    TCTTGATAAAGCTACTGCAAATACAGATGATGCGGCATTTTTGGAAATTC
    CAGTTGCATCAACTATTAATGAAAAAGCAGTTTTAGGAAAAGCAATTGAA
    AATACTTTTGAACTTCAATATGACCATACTCCTGATAAAGCTGACAATCC
    AAAACCATCTAATCCTCCAAGAAAACCAGAAGTTCATACTGGTGGGAAAC
    GATTTGTAAAGAAAGACTCAACAGAAACACAAACACTAGGTGGTGCTGAG
    TTTGATTTGTTGGCTTCTGATGGGACAGCAGTAAAATGGACAGATGCTCT
    TATTAAAGCGAATACTAATAAAAACTATATTGCTGGAGAAGCTGTTACTG
    GGCAACCAATCAAATTGAAATCACATACAGACGGTACGTTTGAGATTAAA
    GGTTTGGCTTATGCAGTTGATGCGAATGCAGAGGGTACAGCAGTAACTTA
    CAAATTAAAAGAAACAAAAGCACCAGAAGGTTATGTAATCCCTGATAAAG
    AAATCGAGTTTACAGTATCACAAACATCTTATAATACAAAACCAACTGAC
    ATCACGGTTGATAGTGCTGATGCAACACCTGATACAATTAAAAACAACAA
    ACGTCCTTCAATCCCTAATACTGGTGGTATTGGTACGGCTATCTTTGTCG
    CTATCGGTGCTGCGGTGATGGCTTTTGCTGTTAAGGGGATGAAGCGTCGT
    ACAAAAGATAAC
    SEQ ID NO: 2
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIPANLGDYEKFEITD
    KFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQNTLKITFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPSIPNTGGIGTAIFVAIGAAVMAFAVKGMKRR
    TKDN
  • As described above, the compositions of the invention may include fragments of AI proteins. In some instances, removal of one or more domains, such as a leader or signal sequence region, a transmembrane region, a cytoplasmic region or a cell wall anchoring motif, may facilitate cloning of the gene encoding the protein and/or recombinant expression of the GBS AI protein. In addition, fragments comprising immunogenic epitopes of the cited GBS AI proteins may be used in the compositions of the invention.
  • For example, GBS 80 contains an N-terminal leader or signal sequence region which is indicated by the underlined sequence at the beginning of SEQ ID NO: 2 above. In one embodiment, one or more amino acids from the leader or signal sequence region of GBS 80 are removed. An example of such a GBS 80 fragment is set forth below as SEQ ID NO: 3:
    SEQ ID NO: 3
    AEVSQERPAKTTVNIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDN
    VKGLQGVQFKRYKVKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKT
    NAQGLVVDALDSKSNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTG
    TGFLSEINIYPKNVVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIP
    ANLGDYEKFEITDKFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQ
    NTLKITFKPEKFKEIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVAS
    TINEKAVLGKAIENTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVK
    KDSTETQTLGGAEFDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPI
    KLKSHTDGTFEIKGLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEF
    TVSQTSYNTKPTDITVDSADATPDTIKNNKRPSIPNTGGIGTAIFVAIGA
    AVMAFAVKGMKRRTKDN
  • GBS 80 contains a C-terminal transmembrane region which is indicated by the underlined sequence near the end of SEQ ID NO: 2 above. In one embodiment, one or more amino acids from the transmembrane region and/or a cytoplasmic region are removed. An example of such a GBS 80 fragment is set forth below as SEQ ID NO: 4:
    SEQ ID NO: 4
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIPANLGDYEKFEITD
    KFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQNTLKITFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPSIPNTG
  • GBS 80 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 5 IPNTG (shown in italics in SEQ ID NO: 2 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 80 protein from the host cell. Accordingly, in one preferred fragment of GBS 80 for use in the invention, the transmembrane and/or cytoplasmic regions and the cell wall anchor motif are removed from GBS 80. An example of such a GBS 80 fragment is set forth below as SEQ ID NO: 6.
    SEQ ID NO: 6
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEPKTDKDVKKLGQDDAGYTTGEEFKWFLKSTTPANLGDYEKFEITD
    KFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQNTLKTTFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAELEIPVASTLNEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPS
  • Alternatively, in some recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • In one embodiment, the leader or signal sequence region, the transmembrane and cytoplasmic regions and the cell wall anchor motif are removed from the GBS 80 sequence. An example of such a GBS 80 fragment is set forth below as SEQ ID NO: 7.
    SEQ ID NO: 7
    AEVSQERPAKTTVNIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDN
    VKGLQGVQFKRYKVKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKT
    NAQGLVVDALDSKSNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTG
    TGFLSEINIYPKNVVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIP
    ANLGDYEKFEITDKFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQ
    NTLKITFKPEKFKEIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVAS
    TINEKAVLGKAIENTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVK
    KDSTETQTLGGAEFDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPI
    KLKSHTDGTFEIKGLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEF
    TVSQTSYNTKPTDITVDSADATPDTIKNNKRPS
  • Applicants have identified a particularly immunogenic fragment of the GBS 80 protein. This immunogenic fragment is located towards the N-terminus of the protein and is underlined in the GBS 80 SEQ ID NO: 2 sequence below. The underlined fragment is set forth below as SEQ ID NO: 8.
    SEQ ID NO: 2
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIPANLGDYEKFEITD
    KFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQNTLKTTFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPSIPNTGGIGTAIFVAIGAAVMAFAVKGMKRR
    TKDN
    SEQ ID NO: 8
    AEVSQERPAKTTVNIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDN
    VKGLQGVQFKRYKVKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKT
    NAQGLVVDALDSKSNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTG
    TGFLSEINIYPKNVVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTIP
    ANLGDYEKFEITDKFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQ
    NTLKITFKPEKFKEIAELLKG
  • The immunogenicity of the protein encoded by SEQ ID NO: 7 was compared against PBS, GBS whole cell, GBS 80 (full length) and another fragment of GBS 80, located closer to the C-terminus of the peptide (SEQ ID NO: 9, below).
    SEQ ID NO: 9
    MTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIENTFELQYD
    HTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAEFDLLASDG
    TAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIKGLAYAVDA
    NAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTDITVDSADA
    TPDTIKNNKRPS
  • Both an Active Maternal Immunization Assay and a Passive Maternal Immunization Assay were conducted on this collection of proteins.
  • As used herein, an Active Maternal Immunization assay refers to an in vivo protection assay where female mice are immunized with the test antigen composition. The female mice are then bred and their pups are challenged with a lethal dose of GBS. Serum titers of the female mice during the immunization schedule are measured as well as the survival time of the pups after challenge.
  • Specifically, the Active Maternal Immunization assays referred to herein used groups of four CD-1 female mice (Charles River Laboratories, Calco Italy). These mice were immunized intraperitoneally with the selected proteins in Freund's adjuvant at days 1, 21 and 35, prior to breeding. 6-8 weeks old mice received 20 μg protein/dose when immunized with a single antigen, 30-45 μg protein/dose (15 μg each antigen) when immunized with combination of antigens. The immune response of the dams was monitored by using serum samples taken on day 0 and 49. The female mice were bred 2-7 days after the last immunization (at approximately t=36-37), and typically had a gestation period of 21 days. Within 48 hours of birth, the pups were challenged via I.P. with GBS in a dose approximately equal to a amount which would be sufficient to kill 70-90% of unimmunized pups (as determined by empirical data gathered from PBS control groups). The GBS challenge dose is preferably administered in 50 μl of THB medium. Preferably, the pup challenge takes place at 56 to 61 days after the first immunization. The challenge inocula were prepared starting from frozen cultures diluted to the appropriate concentration with THB prior to use. Survival of pups was monitored for 5 days after challenge.
  • As used herein, the Passive Maternal Immunization Assay refers to an in vivo protection assay where pregnant mice are passively immunized by injecting rabbit immune sera (or control sera) approximately 2 days before delivery. The pups are then challenged with a lethal dose of GBS.
  • Specifically, the Passive Maternal Immunization Assay referred to herein used groups of pregnant CD1 mice which were passively immunized by injecting 1 ml of rabbit immune sera or control sera via I.P., 2 days before delivery. Newborn mice (24-48 hrs after birth) are challenged via I.P. with a 70-90% lethal dose of GBS serotype II COH1. The challenge dose, obtained by diluting a frozen mid log phase culture, was administered in 50 μl of THB medium.
  • For both assays, the number of pups surviving GBS infection was assessed every 12 hrs for 4 days. Statistical significance was estimated by Fisher's exact test.
  • The results of each assay for immunization with SEQ ID NO: 7, SEQ ID NO: 8, PBS and GBS whole cell are set forth in Tables 1 and 2 below.
    TABLE 1
    Immunization
    Antigen Alive/total % Survival Fisher's exact test
    PBS (neg control) 13/80 16%
    GBS (whole cell) 54/65 83% P < 0.00000001
    GBS80 (intact) 62/70 88% P < 0.00000001
    GBS80 (fragment) 35/64 55% P = 0.0000013
    SEQ ID 7
    GBS80 (fragment) 13/67 19% P = 0.66
    SEQ ID 8
  • TABLE 2
    Passive Maternal Immunization
    Antigen Alive/total % Survival Fisher's exact test
    PBS (neg control) 12/42 28%
    GBS (whole cell) 48/52 92% P < 0.00000001
    GBS80 (intact) 48/55 87% P < 0.00000001
    GBS80 (fragment) 45/57 79% P = 0.0000006
    SEQ ID 7
    GBS80 (fragment) 13/54 24% P = 1
    SEQ ID 8
  • As shown in Tables 1 and 2, immunization with the SEQ ID NO: 7 GBS 80 fragment provided a substantially improved survival rate for the challenged pups than the comparison SEQ ID NO: 8 GBS 80 fragment. These results indicate that the SEQ ID NO: 7 GBS 80 fragment may comprise an important immunogenic epitope of GBS 80.
  • As discussed above, pilin motifs, containing conserved lysine (K) residues have been identified in GBS 80. The pilin motif sequences are underlined in SEQ ID NO: 2, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 199 and 207 and at amino acid residues 368 and 375. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of GBS 80. Preferred fragments of GBS 80 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 2
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEP K TDKDVKKLGQDDAGYTIGEEFKWFLKSTIPANLGDYEKFEITD
    KFADGLTYKSVGKIKIGSKTLNRDEHYTIDEPTVDNQNTLKITFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRK PEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPSIPNTGGIGTAIFVAIGAAVMAFAVKGMKRR
    TKDN
  • E boxes containing conserved glutamic residues have also been identified in GBS 80. The E box motifs are underlined in SEQ ID NO: 2 below. The conserved glutamic acid (E) residues, at amino acid residues 392 and 471, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of GBS 80. Preferred fragments of GBS 80 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 2
    MKLSKKLLFSAAVLTMVAGSTVEPVAQFATGMSIVRAAEVSQERPAKTTV
    NIYKLQADSYKSEITSNGGIENKDGEVISNYAKLGDNVKGLQGVQFKRYK
    VKTDISVDELKKLTTVEAADAKVGTILEEGVSLPQKTNAQGLVVDALDSK
    SNVRYLYVEDLKNSPSNITKAYAVPFVLELPVANSTGTGFLSEINIYPKN
    VVTDEPKTDKDVKKLGQDDAGYTIGEEFKWFLKSTTPANLGDYEKEEITD
    KFADGLTYKSVGKIKIGSKTLNRDLHYTIDEPTVDNQNTLKITFKPEKFK
    EIAELLKGMTLVKNQDALDKATANTDDAAFLEIPVASTINEKAVLGKAIE
    NTFELQYDHTPDKADNPKPSNPPRKPEVHTGGKRFVKKDSTETQTLGGAE
    FDLLASDGTAVKWTDALIKANTNKNYIAGEAVTGQPIKLKSHTDGTFEIK
    GLAYAVDANAEGTAVTYKLKETKAPEGYVIPDKEIEFTVSQTSYNTKPTD
    ITVDSADATPDTIKNNKRPSIPNTGGIGTAIFVAIGAAVMAFAVKGMKRR
    TKDN
  • Similarly, the following offers examples of preferred GBS 104 fragments. Nucleotide and amino acid sequences of GBS 104 sequenced from serotype V isolated strain 2603 are set forth below as SEQ ID NOS 10 and 11:
    SEQ ID NO. 10
    ATGAAAAAGAGACAAAAAATATGGAGAGGGTTATCAGTTACTTTACTAAT
    CCTGTCCCAAATTCCATTTGGTATATTGGTACAAGGTGAAACCCAAGATA
    CCAATCAAGCACTTGGAAAAGTAATTGTTAAAAAAACGGGAGACAATGCT
    ACACCATTAGGCAAAGCGACTTTTGTGTTAAAAAATGACAATGATAAGTC
    AGAAACAAGTCACGAAACGGTAGAGGGTTCTGGAGAAGCAACCTTTGAAA
    ACATAAAACCTGGAGACTACACATTAAGAGAAGAAACAGCACCAATTGGT
    TATAAAAAAACTGATAAAACCTGGAAAGTTAAAGTTGCAGATAACGGAGC
    AACAATAATCGAGGGTATGGATGCAGATAAAGCAGAGAAACGAAAAGAAG
    TTTTGAATGCCCAATATCCAAAATCAGCTATTTATGAGGATACAAAAGAA
    AATTACCCATTAGTTAATGTAGAGGGTTCCAAAGTTGGTGAACAATACAA
    AGCATTGAATCCAATAAATGGAAAAGATGGTCGAAGAGAGATTGCTGAAG
    GTTGGTTATCAAAAAAAATTACAGGGGTCAATGATCTCGATAAGAATAAA
    TATAAAATTGAATTAACTGTTGAGGGTAAAACCACTGTTGAAACGAAAGA
    ACTTAATCAACCACTAGATGTCGTTGTGCTATTAGATAATTCAAATAGTA
    TGAATAATGAAAGAGCCAATATATTCTCAAAGAGCATTAAAAGCTGGGAA
    GCAGTTGAAAAGCTGATTGATAAAATTACATCAAATAAAGACAATAGAGT
    AGCTCTTGTGACATATGCCTCAACCATTTTTGATGGTACTGAAGCGACCG
    TATCAAAGGGAGTTGCCGATCAAAATGGTAAAGCGCTGAATGATAGTGTA
    TCATGGGATTATCATAAAACTACTTTTACAGCAACTACACATAATTACAG
    TTATTTAAATTTAACAAATGATGCTAACGAAGTTAATATTCTAAAGTCAA
    GAATTCCAAAGGAAGCGGAGCATATAAATGGGGATCGCACGCTCTATCAA
    TTTGGTGCGACATTTACTCAAAAAGCTCTAATGAAAGCAAATGAAATTTT
    AGAGACACAAAGTTCTAATGCTAGAAAAAAACTTATTTTTCACGTAACTG
    ATGGTGTCCCTACGATGTCTTATGCCATAAATTTTAATCCTTATATATCA
    ACATCTTACCAAAACCAGTTTAATTCTTTTTTAAATAAAATACCAGATAG
    AAGTGGTATTCTCCAAGAGGATTTTATAATCAATGGTGATGATTATCAAA
    TAGTAAAAGGAGATGGAGAGAGTTTTAAACTGTTTTCGGATAGAAAAGTT
    CCTGTTACTGGAGGAACGACACAAGCAGCTTATCGAGTACCGCAAAATCA
    ACTCTCTGTAATGAGTAATGAGGGATATGCAATTAATAGTGGATATATTT
    ATCTCTATTGGAGAGATTACAACTGGGTCTATCCATTTGATCCTAAGACA
    AAGAAAGTTTCTGCAACGAAACAAATCAAAACTCATGGTGAGCCAACAAC
    ATTATACTTTAATGGAAATATAAGACCTAAAGGTTATGACATTTTTACTG
    TTGGGATTGGTGTAAACGGAGATCCTGGTGCAACTCCTCTTGAAGCTGAG
    AAATTTATGCAATCAATATCAAGTAAAACAGAAAATTATACTAATGTTGA
    TGATACAAATAAAATTTATGATGAGCTAAATAAATACTTTAAAACAATTG
    TTGAGGAAAAACATTCTATTGTTGATGGAAATGTGACTGATCCTATGGGA
    GAGATGATTGAATTCCAATTAAAAAATGGTCAAAGTTTTACACATGATGA
    TTACGTTTTGGTTGGAAATGATGGCAGTCAATTAAAAAATGGTGTGGCTC
    TTGGTGGACCAAACAGTGATGGGGGAATTTTAAAAGATGTTACAGTGACT
    TATGATAAGACATCTCAAACCATCAAAATCAATCATTTGAACTTAGGAAG
    TGGACAAAAAGTAGTTCTTACCTATGATGTACGTTTAAAAGATAACTATA
    TAAGTAACAAATTTTACAATACAAATAATCGTACAACGCTAAGTCCGAAG
    AGTGAAAAAGAACCAAATACTATTCGTGATTTCCCAATTCCCAAAATTCG
    TGATGTTCGTGAGTTTCCGGTACTAACCATCAGTAATCAGAAGAAAATGG
    GTGAGGTTGAATTTATTAAAGTTAATAAAGACAAACATTCAGAATCGCTT
    TTGGGAGCTAAGTTTCAACTTCAGATAGAAAAAGATTTTTCTGGGTATAA
    GCAATTTGTTCCAGAGGGAAGTGATGTTACAACAAAGAATGATGGTAAAA
    TTTATTTTAAAGCACTTCAAGATGGTAACTATAAATTATATGAAATTTCA
    AGTCCAGATGGCTATATAGAGGTTAAAACGAAACCTGTTGTGACATTTAC
    AATTCAAAATGGAGAAGTTACGAACCTGAAAGCAGATCCAAATGCTAATA
    AAAATCAAATCGGGTATCTTGAAGGAAATGGTAAACATCTTATTACCAAC
    ACTCCCAAACGCCCACCAGGTGTTTTTCCTAAAACAGGGGGAATTGGTAC
    AATTGTCTATATATTAGTTGGTTCTACTTTTATGATACTTACCATTTGTT
    CTTTCCGTCGTAAACAATTG
    SEQ ID NO. 11
    MKKRQKIWRGLSVTLLILSQIPFGILVQGETQDTNQALGKVIVKKTGDNA
    TPLGKATFVLKNDNDKSETSHETVEGSGEATFENIKPGDYTLREETAPIG
    YKKTDKTWKVKVADNGATIIEGMDADKAEKRKEVLNAQYPKSAIYEDTKE
    NYPLVNVEGSKVGEQYKALNPINGKDGRREIAEGWLSKKTTGVNDLDKNK
    YKIELTVEGKTTVETKELNQPLDVVVLLDNSNSMNNERANNSQRALKAGE
    AVEKLIDKTTSNKDNRVALVTYASTIFDGTEATVSKGVADQNGKALNDSV
    SWDYHKTTFTATTHNYSYLNLTNDANEVNILKSRIPKEAEHINGDRTLYQ
    FGATFTQKALMKANEILETQSSNARKKLIFHVTDGVPTMSYAINFNPYTS
    TSYQNQFNSFLNKIPDRSGILQEDFIINGDDYQIVKGDGESFKLFSDRKV
    PVTGGTTQAAYRVPQNQLSVMSNEGYAINSGYIYLYWRDYNWVYPFDPKT
    KKVSATKQIKTHGEPTTLYFNGNIRPKGYDIFTVGIGVNGDPGATPLEAE
    KFMQSISSKTENYTNVDDTNKIYDELNKYFKTIVEEKHSIVDGNVTDPMG
    EMIEFQLKNGQSFTHDDYVLVGNDGSQLKNGVALGGPNSDGGILKDVTVT
    YDKTSQTIKINHLNLGSGQKVVLTYDVRLKDNYISNKEYNTNNRTTLSPK
    SEKEPNTIRDFPIPKIRDVREFPVLTISNQKKMGEVEFIKVNKDKHSESL
    LGAKFQLQIEKDFSGYKQFVPEGSDVTTKNDGKIYFKALQDGNYKLYEIS
    SPDGYIEVKTKPVVTFTIQNGEVTNLKADPNANKNQIGYLEGNGKHLITN
    TPKRPPGVFPKTGGIGTIVYILVGSTFMILTICSFRRKQL
  • GBS 104 contains an N-terminal leader or signal sequence region which is indicated by the underlined sequence at the beginning of SEQ ID NO 11 above. In one embodiment, one or more amino acid sequences from the leader or signal sequence region of GBS 104 are removed. An example of such a GBS 104 fragment is set forth below as SEQ ID NO 12.
    SEQ ID NO: 12
    GETQDTNQALGKVIVKKTGDNATPLGKATFVLKNDNDKSETSHETVEGSG
    EATFENIKPGDYTLREETAPIGYKKTDKTWKVKVADNGATIIEGMDADKA
    EKRKEVLNAQYPKSAIYEDTKENYPLVNVEGSKVGEQYKALNPINGKDGR
    REIAEGWLSKKITGVNDLDKNKYKIELTVEGKTTVETKELNQPLDVVVLL
    DNSNSMNNERANNSQPALKAGEAVEKLIDKITSNKDNRVALVTYASTIFD
    GTEATVSKGVADQNGKALNDSVSWDYHKTTFTATTHNYSYLNLTNDANEV
    NILKSRIPKEAEHINGDRTLYQFGATFTQKALMIANEILETQSSNARKKL
    IFHVTDGVPTMSYAINFNPYISTSYQNQFNSFLNKIPDRSGILQEDFIIN
    GDDYQIVKGDGESFKLFSDRKVPVTGGTTQAAYRVPQNQLSVMSNEGYAT
    NSGYIYLYWRDYNWVYPFDPKTKKVSATKQIKTHGEPTTLYPNGNTRPKG
    YDIFTVGIGVNGDPGATPLEAEKFMQSISSKTENYTNVDDTNKIYDELNK
    YEKTIVEEKHSIVDGNVTDPMGEMIEFQLKNGQSPTHDDYVLVGNDGSQL
    KNGVALGGPNSDGGILKDVTVTYDKTSQTIKINHLNLGSGQKVVLTYDVR
    LKDNYISNKFYNTNNRTTLSPKSEKEPNTIRDFPIPKIRDVREFPVLTIS
    NQKKMGEVEFIKVNKDKHSESLLGAKFQLQIEKDESGYKQFVPEGSDVTT
    KNDGKIYFKALQDGNYKLYEISSPDGYIEVKTKPVVTFTIQNGEVTNLKA
    DPNANKNQIGYLEGNGKHLITNTPKRPPGVFPKTGGIGTIVYILVGSTFM
    ILTICSFRRKQL
  • GBS 104 contains a C-terminal transmembrane and/or cytoplasmic region which is indicated by the underlined region near the end of SEQ ID NO 11 above. In one embodiment, one or more amino acids from the transmembrane or cytomplasmic regions are removed. An example of such a GBS 104 fragment is set forth below as SEQ ID NO 13.
    SEQ ID NO: 13
    MKKRQKIWRGLSVTLLILSQIPFGILVQGETQDTNQALGKVIVKKTGDNA
    TPLGKATFVLKNDNDKSETSHETVEGSGEATFENIKPGDYTLREETAPIG
    YKKTDKTWKVKVADNGATIIEGMDADKAEKRKEVLNAQYPKSAIYEDTKE
    NYPLVNVEGSKVGEQYKALNPINGKDGRREIAEGWLSKKITGVNDLDKNK
    YKIELTVEGKTTVETKELNQPLDVVVLLDNSNSMNNERANNSQRALKAGE
    AVEKLIDKITSNKDNRVALVTYASTIFDGTEATVSKGVADQNGKALNDSV
    SWDYHKTTFTATTHNYSYLNLTNDANEVNILKSRIPKEAEHINGDRTLYQ
    FGATFTQKALMKANEILETQSSNARKKLIFHVTDGVPTMSYAINFNPYIS
    TSYQNQFNSFLNKIPDRSGTLQEDFIINGDDYQIVKGDGESFKLFSDRKV
    PVTGGTTQAAYRVPQNQLSVMSNEGYAINSGYIYLYWRDYNWVYPFDPKT
    KKVSATKQIKTHGEPTTLYFNGNTRPKGYDIFTVGIGVNGDPGATPLEAE
    KFMQSISSKTENYTNVDDTNKIYDELNKYFKTIVEEKHSIVDGNVTDPMG
    EMIEFQLKNGQSFTHDDYVLVGNDGSQLKNGVALGGPNSDGGILKDVTVT
    YDKTSQTIKINHLNLGSGQKVVLTYDVRLKDNYISNKFYNTNNRTTLSPK
    SEKEPNTIRDFPIPKIRDVREFPVLTISNQKKMGEVEFIKVNKDKHSESL
    LGAKFQLQIEKDFSGYKQFVPEGSDVTTKNDGKTYFKALQDGNYKLYEIS
    SPDGYIEVKTKPVVTFTIQNGEVTNLKADPNANKNQIGYLEGNGKHLITN
    T
  • In one embodiment, one or more amino acids from the leader or signal sequence region and one or more amino acids from the transmembrane or cytoplasmic regions are removed. An example of such a GBS 104 fragment is set forth below as SEQ ID NO 14.
    SEQ ID NO: 14
    GETQDTNQALGKVIVKKTGDNATPLGKATFVLKNDNDKSETSHETVEGSG
    EATFENIKPGDYTLREETAPIGYKKTDKTWKVKVADNGATIIEGMDADKA
    EKRKEVLNAQYPKSAIYEDTKENYPLVNVEGSKVGEQYKALNPINGKDGR
    REIAEGWLSKKITGVNDLDKNKYKIELTVEGKTTVETKELNQPLDVVVLL
    DNSNSMNNERANNSQRALKAGEAVEKLIDKITSNKDNRVALVTYASTIFD
    GTEATVSKGVADQNGKALNDSVSWDYHKTTFTATTHNYSYLNLTNDANEV
    NILKSRIPKEAEHINGDRTLYQFGATFTQKALMKANEILETQSSNARKKL
    IFHVTDGVPTMSYAINFNPYISTSYQNQFNSFLNKIPDRSGILQEDFIIN
    GDDYQIVKGDGESFKLFSDRKVPVTGGTTQAAYRVPQNQLSVMSNEGYAI
    NSGYIYLYWRDYNWVYPFDPKTKKVSATKQIKTHGEPTTLYFNGNIRPKG
    YDIFTVGIGVNGDPGATPLEAEKFMQSISSKTENYTNVDDTNKIYDELNK
    YFKTIVEEKHSIVDGNVTDPMGEMTEFQLKNGQSFTHDDYVLVGNDGSQL
    KNGVALGGPNSDGGILKDVTVTYDKTSQTIKINHLNLGSGQKVVLTYDVR
    LKDNYISNKFYNTNNRTTLSPKSEKEPNTIRDFPIPKIRDVREFPVLTIS
    NQKKMGEVEFIKVNKDKHSESLLGAKFQLQIEKDFSGYKQFVPEGSDVTT
    KNDGKIYEKALQDGNYKLYEISSPDGYIEVKTKPVVTFTTQNGEVTNLKA
    DPNANKNQIGYLEGNGKHLITNT
  • GBS 104, like GBS 80, contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 123 FPKTG (shown in italics in SEQ ID NO: 11 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 104 protein from the host cell. Accordingly, in one preferred fragment of GBS 104 for use in the invention, only the transmembrane and/or cytoplasmic regions and the cell wall anchor motif are removed from GBS 104. Alternatively, in some recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, containing conserved lysine (K) residues, have been identified in GBS 104. The pilin motif sequences are underlined in SEQ ID NO: 11, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 141 and 149 and at amino acid residues 499 and 507. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of GBS 104. Preferred fragments of GBS 104 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO. 11
    MKKRQKIWRGLSVTLLILSQIPFGILVQGETQDTNQALGKVIVKKTGDNA
    TPLGKATFVLKNDNDKSETSHETVEGSGEATFENIKPGDYTLREETAPIG
    YKKTDKTWKVKVADNGATIIEGMDADKAEKRKEVLNAQYPKSAIYEDTK E
    NYPLVNVEGSKVGEQYKALNPINGKDGRREIAEGWLSKKITGVNDLDKNK
    YKIELTVEGKTTVETKELNQPLDVVVLLDNSNSMNNERANNSQRALKAGE
    AVEKLIDKITSNKDNRVALVTYASTIFDGTEATVSKGVADQNGKALNDSV
    SWDYHKTTFTATTHNYSYLNLTNDANEVNILKSRIPKEAEHINGDRTLYQ
    FGATFTQKALMKANEILETQSSNARKKLIFHVTDGVPTMSYAINFNPYIS
    TSYQNQFNSFLNKIPDRSGILQEDFIINGDDYQIVKGDGESEKLFSDRKV
    PVTGGTTQAAYRVPQNQLSVMSNEGYAINSGYIYLYWRDYNWVYPFDPKT
    KKVSATK QIKTHGEPTTLYFNGNIRPKGYDIFTVGIGVNGDPGATPLEAE
    KFMQSISSKTENYTNVDDTNKIYDELNKYFKTIVEEKHSIVDGNVTDPMG
    EMIEFQLKNGQSFTHDDYVLVGNDGSQLKNGVALGGPNSDGGILKDVTVT
    YDKTSQTTKINHLNLGSGQKVVLTYDVRLKDNYISNKFYNTNNRTTLSPK
    SEKEPNTIRDFPIPKIRDVREFPVLTISNQKKMGEVEFIKVNKDKHSESL
    LGAKFQLQIEKDFSGYKQFVPEGSDVTTKNDGKTYFKALQDGNYKLYEIS
    SPDGYIEVKTKPVVTFTIQNGEVTNLKADPNANKNQIGYLEGNGKHLITN
    TPKRPPGVFPKTGGIGTIVYILVGSTFMILLTICSFRRKQL
  • Two E boxes containing a conserved glutamic residues have also been identified in GBS 104. The E box motifs are underlined in SEQ ID NO: 11 below. The conserved glutamic acid (E) residues, at amino acid residues 94 and 798, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of GBS 104. Preferred fragments of GBS 104 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO. 11
    MKKRQKIWRGLSVTLLILSQIPFGILVQGETQDTNQALGKVIVKKTGDNA
    TPLGKATFVLKNDNDKSETSHETVEGSGEATFENIKPGDYTLREETAPIG
    YKKTDKTWKVKVADNGATIIEGMDADKAEKRKEVLNAQYPKSAIYEDTKE
    NYPLVNVEGSKVGEQYKALNPINGKDGRREIAEGWLSKKITGVNDLDKNK
    YKIELTVEGKTTVETKELNQPLDVVVLLDNSNSMNNERANNSQRALKAGE
    AVEKLIDKITSNKDNRVALVTYASTIFDGTEATVSKGVADQNGKALNDSV
    SWDYHKTTFTATTHNYSYLNLTNDANEVNILKSRIPKEAEHINGDRTLYQ
    FGATFTQKALMKANEILETQSSNARKKLIFHVTDGVPTMSYAINFNPYIS
    TSYQNQFNSFLNKIPDRSGILQEDFIINGDDYQIVKGDGESEKLFSDRKV
    PVTGGTTQAAYRVPQNQLSVMSNEGYAINSGYIYLYWRDYNWVYPFDPKT
    KKVSATKQIKTHGEPTTLYFNGNIRPKGYDIFTVGIGVNGDPGATPLEAE
    KFMQSISSKTENYTNVDDTNKIYDELNKYFKTIVEEKHSIVDGNVTDPMG
    EMIEFQLKNGQSFTHDDYVLVGNDGSQLKNGVALGGPNSDGGILKDVTVT
    YDKTSQTTKINHLNLGSGQKVVLTYDVRLKDNYISNKFYNTNNRTTLSPK
    SEKEPNTIRDFPIPKIRDVREFPVLTISNQKKMGEVEFIKVNKDKHSESL
    LGAKFQLQIEKDFSGYKQFVPEGSDVTTKNDGKTYFKALQDGNYKLYEIS
    SPDGYIEVKTKPVVTFTIQNGEVTNLKADPNANKNQIGYLEGNGKHLITN
    TPKRPPGVFPKTGGIGTIVYILVGSTFMILLTICSFRRKQL

    GBS 067
  • The following offers examples of preferred GBS 067 fragments. Nucleotide and amino acid sequence of GBS 067 sequences from serotype V isolated strain 2603 are set forth below as SEQ ID NOS: 15 and 16.
    SEQ ID NO: 15
    ATGAGAAAATACCAAAAATTTTCTAAAATATTGACGTTAAGTCTTTTTTG
    TTTGTCGCAAATACCGCTTAATACCAATGTTTTAGGGGAAAGTACCGTAC
    CGGAAAATGGTGCTAAAGGAAAGTTAGTTGTTAAAAAGACAGATGACCAG
    AACAAACCACTTTCAAAAGCTACCTTTGTTTTAAAAACTACTGCTCATCC
    AGAAAGTAAAATAGAAAAAGTAACTGCTGAGCTAACAGGTGAAGCTACTT
    TTGATAATCTCATACCTGGAGATTATACTTTATCAGAAGAAACAGCGCCC
    GAAGGTTATAAAAAGACTAACCAGACTTGGCAAGTTAAGGTTGAGAGTAA
    TGGAAAAACTACGATACAAAATAGTGGTGATAAAAATTCCACAATTGGAC
    AAAATCAGGAAGAACTAGATAAGCAGTATCCCCCCACAGGAATTTATGAA
    GATACAAAGGAATCTTATAAACTTGAGCATGTTAAAGGTTCAGTTCCAAA
    TGGAAAGTCAGAGGCAAAAGCAGTTAACCCATATTCAAGTGAAGGTGAGC
    ATATAAGAGAAATTCCAGAGGGAACATTATCTAAACGTATTTCAGAAGTA
    GGTGATTTAGCTCATAATAAATATAAAATTGAGTTAACTGTCAGTGGAAA
    AACCATAGTAAAACCAGTGGACAAACAAAAGCCGTTAGATGTTGTCTTCG
    TACTCGATAATTCTAACTCAATGAATAACGATGGCCCAAATTTTCAAAGG
    CATAATAAAGCCAAGAAAGCTGCCGAAGCTCTTGGGACCGCAGTAAAAGA
    TATTTTAGGAGCAAACAGTGATAATAGGGTTGCATTAGTTACCTATGGTT
    CAGATATTTTTGATGGTAGGAGTGTAGATGTCGTAAAAGGATTTAAAGAA
    GATGATAAATATTATGGCCTTCAAACTAAGTTCACAATTCAGACAGAGAA
    TTATAGTCATAAACAATTAACAAATAATGCTGAAGAGATTATAAAAAGGA
    TTCCGACAGAAGCTCCTAAAGCTAAGTGGGGATCTACTACCAATGGATTA
    ACTCCAGAGCAACAAAAGGAGTACTATCTTAGTAAAGTAGGAGAAACATT
    TACTATGAAAGCCTTCATGGAGGCAGATGATATTTTGAGTCAAGTAAATC
    GAAATAGTCAAAAAATTATTGTTCATGTAACTGATGGTGTTCCTACGAGA
    TCATATGCTATTAATAATTTTAAACTGGGTGCATCATATGAAAGCCAATT
    TGAACAAATGAAAAAAAATGGATATCTAAATAAAAGTAATTTTCTACTTA
    CTGATAAGCCCGAGGATATAAAAGGAAATGGGGAGAGTTACTTTTTGTTT
    CCCTTAGATAGTTATCAAACACAGATAATCTCTGGAAACTTACAAAAACT
    TCATTATTTAGATTTAAATCTTAATTACCCTAAAGGTACAATTTATCGAA
    ATGGACCAGTGAAAGAACATGGAACACCAACCAAACTTTATATAAATAGT
    TTAAAACAGAAAAATTATGACATTTTTAATTTTGGTATCGATATATCTGG
    TTTTAGACAAGTTTATAATGAGGAGTATAAGAAAAATCAAGATGGTACTT
    TTCAAAAATTGAAAGAGGAAGCTTTTAAACTTTCAGATGGAGAAATCACA
    GAACTAATGAGGTCGTTCTCTTCCAAACCTGAGTACTACACCCCTATCGT
    AACTTCAGCCGATACATCTAACAATGAAATTTTATCTAAAATTCAGCAAC
    AATTTGAAACGATTTTAACAAAAGAAAACTCAATTGTTAATGGAACTATC
    GAAGATCCTATGGGTGATAAAATCAATTTACAGCTTGGTAATGGACAAAC
    ATTACAGCCAAGTGATTATACTTTACAGGGAAATGATGGAAGTGTAATGA
    AGGATGGTATTGCAACTGGTGGGCCTAATAATGATGGTGGAATACTTAAG
    GGGGTTAAATTAGAATACATCGGAAATAAACTCTATGTTAGAGGTTTGAA
    TTTAGGAGAAGGTCAAAAAGTAACACTCACATATGATGTGAAACTAGATG
    ACAGTTTTATAAGTAACAAATTCTATGACACTAATGGTAGAACAACATTG
    AATCCTAAGTCAGAGGATCCTAATACACTTAGAGATTTTCCAATCCCTAA
    AATTCGTGATGTGAGAGAATATCCTACAATAACGATTAAAAACGAGAAGA
    AGTTAGGTGAAATTGAATTTATAAAAGTTGATAAAGATAATAATAAGTTG
    CTTCTCAAAGGAGCTACGTTTGAACTTCAAGAATTTAATGAAGATTATAA
    ACTTTATTTACCAATAAAAAATAATAATTCAAAAGTAGTGACGGGAGAAA
    ACGGCAAAATTTCTTACAAAGATTTGAAAGATGGCAAATATCAGTTAATA
    GAAGCAGTTTCGCCGGAGGATTATCAAAAAATTACTAATAAACCAATTTT
    AACTTTTGAAGTGGTTAAAGGATCGATAAAAAATATAATAGCTGTTAATA
    AACAGATTTCTGAATATCATGAGGAAGGTGACAAGCATTTAATTACCAAC
    ACGCATATTCCACCAAAAGGAATTATTCCTATGACAGGTGGGAAAGGAAT
    TCTATCTTTCATTTTAATAGGTGGAGCTATGATGTCTATTGCAGGTGGAA
    TTTATATTTGGAAAAGGTATAAGAAATCTAGTGATATGTCCATCAAAAAA
    GAT
    SEQ ID NO: 16
    MRKYQKFSKILTLSLFCLSQIPLNTNVLGESTVPENGAKGKLVVKKTDDQ
    NKPLSKATFVLKTTAHPESKIEKVTAELTGEATFDNLIPGDYTLSEETAP
    EGYKKTNQTWQVKVESNGKTTIQNSGDKNSTIGQNQEELDKQYPPTGIYE
    DTKESYKLEHVKGSVPNGKSEAKAVNPYSSEGEHIREIPEGTLSKRISEV
    GDLAHNKYKIELTVSGKTIVKPVDKQKPLDVVFVLDNSNSMNNDGPNFQR
    HNKAKKAAEALGTAVKDILGANSDNRVALVTYGSDIFDGRSVDVVKGFKE
    DDKYYGLQTKFTIQTENYSHKQLTNNAEEIIKRIPTEAPKAKWGSTTNGL
    TPEQQKEYYLSKVGETFTMKAFMEADDILSQVNRNSQKIIVHVTDGVPTR
    SYAINNFKLGASYESQFEQMKKNGYLNKSNFLLTDKPEDIKGNGESYFLF
    PLDSYQTQIISGNLQKLHYLDLNLNYPKGTIYRNGPVKEHGTPTKLYINS
    LKQKNYDIFNFGIDISGFRQVYNEEYKKNQDGTFQKLKEEAFKLSDGEIT
    ELMRSFSSKPEYYTPIVTSADTSNNEILSKIQQQFETILTKENSIVNGTI
    EDPMGDKTNLQLGNGQTLQPSDYTLQGNDGSVMKDGIATGGPNNDGGILK
    GVKLEYIGNKLYVRGLNLGEGQKVTLTYDVKLDDSFTSNKFYDTNGRTTL
    NPKSEDPNTLRDFPIPKIRDVREYPTITIKNEKKLGEIEFIKVDKDNNKL
    LLKGATFELQEFNEDYRLYLPIKNNNSKVVTGENGKISYKDLKDGKYQLI
    EAVSPEDYQKITNKPILTFEVVKGSIKNIIAVNKQISEYHEEGDKHLITN
    THIPPKGIIPMTGGKGILSFILIGGAMMSIAGGIYIWKRYKKSSDMSIKK
    D
  • GBS 067 contains a C-terminus transmembrane region which is indicated by the underlined region closest to the C-terminus of SEQ ID NO: 16 above. In one embodiment, one or more amino acids from the transmembrane region is removed and or the amino acid is truncated before the transmembrane region. An example of such a GBS 067 fragment is set forth below as SEQ ID NO: 17.
    SEQ ID NO: 17
    MRKYQKFSKILTLSLFCLSQIPLNTNVLGESTVPENGAKGKLVVKKTDDQ
    NKPLSKATFVLKTTAHPESKIEKVTAELTGEATPDNLIPGDYTLSEETAP
    EGYKKTNQTWQVKVESNGKTTIQNSGDKNSTIGQNQEELDKQYPPTGIYE
    DTKESYKLEHVKGSVPNGKSEAKAVNPYSSEGEHIRETPEGTLSKRISEV
    GDLAHNKYKTELTVSGKTIVKPVDKPKPLDVVFVLDNSNSMNNDGPNFQR
    HNKAKKAAEALGTAVKDILGANSDNRVALVTYGSDIFDGRSVDVVKGFKE
    DDKYYGLQTKETIQTENYSHKQLTNNAEEIIKRIPTEAPKAKWGSTTNGL
    TPEQQKEYYLSKVGETFTMKAFMEADDILSQVNRNSQKIIVHVTDGVPTR
    SYAINNFKLGASYESQFEQMKKNGYLNKSNFLLTDKPEDIKGNGESYFLF
    PLDSYQTQIISGNLQKLHYLDLNLNYPKGTIYRNGPVKEHGTPTKLYINS
    LKQKNYDIFNFGTDISGFRQVYNEEYKKNQDGTFQKLKEEAFKLSDGEIT
    ELMRSESSKPEYYTPIVTSADTSNNEILSKIQQQFETILTKENSIVNGTI
    EDPMGDKINLQLGNGQTLQPSDYTLQGNDGSVMKDGIATGGPNNDGGILK
    GVKLEYIGNKLYVRGLNLGEGQKVTLTYDVKLDDSFISNKFYDTNGRTTL
    NPKSEDPNTLRDFPIPKIRDVREYPTITIKNEKKLGEIEFIKVDKDNNKL
    LLKGATFELQEFNEDYKLYLPIKNNNSKVVTGENGKISYKDLKDGKYQLI
    EAVSPEDYQKITNKPILTFEVVKGSIKNIIAVNKQISEYHEEGDKHLITN
    THIPPKGIIPMTGGKGILS
  • GBS 067 contains an amino acid motif indicative of a cell wall anchor (an LPXTG (SEQ ID NO: 122) motif): SEQ ID NO: 18 IPMTG. (shown in italics in SEQ ID NO: 16 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 067 protein from the host cell. Accordingly, in one preferred fragment of GBS 067 for use in the invention, the transmembrane and the cell wall anchor motif are removed from GBS 67. An example of such a GBS 067 fragment is set forth below as SEQ ID NO: 19.
    SEQ ID NO: 19
    MRKYQKFSKILTLSLFCLSQIPLNTNVLGESTVPENGAKGKLVVKKTDDQ
    NKPLSKATFVLKTTAHPESKIEKVTAELTGEATFDNLIPGDYTLSEETAP
    EGYKKTNQTWQVKVESNGKTTIQNSGDKNSTIGQNQEELDKQYPPTGIYE
    DTKESYKLEHVKGSVPNGKSEAKAVNPYSSEGEHIREIPEGTLSKRTSEV
    GDLAHNKYKIELTVSGKTIVKPVDKQKPLDVVFVLDNSNSMNNDGPNFQR
    HNKAKKPAEALGTAVKDILGANSDNRVALVTYGSDIFDGRSVDVVKGFKE
    DDKYYGLQTKFTIQTENYSHKQLTNNAEEIIKRIPTEAPKAKWGSTTNGL
    TPEQQKEYYLSKVGETFTMKAFMEADDILSQVNRNSQKIIVHVTDGVPTR
    SYAINNFKLGASYESQFEQMKKNGYLNKSNFLLTDKPEDIKGNGESYFLF
    PLDSYQTQIISGNLQKLHYLDLNLNYPKGTIYRNGPVKEHGTPTKLYINS
    LKQKNYDIFNFGIDISGERQVYNEEYKKNQDGTFQKLKEEAFKLSDGEIT
    ELMRSFSSKPEYYTPIVTSADTSNNEILSKIQQQFETILTKENSIVNGTI
    EDPMGDKINLQLGNGQTLQPSDYTLQGNDGSVMKDGIATGGPNNDGGILK
    GVKLEYIGNKLYVRGLNLGEGQKVTLTYDVKLDDSFTSNKFYDTNGRTTL
    NPKSEDPNTLRDFPIPKIRDVREYPTITIKNEKKLGEIEFIKVDKDNNKL
    LLKGATFELQEFNEDYKLYLPIKNNNSKVVTGENGKISYKDLKDGKYQLI
    EAVSPEDYQKITNKPILTFEVVKGSIKNIIAVNKQISEYHEEGDKHLITN
    THIPPKGI
  • Alternatively, in some recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Three pilin motifs, containing conserved lysine (K) residues have been identified in GBS 67. The pilin motif sequences are underlined in SEQ ID NO: 16, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 478 and 488, at amino acid residues 340 and 342, and at amino acid residues 703 and 717. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of GBS 67. Preferred fragments of GBS 67 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 16
    MRKYQKFSKILTLSLFCLSQIPLNTNVLGESTVPENGAKGKLVVKKTDDQ
    NKPLSKATFVLKTTAHPESKIEKVTAELTGEATFDNLIPGDYTLSEETAP
    EGYKKTNQTWQVKVESNGKTTIQNSGDKNSTIGQNQEELDKQYPPTGIYE
    DTKESYKLEHVKGSVPNGKSEAKAVNPYSSEGEHIREIPEGTLSKRISEV
    GDLAHNKYKIELTVSGKTIVKPVDKQKPLDVVFVLDNSNSMNNDGPNFQR
    HNKAKKAAEALGTAVKDILGANSDNRVALVTYGSDIFDGRSVDVVKGFKE
    DDKYYGLQTKFTIQTENYSHKQLTNNAEEIIKRIPTEAPKAK WGSTTNGL
    TPEQQKEYYLSKVGETFTMKAFMEADDILSQVNRNSQKIIVHVTDGVPTR
    SYAINNFKLGASYESQFEQMKKNGYLNKSNFLLTDKPEDIKGNGESYFLF
    PLDSYQTQIISGNLQKLHYLDLNLNYPKGTIYRNGPVK EHGTPTKLYINS
    LKQKNYDIFNFGIDISGFRQVYNEEYKKNQDGTFQKLKEEAFKLSDGEIT
    ELMRSFSSKPEYYTPIVTSADTSNNEILSKIQQQFETILTKENSIVNGTI
    EDPMGDKINLQLGNGQTLQPSDYTLQGNDGSVMKDGIATGGPNNDGGILK
    GVKLEYIGNKLYVRGLNLGEGQKVTLTYDVKLDDSFISNKFYDTNGRTTL
    NPKSEDPNTLRDFPIPK IRDVREYPTITIKNEKKLGEIEFIKVDKDNNKL
    LLKGATFELQEFNEDYKLYLPIKNNNSKVVTGENGKISYKDLKDGKYQLI
    EAVSPEDYQKITNKPILTFEVVKGSIKNIIAVNKQISEYHEEGDKHLITN
    THIPPKGIIPMTGGKGILSFILIGGAMMSIAGGIYIWKRYKKSSDMSIKK
    D
  • Two E boxes containing conserved glutamic residues have also been identified in GBS 67. The E box motifs are underlined in SEQ ID NO: 16 below. The conserved glutamic acid (E) residues, at amino acid residues 96 and 801, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of GBS 67. Preferred fragments of GBS 67 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 16
    MRKYQKFSKILTLSLFCLSQIPLNTNVLGESTVPENGAKGKLVVKKTDDQ
    NKPLSKATFVLKTTAHPESKIEKVTAELTGEARFDNLIPGDYTLSEETAP
    EGYKKTNQTWQVKVESNGKTTIQNSGDKNSTIGQNQEELDKQYPPTGIYE
    DTKESYKLEHVKGSVPNGKSEAKAVNPYSSEGEHIREIPEGTLSKRISEV
    GDLAHNKYKIELTVSGKTIVKPVDKQKPLDVVFVLDNSNSMNNDGPNFQR
    HNKAKKAAEALGTAVKDILGANSDNRVALVTYGSDIFDGRSVDVVKGFKE
    DDKYYGLQTKFTIQTENYSHKQLTNNAEEIIKRIPTEAPKAKWGSTTNGL
    TPEQQKEYYLSKVGETFTMKAFMEADDILSQVNRNSQKIIVHVTDGVPTR
    SYAINNFKLGASYESQFEQMKKNGYLNKSNFLLTDKPEDIKGNGESYFLF
    PLDSYQTQIISGNLQKLHYLDLNLNYPKGTIYRNGPVKEHGTPTKLYINS
    LKQKNYDIFNFGIDISGFRQVYNEEYKKNQDGTFQKLKEEAFKLSDGEIT
    ELMRSFSSKPEYYTPIVTSADTSNNEILSKIQQQFETILTKENSIVNGTI
    EDPMGDKINLQLGNGQTLQPSDYTLQGNDGSVMKDGIATGGPNNDGGILK
    GVKLEYIGNKLYVRGLNLGEGQKVTLTYDVKLDDSFISNKFYDTNGRTTL
    NPKSEDPNTLRDFPIPKIRDVREYPTITIKNEKKLGEIEFIKVDKDNNKL
    LLKGATFELQEFNEDYKLYLPIKNNNSKVVTGENGKISYKDLKDGKYQLI
    EAVSPEDYQKITNKPILTFEVVKGSIKNIIAVNKQISEYHEEGDKHLITN
    THIPPKGIIPMTGGKGILSFILIGGAMMSIAGGIYIWKRYKKSSDMSIKK
    D
  • Predicted secondary structure for the GBS 067 amino acid sequence is set forth in FIG. 33. As shown in this figure, GBS 067 contains several regions predicted to form alpha helical structures. Such alpha helical regions are likely to form coiled-coil structures and may be involved in oligomerization of GBS 067.
  • The amino acid sequence for GBS 067 also contains a region which is homologous to the Cna_B domain of the Staphylococcus aureus collagen-binding surface protein (pfam05738). Although the Cna_B region is not thought to mediate collagen binding, it is predicted to form a beta sandwich structure. In the Staph aureus protein, this beta sandwich structure is through to form a stalk that presents the ligand binding domain away from the bacterial cell surface. This same amino acid sequence region is also predicted to be an outer membrane protein involved in cell envelope biogenesis.
  • The amino acid sequence for GBS 067 contains a region which is homologous to a von Willebrand factor (vWF) type A domain. The vWF type A domain is present at amino acid residues 229-402 of GBS 067 as shown in SEQ ID NO: 16. This type of sequence is typically found in extracellular proteins such as integrins and it thought to mediate adhesion, including adhesion to collagen, fibronectin, and fibrinogen, discussed above.
  • Because applicants have identified GBS 67 as a surface exposed protein on GBS and because GBS 67 may be involved in GBS adhesion, the immunogenicity of the GBS 67 protein was examined in mice. The results of an immunization assay with GBS 67 are set forth in Table 48, below.
    TABLE 48
    GBS 67 Protects Mice in an Immunization Assay
    Challenge GBS
    67 immungen
    GBS strain dead/ PBS immunogen FACS
    (serotype) treated % survival dead/treated % survival Δmean
    3050 (II) 0/30 100 29/49 41 460
    CJB111 (V) 76/185 59 143/189 24 481
    7357 b (Ib) 34/56  39 65/74 12 316
  • As shown in Table 48, immunization with GBS 67 provides a substantially improved survival rate for challenged mice relative to negative control, PBS, immunized mice. These results indicate that GBS 67 may comprise an immunogenic composition of the invention.
  • GBS 59
  • The following offers examples of GBS 59 fragments. Nucleotide and amino acid sequences of GBS 59 sequenced from serotype V isolated strain 2603 are set forth below as SEQ ID NOS: 125 and 126. The GBS 59 polypeptide of SEQ ID NO: 126 is referred to as SAG1407.
    SEQ ID NO: 125
    ttaagcttcctttgattggcgtcttttcatgataactactgctccaagca
    taatgcttaaaccaataattgtgaaaagaattgtaccaataccacctgtt
    tgtgggattgttacctttttattttctacacgtgtcgcatctttttggtt
    gctgttagcaacgtagtcaatgttaccacctgttatgtatgacccttgat
    taactacaaacttaatattacctgccaacttagcaaatcctgctggagca
    agtgtttcttcaaggttgtaagtaccgtctgcaagacctgtaacttcaaa
    ttgaccttgatcgtttgaagtgtaggtaatggctctagccttatctgtta
    tccactcataagctgtacgagcctcaatgaaggctgcatcgtaatctgct
    tgtttagttttgataagttcttttgcagtaattcctttttcacctttttg
    gtctgttgcagacaacttgttataagcagcgatagcttcatctaaagcta
    ttttcttagcagctaaagttttttgaccttctgattgatctgctttaaga
    gcaaggtatttacctgctgagtttttcacaacgaattgtgcaccagccaa
    acggtcaccttgttcattagttttgacaaatttcttaccatgagtttcaa
    cttttggttcagttgggttcaatggtgttgggttatcagaatctttggta
    ttggtaatggttactttaccattttctagatttattgcacttccgtaacc
    agaaacacgttctgagatcatgtatgatttgttttctagaccagtgaatt
    tacccgagaagttaccagatacttcaaatttgataccatttccaaggtcg
    attgtacctttagatgtttttgtcaatgatactgaagcaacagttttatc
    tttatctttcaatgtgtaaacaacgtttacaccatcaggtgcaattccgt
    cagaccaagttttagcaactgttacttcaccctttgaaggtgtaacagga
    agttcagtcaagtctttacctggtttgttaccatacgacaatttgatatc
    attggattctggattatcaataattgcttgaccattaacagtagcactat
    aagtcaatgtaaattcaatatcagctgttttagctgctttttccaatttg
    cccaatccatcagctgtgaattttaatgtgaaaccacgggcatcaatgct
    aagttcatagtctgtatccttagcaaaagtttctgtagttcctgaagctt
    taaggctaacagttgaacccattgtcaaaccatttgacattatatctgtc
    caaaccaagttttcgtatttagaacctttgtgaatttttgttttaacttc
    ataaggaacaactttaccgatttcagcagtagcagttgctttgtcacgtg
    cataattaccataatttgcgccagctgtcaaaagtctattaacatctgtc
    aatgctgtcaaatcgtttgttttagcaaagtttttatcaatttctggttt
    ttcttcagtgttctttggataaacatgggcatcagcaacaacaccatctt
    catttaccaatggaagagtgatgttaactggaaccgcttttgaagcagcc
    aggagggaaccattattgttgtaagtagattttgatttaacttcaacaat
    tttaaactcgcctttcaatcctttggtgttgaaaacaagtccagtatctc
    cctctggtgtcaatccagacacggcctcatcaatatttactgttatttca
    ggagtaccatctttattaattaaggctggtgttaatttgttaccttcttt
    tgccttaacatattgcactttaccacttttatcttctttcaaagctaaag
    caaagaacgcaccttcgatttctttagatccctcgccaaagtaaccagca
    aggtcagaaatagctccacctttgtagtcttttccgttaagacctgtagt
    tcctgggaagttacttttgttaagatttgattcggtttgcaaaatcttgt
    gcaaagtcactgtattagttgttgcttcatccgcaaacgctggtgcaact
    gagagcaatgacgttaaagtcagtaacaatgccgagaacattgcaaaata
    tttgttgattcttttcat
    SEQ ID NO: 126
    MKRINKYFAMFSALLLTLTSLLSVAPAFADEATTNTVTLHKILQTESNLN
    KSNFPGTTGLNGKDYKGGAISDLAGYFGEGSKEIEGAFFALALKEDKSGK
    VQYVKAKEGNKLTPALINKDGTPEITVNIDEAVSGLTPEGDTGLVFNTKG
    LKGEFKIVEVKSKSTYNNNGSLLAASKAVPVNITLPLVNEDGVVADAHVY
    PKNTEEKPEIDKNFAKTNDLTALTDVNRLLTAGANYGNYARDKATATAEI
    GKVVPYEVKTKIHKGSKYENLVWTDIMSNGLTMGSTVSLKASGTTETFAK
    DTDYELSIDARGFTLKFTADGLGKLEKAAKTADIEFTLTYSATVNGQAII
    DNPESNDIKLSYGNKPGKDLTELPVTPSKGEVTVAKTWSDGIAPDGVNVV
    YTLKDKDKTVASVSLTKTSKGTIDLGNGIKPEVSGNFSGKETGLENKSYM
    ISERVSGYGSAINLENGKVTITNTKDSDNPTPLNPTEPKVETHGKKFVKT
    NEQGDRLAGAQFVVKNSAGKYLALKADQSEGQKTLAAKKIALDEAIAAYN
    KLSATDQKGEKGITAKELIKTKQADYDAAFIEARTAYEWITDKARAITYT
    SNDQGQFEVTGLADGTYNLEETLAPAGFAKLAGNIKFVVNQGSYITGGNI
    DYVANSNQKDATRVENKKVTIPQTGGIGTILFTIIGLSIMLGAVVIMKRR
    QSKEA
  • Nucleotide and amino acid sequences of GBS 59 sequenced from serotype V isolated strain CJB111 are set forth below as SEQ ID NOS: 127 and 128. The GBS 59 polypeptide of SEQ ID NO: 128 is referred to as BO1575.
    SEQ ID NO: 127
    ATGAAAAAAATCAACAAATGTCTTACAATGTTCTCGACACTGCTATTGAT
    CTTAACGTCACTATTCTCAGTTGCACCAGCGTTTGCGGACGACGCAACAA
    CTGATACTGTGACCTTGCACAAGATTGTCATGCCACAAGCTGCATTTGAT
    AACTTTACTGAAGGTACAAAAGGTAAGAATGATAGCGATTATGTTGGTAA
    ACAAATTAATGACCTTAAATCTTATTTTGGCTCAACCGATGCTAAAGAAA
    TCAAGGGTGCTTTCTTTGTTTTCAAAAATGAAACTGGTACAAAATTCATT
    ACTGAAAATGGTAAGGAAGTCGATACTTTGGAAGCTAAAGATGCTGAAGG
    TGGTGCTGTTCTTTCAGGGTTAACAAAAGACAATGGTTTTGTTTTTAACA
    CTGCTAAGTTAAAAGGAATTTACCAAATCGTTGAATTGAAAGAAAAATCA
    AACTACGATAACAACGGTTCTATCTTGGCTGATTCAAAAGCAGTTCCAGT
    TAAAATCACTCTGCCATTGGTAAACAACCAAGGTGTTGTTAAAGATGCTC
    ACATTTATCCAAAGAATACTGAAACAAAACCACAAGTAGATAAGAACTTT
    GCAGATAAAGATCTTGATTATACTGACAACCGAAAAGACAAAGGTGTTGT
    CTCAGCGACAGTTGGTGACAAAAAAGAATACATAGTTGGAACAAAAATTC
    TTAAAGGCTCAGACTATAAGAAACTGGTTTGGACTGATAGCATGACTAAA
    GGTTTGACGTTCAACAACAACGTTAAAGTAACATTGGATGGTGAAGATTT
    TCCTGTTTTAAACTACAAACTCGTAACAGATGACCAAGGTTTCCGTCTTG
    CCTTGAATGCAACAGGTCTTGCAGCAGTAGCAGCAGCTGCAAAAGACAAA
    GATGTTGAAATCAAGATCACTTACTCAGCTACGGTGAACGGCTCCACTAC
    TGTTGAAATTCCAGAAACCAATGATGTTAAATTGGACTATGGTAATAACC
    CAACGGAAGAAAGTGAACCACAAGAAGGTACTCCAGCTAACCAAGAAATT
    AAAGTCATTAAAGACTGGGCAGTAGATGGTACAATTACTGATGCTAATGT
    TGCAGTTAAAGCTATCTTTACCTTGCAAGAAAAACAAACGGATGGTACAT
    GGGTGAACGTTGCTTCACACGAAGCAACAAAACCATCACGCTTTGAACAT
    ACTTTCACAGGTTTGGATAATGCTAAAACTTACCGCGTTGTCGAACGTGT
    TAGCGGCTACACTCCAGAATACGTATCATTTAAAAATGGTGTTGTGACTA
    TCAAGAACAACAAAAACTCAAATGATCCAACTCCAATCAACCCATCAGAA
    CCAAAAGTGGTGACTTATGGACGTAAATTTGTGAAAACAAATCAAGCTAA
    CACTGAACGCTTGGCAGGAGCTACCTTCCTCGTTAAGAAAGAAGGCAAAT
    ACTTGGCACGTAAAGCAGGTGCAGCAACTGCTGAAGCAAAGGCAGCTGTA
    AAAACTGCTAAACTAGCATTGGATGAAGCTGTTAAAGCTTATAACGACTT
    GACTAAAGAAAAACAAGAAGGCCAAGAAGGTAAAACAGCATTGGCTACTG
    TTGATCAAAAACAAAAAGCTTACAATGACGCTTTTGTTAAAGCTAACTAC
    TCATATGAATGGGTTGCAGATAAAAAGGCTGATAATGTTGTTAAATTGAT
    CTCTAACGCCGGTGGTCAATTTGAAATTACTGGTTTGGATAAAGGCACTT
    ATGGCTTGGAAGAAACTCAAGCACCAGCAGGTTATGCGACATTGTCAGGT
    GATGTAAACTTTGAAGTAACTGCCACATCATATAGCAAAGGGGCTACAAC
    TGACATCGCATATGATAAAGGCTCTGTAAAAAAAGATGCCCAAGAAGTTC
    AAAACAAAAAAGTAACCATCCCACAAACAGGTGGTATTGGTACAATTCTT
    TTCACAATTATTGGTTTAAGCATTATGCTTGGAGCAGTAGTTATCATGAA
    AAAACGTCAATCAGAGGAAGCTTAA
    SEQ ID NO: 128
    MKKINKCLTMFSTLLLILTSLFSVAPAFADDATTDTVTLHKIVMPQAAFD
    NFTEGTKGKNDSDYVGKQINDLKSYFGSTDAKEIKGAFFVFKNETGTKFI
    TENGKEVDTLEAKDAEGGAVLSGLTKDNGFVFNTAKLKGIYQIVELKEKS
    NYDNNGSILADSKAVPVKITLPLVNNQGVVKDAHIYPKNTETKPQVDKNF
    ADKDLDYTDNRKDKGVVSATVGDKKEYIVGTKILKGSDYKKLVWTDSMTK
    GLTFNNNVKVTLDGEDFPVLNYKLVTDDQGFRLALNATGLAAVAAAAKDK
    DVEIKITYSATVNGSTTVEIPETNDVKLDYGNNPTEESEPQEGTPANQEI
    KVIKDWAVDGTITDANVAVKAIFTLQEKQTDGTWVNVASHEATKPSRFEH
    TFTGLDNAKTYRVVERVSGYTPEYVSFKNGVVTIKNNKNSNDPTPINPSE
    PKVVTYGRKFVKTNQANTERLAGATFLVKKEGKYLARKAGAATAEAKAAV
    KTAKLALDEAVKAYNDLTKEKQEGQEGKTALATVDQKQKAYNDAFVKANY
    SYEWVADKKADNVVKLISNAGGQFEITGLDKGTYGLEETQAPAGYATLSG
    DVNFEVTATSYSKGATTDIAYDKGSVKKDAQQVQNKKVTIPQTGGIGTIL
    FTIIGLSIMLGAVVIMKKRQSEEA
  • The GBS 59 polypeptides contain an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 129 IPQTG (shown in italics in SEQ ID NOs: 126 and 128 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 59 protein from the host cell. Alternatively, in some recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Pilin motifs, containing conserved lysine (K) residues have been identified in the GBS 59 polypeptides. The pilin motif sequences are underlined in each of SEQ ID NOs: 126 and 128, below. Conserved lysine (K) residues are marked in bold. The conserved lysine (K) residues are located at amino acid residues 202 and 212 and amino acid residues 489 and 495 of SEQ ID NO: 126 and at amino acid residues 188 and 198 of SEQ ID NO: 128. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of GBS 59. Preferred fragments of GBS 59 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 126
    MKRINKYFAMFSALLLTLTSLLSVAPAFADEATTNTVTLHKILQTESNLN
    KSNFPGTTGLNGKDYKGGATSDLAGYFGEGSKSIEGAFFALALKEDKSGK
    VQYVKAKEGNKLTPALINKDGTPEITVNIDEAVSGLTPEGDTGLVFNTKG
    LKGEFKIVEVKSKSTYNNNGSLLAASKAVPVNITLPLVNEDGVVADAHVY
    PKNTEEKPEIDK NFAKTNDLTALTDVNRLLTAGANYGNYARDKATATAET
    GKVVPYEVKTKIHKGSKYENLVWTDIMSNGLTMGSTVSLKASGTTETFAK
    DTDYELSIDARGFTLKFTADGLGKLEKAAKTADIEFTLTYSATVNGQAII
    DNPESNDIKLSYGNKPGKDLTELPVTPSKGEVTVAKTWSDGIAPDGVNVV
    YTLKDKDKTVASVSLTKTSKGTIDLGNGIKEEVSGNFSGKFTGLENKSYM
    ISERVSGYGSAINLENGKVTITNTKDSDNPTPLNPTEPKVETHGK KFVKT
    NEQGDRLAGAQFVVKNSAGKYLALKADQSEGQKTLAAKKIALDEAIAAYN
    KLSATDQKGEKGITAKELIKTKQADYDAAFIEARTAYEWITDKARAITYT
    SNDQGQFEVTGLADGTYNLEETLAPAGFAKLAGNIKFVVNQGSYITGGNI
    DYVANSNQKDATRVENKKVTIPQTGGIGTILFTIIGLSIMLGAVVIMKRR
    QSKEA
    SEQ ID NO: 128
    MKKINKCLTMFSTLLLILTSLFSVAPAFADDATTDTVTLHKIVMPQPAFD
    NFTEGTKGKNDSDYVGKQINDLKSYFGSTDAKEIKGAFFVFKNETGTKFI
    TENGKEVDTLEAKDAEGGAVLSGLTKDNGFVFNTAKLKGIYQIVELKEKS
    NYDNNGSILADSKAVPVKITLPLVNNQGVVKDAHIYPKNTETKPQVDK NF
    ADKDLDYTDNRKDKGVVSATVGDKKEYIVGTKILKGSDYKKLVWTDSMTK
    GLTFNNNVKVTLDGEDFPVLNYKLVTDDQGFRLALNATGLAAVAAAAKDK
    DVEIKITYSATVNGSTTVEIPETNDVKLDYGNNPTEESEPQEGTPANQEI
    KVIKDWAVDGTITDANVAVKAIFTLQEKQTDGTWVNVASHEATKPSRFEH
    TFTGLDNAKTYRVVERVSGYTPEYVSFKNGVVTIKNNKNSNDPTPINPSE
    PKVVTYGRKFVKTNQANTERLAGATFLVKKEGKYLARKAGAATAEAKAAV
    KTAKLALDEAVKAYNDLTKEKQEGQEGKTALATVDQKQKAYNDAFVKANY
    SYEWVADKKADNVVKLISNAGGQFEITGLDKGTYGLEETQAPAGYATLSG
    DVNFEVTATSYSKGATTDIAYDKGSVKKDAQQVQNKKVTIPQTGGIGTIL
    FTIIGLSIMLGAVVIMKKRQSEEA
  • An E box containing a conserved glutamic residue has also been identified in each of the GBS 59 polypeptides. The E box motif is underlined in each of SEQ ID NOs: 126 and 128 below. The conserved glutamic acid (E) is marked in bold at amino acid residue 621 in SEQ ID NO: 126 and at amino acid residue 588 in SEQ ID NO: 128. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of GBS 59. Preferred fragments of GBS 59 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 126
    MKRINKYFAMFSALLLTLTSLLSVAPAFADEATTNTVTLHKILQTESNLN
    KSNFPGTTGLNGKDYKGGAISDLAGYFGEGSKEIEGAFFALALKEDKSGK
    VQYVKAKEGNKLTPALINKDGTPEITVNIDEAVSGLTPEGDTGLVFNTKG
    LKGEFKIVEVKSKSTYNNNGSLLAASKAVPVNITLPLVNEDGVVADAHVY
    PKNTEEKPEIDKNFAKTNDLTALTDVNRLLTAGANYGNYARDKATATAEI
    GKVVPYEVKTKIHKGSKYENLVWTDIMSNGLTMGSTVSLKASGTTETFAK
    DTDYELSIDARGFTLKFTADGLGKLEKAAKTADIEFTLTYSATVNGQAII
    DNPESNDIKLSYGNKPGKDLTELPVTPSKGEVTVAKTWSDGIAPDGVNVV
    YTLKDKDKTVASVSLTKTSKGTIDLGNGIKFEVSGNFSGKFTGLENKSYM
    ISERVSGYGSAINLENGKVTITNTKDSDNPTPLNPTEPKVETHGKKEVKT
    NEQGDRLAGAQFVVKNSAGKYLALKADQSEGQKTLAAKKIALDEAIAAYN
    KLSATDQKGEKGITAKELIKTKQADYDAAFIEARTAYEWITDKARAITYT
    SNDQGQFEVTGLADGTYNLEETLAPAGFAKLAGNIKEVVNQGSYITGGNI
    DYVANSNQKDATRVENKKVTIPQTGGIGTILFTIIGLSIMLGAVVIMKRR
    QSKEA
    SEQ ID NO: 128
    MKKINKCLTMFSTLLLILTSLFSVAPAFADDATTDTVTLHKIVMPQAAFD
    NFTEGTKGKNDSDYVGKQINDLKSYFGSTDAKEIKGAFFVFKNETGTKFI
    TENGKEVDTLEAKDAEGGAVLSGLTKDNGFVFNTAKLKGIYQIVELKEKS
    NYDNNGSILADSKAVPVKITLPLVNNQGVVKDAHIYPKNTETKPQVDKNF
    ADKDLDYTDNRKDKGVVSATVGDKKEYTVGTKILKGSDYKKLVWTDSMTK
    GLTFNNNVKVTLDGEDFPVLNYKLVTDDQGFRLALNATGLAAVAAAAKDK
    DVEIKITYSATVNGSTTVEIPETNDVKLDYGNNPTEESEPQEGTPANQEI
    KVIKDWAVDGTITDANVAVKAIFTLQEKQTDGTWVNVASHEATKPSRFEH
    TFTGLDNAKTYRVVERVSGYTPEYVSFKNGVVTIKNNKNSNDPTPINPSE
    PKVVTYGRKFVKTNQANTERLAGATFLVKKEGKYLARKAGAATAEAKAAV
    KTAKLALDEAVKAYNDLTKEKQEGQEGKTALATVDQKQKAYNDAFVKANY
    SYEWVADKKADNVVKLISNAGGQFEITGLDKGTYGLEETQAPAGYATLSG
    DVNFEVTATSYSKGATTDIAYDKGSVKKKAQQVQNKKVTIPQTGGIGTIL
    FTIIGLSIMLGAVVIMKKRQSEEA
  • Female mice were immunized with either SAG1407 (SEQ ID NO: 126) or BO1575 (SEQ ID NO: 128) in an active maternal immunization assay. Pups bred from the immunized female mice survived GBS challenge better than control (PBS) treated mice. Results of the active maternal immunization assay using the GBS 59 immunogenic compositions are shown in Table 17, below.
    TABLE 17
    Active maternal immunization assay for GBS 59
    Challenge GBS 59 PBS
    GBS strain Survival Survival
    (serotype) Dead/treated (%) Dead/treated (%) FACS
    CJB111 (V)*  7/20 65 41/49 16 493
    18RS21 (II)** 18/30 40 39/40 2.5 380

    *immunized with BO1575

    **immunized with SAG1407
  • Opsonophagocytosis assays also demonstrated that antibodies against BO 1575 are opsonic for GBS serotype V, strain CJB111. See FIG. 67.
  • GBS 52
  • Examples of polynucleotide and amino acid sequences for GBS 52 are set forth below. SEQ ID NO: 20 and 21 represent GBS 52 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 20
    ATGAAACAAACATTAAAACTTATGTTTTCTTTTCTGTTGATGTTAGGGAC
    TATGTTTGGAATTAGCCAAACTGTTTTAGCGCAAGAAACTCATCAGTTGA
    CGATTGTTGATCTTGAAGCAAGGGATATTGATCGTCCAAATCCACAGTTG
    GAGATTGCCCCTAAAGAAGGGACTCCAATTGAAGGAGTACTCTATCAGTT
    GTACCAATTAAAATCAACTGAAGATGGGGATTTGTTGGCAGATTGGAATT
    CCCTAACTATCACAGAATTGAAAAAACAGGCGCAGCAGGTTTTTGAAGCG
    ACTACTAATCAACAAGGAAAGGCTACATTTAACCAACTACCAGATGGAAT
    TTATTATGGTCTGGCGGTTAAAGCCGGTGAAAAAAATCGTAATGTCTCAG
    CTTTCTTGGTTGACTTGTCTGAGGATAAAGTGATTTATCCTAAAATCATC
    TGGTCCACAGGTGAGTTGGACTTGCTTAAAGTTGGTGTGGATGGTGATAC
    CAAAAAACCACTAGCAGGCGTTGTCTTTGAACTTTATGAAAAGAATGGTA
    GGACTCCTATTCGTGTGAAAAATGGGGTGCATTCTCAAGATATTGACGCT
    GCAAAACATTTAGAAACAGATTCATCAGGGCATATCAGAATTTCCGGGCT
    CATCCATGGGGACTATGTCTTAAAAGAAATCGAGACACAGTCAGGATATC
    AGATCGGACAGGCAGAGACTGCTGTGACTATTGAAAAATCAAAAACAGTA
    ACAGTAACGATTGAAAATAAAAAAGTTCCGACACCTAAAGTGCCATCTCG
    AGGAGGTCTTATTCCCAAAACAGGTGAGCAACAGGCAATGGCACTTGTAA
    TTATTGGTGGTATTTTAATTGCTTTAGCCTTACGATTACTATCAAAACAT
    CGGAAACATCAAAATAAGGAT
    SEQ ID NO: 21
    MKQTLKLMFSFLLMLGTMFGISQTVLAQETHQLTIVHLEARDIDRPNPQL
    EIAPKEGTPIEGVLYQLYQLKSTEDGDLLAHWNSLTITELKKQAQQVFEA
    TTNQQGKATFNQLPDGIYYGLAVKAGEKNRNVSAFLVDLSEDKVIYPKII
    WSTGELDLLKVGVDGDTKKPLAGVVFELYEKNGRTPTRVKNGVHSQDIDA
    AKHLETDSSGHIRISGLIHGDYVLKEIETQSGYQIGQAETAVTIEKSKTV
    TVTIENKKVPTPKVPSRGGLIPKTGEQQAMALVIIGGILIALALRLLSKH
    RKHQNKD
  • GBS 52 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 124 IPKTG (shown in italics in SEQ ID NO: 21, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 52 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in GBS 52. The pilin motif sequence is underlined in SEQ ID NO: 21, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 148 and 160. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of GBS 52 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 21
    MKQTLKLMFSFLLMLGTMFGISQTVLAQETHQLTIVHLEARDIDRPNPQL
    EIAPKEGTPIEGVLYQLYQLKSTEDGDLLAHWNSLTITELKKQAQQVFEA
    TTNQQGKATFNQLPDGIYYGLAVKAGEKNRNVSAFLVDLSEDKVIYPKII
    WSTGELDLLK VGVDGDTKKPLAGVVFELYEKNGRTPTRVKNGVHSQDIDA
    AKHLETDSSGHIRISGLIHGDYVLKEIETQSGYQIGQAETAVTIEKSKTV
    TVTIENKKVPTPKVPSRGGLIPKTGEQQAMALVIIGGILIALALRLLSKH
    RKHQNKD
  • An E box containing a conserved glutamic residue has been identified in GBS 52. The E-box motif is underlined in SEQ ID NO: 21, below. The conserved glutamic acid (E), at amino acid residue 226, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of GBS 52. Preferred fragments of GBS 52 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 21
    MKQTLKLMFSFLLMLGTMFGISQTVLAQETHQLTIVHLEARDIDRPNPQL
    EIAPKEGTPIEGVLYQLYQLKSTEDGDLLAHWNSLTITELKKQAQQVFEA
    TTNQQGKATFNQLPDGIYYGLAVKAGEKNRNVSAFLVDLSEDKVIYPKII
    WSTGELDLLKVGVDGDTKKPLAGVVFELYEKNGRTPTRVKNGVHSQDIDA
    AKHLETDSSGHIRISGLIHGDYVLKEIETQSGYQIGQAETAVTIEKSKTV
    TVTIENKKVPTPKVPSRGGLIPKTGEQQAMALVIIGGILIALALRLLSKH
    RKHQNKD

    SAG0647
  • Examples of polynucleotide and amino acid sequences for SAG0647 are set forth below. SEQ ID NO: 22 and 23 represent SAG0647 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 22
    ATGGGACAAAAATCAAAAATATCTCTAGCTACGAATATTCGTATATGGAT
    TTTTCGTTTAATTTTCTTAGCGGGTTTCCTTGTTTTGGCATTTCCCATCG
    TTAGTCAGGTCATGTACTTTCAAGCCTCTCACGCCAATATTAATGCTTTT
    AAAGAAGGTGTTACCAAGATTGACCGGGTGGAGATTAATCGGCGTTTAGA
    ACTTGCTTATGCTTATAAGGCCAGTATAGCAGGTGCCAAAACTAATGGCG
    AATATCCAGCGCTTAAAGACCCCTACTCTGGTGAACAAAAGCAGGCAGGG
    GTCGTTGAGTACGCCCGCATGCTTGAAGTCAAAGAACAAATAGGTCATGT
    GATTATTCCAAGAATTAATCAGGATATCCCTATTTACGCTGGCTCTGCTG
    AAGAAAATCTTCAGAGGGGCGTTGGACATTTAGAGGGGACCAGTCTTCCA
    GTCGGTGGTGAGTCAACTCATGCCGTTGTAACTGCCCATCGAGGGCTACC
    AACGGCCAAGCTATTTACGAATTTAGACAAGGTAACAGTAGGTGACCGTT
    TTTACATTGAAGACATCGGCGGAAAGATTGCTTATCAGGTAGACCAAATC
    AAAGTTATCGGCCCTGATCAGTTAGAGGATTTGTACGTGATTCAAGGAGA
    AGATCACGTCACCCTATTAACTTGCACAGCTTATATGATAAATAGTCATC
    GCCTCCTCGTTCGAGGCAAGCGAATTCCTTATGTGGAAAAAACAGTGCAG
    AAAGATTCAAAGACCTTCAGGCAACAACAATACCTAACCTATGCTATGTG
    GGTAGTCGTTGGACTTATGTTGCTGTCGCTTCTCATTTGGTTTAAAAAGA
    CGAAACAGAAAAAGCGGAGAAAGAATGAAAAAGCGGCTAGTCAAAATAGT
    CACAATAATTCGAAATAA
    SEQ ID NO: 23
    MGQKSKISLATNIRIWIERLIFLAGFLVLAFPIVSQVMYFQASHANINAF
    KEAVTKIDRVEINRRLELAYAYNASIAGAKTNGEYPALKDPYSAEQKQAG
    VVEYARMLEVKEQIGHVIIPRINQDIPIYAGSAEENLQRGVGHLEGTSLP
    VGGESTHAVLTAHRGLPTAKLFTNLDKVTVGDRFYIEHIGGKIAYQVDQI
    KVIAPDQLEDLYVIQGEDHVTLLTCTPYMINSHRLLVRGKRIPYVEKTVQ
    KDSKTFRQQQYLTYAMWVVVGLILLSLLIWFKKTKQKKRRKNEKAASQNS
    HNNSK

    SAG0648
  • Examples of polynucleotide and amino acid sequences for SAG0648 are set forth below. SEQ ID NO: 24 and 25 represent SAG0648 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 24
    ATGGGAAGTCTGATTCTCTTATTTCCGATTGTGAGCCAGGTAAGTTACTA
    CCTTGCTTCGCATCAAAATATTAATCAATTTAAGCGGGAAGTCGCTAAGA
    TTGATACTAATACGGTTGAACGACGCATCGCTTTAGCTAATGCTTACAAT
    GAGACGTTATCAAGGAATCCCTTGCTTATAGACCCTTTTACCAGTAAGCA
    AAAAGAAGGTTTGAGAGAGTATGCTCGTATGCTTGAAGTTCATGAGCAAA
    TAGGTCATGTGGCAATCCCAAGTATTGGGGTTGATATTCCAATTTATGCT
    GGAACATCCGAAACTGTGCTTCAGAAAGGTAGTGGGCATTTGGAGGGAAC
    CAGTCTTCCAGTGGGAGGTTTGTCAACCCATTCAGTACTAACTGCCCACC
    GTGGCTTGCCAACAGCTAGGCTATTTACCGACTTAAATAAAGTTAAAAAA
    GGCCAGATTTTCTATGTGACGAACATCAAGGAAACACTTGCCTACAAAGT
    CGTGTCTATCAAAGTTGTGGATCCAACAGCTTTAAGTGAGGTTAAGATTG
    TCAATGGTAAGGATTATATAACCTTGCTGACTTGCACACCTTACATGATC
    AATAGTCATGGTCTCTTGGTAAAAGGAGAGCGTATTCCTTATGATTCTAC
    CGAGGCGGAAAAGCACAAAGAACAAACGGTACAAGATTATCGTTTGTCAC
    TAGTGTTGAAGATACTACTAGTATTATTAATTGGACTCTTCATCGTGATA
    ATGATGAGAAGATGGATGGAACATCGTCAATAA
    SEQ ID NO: 25
    MGSLILLFPIVSQVSYYLASHQNINQFKREVAKIDTNTVERRIALANAYN
    ETLSRNPLLIDPFTSKQKEGLREYARMLEVHEQTGHVATPSIGVDIPIYA
    GTSETVLQKGSGHLEGTSLPVGGLSTHSVLTAHRGLPTARLFTDLNKVKK
    GQIFYVTNIKETLAYKVVSIKVVDPTALSEVKIVNGKDYITLLTCTPYMI
    NSHRLLVKGERIPYDSTEAEKHKEQTVQDYRLSLVLKILLVLLIGLFIVI
    MMRRWMQHRQ

    GBS 150
  • Examples of polynucleotide and amino acid sequences for GBS 150 are set forth below. SEQ ID NO: 26 and 27 represent GBS 150 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 26
    ATGAAAAAGATTAGAAAAAGTTTAGGACTTCTAGTATGTTGGTTTTTAGG
    ATTGGTACAATTAGCGTTTTTTTCGGTAGGCAGTGTAAATGCTGATACCC
    CTAATCAACTAACAATCACAGAGATAGGACTTCAGCGAAATACTACAGAG
    GAGGGGATTTCTTATGGTTTATGGACTGTGACTGACAAGTTAAAAGTTGA
    TTTATTGAGGCAAATGACAGATAGCGAATTGAAGGAGAAGTATAAGAGTA
    TGTTGACTTCTCCTAGTGATACTAATGGTCAGACAAAGATAGGACTGGGA
    AATGGTTCGTACTTTGGTCGTGCTTATAAAGCTGATGAAAGGGTTTCAAC
    AATAGTACCTTTTTATATTGAATTAGGAGATGATAAGTTATGAAATCAAT
    TACAGATAAATCGTAAGCGAAAAGTTGAAACAGGCGGATTAAAACTTATT
    AAATATACAAAAGAAGGAAAGATAAAGAAAAGGCTATCCGGAGTAATATT
    TGTATTATACGATAACCAGAATGAGGGAGTTCGCTTTAAAAATGGACGAT
    TTACGACGGATCAAGATGGGATTAGTTGATTAGTAACTGATGATAAGGGA
    GAAATTGAGGTTGAAGGTTTATTACGTGGTAAGTATATTTTTCGAGAAGC
    AAAAGCACTAACTGGTTACCGTATATGTATGAAGGATGGTGTAGTTGCTG
    TAGTTGCTAATAAAACACAGGAAGTAGAGGTAGAAAAcGAAAAAGAAACT
    CCTCCACCAACAAATCCTAAACCATCACAACCGCTTTTTGCACAATCATT
    TCTTCCTAAAACAGGAATGATTATTGGTGGAGGACTGACAATTCTTGGTT
    GTATTATTTTGGGAATTTTGTTTATCTTTTTAAGAAAAACTAAAAATAGC
    AAATCTGAAAGAAACGATACAGTA
    SEQ ID NO: 27
    MKKIRKSLGLLLCCFLGLVQLAFESVASVNADTPNQLTITQIGLQPNTTE
    EGISYRLWTVTDNLKVDLLSQMTDSELNQKYKSILTSPTDTNGQTKIALP
    NGSYFGRAYKADQSVSTIVPEYIELPDDKLSNQLQINPKRKVETGRLKLI
    KYTKEGKIKKRLSGVIFVLYDNQNQPVRFKNGRFTTDQDGITSLVTDDKG
    EIEVEGLLPGKYIFREAKALTGYRISMKDAVVAVVANKTQEVEVENEKET
    PPPTNPKPSQPLFPQSFLPKTGMIIGGGLTILGCIILGILFIFLRKTKNS
    KSERNDTV
  • GBS 150 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 130 LPKTG (shown in italics in SEQ ID NO: 27 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GBS 150 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • As discussed above, a pilin motif, containing a conserved lysine (K) residue has been identified in GBS 150. The pilin motif sequence is underlined in SEQ ID NO: 27, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 139 and 148. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of GBS 150. Preferred fragments of GBS 150 include a conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 27
    MKKIRKSLGLLLCCFLGLVQLAFFSVASVNADTPNQLTITQIGLQPNTTE
    EGISYRLWTVTDNLKVDLLSQMTDSELNQKYKSILTSPTDTNGQTKIALP
    NGSYFGRAYKADQSVSTIVPFYIELPDDKLSNQLQINPKRKVETGRLK LI
    KYTKEGKIKKRLSGVIFVLYDNQNQPVRFKNGRFTTDQDGITSLVTDDKG
    EIEVEGLLPGKYIFREAKALTGYRISMKDAVVAVVANKTQEVEVENEKET
    PPPTNPKPSQPLFPQSFLPKTGMIIGGGLTILGCTTLGILFIFLRKTKNS
    KSERNDTV
  • An E box containing a conserved glutamic residue has also been identified in GBS 150. The E box motif is underlined in SEQ ID NO: 27 below. The conserved glutamic acid (E), at amino acid residue 216, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of GBS 150. Preferred fragments of GBS 150 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 27
    MKKIRKSLGLLLCCFLGLVQLAFFSVASVNADTPNQLTITQIGLQPNTTE
    EGISYRLWTVTDNLKVDLLSQMTDSELNQKYKSILTSPTDTNGQTKIALP
    NGSYFGRAYKADQSVSTIVPFYIELPDDKLSNQLQINPKRKVETGRLKLI
    KYTKEGKIKKRLSGVIFVLYDNQNQPVRFKNGRFTTDQDGITSLVTDDKG
    EIEVEGLLPGKYIFREAKALTGYRISMKDAVVAVVANKTQEVEVENEKET
    PPPTNPKPSQPLFPQSFLPKTGMIIGGGLTILGCIILGILFIPLRKTKNS
    KSERNDTV
  • Examples of polynucleotide and amino acid sequences for SAG1405 are set forth below. SEQ ID NO: 28 and 29 represent SAG1405 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 28
    ATGGGAGGAAAATTTCAGAAAAACCTTAAGAAATCGGTCGTTTTAAATCG
    ATGGATGAATGTAGGCTTGATACTATTGTTCTTAGTTGGTCTTTTGATAA
    CCTCATATCCTTTTATTTCAAATTGGTACTATAATATTAAAGCTAATAAT
    CAAGTAACTAACTTTGATAATCAAACCCAAAAATTAAATACTAAAGAGAT
    TAATAGACGATTTGAGTTAGCAAAAGCTTATAATAGAACACTGGACCCAA
    GCCGCCTATCAGATCCCTATACTGAAAAAGAAAAAAAAGGTATTGCTGAA
    TACGCCCACATGCTTGAGATTGCTGAAATGATTGGATATATTGATATACC
    GTCTATCAAGCAAAAATTACCTATCTATGCGGGGACTACCAGTAGTGTTC
    TTGAAAAAGGAGCAGGACACCTTGAAGGAACCTCCTTGCCAATTGGTGGA
    AAAAGTTCACATACTGTTATCACAGCTCATCGCGGCTTACCTAAAGCTAA
    GTTATTTACAGATTTAGATAAACTTAAAAAAGGAAAAATTTTTTATATTC
    ATAATATCAAAGAAGTTTTAGCCTATAAGGTTGATCAAATAAGTGTTGTA
    AAGCCAGATAATTTTTCTAAATTATTGGTTGTTAAAGGTAAGGATTATGC
    GACTTTGCTAACATGTACACCTTATTCGATTAATTCACATCGTTTACTAG
    TTAGAGGGCATCGAATCAAGTATGTACCTCCTGTTAAAGAAAAGAACTAT
    TTAATGAAAGAATTGCAAACACACTATAAACTTTATTTCCTCTTATCAAT
    CCTAGTTATTCTTATATTAGTCGCTTTACTATTATATTTAAAACGAAAAT
    TTAAAGAGAGAAAGAGAAAGGGAAATCAAAAATGA
    SEQ ID NO: 29
    MGGKFQKNLKKSVVLNRWMNVGLILLFLVGLLITSYPFISNWYYNIKANN
    QVTNFDNQTQKLNTKEINRRFELAKAYNRTLDPSRLSDPYTEKEKKGIAE
    YAHMLEIAEMIGYIDIPSIKQKLPIYAGTTSSVLEKGAGHLEGTSLPIGG
    KSSHTVITAHRGLPKAKLFTDLDKLKKGKIFYIHNIKEVLAYKVDQISVV
    KPDNFSKLLVVKGKDYATLLTCTPYSINSHRLLVRGHRIKYVPPVKEKNY
    LMKELQTRYKLYFLLSILVILILVALLLYLKRKPKERKRKGNQK

    SAG1406
  • Examples of polynucleotide and amino acid sequences for SAG1405 are set forth below. SEQ ID NO: 30 and 31 represent SAG1405 sequences from GBS serotype V, strain isolate 2603.
    SEQ ID NO: 30
    GTGAAGACTAAAAAAATCATCAAAAAAACAAAAAAAAAGAAGAAGTCAAA
    TCTTCCTTTTATCATTCTTTTTCTAATAGGTCTATCTATTTTATTGTATC
    CAGTGGTATCACGTTTTTACTATACGATAGAATCTAATAATCAAACACAG
    GATTTTGAGAGAGCTGCTAAAAAACTTAGTCAGAAAGAAATCAATCGACG
    TATGGCTCTAGCACAAGCTTATAATGATTCTTTAAATAATGTCCATCTTG
    AAGATCCTTATGAGAAAAAACGAATTCAAAAGGGGGTAGCAGAGTACGCC
    CGTATGTTAGAGGTAAGTGAAAAAATCGGAACAATTTCAGTTCCTAAGAT
    AGGTCAAAAACTCCCTATATTTGCAGGTTCAAGTCAAGAAGTTCTATCTA
    AAGGAGCAGGGCATTTAGAAGGTACCTCTCTTCCAATTGGGGGCAATAGT
    ACACATACTGTTATAACAGCGCATTCAGGAATTCCAGATAAAGAACTCTT
    TTCTAACCTTAAAAAGTTAAAAAAAGGAGATAAGTTTTATATTCAAAACA
    TAAAAGAAACGATAGCATATCAAGTAGATCAGATAAAAGTCGTTACACCC
    GATAACTTTTCAGATTTGTTGGTTGTTCCTGGACATGATTATGCAACCTT
    ATTGACTTGCACCCCGATTATGATCAATACACACAGACTTTTAGTAAGGG
    GACATCGTATCCCTTATAAAGGTCCTATTGATGAAAAATTAATAAAAGAC
    GGTCATTTAAACACGATTTATAGATATCTATTCTATATATCTTTAGTTAT
    TATTGCTTGGTTACTTTGGTTAATAAAACGTCAACGTCAAAAAAATCGTT
    TAGCAAGTGTTAGAAAAGGAATTGAATCATAA
    SEQ ID NO: 31
    MKTKKIIKKTKKKKKSNLPFIILFLIGLSILLYPVVSRFYYTIESNNQTQ
    DFERAAKKLSQKEINRRMALAQAYNDSLNNVHLEDPYEKKRIQKGVAEYA
    RMLEVSEKIGTISVPKIGQKLPIFAGSSQEVLSKGAGHLEGTSLPIGGNS
    THTVITAHSGIPDKELFSNLKKLKKGDKFYIQNIKETIAYQVDQIKVVTP
    DNFSDLLVVPGHDYATLLTCTPTMINTHRLLVRGHRIPYKGPIDEKLIKD
    GHLNTIYRYLFYISLVIIAWLLWLIKRQRQKNRLASVRKGIES

    01520
  • An example of an amino acid sequence for 01520 is set forth below. SEQ ID NO: 32 represents a 01520 sequence from GBS serotype III, strain isolate COH1.
    SEQ ID NO: 32
    MIRRYSANFLAILGIILVSSGIYWGWYNINQAHQADLTSQHIVKVLDKSI
    THQVKGSENGELPVKKLDKTDYLGTLDIPNLKLHLPVAANYSFEQLSKTP
    TRYYGSYLTNNMVICAHNFPYHFDALKNVDMGTDVYFTTTTGQIYHYKIS
    NREIIEPTAIEKVYKTATSDNDWDLSLFTCTKAGVARVLVRCQLIDVKN

    01521
  • An example of an amino acid sequence for 01521 is set forth below. SEQ ID NO: 33 represents a 01521 sequence from GBS serotype III, strain isolate COH1.
    SEQ ID NO: 33
    MIYKKILKITLLLLFSLSTQLVSADTNDQMKTGSITIQNKYNNQGIAGGN
    LLVYQVAQAKDVDGNQVFTLTTPFQGIGIKDDDLTQVNLDSNQAKYVNLL
    TKAVHKTQPLQTFDNLPAEGIVANNLPQGIYLFIQTKTAQGYELMSPFIL
    SIPKDGKYDITAFEKMSPLNAKPKKEETITPTVTHQTKGKLPFTGQVWWP
    IPILIMSGLLCLIIALKWRRRRD
  • 01521 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 132 LPFTG (shown in italics in SEQ ID NO: 33 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 01521 protein from the host cell. Alternatively, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, containing conserved lysine (K) residues have been identified in 01521. The pilin motif sequences are underlined in SEQ ID NO: 33, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 154 and 165 and at amino acid residues 174 and 188. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of 01521. Preferred fragments of 01521 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 33
    MIYKKILKITLLLLFSLSTQLVSADTNDQMKTGSITIQNKYNNQGIAGGN
    LLVYQVAQAKDVDGNQVFTLTTPFQGIGIKDDDLTQVNLDSNQAKYVNLL
    TKAVHKTQPLQTFDNLPAEGIVANNLPQGIYLFIQTKTAQGYELMSPFIL
    SIPKDGKYDITAFEK MSPLNAKPKKEETITPTVTHQTK GKLPFTGQVWWP
    TPILIMSGLLCLIIALKWRRRRD
  • An E box containing a conserved glutamic residue has also been identified in 01521. The E box motif is underlined in SEQ ID NO: 33 below. The conserved glutamic acid (E), at amino acid residue 177, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of 01521. Preferred fragments of 01521 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 33
    MIYKKILKTTLLLLFSLSTQLVSADTNDQMKTGSTTIQNKYNNQGIAGGN
    LLVYQVAQAKDVDGNQVFTLTTPFQGIGIKDDDLTQVNLDSNQAKYVNLL
    TKAVHKTQPLQTFDNLPAEGIVANNLPQGIYLFIQTKTAQGYELMSPFTL
    SIPKDGKYDITAFEKMSPLNAKPKKEETITPTVTHQTKGKLPFTGQWPIP
    ILIMSGLLCLIIALKWRRRRD

    01522
  • An example of an amino acid sequence for 01522 is set forth below. SEQ ID NO: 34 represents a 01522 sequence from GBS serotype III, strain isolate COH1.
    SEQ ID NO: 34
    MAYPSLANYWNSFHQSRAIMDYQDRVTHMDENDYKKITNRAKEYNKQFKT
    SGMKWHMTSQERLDYNSQLAIDKTGNMGYISIPKINIKLPLYHGTSEKVL
    QTSIGHLEGSSLPIGGDSTHSILSGHRGLPSSRLFSDLDKLKVGDHWTVS
    ILNETYTYQVDQIRTVKPDDLRDLQIVKGKDYQTLVTCTPYGVNTHRLLV
    RGHRVPNDNGNALVVAEAIQIEPIYIAPFIAIFLTLILLLISLEVTRRAR
    QRKKILKQAMRKEENNDL

    01523
  • An example of an amino acid sequence for 01523 is set forth below. SEQ ID NO: 35 represents a 01523 sequence from GBS serotype III, strain isolate COH1.
    SEQ ID NO: 35
    MKKKMIQSLLVASLAFGMAVSPVTPIAFAAETGTITVQDTQKGATYKAYK
    VFDAEIDNANVSDSNKDGASYLIPQGKEAEYKASTDFNSLFTTTTNGGRT
    YVTKKDTASANEIATWAKSISANTTPVSTVTESNNDGTEVINVSQYGYYY
    VSSTVNNGAVIMVTSVTPNATIHEKNTDATWGDGGGKTVDQKTYSVGDTV
    KYTITYKNAVNYHGTEKVYQYVIKDTMPSASVVDLNEGSYEVTITDGSGN
    ITTLTQGSEKATGKYNLLEENNNFTITIPWAATNTPTGNTQNGANDDFFY
    KGINTITVTYTGVLKSGAKPGSADLPENTNIATINPNTSNDDPGQKVTVR
    DGQITIKKIDGSTKASLQGAIFVLKNATGQFLNFNDTNNVEWGTEANATE
    YTTGADGIITITGLKEGTYYLVEKKAPLGYNLLDNSQKVILGDGATDTTN
    SDNLLVNPTVENNKGTELPSTGGIGTTIEYIIGAILVIGAGIVLVARRRL
    RS
  • 01523 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 131 LPSTG (shown in italics in SEQ ID NO: 35 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 01523 protein from the host cell. Alternatively, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • An E box containing a conserved glutamic residue has also been identified in 01523. The E box motif is underlined in SEQ ID NO: 35 below. The conserved glutamic acid (E), at amino acid residue 423, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of 01523. Preferred fragments of 01523 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 35
    MKKKMIQSLLVASLAFGMAVSPVTPIAFAAETGTITVQDTQKGATYKAYK
    VFDAEIDNANVSDSNKDGASYLIPQGKEAEYKASTDFNSLFTTTTNGGRT
    YVTKKDTASANEIATWAKSISANTTPVSTVTESNNDGTEVINVSQYGYYY
    VSSTVNNGAVIMVTSVTPNATIHEKNTDATWGDGGGKTVDQKTYSVGDTV
    KYTITYKNAVNYHGTEKVYQYVIKDTMPSASVVDLNEGSYEVTITDGSGN
    ITTLTQGSEKATGKYNLLEENNNFTITIPWAATNTPTGNTQNGANDDFFY
    KGINTITVTYTGVLKSGAKPGSADLPENTNIATINPNTSNDDPGQKVTVR
    DGQITIKKIDGSTKASLQGAIFVLKNATGQFLNFNDTNNVEWGTEANATE
    YTTGADGIITITGLKEGTYYLVEKKAPLGYNLLDNSQKVILGDGATDTTN
    SDNLLVNPTVENNKGTELPSTGGIGTTIEYIIGAILVIGAGIVLVARRRL
    RS

    01524
  • An example of an amino acid sequence for 01524 is set forth below. SEQ ID NO: 36 represents a 01524 sequence from GBS serotype m, strain isolate COH1.
    SEQ ID NO: 36
    MLKKCQTFIIESLKKKKHPKEWKIIMWSLMILTTFLTTYFLILPAITVEE
    TKTDDVGITLENKNSSQVTSSTSSSQSSVEQSKPQTPASSVTETSSSEEA
    AYREEPLMFRGADYTVTVTLTKEAKIPKNADLKVTELKDNSATFKDYKKK
    ALTEVAKQDSEIKNFKLYDITIESNGKEAEPQAPVKVEVNYDKPLEASDE
    NLKVVHFKDDGQTEVLKSKDTAETKNTSSDVAFKTDSFSTYAIVQEDNTE
    VPRLTYHFQNNDGTDYDPLTASGMQVHHQIIKDGESLGEVGIPTIKAGEH
    FNGWYTYDPTTGKYGDPVKEGEPITVTETKEICVRPFMSKVATVTLYDDS
    AGKSILERYQVPLDSSGNGTADLSSFKVSPPTSTLLFVGWSKTQNGAPLS
    ESEIQALPVSSDISLYPVFKESYGVEFNTGDLSTGVTYIAPRRVLTGQPA
    STIKPNDPTRPGYTFAGWYTAASGGAAFDFNQVLTKDTTLYAHWSPAQTT
    YTINYWQQSATDNKNATDAQKTYEYAGQVTRSGLSLSNQTLTQQDINDKL
    PTGFKVNNTRTETSVMIKDDGSSVVNVYYDRKLITIKFAKYGGYSLPEYY
    YSYNWSSDADTYTGLYGTTLAANGYQWKTGAWGYLANVGNNQVGTYGMSY
    LGEFILPNDTVDSDVIKLFPKGNIVQTYRFTKQGLDGTYSLADTGGGAGA
    DEFTFTEKYLGFNVKYYQRLYPDNYLFDQYASQTSAGVKVPISDEYYDRY
    GAYHKDYLNLVVWYERNSYKIKYLDPLDNTELPNFPVKDVLYEQNLSSYA
    PDTTTVQPKPSRPGYVWDGKWYKDQAQTQVFDFNTTMPPHDVKVYAGWQK
    VTYRVNIDPNGGRLSKTDDTYLDLHYGDRIPDYTDITRDYIQDPSGTYYY
    KYDSRDKDPDSTKDAYYTTDTSLSNVDTTTKYKYVKDAYKLVGWYYVNPD
    GSIRPYNFSGAVTQDINLRAIWRKAGDYHIIYSNDAVGTDGKPALDASGQ
    QLQTSNEPTDPDSYDDGSHSALLRRPTMPDGYRFRGWWYNGKIYNPYDSI
    DIDAHLADANKNITIKPVIIPVGDIKLEDTSIKYNGNGGTRVENGNVVTQ
    VETPRMELNSTTTIPENQYFTRTGYNLIGWHHDKDLADTGRVEFTAGQSI
    GIDNNPDATNTLYAVWQPKEYTVRVSKTVVGLDEDKTKDFLFNPSETLQQ
    ENFPLRDGQTKEEKVPYGTSISIDEQAYDEFKVSESITEKNLATGEADKT
    YDATGLQSLTVSGDVDISFTNTRIKQKVRLQKVNVENDNNFLAGAVFDIY
    ESDANGNKASHPMYSGLVTNDKGLLLVDANNYLSLPVGKYYLTETKAPPG
    YLLPKNDISVLVISTGVTFEQNGNNATPIKENLVDGSTVYTFKITNSKGT
    ELPSTGGIGTHIYILVGLALALPSGLILYYRKKI
  • 01524 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 131 LPSTG (shown in italics in SEQ ID NO: 36 above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 01524 protein from the host cell. Alternatively, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Three pilin motifs, containing conserved lysine (K) residues have been identified in 01524. The pilin motif sequences are underlined in SEQ ID NO: 36, below. Conserved lysine (K) residues are marked in bold, at amino acid residues 128 and 138, amino acid residues 671 and 682, and amino acid residues 809 and 820. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures of 01524. Preferred fragments of 01524 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 36
    MLKKCQTFIIESLKKKKHPKEWKIIMWSLMILTTFLTTYFLILPAITVEE
    TKTDDVGITLENKNSSQVTSSTSSSQSSVEQSKPQTPASSVTETSSSEEA
    AYREEPLMFRGADYTVTVTLTKEAKIPKNADLKVTELK DNSATFKDYKKK
    ALTEVAKQDSEIKNFKLYDITIESNGKEAEPQAPVKVEVNYDKPLEASDE
    NLKVVHFKDDGQTEVLKSKDTAETKNTSSDVAFKTDSFSIYAIVQEDNTE
    VPRLTYHFQNNDGTDYDPLTASGMQVHHQIIKDGESLGEVGIPTIKAGEH
    FNGWYTYDPTTGKYGDPVKFGEPITVTETKEICVRPFMSKVATVTLYDDS
    AGKSILERYQVPLDSSGNGTADLSSFKVSPPTSTLLFVGWSKTQNGAPLS
    ESEIQALPVSSDISLYPVFKESYGVEFNTGDLSTGVTYIAPRRVLTGQPA
    STIKPNDPTRPGYTFAGWYTAASGGAAFDFNQVLTKDTTLYAHWSPAQTT
    YTINYWQQSATDNKNATDAQKTYEYAGQVTRSGLSLSNQTLTQQDINDKL
    PTGFKVNNTRTETSVMIKDDGSSVVNVYYDRKLITIKFAKYGGYSLPEYY
    YSYNWSSDADTYTGLYGTTLAANGYQWKTGAWGYLANVGNNQVGTYGMSY
    LGEFILPNDTVDSDVIKLFPKGNIVQTYRFFK QGLDGTYSLADTGGGAGA
    DEFTFTEKYLGFNVKYYQRLYPDNYLFDQYASQTSAGVKVPISDEYYDRY
    GAYHKDYLNLVVWYERNSYKIKYLDPLDNTELPNFPVKDVLYEQNLSSYA
    PDTTTVQPKPSRPGYVWDGK WYKDQAQTQVFDFNTTMPPHDVKVYAGWQK
    VTYRVNIDPNGGRLSKTDDTYLDLHYGDRIPDYTDITRDYIQDPSGTYYY
    KYDSRDKDPDSTKDAYYTTDTSLSNVDTTTKYKYVKDAYKLVGWYYVNPD
    GSIRPYNFSGAVTQDINLRAIWRKAGDYHIIYSNDAVGTDGKPALDASGQ
    QLQTSNEPTDPDSYDDGSHSALLRRPTMPDGYRFRGWWYNGKIYNPYDSI
    DIDAHLADANKNITIKPVIIPVGDIKLEDTSIKYNGNGGTRVENGNVVTQ
    VETPRMELNSTTTIPENQYFTRTGYNLIGWHHDKDLADTGRVEFTAGQSI
    GIDNNPDATNTLYAVWQPKEYTVRVSKTVVGLDEDKTKDFLFNPSETLQQ
    ENFPLRDGQTKEEKVPYGTSISIDEQAYDEFKVSESITEKNLATGEADKT
    YDATGLQSLTVSGDVDISFTNTRIKQKVRLQKVNVENDNNFLAGAVFDIY
    ESDANGNKASHPMYSGLVTNDKGLLLVDANNYLSLPVGKYYLTETKAPPG
    YLLPKNDISVLVISTGVTFEQNGNNATPIKENLVDGSTVYTFKITNSKGT
    ELPSTGGIGTHIYILVGLALALPSGLILYYRKKI
  • An E box containing a conserved glutamic residue has also been identified in 01524. The E box motif is underlined in SEQ ID NO: 36 below. The conserved glutamic acid (E), at amino acid residue 1344, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of 01524. Preferred fragments of 01524 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 36
    MLKKCQTFIIESLKKKKHPKEWKIIMWSLMILTTFLTTYFLILPAITVEE
    TKTDDVGITLENKNSSQVTSSTSSSQSSVEQSKPQTPASSVTETSSSEEA
    AYREEPLMFRGADYTVTVTLTKEAKIPKNADLKVTELKDNSATFKDYKKK
    ALTEVAKQDSEIKNFKLYDITIESNGKEAEPQAPVKVEVNYDKPLEASDE
    NLKVVHFKDDGQTEVLKSKDTAETKNTSSDVAFKTDSFSTYAIVQEDNTE
    VPRLTYHFQNNDGTDYDPLTASGMQVHHQIIKDGESLGEVGIPTIKAGEH
    FNGWYTYDPTTGKYGDPVKEGEPITVTETKEICVRPFMSKVATVTLYDDS
    AGKSILERYQVPLDSSGNGTADLSSFKVSPPTSTLLFVGWSKTQNGAPLS
    ESEIQALPVSSDISLYPVFKESYGVEFNTGDLSTGVTYIAPRRVLTGQPA
    STIKPNDPTRPGYTFAGWYTAASGGAAFDFNQVLTKDTTLYAHWSPAQTT
    YTINYWQQSATDNKNATDAQKTYEYAGQVTRSGLSLSNQTLTQQDINDKL
    PTGFKVNNTRTETSVMIKDDGSSVVNVYYDRKLITIKFAKYGGYSLPEYY
    YSYNWSSDADTYTGLYGTTLAANGYQWKTGAWGYLANVGNNQVGTYGMSY
    LGEFILPNDTVDSDVIKLFPKGNIVQTYRFTKQGLDGTYSLADTGGGAGA
    DEFTFTEKYLGFNVKYYQRLYPDNYLFDQYASQTSAGVKVPISDEYYDRY
    GAYHKDYLNLVVWYERNSYKIKYLDPLDNTELPNFPVKDVLYEQNLSSYA
    PDTTTVQPKPSRPGYVWDGKWYKDQAQTQVFDFNTTMPPHDVKVYAGWQK
    VTYRVNIDPNGGRLSKTDDTYLDLHYGDRIPDYTDITRDYIQDPSGTYYY
    KYDSRDKDPDSTKDAYYTTDTSLSNVDTTTKYKYVKDAYKLVGWYYVNPD
    GSIRPYNFSGAVTQDINLRAIWRKAGDYHIIYSNDAVGTDGKPALDASGQ
    QLQTSNEPTDPDSYDDGSHSALLRRPTMPDGYRFRGWWYNGKIYNPYDSI
    DIDAHLADANKNITIKPVIIPVGDIKLEDTSIKYNGNGGTRVENGNVVTQ
    VETPRMELNSTTTIPENQYFTRTGYNLIGWHHDKDLADTGRVEFTAGQSI
    GIDNNPDATNTLYAVWQPKEYTVRVSKTVVGLDEDKTKDFLFNPSETLQQ
    ENFPLRDGQTKEEKVPYGTSISIDEQAYDEFKVSESITEKNLATGEADKT
    YDATGLQSLTVSGDVDISFTNTRIKQKVRLQKVNVENDNNFLAGAVFDIY
    ESDANGNKASHPMYSGLVTNDKGLLLVDANNYLSLPVGKYYLTETKAPPG
    YLLPKNDISVLVISTGVTFEQNGNNATPIKENLVDGSTVYTFKITNSKGT
    ELPSTGGIGTHIYILVGLALALPSGLILYYRKKI

    01525
  • An example of an amino acid sequence for 01525 is set forth below. SEQ ID NO: 37 represents a 01525 sequence from GBS serotype III, strain isolate COH1.
    SEQ ID NO: 37
    MKRQISSDKLSQELDRVTYQKRFWSVIKNTIYILMAVASIAILIAVLWLP
    VLRIYGHSMNKTLSAGDVVFTVKGSNFKTGDVVAFYYNNKVLVKRVTAES
    GDWVNIDSQGDVYVNQHKLKEPYVIHKALGNSNIKYPYQVPDKKIFVLGD
    NRKTSIDSRSTSVGDVSEEQIVGKTSFRIWPLGKISSIN

    GBS 322
  • GBS 322 refers to a surface immunogenic protein, also referred to as “sip”. Nucleotide and amino acid sequences of GBS 322 sequenced from serotype V isolated strain 2603 V/R are set forth in Ref. 3 as SEQ ID 8539 and SEQ OD 8540. These sequences are set forth below as SEQ ID NOS 38 and 39:
    SEQ ID NO.38
    ATGAATAAAAAGGTACTATTGACATCGACAATGGCAGCTTCGCTATTATC
    AGTCGCAAGTGTTCAAGCACAAGAAACAGATACGACGTGGACAGCACGTA
    CTGTTTCAGAGGTAAAGGCTGATTTGGTAAAGCAAGACAATAAATCATCA
    TATACTGTGAAATATGGTGATACACTAAGCGTTATTTCAGAAGCAATGTC
    AATTGATATGAATGTCTTAGCAAAAATAAATAACATTGCAGATATCAATC
    TTATTTATCCTGAGACAACACTGACAGTAACTTACGATCAGAAGAGTCAT
    ACTGCCACTTCAATGAAAATAGAAACACCAGCAACAAATGCTGCTGGTCA
    AACAACAGCTACTGTGGATTTGAAAACCAATCAAGTTTCTGTTGCAGACC
    AAAAAGTTTCTCTCAATACAATTTCGGAAGGTATGACACCAGAAGCAGCA
    ACAACGATTGTTTGGCCAATGAAGACATATTCTTCTGCGCCAGCTTTGAA
    ATCAAAAGAAGTATTAGCACAAGAGCAAGCTGTTAGTCAAGCAGCAGCTA
    ATGAACAGGTATCACGAGCTCCTGTGAAGTCGATTACTTCAGAAGTTCCA
    GCAGCTAAAGAGGAAGTTAAACCAACTCAGACGTCAGTCAGTCAGTCAAC
    AACAGTATCACCAGCTTCTGTTGCCGCTGAAACACCAGCTCCAGTAGCTA
    AAGTAGCACCGGTAAGAACTGTAGCAGCCCCTAGAGTGGCAAGTGTTAAA
    GTAGTCACTCCTAAAGTAGAAACTGGTGCATCACCAGAGCATGTATCAGC
    TCCAGCAGTTCCTGTGACTACGACTTCACCAGCTACAGACAGTAAGTTAC
    AAGCGACTGAAGTTAAGAGCGTTCCGGTAGCACAAAAAGCTCCAACAGCA
    ACACCGGTAGCACAACCAGCTTCAACAACAAATGCAGTAGCTGCACATCC
    TGAAAATGCAGGGCTCCAACCTCATGTTGCAGCTTATAAAGAAAAAGTAG
    CGTCAACTTATGGAGTTAATGAATTCAGTACATACCGTGCGGGAGATCCA
    GGTGATCATGGTAAAGGTTTAGCAGTTGACTTTATTGTAGGTACTAATCA
    AGCACTTGGTAATAAAGTTGCACAGTACTCTACACAAAATATGGCAGCAA
    ATAACATTTCATATGTTATCTGGCAACAAAAGTTTTACTCAAATACAAAC
    AGTATTTATGGACCTGCTAATACTTGGAATGCAATGCCAGATCGTGGTGG
    CGTTACTGCCAACCACTATGACCACGTTCACGTATCATTTAACAAATAAT
    ATAAAAAAGGAAGCTATTTGGCTTCTTTTTTATATGCCTTGAATAGACTT
    TCAAGGTTCTTATATAATTTTTATTA
    SEQ ID NO.39
    MNKKVLLTSTMAASLLSVASVQAQETDTTWTARTVSEVKADLVKQDNKSS
    YTVKYGDTLSVISEAMSIDMNVLAKINNIADINLIYPETTLTVTYDQKSH
    TATSMKIETPATNAAGQTTATVDLKTNQVSVADQKVSLNTISEGMTPEAA
    TTIVSPMKTYSSAPALKSKEVLAQEQAVSQAAANEQVSPAPVKSITSEVP
    AAKEEVKPTQTSVSQSTTVSPASVAAETPAPVAKVAPVRTVAAPRVASVK
    VVTPKVETGASPEHVSAPAVPVTTTSPATDSKLQATEVKSVPVAQKAPTA
    TPVAQPASTTNAVAAHPENAGLQPHVAAYKEKVASTYGVNEFSTYRAGDP
    GDHGKGLAVDFIVGTNQALGNKVAQYSTQNMAANNISYVIWQQKFYSNTN
    SIYGPANTWNAMPDRGGVTANHYDHVHVSFNK
  • GBS 322 contains an N-terminal leader or signal sequence region which is indicated by the underlined sequence near the beginning of SEQ ID NO: 39. In one embodiment, one or more amino acids from the leader or signal sequence region of GBS 322 are removed. An example of such a GBS 322 fragment is set forth below as SEQ ID NO: 40.
    SEQ ID NO: 40
    DLVKQDNKSSYTVKYGDTLSVISEAMSIDMNVLAKINNIADINLIYPETT
    LTVTYDQKSHTATSMKIETPATNAAGQTTATVDLKTNQVSVADQKVSLNT
    ISEGMTPEAATTIVSPMKTYSSAPALKSKEVLAQEQAVSQAAANEQVSPA
    PVKSITSENPAAKEEVKPTQTSVSQSTTVSPASVAAETPAPVAKVAPVRT
    VAAPRVASVKVVTPKVETGASPEHVSAPAVPVTTTSPATDSKLQATEVKS
    VPVAQKAPTATPVAQPASTTNAVAAHPENAGLQPHVAAYKEKVASTYGVN
    EFSTYRAGDPGDHGKGLAVDFIVGTNQALGNKVAQYSTQNMAANNISYVI
    WQQKFYSNTNSIYGPANTWNAMPDRGGVTANHYDHVHVSFNK
  • Additional preferred fragments of GBS 322 comprise the immunogenic epitopes identified in WO 03/068813, each of which are specifically incorporated by reference herein.
  • There may be an upper limit to the number of GBS proteins which will be in the compositions of the invention. Preferably, the number of GBS proteins in a composition of the invention is less than 20, less than 19, less than 18, less than 17, less than 16, less than 15, less than 14, less than 13, less than 12, less than 11, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, or less than 3. Still more preferably, the number of GBS proteins in a composition of the invention is less than 6, less than 5, or less than 4. Still more preferably, the number of GBS proteins in a composition of the invention is 3.
  • The GBS proteins and polynucleotides used in the invention are preferably isolated, i.e., separate and discrete, from the whole organism with which the molecule is found in nature or, when the polynucleotide or polypeptide is not found in nature, is sufficiently free of other biological macromolecules so that the polynucleotide or polypeptide can be used for its intended purpose.
  • Group A Streptococcus Adhesin Island Sequences
  • The GAS AI polypeptides of the invention can, of course, be prepared by various means (e.g. recombinant expression, purification from GAS, chemical synthesis etc.) and in various forms (e.g. native, fusions, glycosylated, non-glycosylated etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other streptococcal or host cell proteins) or substantially isolated form.
  • The GAS AI proteins of the invention may include polypeptide sequences having sequence identity to the identified GAS proteins. The degree of sequence identity may vary depending on the amino acid sequence (a) in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more). Polypeptides having sequence identity include homologs, orthologs, allelic variants and functional mutants of the identified GBS proteins. Typically, 50% identity or more between two proteins is considered to be an indication of functional equivalence. Identity between proteins is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affinity gap search with parameters gap open penalty=12 and gap extension penalty=1.
  • The GAS adhesin island polynucleotide sequences may include polynucleotide sequences having sequence identity to the identified GAS adhesin island polynucleotide sequences. The degree of sequence identity may vary depending on the polynucleotide sequence in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more).
  • The GAS adhesin island polynucleotide sequences of the invention may include polynucleotide fragments of the identified adhesin island sequences. The length of the fragment may vary depending on the polynucleotide sequence of the specific adhesin island sequence, but the fragment is preferably at least 10 consecutive polynucleotides, (e.g. at least 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more).
  • The GAS adhesin island amino acid sequences of the invention may include polypeptide fragments of the identified GAS proteins. The length of the fragment may vary depending on the amino acid sequence of the specific GAS antigen, but the fragment is preferably at least 7 consecutive amino acids, (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more). Preferably the fragment comprises one or more epitopes from the sequence. Other preferred fragments include (1) the N-terminal signal peptides of each identified GAS protein, (2) the identified GAS protein without their N-terminal signal peptides, and (3) each identified GAS protein wherein up to 10 amino acid residues (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) are deleted from the N-terminus and/or the C-terminus e.g. the N-terminal amino acid residue may be deleted. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • GAS AI-1 Sequences
  • As discussed above, a GAS AI-1 sequence is present in an M6 strain isolate (MGAS10394). Examples of GAS AI-1 sequences from M6 strain isolate MGAS10394 are set forth below.
  • M6_Spy0156: Spy0156 is a rofA transcriptional regulator. An example of an amino acid sequence for M6_Spy0156 is set forth in SEQ ID NO: 41.
    SEQ ID NO: 41
    MIEKYLESSIESKCQLVVLEFKTSYLPITEVAEKTGLTFLQLNHYCEELN
    AFFPDSLSMTIQKRMISCQFTHPFKETYLYQLYASSNVLQLLAFLIKNGS
    HSRPLTDFARSHFLSNSSAYRMREALIPLLRNFELKLSKNKIVGEEYRIR
    YLIALLYSKFGIKVYDLTQQDKNTIHSFLSHSSTHLKTSPWLSESFSFYD
    ILLALSWKRHQFSVTIPQTRIFQQLKKLFIYDSLKKSSRDIIETYCQLNF
    SAGDLDYLYLIYITANNSFASLQWTPEHIRQCCQLFEENDTFRLLLKPII
    TLLPNLKEQKPSLVKALMFESKSFLENLQHFIPETNLFVSPYYKGNQKLY
    TSLKLIVEEWLAKLPGKRYLNHKHFHLFCHYVEQILRNIQPPLVVVFVAS
    NFINAHLLTDSFPRYFSDKSIDFHSYIAR
  • M6_Spy0157: M6_Spy0157 is a fibronectin binding protein. It contains a sortase substrate motif LPXTG (SEQ ID NO: 122), shown in italics in the amino acid sequence SEQ ID NO: 42.
    SEQ ID NO: 42
    MVSSYMFVRGEKMNNKIFLNKEASFLAHTKRKRRFAVTLVGVFFMLLACA
    GAIGFGQVAYAADEKTVPSHSSPNPEFPWYGYDAYGKEYPGYNIWTRYHD
    LRVNLNGSRSYQVYCFNIQSNYPSQKNSEIKNWFKKIEGNGKSFVDYAHT
    TKLGKEELEQRLLSLLYNAYPNDANGYMKGLEHLNAITVTQYAVWHYSDN
    SQYQFETLWESEAKEGKISRSQVTLMREALKKLIDPNLEATAVNKIPSGY
    RLNIFESENEAYQNLLSAEYVPDDPPKPGETSEHNPKTPELDGTPIPEDP
    KHPDDNLEPTLPPVMLDGEEVPEVPSESLEPALPPLMPELDGQEVPEKPS
    IDLPIEVPRYEFNNKDQSPLAGESGETEYITEVYGNQQNPVDIDKKLPNE
    TGFSGNMVETEDTKEPEVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDT
    KEPEVLMGGQSESVEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSES
    VEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSESVEFTKDTQTGMSG
    FSETATVVEDTRPKLVFHFDNNEPKVEENREKPTKNITPILPATGDIENV
    LAFLGILILSVLSIFSLLKNKQSNKKV
  • M6_Spy0157 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 180 LPATG (shown in italics in SEQ ID NO: 42, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant M6_Spy0157 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in M6_Spy0157. The pilin motif sequence is underlined in SEQ ID NO: 42, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 277, 287, and 301. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of M6_Spy0157 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 42
    MVSSYMFVRGEKMNNKTFLNKEASFLAHTKRKRRFAVTLVGVFFMLLACA
    GAIGFGQVAYAADEKTVPSHSSPNPEFPWYGYDAYGKEYPGYNIWTRYHD
    LRVNLNGSRSYQVYCFNIQSNYPSQKNSFIKNWFKKIEGNGKSFVDYAHT
    TKLGKEELEQRLLSLLYNAYPNDANGYMKGLEHLNAITVTQYAVWHYSDN
    SQYQFETLWESEAKEGKISRSQVTLMREALKKLIDPNLEATAVNKIPSGY
    RLNIFESENEAYQNLLSAEYVPDDPPKPGETSEHNPKTPELDGTPTPEDP
    K HPDDNLEPTLPPVMLDGEEVPEVPSESLEPALPPLMPELDGQEVPEKPS
    TDLPIEVPRYEFNNKDQSPLAGESGETEYITEVYGNQQNPVDIDKKLPNE
    TGFSGNMVETEDTKEPEVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDT
    KEPEVLMGGQSESVEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSES
    VEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSESVEFTKDTQTGMSG
    ESETATVVEDTRPKLVFHFDNNEPKVEENREKPTKNITPILPATGDIENV
    LAFLGILILSVLSIFSLLKNKQSNKKV
  • A repeated series of four E boxes containing a conserved glutamic residue have been identified in M6_Spy0157. The E-box motifs are underlined in SEQ ID NO: 42, below. The conserved glutamic acid (E) residues, at amino acid residues 415, 452, 489, and 526 are marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of M6_Spy0157. Preferred fragments of M6_Spy0157 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 42
    MVSSYMFVRGEKMNNKIFLNKEASFLAHTKRKRRFAVTLVGVFFMLLACA
    GAIGFGQVAYAADEKTVPSHSSPNPEFPWYGYDAYGKEYPGYNIWTRYHD
    LRVNLNGSRSYQVYCFNIQSNYPSQKNSFIKNWFKKIEGNGKSFVDYAHT
    TKLGKEELEQRLLSLLYNAYPNDANGYMKGLEHLNAITVTQYAVWHYSDN
    SQYQFETLWESEAKEGKISRSQVTLMREALKKLIDPNLEATAVNKIPSGY
    RLNIFESENEAYQNLLSAEYVPDDPPKPGETSEHNPKTPELDCTPIPEDP
    KHPDDNLEPTLPPVMLDGEEVPEVPSESLEPALPPLMPELDGQEVPEKPS
    IDLPIEVPRYEFNNKDQSPLAGESGETEYITEVYGNQQNPVDIDKKLPNE
    TGFSGNMVETEDTKEPEVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDT
    KEPEVLMGGQSESVEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSES
    VEFTKDTQTGMSGQTTPQIETEDTKEPEVLMGGQSESVEFTKDTQTGMSG
    FSETATVVEDTRPKLVFHFDNNEPKVEENREKPTKNITPILPATGDIENV
    LAFLGILILSVLSIFSLLKNKQSNKKV
  • M6_Spy0158: M6_Spy0158 is a reverse transcriptase. An example of Spy0158 is shown in the amino acid sequence SEQ ID NO 43.
  • SEQ ID NO: 43
  • MSLRHQNKKGIRKEGWKSRPQSRWSDHCQLVAQKSVLKQAISKTVLAERGLFSCLDDYLERHALKVN
  • M6_Spy0159: M6_Spy0159 is a collagen adhesion protein. It contains a sortase substrate motif LPXSG, shown in italics in the amino acid sequence SEQ ID NO: 44.
    SEQ ID NO: 44
    MYSRLKRELVIVINRKKKYKLIRLMVTVGLIFSQLVLPIRRLGLQMISTQ
    TKVIPQEIVTQTETQGTQVVATKQKLESENSSLKVALKRESGFEHNATID
    ASLDTESQGDNSQRSVTQAIVTMALELRKQGLSIVDTKIVRIQSSTNQRN
    DITTTLTFKNGLSLEGASTEANDPNVRVGIVNPNDTVQTITPTIKQDADG
    KVKNLVFTGRLGKQVIIVSTTRLKEEQTISLDSYGELVIDGAVGLSQKDR
    PPYSKPITVNILKPKLSSIESSLDSKDFETVKTIDNLYTWDDQFYLLDFI
    SKQYEVLKTDYQSAKDSTPQTRKILFGEYTVEPLVMNKGHNNTINIYIRS
    TRPLGLKPIGAAPALIQPRSFRSLTPRSTRMKRSAPVEKFEGELEHHKRI
    DYLGDNQNNPDTTIDDKEDEHDTSDLYRLYLDMTGKKNPLDILVVVDKSG
    SMQEGIGSVQRYRYYAQRWDDYYSQWVYHGTFDYSSYQGESFNRGQIHYR
    YRGIVSVSDGIRRDDAVKNSLLGVNGLLQRFVNINPENKLSVIGFQGSAD
    YHAGKWYPDQSPRGGFYQPNLNNSRDAELLKGWSTNSLLDPNTLTALHNN
    GTNYHAALLKAKEILNEVKDDGRRKIMIFISDGVPTEYFGEDGYRSGNGS
    SNDRNNVTRSQEGSKLAIDEFKARYPNLSIYSLGVSKDINSDTASSSPVV
    LKYLSGEEHYYGITDTAELEKTLNKIVEDSKLSQLGISDSLSQYVDYYDK
    QPDVLVTRKSKVNDETEILYQKDQVQEAGKDIIDKVVFTPKTTSQPKGKV
    TLTFKSDYKVDDEYTYTLSFNVKASDEAYEKYKDNEGRYSEMGDSDTDYG
    TNQTSSGKGGLPSNSDASVNYMADGREQKLPYKHPVIQVKTVPITFTKVD
    ADNNQKKLAGVEFELRKEDKKIVWEKGTTGSNGQLNFKYLQKGKTYYLYE
    TKAKLGYTLPENPWEVAVANNGDIKVKHPIEGELKSKDGSYMIKNYKIYQ
    LPSSGGRGSQIFIIVGSMTATVALLFYRRQHRKKQY
  • M6_Spy0159 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 181 LPSSG (shown in italics in SEQ ID NO: 44, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant M6_Spy0159 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in M6_Spy0159. The pilin motif sequence is underlined in SEQ ID NO: 44, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 265 and 276. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of M6_Spy0159 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 44
    MYSRLKRELVIVINRKKKYKLIRLMVTVGLIFSQLVLPIRRLGLQMISTQ
    TKVIPQEIVTQTETQGTQVVATKQKLESENSSLKVALKRESGFEHNATID
    ASLDTESQGDNSQRSVTQAIVTMALELRKQGLSIVDTKIVRIQSSTNQRN
    DITTTLTFKNGLSLEGASTEANDPNVRVGIVNPNDTVQTITPTIKQDADG
    KVKNLVFTGRLGKQVIIVSTTRLKEEQTISLDSYGELVIDGAVGLSQKDR
    PPYSKPITVNILKPKLSSIESSLDSK DFETVKTIDNLYTWDDQFYLLDFI
    SKQYEVLKTDYQSAKDSTPQTRKILFGEYTVEPLVMNKGHNNTINIYIRS
    TRPLGLKPIGAAPALIQPRSFRSLTPRSTRMKRSAPVEKFEGELEHHKRI
    DYLGDNQNNPDTTIDDKEDEHDTSDLYRLYLDMTGKKNPLDILVVVDKSG
    SMQEGIGSVQRYRYYAQRWDDYYSQWVYHGTFDYSSYQGESFNRGQIHYR
    YRGIVSVSDGIRRDDAVKNSLLGVNGLLQRFVNINPENKLSVIGFQGSAD
    YHAGKWYPDQSPRGGFYQPNLNNSRDAELLKGWSTNSLLDPNTLTALHNN
    GTNYHAALLKAKEILNEVKDDGRRKIMIFISDGVPTEYFGEDGYRSGNGS
    SNDRNNVTRSQEGSKLAIDEFKARYPNLSIYSLGVSKDINSDTASSSPVV
    LKYLSGEEHYYGITDTAELEKTLNKIVEDSKLSQLGISDSLSQYVDYYDK
    QPDVLVTRKSKVNDETEILYQKDQVQEAGKDIIDKVVFTPKTTSQPKGKV
    TLTFKSDYKVDDEYTYTLSFNVKASDEAYEKYKDNEGRYSEMGDSDTDYG
    TNQTSSGKGGLPSNSDASVNYMADGREQKLPYKHPVIQVKTVPITFTKVD
    ADNNQKKLAGVEFELRKEDKKIVWEKGTTGSNGQLNFKYLQKGKTYYLYE
    TKAKLGYTLPENPWEVAVANNGDIKVKHPIEGELKSKDGSYMIKNYKIYQ
    LPSSGGRGSQIFIIVGSMTATVALLFYRRQHRKKQY
  • An E box containing a conserved glutamic residue has been identified in M6_Spy0159. The E-box motif is underlined in SEQ ID NO: 44, below. The conserved glutamic acid (E), at amino acid residue 950, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of M6_Spy0159. Preferred fragments of M6_Spy0159 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 44
    MYSRLKRELVIVINRKKKYKLIRLMVTVGLIFSQLVLPIRRLGLQMISTQ
    TKVIPQEIVTQTETQGTQVVATKQKLESENSSLKVALKRESGFEHNATID
    ASLDTESQGDNSQRSVTQAIVTMALELRKQGLSIVDTKIVRIQSSTNQRN
    DITTTLTFKNGLSLEGASTEANDPNVRVGIVNPNDTVQTITPTIKQDADG
    KVKNLVFTGRLGKQVIIVSTTRLKEEQTISLDSYGELVIDGAVGLSQKDR
    PPYSKPITVNILKPKLSSIESSLDSKDFETVKTIDNLYTWDDQFYLLDFI
    SKQYEVLKTDYQSAKDSTPQTRKILFGEYTVEPLVMNKGHNNTINIYIRS
    TRPLGLKPIGAAPALIQPRSFRSLTPRSTRMKRSAPVEKFEGELEHHKRI
    DYLGDNQNNPDTTIDDKEDEHDTSDLYRLYLDMTGKKNPLDILVVVDKSG
    SMQEGIGSVQRYRYYAQRWDDYYSQWVYHGTFDYSSYQGESFNRGQIHYR
    YRGIVSVSDGIRRDDAVKNSLLGVNGLLQRFVNINPENKLSVIGFQGSAD
    YHAGKWYPDQSPRGGFYQPNLNNSRDAELLKGWSTNSLLDPNTLTALHNN
    GTNYHAALLKAKEILNEVKDDGRRKIMIFISDGVPTEYFGEDGYRSGNGS
    SNDRNNVTRSQEGSKLAIDEFKARYPNLSIYSLGVSKDINSDTASSSPVV
    LKYLSGEEHYYGITDTAELEKTLNKIVEDSKLSQLGISDSLSQYVDYYDK
    QPDVLVTRKSKVNDETEILYQKDQVQEAGKDIIDKVVFTPKTTSQPKGKV
    TLTFKSDYKVDDEYTYTLSFNVKASDEAYEKYKDNEGRYSEMGDSDTDYG
    TNQTSSGKGGLPSNSDASVNYMADGREQKLPYKHPVIQVKTVPITFTKVD
    ADNNQKKLAGVEFELRKEDKKIVWEKGTTGSNGQLNFKYLQKGKTYYLYE
    TKAKLGYTLPENPWEVAVANNGDIKVKHPIEGELKSKDGSYMIKNYKIYQ
    LPSSGGRGSQIFIIVGSMTATVALLFYRRQHRKKQY
  • M6_Spy0160: M6_Spy0160 is a fimbrial structural subunit. It contains a sortase substrate motif LPXTG (SEQ ID NO: 122), shown in italics in amino acid sequence SEQ ID NO: 45.
    SEQ ID NO: 45
    MTNRRETVREKILITAKKLMLACLATLAVVGLGMTRVSALSKDDTAQLKI
    TNIEGGPTVTLYKIGEGVYNTNGDSFINFKYAEGVSLTETGPTSQEITTI
    ANGINTGKIKPFSTENVSISNGTATYNARGASVYIALLTGATDGRTYNPI
    LLAASYNGEGNLVTKNIDSKSNYLYGQTSVAKSSLPSITKKVTGTIDDVN
    KKTTSLGSVLSYSLTFELPSYTKEAVNKTVYVSDNMSEGLTFNFNSLTVE
    WKGKMANITEDGSVMVENTKIGIAKEVNNGFNLSFIYDSLESISPNISYK
    AVVNNKAIVGEEGNPNKAEFFYSNNPTKGNTYDNLDKKPDKGNGITSKED
    SKIVYTYQIAFRKVDSVSKTPLTGAIFGVYDTSNKLIDIVTTNKNGYAIS
    TQVSSGKYKIKELKAPKGYSLNTETYEITANWVTATVKTSANSKSTTYTS
    DKNKATDNSEQVGWLKNGIFYSIDSRPTGNDVKEAYIESTKALTDGTTFS
    KSNEGSGTVLLETDIPNTKLGELPSTGSIGTYLEKAIGSAAMIGAIGIYI
    VKRRKA
  • M6_Spy0160 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 131 LPSTG (shown in italics in SEQ ID NO: 45, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant M6_Spy0160 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • An E box containing a conserved glutamic residue has been identified in M6_Spy0160. The E-box motif is underlined in SEQ ID NO: 45, below. The conserved glutamic acid (E), at amino acid residue 412, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of M6_Spy0160. Preferred fragments of M6_Spy0160 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 45
    MTNRRETVREKILITAKKLMLACLATLAVVGLGMTRVSALSKDDTAQLKI
    TNIEGGPTVTLYKIGEGVYNTNGDSFINFKYAEGVSLTETGPTSQEITTI
    ANGINTGKIKPFSTENVSISNGTATYNARGASVYIALLTGATDGRTYNPI
    LLAASYNGEGNLVTKNIDSKSNYLYGQTSVAKSSLPSITKKVTGTIDDVN
    KKTTSLGSVLSYSLTFELPSYTKEAVNKTVYVSDNMSEGLTFNFNSLTVE
    WKGKMANITEDGSVMVENTKIGIAKEVNNGFNLSFIYDSLESISPNISYK
    AVVNNKAIVGEEGNPNKAEFFYSNNPTKGNTYDNLDKKPDKGNGITSKED
    SKIVYTYQIAFRKVDSVSKTPLTGAIFGVYDTSNKLIDIVTTNKNGYAIS
    TQVSSGKYKIKELKAPKGYSLNTETYEITANWVTATVKTSANSKSTTYTS
    DKNKATDNSEQVGWLKNGIFYSIDSRPTGNDVKEAYIESTKALTDGTTFS
    KSNEGSGTVLLETDIPNTKLGELPSTGSIGTYLFKAIGSAAMIGAIGIYI
    VKRRKA
  • M6_Spy0161 is a srtB type sortase. An example of an amino acid sequence of M6_Spy-161 is shown in SEQ ID NO: 46.
    SEQ ID NO: 46
    MTERLKNLGILLLFLLGTAIFLYPTLSSQWNAYRDRQLLSTYHKQVIQKK
    PSEMEEVWQKAKAYNARLGIQPVPDAFSFRDGIHDKNYESLLQIENNDIM
    GYVEVPSIKVTLPIYHYTTDEVLTKGAGHLFGSALPVGGDGTHTVISAHR
    GLPSAEMFTNLNLVKKGDTFYFRVLNKVLAYKVDQILIVEPDQATSLSGV
    MGKDYATLVTCTPYGVNTKRLLVRGHRIAYHYKKYQQAKKAMKLVDKSRM
    WAEVVCAAFGVVIAIILVFMYSRVSAKKSK
  • As discussed above, applicants have also determined the nucleotide and encoded amino acid sequence of fimbrial structural subunits in several other GAS AI-1 strains of bacteria. Examples of sequences of these fimbrial structural subunits are set forth below.
  • M6 strain isolate CDC SS 410 is a GAS AI-1 strain of bacteria. CDC SS 410_fimbrial is thought to be a fimbrial structural subunit of M6 strain isolate CDC SS 410. An example of a nucleotide sequence encoding the CDC SS 410_fimbrial protein (SEQ ID NO: 267) and a CDC SS 410_fimbrial protein amino acid sequence (SEQ ID NO: 268) are set forth below.
    SEQ ID NO: 267
    aaagatgatactgcacaactaaagataacaaatattgaaggtgggccaac
    agtaacactttataaaataggagaaggtgtttacaacactaatggtgatt
    cttttattaactttaaatatgctgagggggtttctttaactgaaacagga
    cctacatcacaagaaattactactattgcaaatggtattaatacgggtaa
    aataaagccttttagtactgaaaacgttagtatttctaatggaacagcaa
    cttataatgcgagaggtgcatctgtttatattgcattattaacaggtgcg
    acagatggccgtacctacaatcctattttattagctgcatcttataatgg
    tgagggaaatttagttactaaaaatattgattccaaatctaattatttat
    atggacaaacaagtgttgcaaaatcatcattaccatctattacaaagaaa
    gtaaccgggacaatagatgacgtgaataaaaagactacctcgttaggaag
    tgtattgtcttattcgctgacatttgaattaccaagttataccaaagaag
    cagtcaataaaacagtatatgtttctgataatatgtcggaaggtcttact
    tttaactttaatagtcttacagtagaatggaaaggtaagatggctaatat
    tactgaagatggttcagtaatggtagaaaatacaaaaatcggaatagcta
    aggaggttaataacggttttaatttaagttttatttatgatagtttagaa
    tctatatcaccaaatataagttataaagctgttgtaaacaataaagctat
    tgttggtgaagagggtaatcctaataaagctgaattcttctattcaaata
    atccaacaaaaggtaatacatacgataatttagataagaagcctgataaa
    gggaatggtattacatccaaagaagattctaaaattgtttatacttatca
    aatagcgtttagaaaagttgatagtgttagtaagaccccacttattggtg
    caatttttggagtttatgatactagtaataaattaattgatattgttaca
    accaataaaaatggatatgctatttcaacacaagtatcttcaggaaaata
    taaaattaaggaattaaaagctcctaaaggttattcattgaatacagaaa
    cttatgaaattacggcaaattgggtaactgctacagtcaagacaagtgct
    aattcaaaaagtactacttatacatctgataaaaataaggcgacagataa
    ttcagagcaagtaggatggttaaaaaatggtatattctattctatagata
    gtagacctacaggaaatgatgttaaagaggcttatattgaatctactaag
    gctttaactgatggaacaactttctcaaaatcgaatgaaggttcaggtac
    agtattattagaaactgacatccctaacaccaagctaggtgaactc
    SEQ ID NO: 268
    KDDTAQLKTTNIEGGPTVTLYKIGEGVYNTNGDSFINFKYAEGVSLTETG
    PTSQEITTIANGINTGKIKPFSTENVSISNGTATYNARGASVYIALLTGA
    TDGRTYNPILLAASYNGEGNLVTKNIDSKSNYLYGQTSVAKSSLPSITKL
    VTGTIDDVNKKTTSLGSVLSYSLTFELPSYTKEAVNKTVYVSDNMSEGLT
    FNFNSLTVEWKKGMANITEDGSVMVENTKIGIAKEVNNGFNLSFIYDSLE
    SISPNISYKAVVNNKAIVGEEGNPNKAEFFYSNNPTKGNTYDNLDKKPDK
    GNGITSKEDSKIVYTYQIAFRKVDSVSKTPLIGAIFGVYDTSNKLIDIVT
    TNKNGYAISTQVSSGKYKIKELKAPKGYSLNTETYEITANWVTATVKTSA
    NSKSTTYTSDKNKATDNSEQVGWLKNGIFYSIDSRPTGNDVKEAYTESTK
    ALTDGTTFSKSNEGSGTVLLETDIPNTKLGEL
  • M6 strain isolate ISS 3650 is a GAS AI-1 strain of bacteria. ISS3650_fimbrial is thought to be a fimbrial structural subunit of M6 strain isolate ISS 3650. An example of a nucleotide sequence encoding the ISS3650_fimbrial protein (SEQ ID NO: 269) and an ISS3650_fimbrial protein amino acid sequence (SEQ ID NO: 270) are set forth below.
    SEQ ID NO: 269
    gaatggaaaggtaagatggctaatattactgaagatggttcagtaatggt
    agaaaatacaaaaatcggaatagctaaggaggttaataacggttttaatt
    taagttttatttatgatagtttagaatctatatcaccaaatataagttat
    aaagctgttgtaaacaataaagctattgttggtgaagagggtaatcctaa
    taaagctgaattcttctattcaaataatccaacaaaaggtaatacatacg
    ataatttagataagaagcctgataaagggaatggtattacatccaaagaa
    gattctaaaattgtttatacttatcaaatagcgtttagaaaagttgatag
    tgttagtaagaccccacttattggtgcaatttttggagtttatgatacta
    gtaataaattaattgatattgttacaaccaataaaaatggatatgctatt
    tcaacacaagtatcttcaggaaaatataaaattaaggaattaaaagctcc
    taaaggttattcattgaatacagaaacttatgaaattacggcaaattggg
    taactgctacagtcaagacaagtgctaattcaaaaagtactacttataca
    tctgataaaaataaggcgacagataattcagagcaagtaggatggttaaa
    aaatggtatattctattctatagatagtagacctacaggaaatgatgtta
    aagaggcttatattgaatctactaaggctttaactgatggaacaactttc
    tcaaaatcgaatgaaggttcaggtacagtattattagaaactgacatcc
    SEQ ID NO: 270
    EWKGKMANITEDGSVMVENTKIGIAKEVNNGFNLSFIYDSLESISPNISY
    KAVVNNKAIVGEEGNPNKAEFFYSNNPTKGNTYDNLDKKPDKGNGITSKE
    DSDIVYTYQIAFRKVDSVSKTPLIGAIFGVYDTSNKLIDIVTTNKNGYAI
    STQVSSGKYKIKELKAPKGYSLNTETYEITANWVTATVKTSANSKSTTYT
    SDKNKATDNSEQVGWLKNGIFYSIDSRPTGNDVKEAYIESTKALTDGTTF
    SKSNEGSGTVLLETDI
  • M23 strain isolate DSM2071 is a GAS AI-1 strain of bacteria. DSM2071_fimbrial is thought to be a fimbrial structural subunit of M23 strain DSM2071. An example of a nucleotide sequence encoding the DSM2071_fimbrial protein (SEQ ID NO: 251) and a DSM2071_fimbrial protein amino acid sequence (SEQ ID NO: 252) are set forth below.
    SEQ ID NO: 251
    atgagagagaaaatattaatagcagcaaaaaaactaatgctagcttgttt
    agctatcttagctgtagtagggcttggaatgacaagagtatcagctttat
    caaaagatgataaggcggagttgaagataacaaatatcgaaggtaaaccg
    accgtgacactgtataaaattggtgatggaaaatacagtgagcgagggga
    ttcttttattggatttgagttaaagcaaggtgtggagctaaataaggcaa
    aacctacatctcaagaaataaataaaatcgctaatggtattaataaaggt
    agtgttaaggctgaagtagttaatataaaagaacatgctagtacaactta
    tagttatacaacaactggtgcaggtatttacttggctatattgactggag
    ctactgatggacgtgcctataatcctatcttactgacagcttcttacaat
    gaggaaaatccacttaagggagggcagattgacgcaactagtcattatct
    ttttggagaagaagcagttgctaaatctagccaaccaacaattagcaagt
    caattacaaaatccacaaaagatggtgataaagatacagcatctgtaggt
    gaaaaagttgattacaaattaactgttcagttaccaagttattcgaaaga
    tgctatcaataaaacggtgtttatcactgacaaattgtctcagggactta
    ctttccttccaaaaagtttaaagattatctggaatggtcaaacgttaaca
    aaggtgaatgaagaatttaaagctggagataaggtaattgctcaacttaa
    ggttgaaaataatggatttaatctgaactttaattatgataaccttgata
    atcatgccccagaagttaactatagtgctctactaaatgaaaacgcagtt
    gttggtaaaggtggtaatgacaataatgtagactattactattcaaataa
    tccgaataaaggagagacccataaaacaactgagaagcctaaagagggtg
    aaggtactggtatcactaaaaagacggataaaaaaaccgtctacacctat
    cgtgtagcctttaagaaaacaggcaaagatcatgccccactagctggtgc
    tgttttcggtatctattcagataaggaagcgaaacaattagtcgatattg
    ttgtgacaaatgcacagggttatgcagcatcaagcgaagttgggaaaggg
    acttattacattaaagaaattaaatcccctaagggttactctttaaatac
    aaatatttatgaagtggaaacttcatgggaaaaagctacaacgacttcta
    caactaatcgtttagagacaatttatacaacagatgataatcaaaagtct
    ccaggaactaatacagttggttggttggaagatggtgtcttttacaaaga
    aaatccaggtggtgatgctaaacttgcctatatcaaacaatcaacagagg
    agacttctacaactatagaagtcaaagaaaatcaagctgaaggttcaggt
    acggtattattagaaactgaaattcctaacaccaaattaggtgaattacc
    ttcgacaggtagcattggtacttacctctttaaagctattggttcggctg
    ctatgatcggtgcaattggtatttatattgttaaacgtcgtaaagcttaa
    SEQ ID NO: 252
    MREKILIAAKKLMLACLATLAVVGLGMTRVSALSKDDKAELKITNIEGKP
    TVTLYKIGDGKYSERGDSFIGFELKQGVELNKAKPTSQEINKIANGINKG
    SVKAEVVNIKEHASTTYSYTTTGAGIYLAILTGATDGRAYNPILLTASYN
    EENPLKGGQIDATSHYLFGEEAVAKSSQPTISKSITKSTKDGDKDTASVG
    EKVDYKLTVQLPSYSKDAINKTVFITDKLSQGLTFLPKSLKIIWNGQTLT
    KVNEEFKAGDKVIAQLKVENNGFNLNFNYDNLDNHAPEVNYSALLNENAV
    VGKGGNDNNVDYYYSNNPNKGETHKTTEKPKEGEGTGITKKTDKKTVYTY
    RVAFKKTGKDHAPLAGAVFGIYSDKEAKQLVDIVVTNAQGYAASSEVGKG
    TYYIKEIKSPKGYSLNTNIYEVETSWEKATTTSTTNRLETIYTTDDNQKS
    PGTNTVGWLEDGVFYKENPGGDAKLAYIKQSTEETSTTIEVKENQAEGSG
    TVLLETEIPNTKLGELPSTGSIGTYLFKAIGSAAMIGAIGIYIVKRRKA
  • GAS AI-2 sequences
  • As discussed above, a GAS AI-2 sequence is present in an M1 strain isolate (SF370). Examples of GAS AI-2 sequences from M1 strain isolate SF370 are set forth below.
  • Spy0124 is a rofA transcriptional regulator. An example of an amino acid sequence for
  • Spy0124 is set forth in SEQ ID NO:47.
    SEQ ID NO: 47
    MIEKYLESSIESKGQLIVLFFKTSYLPITEVAEKTGLTFLQLNHYCEELN
    AFFPGSLSMTIQKRMISCQFTHPFKETYLYQLYASSNVLQLLAFLIKNGS
    HSRPLTDPARSHFLSNSSAYRMREALIPLLRNFELKLSKNKIVGEEYRIR
    YLIALLYSKFGIKVYDLTQQDKNTIHSFLSHSSTHLKTSPWLSESESFYD
    ILLALSWKRHQFSVTIPQTRIFQQLKKLFVYDSLKKSSHDIIETYGQLNF
    SAGDLDYLYLIYITANNSFASLQWTPEHIRQYCQLFEENDTFRLLLNPII
    TLLPNLKEQKASLVKALMFFSKSFLFNLQHFIPETNLFVSPYYKGNQKLY
    TSLKLIVEEWMAKLPGKRDLNHKHFHLFCHYVEQSLRNIQPPLVVVFVAS
    NFINAHLLTDSFPRYFSDKSIDFHSYYLLQDNVYQIPDLKPDLVITHSQL
    IPFVHHELTKGIAVAEISFDESILSIQELMYQVKEEKEQADLTKQLT
  • GAS 015 is also referred to as Cpa. It contains a sortase substrate motif VVXTG (SEQ ID NO: 135), shown in italics in SEQ ID NO: 48.
    SEQ ID NO: 48
    LRGEKMKKTRFPNKLNTLNTQRVLSKNSKRFTVTLVGVFLMIEALVTSMV
    GAKTVFGLVESSTPNAINPDSSSEYRWYGYESYVRGHPYYKQFRVAHDLR
    VNLEGSRSYQVYCFNLKKAFPLGSDSSVKKWYKKHDGISTKFEDYAMSPR
    ITGDELNQKLRAVMYNGHPQNANGIMEGLEPLNAIRVTQEAVWYYSDNAP
    ISNPDESFKRESESNLVSTSQLSLMRQALKQLIDPNLATKMPKQVPDDFQ
    LSIFESEDKGDKYNKGYQNLLSGGLVPTKPPTPGDPPMPPNQPQTTSVLI
    RKYAIGDYSKLLEGATLQLTGDNVNSFQARVFSSNDIGERIELSDGTYTL
    TELNSPAGYSIAEPITFKVEAGKVYTIIDGKQIENPNKEIVEPYSVEAYN
    DFEEFSVLTTQNYAKFYYAKNKNGSSQVVYCFNADLKSPPDSEDGGKTMT
    PDFTTGEVKYTHIAGRDLFKYTVKPRDTDPDTFLKHIKKVIEKGYREKGQ
    AIEYSGLTETQLRAATQLATYYFTDSAELDKDKLKDYHGFGDMNDSTLAV
    AKILVEYAQDSNPPQLTDLDFFIPNNNKYQSLIGTQWHPEDLVDTIRMED
    KKEVIPVTHNLTLRKTVTGLAGDRTKDFHFEIELKNNKQELLSQTVKTDK
    TNLEFKDGKATINLKHGESLTLQGLPEGYSYLVKETDSEGYKVKVNSQEV
    ANATVSKTGITSDETLAFENNKEPVVPTGVDQKINGYLALIVIAGISLGI
    WGIHTIRIRKHD
  • GAS 015 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 182 VVPTG (shown in italics in SEQ ID NO: 48, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant GAS 015 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in GAS 015. The pilin motif sequence is underlined in SEQ ID NO: 48, below. Conserved lysine (K) residues are also marked in bold, at amino acid residue 243. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of GAS 015 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 48
    LRGEKMKKTRFPNKLNTLNTQRVLSKNSKRFTVTLVGVFLMIFALVTSMV
    GAKTVFGLVESSTPNAINPDSSSEYRWYGYESYVRGHPYYKQFRVAHDLR
    VNLEGSRSYQVYCFNLKKAFPLGSDSSVKKWYKKHDGISTKFEDYANSPR
    ITGDELNQKLRAVMYNGHPQNANGIMEGLEPLNAIRVTQEAVWYYSDNAP
    ISNPDESFKRESESNLVSTSQLSLMRQALKQLIDPNLATKMPKQVPDDFQ
    LSIFESEDKGDKYNKGYQNLLSGGLVPTKPPTPGDPPMPPNQPQTTSVLI
    RKYAIGDYSKLLEGATLQLTGDNVNSFQARVFSSNDIGERIELSDGTYTL
    TELNSPAGYSIAEPITFKVEAGKVYTIIDGKQIENPNKETVEPYSVEAYN
    DFEEFSVLTTQNYAKFYYAKNKNGSSQVVYCFNADLKSPPDSEDGGKTMT
    PDFTTGEVKYTHIAGRDLFKYTVKPRDTDPDTFLKHIKKVIEKGYREKGQ
    AIEYSGLTETQLRAATQLAIYYFTDSAELDKDKLKDYHGFGDMNDSTLAV
    AKILVEYAQDSNPPQLTDLDFFIPNNNKYQSLIGTQWHPEDLVDTIRMED
    KKEVIPVTHNLTLRKTVTGLAGDRTKDFHPEIELKNNKQELLSQTVKTDK
    TNLEFKDGKATINLKHGESLTLQGLPEGYSYLVKETDSEGYKVKVNSQEV
    ANATVSKTGTTSDETLAFENNKEPVVPTGVDQKINGYLALIVIAGISLGI
    WGIHTIRIRKHD
  • An E box containing a conserved glutamic residue has been identified in GAS 015. The E-box motif is underlined in SEQ ID NO: 48, below. The conserved glutamic acid (E), at amino acid residue 352, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of GAS 015. Preferred fragments of GAS 015 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 48
    LRGEKMKKTRFPNKLNTLNTQRVLSKNSKRFTVTLVGVFLMIFALVTSMV
    GAKTVFGLVESSTPNAINPDSSSEYRWYGYESYVRGHPYYDQFRVAHDLR
    VNLEGSRSYQVYCFNLKKAFPLGSDSSVKKWYKKHDGISTKFEDYAMSPR
    ITGDELNQKLRAVMYNGHPQNANGIMEGLEPLNAIRVTQEAVWYYSDNAP
    ISNPDESFKRESESNLVSTSQLSLMRQALKQLIDPNLATKMPKQVPDDFQ
    LSIFESEDKGDKYNKGYQNLLSGGLVPTKPPTPGDPPMPPNQPQTTSVLI
    RKYAIGDYSKLLEGATLQLTGDNVNSFQARVFSSNDIGERIELSDGTYTL
    TELNSPAGYSIAEPITFKVEAGKVYTIIDGKQIENPNKEIVEPYSVEAYN
    DFEEFSVLTTQNYAKFYYAKNKNGSSQVVYCPNADLKSPPDSEDGGKTMT
    PDFTTGEVKYTHIAGRDLFKYTVKPRDTDPDTFLKHIKKVIEKGYREKGQ
    AIEYSGLTETQLRAATQLAIYYFTDSAELDKDKLKDYHGFGDMNDSTLAV
    AKILVEYAQDSNPPQLTDLDFFIPNNNKYQSLIGTQWHPEDLVDIIRMED
    KKEVIPVTHNLTLRKTVTGLAGDRTKDFHFEIELKNNKQELLSQTVKTDK
    TNLEFKDGKATINLKHGESLTLQGLPEGYSYLVKETDSEGYKVKVNSQEV
    ANATVSKTGITSDETLAFENNKEPVVPTGVDQKINGYLALIVIAGISLGI
    WGIHTIRIRKHD
  • Spy0127 is a LepA putative signal peptidase. An example of an amino acid sequence for Spy0127 is set forth in SEQ ID NO: 49.
    SEQ ID NO: 49
    MIIKRNDMAPSVKAGDAILFYRLSQTYKVEEAVVYEDSKTSITKVGRIIA
    QAGDEVDLTEQGELKINGHIQNEGLTFIKSREANYPYRIADNSYLTLNDY
    YSQESENYLQDAIAKDAIKGTINTLIRLRNH
  • Spy0128 is thought to be a fibrial protein. It contains a sortase substrate motif EVXTG (SEQ ID NO: 136) shown in italics in SEQ ID NO: 50.
    SEQ ID NO: 50
    MKLRHLLLTGAALTSFAATTVHGETVVNGAKLTVTKNLDLVNSNALIPNT
    DFTFKIEPDTTVNEDGNKFKGVALNTPMTKVTYTNSDKGGSNTKTAEFDF
    SEVTFEKPGVYYYKVTEEKIDKVPGVSYDTTSYTVQVHVLWNEEQQKPVA
    TYIVGYKEGSKVPIQFKNSLDSTTLTVKKKVSGTGGDRSKDFNFGLTLKA
    NQYYKASEKVMIEKTTKGGQAPVQTEASIDQLYHFTLKDGESIKVTNLPV
    GVDYVVTEDDYKSEKYTTNVEVSPQDGAVKNIAGNSTEQETSTDKDMTIT
    FTNKKDFEVPTGVAMTVAPYIALGIVAVGGALYFVKKKNA
  • Spy0128 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 183 EVPTG (shown in italics in SEQ ID NO: 50, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Spy0128 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two E boxes containing a conserved glutamic residue have been identified in Spy0128. The E-box motifs are underlined in SEQ ID NO: 50, below. The conserved glutamic acid (E) residues, at amino acid residues 271 and 290, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of Spy0128. Preferred fragments of Spy0128 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 50
    MKLRHLLLTGAALTSFAATTVHGETVVNGAKLTVTKNLDLVNSNALIPNT
    DFTFKIEPDTTVNEDGNKFKGVALNTPMTKVTYTNSDKGGSNTKTAEFDF
    SEVTPEKPGVYYYKVTEEKIDKVPGVSYDTTSYTVQVHVLWNEEQQKPVA
    TYIVGYKEGSKVPIQFKNSLDSTTLTVKKKVSGTGGDRSKDFNFGLTLKA
    NQYYKASEKVMIEKTTKGGQAPVQTEASTDQLYHFTLKDGESIKVTNLPV
    GVDYVVTEDDYKSEKYTTNVEVSPQDGAVKNIAGNSTEQETSTDKDMTIT
    FTNKKDFEVPTGVAMTVAPYIALGIVAVGGALYFVKKKNA
  • Spy0129 is a srtC1 type sortase. An example of an amino acid sequence for Spy0129 is set forth in SEQ ID NO: 51.
    SEQ ID NO: 51
    MIVRLIKLLDKLINVIVLCFFFLCLLIAALGIYDALTVYQGANATNYQQY
    KKKGVQFDDLLAINSDVMAWLTVKGTHIDYPIVQGENNLEYINKSVEGEY
    SLSGSVFLDYRNKVTFEDKYSLIYAHHMAGNVMFGELPNFRKKSFFNKHK
    EESIETKTKQKLKINIFACIQTDAFDSLLFNPIDVDISSKNEFLNHIKQK
    SVQYREILTTNESRFVALSTCEDMTTDGRIIVIGQIE″
  • Spy0130 is referred to as a hypothetical protein. It contains a sortase substrate motif LPXTG (SEQ ID NO: 122), shown in italics in SEQ ID NO: 52.
    SEQ ID NO: 52
    MKKSILRILAIGYLLMSFCLLDSVEAENLTASINIEVINQVDVATNKQSS
    DIDETFMPVIEALDKESPLPNSVTTSVKGNGKTSFEQLTPSEVGQYHYKI
    HQLLGKNSQYHYDETVYEVVIYVLYNEQSGALETNLVSNKLGETEKSELI
    FKQEYSEKTPEPHQPDTTEKEKPQKKRNGILPSTGEMVSYVSALGIVLVA
    TITLYSIYKKLKTSK
  • Spy0130 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 131 LPSTG (shown in italics in SEQ ID NO: 52, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Spy0130 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two E boxes containing conserved glutamic residues have been identified in Spy0130. The E-box motifs are underlined in SEQ ID NO: 52, below. The conserved glutamic acid (E) residues, at amino acid residues 118 and 148, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of Spy0130. Preferred fragments of Spy0130 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO:52
    MKKSILRILAIGYLLMSFCLLDSVEAENLTASINIEVINQVDVATNKQSS
    DTDETFMFVIEALDKESPLPNSVTTSVKGNGKTSFEQLTFSEVGQYHYKI
    HQLLGKNSQYHYDETVYEVVIYVLYNEQSGALETNLVSNKLGETEKSELI
    FKQEYSEKTPEPHQPDTTEKEKPQKKRNGILPSTGEMVSYVSALGIVLVA
    TITLYSIYKKLKTSK
  • Spy0131 is referred to as a conserved hypothetical protein. An example of an amino acid sequence of Spy0131 is set forth in SEQ ID NO: 53
    SEQ ID NO: 53
    MTRTNYQKKRMTCPVETEDITYRRKKIKGRRQAILAQFEPELVHHELIGD
    SCTCPDCHGTLTEIGSVVQRQELVFIPAQLKRINHVQHAYKCQTCSDNSL
    SDKIIKAPVPKAPLAHSLGSASIIAHTVHQKFTLKVPNYRQEEDWNKLGL
    SISRKEIANWHIKSSQYYFEPLYDLLRDILLSQEVIHADETSYRVLESDT
    QLTYYWTFLSGKHEKKGITLYHHDKRRSGLVTQEVLGDYSGYVHCDMHGA
    YRQLEHAKLVGCWAHVRRKFFEATPKQADKTSLGRKGLVYCDKLFALEAE
    WCELPPQERLVKRKEILTPLMTTFFDWCREQVVLSGSKLGLAIAYSLKHE
    RTFRTVLEDGHIVLSNNMAERAIKSLVMGRKNWLFSQSFEGAKAAAIIMS
    LLETAKRHGLNSEKYISYLLDRLPNEETLAKREVLEAYLPWAKKVQTNCQ
  • Spy0133 is referred to as a conserved hypothetical protein. An example of an amino acid sequence of Spy0133 is set forth in SEQ ID NO: 54.
    SEQ ID NO: 54
    MTIRLNDLGQVYLVCGKTDMRQGIDSLAYLVKSQHELDLFSGAVYLFCGG
    RTRDRFKALYWDGQGFWLLYKRFENGKLAWPRNRDEVKCLTAVQVDWLMK
    GFFISPNIKISKSHDFY
  • Spy0135 is a SrtB type sortase. It is also referred to as a putative fibria-associated protein. An example of an amino acid sequence of Spy0135 is set forth in SEQ ID NO: 55.
    SEQ ID NO: 55
    MECYRDRQLLSTYHKQVTQKKPSEMEEVWQKAKAYNARLGIQPVPDAFSF
    RDGIHDKNYESLLQIENNDIMGYVEVPSIKVTLPIYHYTTDEVLTKGAGH
    LFGSALPVGGDGTHTVISAHRGLPSAEMETNLNLVKKGDTFYFRVLNKVL
    AYKVDQILTVEPDQVTSLSGVMGKDYATLVTCTPYGVNTKRLLVRGHRIA
    YHYKKYQQAKKAMKLVDKSRMWAEVVCAAFGVVIAIILVFMYSRVSAKKS
    K
  • GAS AI-3 Sequences
  • As discussed above, a GAS AI-3 sequence is present in a M3, M18 and M5 strain isolates. Examples of GAS AI-3 sequences from M3 strain isolate MGAS315 are set forth below.
  • SpyM30097 is as a negative transcriptional regulator (Nra). An example of an amino acid sequence of SpyM30097 is set forth in SEQ ID NO: 56.
    SEQ ID NO: 56
    MPYVKKKKDSFLVETYLEQSIRDKSELVLLLFKSPTIIFSHVAKQTGLTA
    VQLKYYCKELDDFFGNNLDTTIKKGKIICCFVKPVKEFYLHQLYDTSTIL
    KLLVFFIKNGTSSQPLIKFSKKYFLSSSSAYRLRESLIKLLREFGLRVSK
    NTIVGEEYRIRYLIAMLYSKFGIVIYPLDHLDNQIIYRFLSQSATNLRTS
    PWLEEPFSFYNMLLALSWKRHQFAVSIPQTRIFRQLKKLFIYDCLTRSSR
    QVIENAFSLTFSQGDLEYLFLIYITTNNSFASLQWTPQHIETCCHIFEKN
    DTFRLLLEPILKRLPQLNHSKQDLIKALMYFSKSFLFNLQHFVIEIPSFS
    LPTYTGNSNLYKALKNIVNQWLAQLPGKRHLNEKHLQLFCSHIEQILKNK
    QPALTVVLISSNFINAKLLTDTIPRYPSDKGIHFYSFYLLRDDIYQIPSL
    KPDLVITHSRLIPFVKNDLVKGVTVAEFSFDNPDYSIASIQNLTYQLKDK
    KYQDFLNEQLQ
  • SpyM30098 is thought to be a collagen binding protein (Cpb). It contains a sortase substrate motif VPXTG (SEQ ID NO: 137) shown in italics in SEQ ID NO: 57.
    SEQ ID NO: 57
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    VPNKQSSVQDYPWYGYDSYSKGYPDYSPLKTYHNLKVNLDGSKEYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNDRNGIMKGIDPLNAILVTQNAIWYYTDSSYISDTSKAFQQEETD
    LKLDSQQLQLMRNALKRLINPKEVESLPNQVPANYQLSIFQSSDKTFQNL
    LSAEYVPDTPPKPGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLAQI
    EGSGFQEKIFDSNKSGEKVELPNGTYVLSELKPPQGYGVATPITFKVAAE
    KVLIKNKEGQFVENQNKEIAEPYSVTAFNDEEEIGYLSDFNNYGKFYYAK
    NTNGTNQVVYCFNADLHSPPDSYDHGANIDPDVSESKEIKYTHVSGYDLY
    KYAATPRDKDADFFLKHIKKILDKGYKKKGDTYKTLTEAQFRAATQLAIY
    YYTDSADLTTLKTYNDNKGYHGFDKLDDATLAVVHELITYAEDVTLPMTQ
    NLDEFVPNSSRYQALIGTQYHPNELIDVISMEDKQAPIIPITHKLTISKT
    VTGTIADKKKEFNFEIHLKSSDGQAISGTYPTNSGELTVTDGKATFTLKD
    GESLIVEGLPSGYSYEITETGASDYEVSVNGKNAPDGKATKASVKEDETV
    AFENRKDLVPPTGLTTDGAIYLWLLLLVPFGLLVWLFGRKGTKK
  • SpyM30098 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 184 VPPTG (shown in italics in SEQ ID NO: 57, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM30098 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM30098. The pilin motif sequence is underlined in SEQ ID NO: 57, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 262 and 270. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM30098 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 57
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    VPNKQSSVQDYPWYGYDSYSKGYPDYSPLKTYHNLKVNLDGSKEYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNDRNGIMKGTDPLNATLVTQNAIWYYTDSSYISDTSKAFQQEETD
    LKLDSQQLQLMRNALKRLINPKEVESLPNQVPANYQLSIFQSSDKTFQNL
    LSAEYVPDTPPKPGEEPPAK TEKTSVIIRKYAEGDYSKLLEGATLKLAQI
    EGSGFQEKIFDSNKSGEKVELPNGTYVLSELKPPQGYGVATPITFKVAAE
    KVLIKNKEGQFVENQNKEIAEPYSVTAFNDFEEIGYLSDFNNYGKFYYAK
    NTNGTNQVVYCFNADLHSPPDSYDHGANIDPDVSESKEIKYTHVSGYDLY
    KYAATPRDKDADFFLKHIKKILDKGYKKKGDTYKTLTEAQFRAATQLAIY
    YYTDSADLTTLKTYNDNKGYHGFDKLDDATLAVVHELITYAEDVTLPMTQ
    NLDFFVPNSSRYQALIGTQYHPNELIDVISMEDKQAPIIPITHKLTISKT
    VTGTIADKKKEFNFEIHLKSSDGQAISGTYPTNSGELTVTDGKATFTLKD
    GESLIVEGLPSGYSYEITETGASDYEVSVNGKNAPDGKATKASVKEDETV
    AFENRKDLVPPTGLTTDGAIYLWLLLLVPFGLLVWLFGRKGTKK
  • An E box containing a conserved glutamic residue has been identified in SpyM30098. The E-box motif is underlined in SEQ ID NO: 57, below. The conserved glutamic acid (E), at amino acid residue 330, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyM30098. Preferred fragments of SpyM30098 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 57
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    VPNKQSSVQDYPWYGYDSYSKGYPDYSPLKTYHNLKVNLDGSKEYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNDRNGIMKGTDPLNATLVTQNAIWYYTDSSYISDTSKAFQQEETD
    LKLDSQQLQLMRNALKRLINPKEVESLPNQVPANYQLSIFQSSDKTFQNL
    LSAEYVPDTPPKPGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLAQI
    EGSGFQEKIFDSNKSGEKVELPNGTYVLSELKPPQGYGVATPITFKVAAE
    KVLIKNKEGQFVENQNKEIAEPYSVTAFNDFEEIGYLSDFNNYGKFYYAK
    NTNGTNQVVYCFNADLHSPPDSYDHGANIDPDVSESKEIKYTHVSGYDLY
    KYAATPRDKDADFFLKHIKKILDKGYKKKGDTYKTLTEAQFRAATQLAIY
    YYTDSADLTTLKTYNDNKGYHGFDKLDDATLAVVHELITYAEDVTLPMTQ
    NLDFFVPNSSRYQALIGTQYHPNELIDVISMEDKQAPIIPITHKLTISKT
    VTGTIADKKKEFNFEIHLKSSDGQAISGTYPTNSGELTVTDGKATFTLKD
    GESLIVEGLPSGYSYEITETGASDYEVSVNGKNAPDGKATKASVKEDETV
    AFENRKDLVPPTGLTTDGAIYLWLLLLVPFGLLVWLFGRKGTKK
  • SpyM30099 is referred to as LepA. An example of an amino acid sequence of SpyM30099 is set forth in SEQ ID) NO: 58.
    SEQ ID NO: 58
    MTNYLNRLNENPLLKAFIRLVLKISTIGFLGYILFQYVFGVMIVNTNQMS
    PAVSAGDGVLYYRLTDRYHINDVVVYEVDDTLKVGRIAAQAGDVENFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKVPTGTYFILNDYREERLDSR
    YYGALPINQIKGKISTLLRVRGI
  • SpyM30100 is thought to be a fimbrial protein. An example of an amino acid sequence of SpyM30100 is set forth in SEQ ID NO: 59.
    SEQ ID NO: 59
    MKKNKLLLATAILATALGTASLNQNVKAETAGVSENAKLIVKKTFDSYTD
    NEVLMPKADYTFKVEADSTASGKTKDGLEIKPGIVNGLTEQIISYTNTDK
    PDSKVKSTEFDFSKVVFPGIGVYRYTVSEKQGDVEGITYDTKKWTVDVYV
    GNKEGGGFEPKFIVSKEQGTDVKKPVNFNNSFATTSLKVKKNVSGNTGEL
    QKEFDFTLTLNESTNFKKDQIVSLQKGNEKFEVKIGTPYKFKLKNGESIQ
    LDKLPVGITYKVNEMEANKDGYKTTASLKEGDGQSKMYQLDMEQKTDESA
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • SpyM30100 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 140 QVPTG (shown in italics in SEQ ID NO: 59, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM30100 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, discussed above, containing conserved lysine (K) residues have also been identified in SpyM30100. The pilin motif sequences are underlined in SEQ ID NO: 59, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 57 and 63 and at amino acid residues 161 and 166. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM30100 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 59
    MKKNKLLLATAILATALGTASLNQNVKAETAGVSENAKLIVKKTFDSYTD
    NEVLMPKADYTFK VEADSTASGKTKDGLEIKPGIVNGLTEQIISYTNTDK
    PDSKVKSTEFDFSKVVFPGIGVYRYTVSEKQGDVEGITYDTKKWTVDVYV
    GNKEGGGFEPKFIVS K EQGTDVKKPVNFNNSFATTSLKVKKNVSGNTGEL
    QKEFDFTLTLNESTNFKKDQIVSLQKGNEKFEVKIGTPYKFKLKNGESIQ
    LDKLPVGITYKVNEMEANKDGYKTTASLKEGDGQSKMYQLDMEQKTDESA
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • Two E boxes, each containing a conserved glutamic residue, have been identified in SpyM30100. The E-box motifs are underlined in SEQ ID NO: 59, below. The conserved glutamic acid (E) residues, at amino acid residues 232 and 264, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyM30100. Preferred fragments of SpyM30100 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 59
    MKKNKLLLATAILATALGTASLNQNVKAETAGVSENAKLIVKKTFDSYTD
    NEVLMPKADYTFKVEADSTASGKTKDGLEIKPGIVNGLTEQIISYTNTDK
    PDSKVKSTEFDFSKVVFPGIGVYRYTVSEKQGDVEGITYDTKKWTVDVYV
    GNKEGGGFEPKFIVSKEQGTDVKKPVNFNNSFATTSLKVKKNVSGNTGEL
    QKEFDFTLTLNESTNFKKDQIVSLQKGNEKFEVKIGTPYKFKLKNGESIQ
    LDKLPVGITYKVNEMEANKDGYKTTASLKEGDGQSKMYQLDMEQKTDESA
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • SpyM30101 is a SrtC2 type sortase. An example of an amino acid sequence of SpyM30101 is set forth in SEQ ID NO: 60.
    SEQ ID NO: 60
    MTIVQVINKAIDTLILIFCLVVLFLAGFGLWDSYHLYQQADASNPKKFKT
    AQQQPKFEDLLALNEDVIGWLNIPGTHIDYPLVQGKTNLEYINKAVDGSV
    ANSGSLFLDTRNHNDFTDDYSLIYGHHMAGNAMFGEIPKFLKKDFPSKHN
    KATIETKERKKLTVTIFACLKTDAFNQLVFNPNAITNQDQQRQLVDYISK
    RSKQFKPVKLKHHTKFVAFSTCENFSTDNRVIVVGTTQE
  • SpyM30102 is referred to as a hypothetical protein. An example of an amino acid sequence of SpyM30102 is set forth in SEQ ID NO: 61.
    SEQ ID NO: 61
    MILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALESIDAMKT
    IEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQASTTVFDVLV
    YVTYDEDGTLVAKVISRRAGDEEKSAITFKPKWLVKPIPPRQPNIPKTPL
    PLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • SpyM30102 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 185 LPLAG (shown in italics in SEQ ID NO: 61, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM30102 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM30102. The pilin motif sequence is underlined in SEQ ID NO: 61, below. The conserved lysine (K) residue is also marked in bold, at amino acid residue 132. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM30102 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 61
    MILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALESIDAMKT
    IEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTTVFDVLV
    YVTYDEDGTLVAKVISRRAGDEEKSAITFKPKWLVKPIPPRQPNIPKTPL
    PLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • Two E boxes containing conserved glutamic residues have been identified in SpyM30102. The E-box motifs are underlined in SEQ ID NO: 61, below. The conserved glutamic acid (E) residues, at amino acid residues 52 and 122, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyM30102. Preferred fragments of SpyM30102 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 61
    MILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALESIDAMKT
    IEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQASTTVFDVLV
    YVTYDEDGTLVAKVISRRAGDEEKSAITFKPKWLVKPIPPRQPNIPKTPL
    PLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • SpyM30103 is referred to as a putative multiple sugar metabolism regulator. An example of an amino acid sequence for SpyM3103 is set forth in SEQ ID NO: 62.
    SEQ ID NO: 62
    MVRFDLKHVQTLHSLSQLPISVMSQDKALIQVYGNDDYLLCYYQFLKHLA
    IPQAAQDVIFYEGLFEESFMIFPLCHYIIAIGPGYPYSLNKDYQEQLANN
    CLKHSSHRSKEELLSYMALVPHFPINNVRNLLIAIDAFFDTQFETTCQQT
    IHQLLQHSKQMTADPDIIHRLKHISKASSQLPPVLEHLNHTMDLVKLGNP
    QLLKQETNRIPLSSITSSSISALRAEKNLTVIYLTRLLEFSFVENTDVAK
    HYSLVKYYMALNEEASDLLKVLRIRCAAIIHFSESLTNKSISDKRQMYNS
    VLHYVDSHLYSKLKVSDIAKRLYVSESHLRSVFKKYSNVSLQHYILSTKI
    KEAQLLLKRGIPVGEVAKSLYFYDTTHFHKIFKKYTGISSKDYLAKYRDN
    I
  • SpyM30104 is thought to be a F2 like fibronectic binding protein. An example of an amino acid sequence for SpyM30104 is set forth in SEQ ID NO: 63.
    SEQ ID NO: 63
    MSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVLTEGYPTNKSDWLNG
    LTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQKMKEVYQKLIDTTD
    IDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTSLKPIAQKDITAKKI
    WVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQINSEGQQEISVTWTNQ
    LVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGLTVTNTYVKPTSGHY
    DIEVTFGNGHIDTTEDTTPDIVSGENQMKQIEGEDSKPIDEVTENNLIEF
    GKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQGQSGDMTIEEDSAT
    HIKFSKRDIDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTF
    VETAAPDGYEVATAITFTVNEQGQVTVNGKATKGKAHIVMVDAYKPTKGS
    GQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGM
    TGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTKIEDS
    KSSDVIVGGQGQIVETTEDTQTGMHGDSGRKTEVEDTKLVQSFHEDNKEP
    ESNSEIPKKDKSKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVISLKSK
    KRLSSC
  • SpyM30104 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 180 LPATG (shown in italics in SEQ ID NO: 63, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM30104 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, discussed above, containing conserved lysine (K) residues have also been identified in SpyM30104. The pilin motif sequences are underlined in SEQ ID NO: 63, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 156 and 227. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM30104 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 63
    MSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVLTEGYPTNKSDWLNG
    LTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQKMKEVYQKLIDTTD
    IDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTSLKPIAQKDITAKKI
    WVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQINSEGQQEISVTWTNQ
    LVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGLTVTNTYVKPTSGHY
    DIEVTFGNGHIDTTEDTTPDIVSGENQMKQIEGEDSKPIDEVTENNLIEF
    GKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQGQSGDMTIEEDSAT
    HIKFSKRDIDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTF
    VETAAPDGYEVATAITFTVNEQGQVTVNGKATKGKAHIVMVDAYKPTKGS
    GQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGM
    TGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTKIEDS
    KSSDVIVGGQGQIVETTEDTQTGMHGDSGRKTEVEDTKLVQSFHEDNKEP
    ESNSEIPKKDKSKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVISLKSK
    KRLSSC
  • An E box containing a conserved glutamic residue has been identified in SpyM30104. The E-box motif is underlined in SEQ ID NO: 63, below. The conserved glutamic acid (E), at amino acid residue 402, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyM30104. Preferred fragments of SpyM30104 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 63
    MSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVLTEGYPTNKSDWLNG
    LTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQKMKEVYQKLIDTTD
    IDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTSLKPIAQKDITAKKI
    WVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQINSEGQQEISVTWTNQ
    LVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGLTVTNTYVKPTSGHY
    DIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDSKPIDEVTENNLIEF
    GKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQGQSGDMTIEEDSAT
    HIKFSKRDIDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTF
    VETAAPDGYEVATAITFTVNEQGQVTVNGKATKGDAHIVMVDAYKPTKGS
    GQVIDTEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGM
    TGHSGSTTEIEDSKSSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTKIEDS
    KSSDVIVGGQGQIVETTEDTQTGMHGDSGRKTEVEDTKLVQSFHFDNKEP
    ESNSEIPKKDKSKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVISLKSK
    KRLSSC
  • Examples of GAS AI-3 sequences from M3 strain isolate SSI-1 are set forth below.
  • Sps0099 is a negative transcriptional regulator (Nra). An example of an amino acid sequence for Sps0099 is set forth in SEQ ID NO: 64.
    SEQ ID NO: 64
    MPYVKKKKDSPLVETYLEQSIRDKSELVLLLFKSPTIIFSHVAKQTGLTA
    VQLKYYCKELDDFFGNNLDITIKKGKIICCFVKPVKEFYLHQLYDTSTIL
    KLLVFFIKNGTSSQPLIKFSKKYFLSSSSAYRLRESLIKLLREFGLRVSK
    NTIVGEEYRIRYLIAMLYSKFGIVIYPLDHLDNQIIYRFLSQSATNLRTS
    PWLEEPFSFYNMLLALSWKRHQFAVSIPQTRIFRQLKKLFIYDCLTRSSR
    QVIENAFSLTESQGDLEYLFLIYITTNNSFASLQWTPQHIETCCHIFEKN
    DTFRLLLEPILKRLPQLNHSKQDLIKALMYFSKSFLFNLQHFVIEIPSFS
    LPTYTGNSNLYKALKNIVNQWLAQLPGKRHLNEKHLQLFCSHIEQILKNK
    QPALTVVLTSSNFINAKLLTDTIPRYFSDKGIHFYSFYLLRDDIYQIPSL
    KPDLVTTHSRLIPFVKNDLVKGVTVAEFSFDNPDYSIASIQNLIYQLKDK
    KYQDFLNEQLQ
  • Sps0100 is thought to be a collagen binding protein (Cbp). It contains a sortase substrate motif VPXTG shown in italics in SEQ ID NO: 65.
    SEQ ID NO: 65
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    VPNKQSSVQDYPWYGYDSYSKGYPDYSPLKTYHNLKVNLDGSKEYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNDRNGIMKGIDPLNAILVTQNAIWYYTDSSYISDTSKAFQQEETD
    LKLDSQQLQLMRNALKRLINPKEVESLPNQVPANYQLSIFQSSDKTFQNL
    LSAEYVPDTPPKPGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLAQI
    EGSGFQEKIFDSNKSGEKVELPNGTYVLSELKPPQGYGVATPITFKVAAE
    KVLIKNKEGQFVENQNKETAEPYSVTAFNDFEEIGYLSDFNNYGKFYYAK
    NTNGTNQVVYCFNADLHSPPDSYDHGANIDPDVSESKEIKYTHVSGYDLY
    KYAATPRDKDADFFLKHIKKILDKGYKKKGDTYKTLTEAQFRAATQLAIY
    YYTDSADLTTLKTYNDNKGYHGFDKLDDATLAVVHELITYAEDVTLPMTQ
    NLDFFVPNSSRYQALIGTQYHPNELIDVISMEDKQAPIIPITHKLTISKT
    VTGTIADKKKEFNFEIHLKSSDGQAISGTYPTNSGELTVTDGKATFTLKD
    GESLIVEGLPSGYSYEITETGASDYEVSVNGKNAPDGKATKASVKEDETV
    AFENRKDLVPPTGLTTDGAIYLWLLLLVPFGLLVWLFGRKGTKK
  • Sps0101 is referred to as a LepA protein. An example of an amino acid sequence of Sps0101 is set forth as SEQ ID NO: 66
    SEQ ID NO: 66
    MTNYLNRLNENPLLKAFIRLVLKISIIGFLGYILFQYVFGVMIVNTNQMS
    PAVSAGDGVLYYRLTDRYHINDVVVYEVDDTLKVGRIAAQAGDEVNFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKVPTGTYFILNDYREERLDSR
    YYGALPINQIKGKISTLLRVRGI
  • Sps0102 is thought to be a fimbrial protein. It contains a sortase substrate motif QVXTG shown in italics in SEQ ID NO: 67.
    SEQ ID NO: 67
    MEREKMKKNKLLLATAILATALGTASLNQNVKAETAGVSENAKLIVKKTF
    DSYTDNEVLMPKADYTFKVEADSTASGKTKDGLEIKPGIVNGLTEQIISY
    TNTDKPDSKVKSTEFDFSKVVFPGIGVYRYTVSEKQGDVEGITYDTKKWT
    VDVYVGNKEGGGFEPKFIVSKEQGTDVKKPVNFNNSFATTSLKVKKNVSG
    NTGELQKEFDFTLTLNESTNFKKDQIVSLQKGNEKFEVKIGTPYKFKLKN
    GESIQLDKLPVGITYKVNEMEANKDGYKTTASLKEGDGQSKMYQLDMEQK
    TDESADEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • Sps0103 is a SrtC2 type sortase. An example of Sps0103 is set forth in SEQ ID NO: 68.
    SEQ ID NO: 68
    MVMTIVQVINKATDTLILTFGLVVLFLAGFGLWDSYHLYQQADASNFKKF
    KTAQQQPKFEDLLALNEDVIGWLNIPGTHIDYPLVQGKTNLEYINKAVDG
    SVAMSGSLFLDTRNHNDFTDDYSLTYGHHMAGNAMFGEIPKFLKKDFFSK
    HNKAIIETKERKKLTVTIFACLKTDAFNQLVFNPNAITNQDQQRQLVDYI
    SKRSKQFKPVKLKHHTKFVAFSTCENFSTDNRVTVVGTIQE
  • Sps0104 is referred to as a hypothetical protein. It contains a sortase substrate motif LPXAG shown in italics in SEQ ID NO: 69.
    SEQ ID NO: 69
    MLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKWLVKPIPPRQPN
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • Sps0105 is referred to as a putative multiple sugar metabolism regulator. An example of Sps0105 is set forth in SEQ ID NO: 70.
    SEQ ID NO: 70
    MALVPHFPINNVRNLLIAIDAFFDTQFETTCQQTIHQLLQHSKQMTADPD
    ITHRLKHISKASSQLPPVLEHLNHIMDLVKLGNPQLLKQEINRIPLSSIT
    SSSISALRAEKNLTVIYLTRLLEPSFVENTDVAKHYSLVKYYMALNEEAS
    DLLKVLRIRCAAIIHFSESLTNKSISDKRQMYNSVLHYVDSHLYSKLKVS
    DIAKRLYVSESHLRSVEKKYSNVSLQHYTLSTKIKEAQLLLKRGIPVGEV
    AKSLYFYDTTHFHKIFKKYTGISSKDYLAKYRDNI
  • Sps0106 is thought to be a F2 like fibronectic binding protein. It contains a sortase substrate LPXTG (SEQ ID NO: 122) shown in italics in SEQ ID NO: 71.
    SEQ ID NO: 71
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDATWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPTIYFKLYRQLPGEKEVAVDDAELKQTN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEFGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTTEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEVATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GEVVDTTEDTQSGMTGHSGSTTKIEDSKSSDVIVGGQGQIVETTEDTQTG
    MHGDSGRKTEVEDTKLVQSFHFDNKEPESNSEIPKKDKSKSNTSLPATGE
    KQHNKFFWMVTSCSLISSVFVISLKSKKRLSSC
  • Examples of GAS AI-3 sequences from M5 isolate Manfredo are set forth below.
  • Orf 77 encodes a negative transcription regulator (Nra). An example of the nucleotide sequence encoding Nra (SEQ ID NO: 88) and an Nra amino acid sequence (SEQ ID NO: 89) are set forth below.
    SEQ ID NO: 88
    ATGCCTTATGTCAAAAAGAAAAAGGATAGTTTCTTAGTAGAAACATATCT
    TGAACAGTCTATTAGAGATAAAAGTGAATTAGTCTTACTGTTATTTAAAT
    CGCCTACTATCATTTTTTCTCATGTTGCTAAACAAACTGGTCTGACGGCT
    GTACAATTAAAATATTACTGTAAAGAACTTGATGACTTTTTTGGAAATAA
    TTTAGACATTACCATTAAAAAGGGCAAAATAATATGTTGTTTTGTCAAAC
    CTGTTAAGGAATTCTACCTTCATCAACTCTATGACACATCAACAATATTA
    AAATTATTAGTTTTCTTTATTAAAAATGGAACGTCATCACAACCTCTGAT
    TAAATTTTCAAAAAAGTATTTTCTATCAAGCTCCTCAGCTTATCGACTAC
    GGGAATCGCTGATCAAATTACTACGGGAATTTGGCTTGAGAGTCTCAAAA
    AATACAATTGTCGGAGAGGAATATCGTATTCGCTATCTTATTGCCATGCT
    ATATAGTAAATTTGGCATTGTCATCTATCCGTTAGATCATCTAGACAATC
    AAATTATTTATCGCTTCTTATCACAAAGTGCAACCAATTTAAGAACATCG
    CCCTGGCTAGAGGAACCTTTTTCTTTTTATAATATGTTACTTGCCTTGTC
    ATGGAAACGTCACCAATTTGCAGTTAGCATTCCTCAAACACGTATTTTTC
    GACAATTAAAAAAGCTTTTTATCTATGATTGTTTAACTCGAAGCAGTCGA
    CAAGTAATCGAAAATGCTTTTTCGTTAATGTTCTCACAAGGAGATCTCGA
    TTATCTTTTTTTAATTTATATTACCACCAATAATTCCTTTGCCAGCCTAC
    AATGGACTCCACAGCATATTGAAACTTGCTGCCATATTTTTGAAAAAAAT
    GACACATTTCGGTTATTGTTAGAGCCCATTCTTAAACGTTTACCGCAATT
    AAACCATTCTAAACAAGACCTTATTAAAGCCCTTATGTATTTTTCAAAAT
    CTTTTCTATTTAACCTCCAACATTTCGTCATCGAGATTCCTTCTTTTTCC
    TTGCCGACCTATACAGGCAACTCTAATCTTTACAAAGCTTTAAAAAATAT
    TGTAAATCAGTGGCTTGCTCAATTACCCGGAAAGCGTCATCTTAACGAAA
    AGCATCTCCAACTTTTTTGCTCTCATATTGAACAAATCTTAAAAAATAAA
    CAACCTGCTTTAACTGTCGTTTTAATATCTAGTAACTTTATAAATGCTAA
    ACTCCTTACAGATACTATCCCACGATATTTTTCTGATAAAGGAATTCATT
    TTTATTCTTTTTACTTATTAAGAGATGATATCTATCAAATTCCAAGCTTA
    AAACCAGATTTAGTTATCACTCATAGCCGATTAATTCCTTTTGTTAAGAA
    TGATCTGGTCAAAGGTGTTACTGTTGCTGAATTTTCTTTTGATAACCCTG
    ACTACTCTATTGCTTCAATTCAAAACTTGATATATCAGCTCAAAGATAAA
    AAATATCAAGATTTTCTAAACGAGCAATTACAA
    SEQ ID NO: 89
    MPYVKKKKDSFLVETYLEQSIRDKSELVLLLFKSPTIIFSHVAKQTGLTA
    VQLKYYGKELDDFPGNNLDITIKKGKIICCFVKPVKEFYLHQLYDTSTIL
    KLLVFFIKNGTSSQPLIKFSKKYFLSSSSAYRLRESLIKLLREFGLRVSK
    NTIVGEEYRIRYLIAMLYSKFGIVIYPLDHLDNQIIYRFLSQSATNLRTS
    PWLEEPFSFYNMLLALSWKRHQFAVSIPQTRIFRQLKKLFIYDCLTRSSR
    QVIENAFSLMFSQGDLDYLFLIYITTNNSFASLQWTPQHIETCCHIFEKN
    DTFRLLLEPILKRLPQLNHSKQDLIKALMYFSKSFLFNLQHFVIEIPSFS
    LPTYTGNSNLYKALKNIVNQWLAQLPGKRHLNEKHLQLFCSHIEQILKNK
    QPALTVVLISSNFINAKLLTDTIPRYFSDKGIHFYSEYLLRDDIYQIPSL
    KPDLVITHSRLIPFVKNDLVKGVTVAEFSFDNPDYSIASIQNLIYQLKDK
    KYQDFLNEQLQ
  • Orf 78 is thought to be a collagen binding protein (Cbp). An example of the nucleotide sequence encoding Cbp (SEQ ID NO: 90) and a Cbp amino acid sequence (SEQ ID NO: 91) are set forth below.
    SEQ ID NO: 90
    TTGCAAAAGAGGGATAAAACCAATTATGGAAGCGCTAACAACAAACGACG
    ACAAACGACGATCGGATTACTGAAAGTATTTTTGACGTTTGTAGCTCTGA
    TAGGAATAGTAGGGTTTTCTATCAGAGCGTTCGGAGCTGAAGAAAAATCT
    ACTGAAACTAAAAAAACGTCAGTCATTATTAGAAAATATGCTGAAGGTGA
    CTACTCTAAACTTCTAGAGGGAGCAACTTTGCGTTTAACAGGGGAAGATA
    TCCCAGATTTTCAAGAAAAAGTCTTCCAAAGTAATGAACAGGAGAAAAAG
    ATTGAATTATCAAATGGGACTTATACCTTAACAGAAACATCATCTCCAGA
    TGGATATAAAATTACGGAGCCGATTAAGTTTAGAGTAGTGAATAAAAAAG
    TATTTATCGTCCAAAAAGATGGTTCTCAAGTGGAAAACCCAAACAAAGAA
    CTAGGTTCTCCATATACTATAGAGGCATACAATGATTTTGATGAATTTGG
    CTTACTGTCAACACAAAATTATGCGAAATTTTATTATGGAAAAAACTATG
    ATGGCAGTTCACAAATTGTTTATTGCTTCAATGCCAACTTGAAATCTCCA
    CCTGACTCGGAAGATCATGGTGCTACAATAAATCCTGACTTTACGACTGG
    TGATATTAGGTACAGTCATATTGCTGGTTCAGATTTGATAAAATACGCTA
    ATACAGCTAGGGATGAAGATCCTCAATTATTTTTAAAACACGTAAAAAAA
    GTAATTGAAAATGGGTATCATAAAAAAGGTCAAGCTATTCCATATAACGG
    TCTGACTGAGGCACAGTTTCGTGCGGCTACTCAACTGGCAATTTATTATT
    TTACAGATAGTGTTGACTTAACTAAGGATAGATTGAAAGACTTCCATGGA
    TTTGGAGATATGAATGATCAAACTTTGGGTGTAGCTAAAAAAATTGTAGA
    ATACGCTTTGAGTGATGAAGATTCAAAACTAACAAATCTTGATTTCTTCG
    TACCTAATAATAGCAAATACCAATCTCTTATTGGGACAGAATACCATCCA
    GATGATTTGGTTGACGTGATTCGTATGGAAGATAAAAAGCAAGAAGTTAT
    TCCAGTAACTCATAGTTTGACGGTGCAAAAAACAGTAGTCGGTGAGTTGG
    GAGATAAGACTAAAGGCTTTCAATTTGAACTTGAGTTGAAAGATAAAACT
    GGACAGCCTATTGTTAACACTCTAAAAACTAATAATCAAGATTTAGTAGC
    TAAAGATGGGAAATATTCATTTAATCTAAAGCATGGTGACACCATAAGAA
    TAGAAGGATTACCGACGGGATATTCTTATACCCTGAAAGAGACTGAAGCT
    AAGGATTATATAGTAACTGTTGATAACAAAGTTAGTCAAGAAGCTCAATC
    AGCAAGTGAGAATGTCACAGCAGACAAAGAAGTCACTTTTGAAAACCGAA
    AAGATCTTGTCCCACCAACTGGTTTGACAACAGATGGGGCTATCTATCTT
    TGGTTATTACTACTTGTTCCATTTGGGTTATTGGTTTGGCTATTTGGTCG
    TAAAGGGTTAAAAAATGAC
    SEQ ID NO: 91
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEKS
    TETKKTSVIIRKYAEGDYSKLLEGATLRLTGEDIPDFQEKVFQSNGTGEK
    IELSNGTYTLTETSSPDGYKITEPEKFRVVNKKVFIVQKDGSQVENPNKE
    LGSPYTIEAYNDFDEFGLLSTQNYAKFYYGKNYDGSSQIVYCFNANLKSP
    PDSEDHGATINPDFTTGDIRYSHIAGSDLIKYANTARDEDPQLFLKHVKK
    VIENGYHKKGQATPYNGLTEAQFRAATQLAIYYETDSVDLTKDRLKDPHG
    FGDMNDQTLGVAKKIVEYALSDEDSKLTNLDFFVPNNSKYQSLIGTEYHP
    DDLVDVIRMEDKKQEVTPVTHSLTVQKTVVGELGDKTKGFQFELELKDKT
    GQPIVNTLKTNNQDLVAKDGKYSFNLKHGDTIRIEGLPTGYSYTLKETEA
    KDYIVTVDNKVSQEAQSASENVTADKEVTFENRKDLVPPTGLTTDGAIYL
    WLLLLVPFGLLVWLFGRKGLKND
  • Orf 78 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 184 VPPTG (shown in italics in SEQ ID NO: 91, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Orf 78 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Three E boxes containing conserved glutamic residues have been identified in Orf 78. The E-box motifs are underlined in SEQ ID NO: 91, below. The conserved glutamic acid (E) residues, at amino acid residues 112, 395, and 447, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of Orf 78. Preferred fragments of Orf 78 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 91
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEKS
    TETKKTSVIIRKYAEGDYSKLLEGATLRLTGEDIPDFQEKVFQSNGTGEK
    IELSNGTYTLTETSSPDGYKITEPEKFRVVNKKVFIVQKDGSQVENPNKE
    LGSPYTIEAYNDFDEFGLLSTQNYAKFYYGKNYDGSSQIVYCFNANLKSP
    PDSEDHGATINPDFTTGDIRYSHIAGSDLIKYANTARDEDPQLFLKHVKK
    VIENGYHKKGQATPYNGLTEAQFRAATQLAIYYETDSVDLTKDRLKDPHG
    FGDMNDQTLGVAKKIVEYALSDEDSKLTNLDFFVPNNSKYQSLIGTEYHP
    DDLVDVIRMEDKKQEVTPVTHSLTVQKTVVGELGDKTKGFQFELELKDKT
    GQPIVNTLKTNNQDLVAKDGKYSFNLKHGDTIRIEGLPTGYSYTLKETEA
    KDYIVTVDNKVSQEAQSASENVTADKEVTFENRKDLVPPTGLTTDGAIYL
    WLLLLVPFGLLVWLFGRKGLKND
  • Orf 79 is thought to be a LepA signal peptidase I. An example of the nucleotide sequence encoding a LepA signal peptidase I (SEQ ID NO: 92) and a LepA signal peptidase I amino acid sequence (SEQ ID NO: 93) are set forth below.
    SEQ ID NO: 92
    ATGACTAATTACCTAAATCGTTTAAATGAGAATTCACTATTTAAAGCTTT
    CATACGGTTAGTACTTAAGATTTCTATTATTGGGTTTCTAGGTTACATTC
    TATTTCAGTATGTTTTTGGTGTTATGATTATTAACACTAATGATATGAGT
    CCTGCTTTAAGTGCAGGTGACGGTGTTTTATATTATCGTTTGACTGATCG
    CTATCATATTAATGATGTGGTGGTCTATGAGGTTGATAACACTTTGAAAG
    TTGGTCGAATTGTCGCTCAAGCTGGCGATGAGGTTAGTTTTACGCAAGAA
    GGAGGACTGTTGATTAATGGGCATCCACCAGAAAAAGAGGTCCCTTACCT
    GACGTATCCTCACTCAAGTGGCCCAAACTTTCCCTATAAAGTTCCTACGG
    GTAAGTATTTCATATTGAATGATTATCGTGAAGAACGTTTGGACAGTCGT
    TATTATGGGGCGTTACCCGTCAATCAAATAAAAGGGAAAATCTCAACTCT
    ATTAAGAGTGAGAGGAATT
    SEQ ID NO: 93
    MTNYLNRLNENSLFKAFIRLVLKISIIGFLGYTLFQYVFGVMIINTNDMS
    PALSAGDGVLYYRLTDRYHINDVVVYEVDNTLKVGRTVAQAGDEVSFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKTPTGKYFILNDYREERLDSR
    YYGALPVNQIKGKISTLLRVRGI
  • Orf 80 is thought to to be a fimbrial protein. An example of the nucleotide sequence encoding the fimbrial protein (SEQ ID NO: 94) and a fimbrial protein amino acid sequence (SEQ ID NO: 95) are set forth below.
    SEQ ID NO: 94
    TTGGAGAGAGAAAAAATGAAAAAAAACAAATTATTACTTGCTACTGCAAT
    CTTAGCAACTGCTTTAGGAACAGCTTCTTTAAATCAAAACGTAAAAGCTG
    AGACGGCAGGGGTTGTAACAGGAAAATCACTACAAGTTACAAAGACAATG
    ACTTATGATGATGAAGAGGTGTTAATGCCCGAAACCGCCTTTACTTTTAC
    TATAGAGCCTGATATGACTGCAAGTGGAAAAGAAGGCAGCCTAGATATTA
    AAAATGGAATTGTAGAAGGCTTAGACAAACAAGTAACAGTAAAATATAAG
    AATACAGATAAAACATCTCAAAAAACTAAAATAGCACAATTTGATTTTTC
    TAAGGTTAAATTTCCAGCTATAGGTGTTTACCGCTATATGGTTTCAGAGA
    AAAACGATAAAAAAGACGGAATTACGTACGATGATAAAAAGTGGACTGTA
    GATGTTTATGTTGGGAATAAGGCCAATAACGAAGAAGGTTTCGAAGTTCT
    ATATATTGTATCAAAAGAAGGTACTTCTAGTACTAAAAAACCAATTGAAT
    TTACAAACTCTATTAAAACTACTTCCTTAAAAATTGAAAAACAAATAACT
    GGCAATGCAGGAGATCGTAAAAAATCATTCAACTTCACATTAACATTACA
    ACCAAGTGAATATTATAAAACTGGATCAGTTGTGAAAATCGAACAGGATG
    GAAGTAAAAAAGATGTGACGATAGGAACGCCTTACAAATTTACTTTGGGA
    CACGGTAAGAGTGTCATGTTATCGAAATTACCAATTGGTATCAATTACTA
    TCTTAGTGAAGACGAAGCGAATAAAGACGGCTACACTACAACGGCAACAT
    TAAAAGAACAAGGCAAAGAAAAGAGTTCCGATTTCACTTTGAGTACTCAA
    AACCAGAAAACAGACGAATCTGCTGACGAAATCGTTGTCACAAATAAGCG
    TGACACTCAAGTTCCAACTGGTGTTGTAGGGACCCTTGCTCCATTTGCAG
    TTCTTAGCATTGTGGCTATTGGTGGAGTTATCTATATTACAAAACGTAAA
    AAAGCT
    SEQ ID NO: 95
    MEREKMKKNKLLLATAILATALGTASLNQNVKAETAGVVTGKSLQVTKTM
    TYDDEEVLMPETAFTPTIEPDMTASGKEGSLDIKNGIVEGLDKQVTVKYK
    NTDKTSQKTKIAQFDFSKVKFPAIGVYRYMVSEKNDKKDGITYDDKKWTV
    DVYVGNKANNEEGPEVLYIVSKEGTSSTKKPIEFTNSIKTTSLKIEKQIT
    GNAGDRKKSFNFTLTLQPSEYYKTGSVVKIEQDGSKKDVTIGTPYKFTLG
    HGKSVMLSKLPIGINYYLSEDEANKDGYTTTATLKEQGKEKSSDFTLSTQ
    NQKTDESADEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRK
    KA
  • Orf 82 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 140 QVPTG (shown in italics in SEQ ID NO: 95, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Orf 82 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • An E box containing a conserved glutamic residue has been identified in Orf 80. The E-box motif is underlined in SEQ ID NO: 95, below. The conserved glutamic acid (E), at amino acid residue 270, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of Orf 80. Preferred fragments of Orf 80 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 95
    MEREKMKKNKLLLATAILATALGTASLNQNVKAETAGVVTGKSLQVTKTM
    TYDDEEVLMPETAFTPTIEPDMTASGKEGSLDIKNGIVEGLDKQVTVKYK
    NTDKTSQKTKIAQFDFSKVKFPAIGVYRYMVSEKNDKKDGITYDDKKWTV
    DVYVGNKANNEEGPEVLYIVSKEGTSSTKKPIEFTNSIKTTSLKIEKQIT
    GNAGDRKKSFNFTLTLQPSEYYKTGSVVKIEQDGSKKDVTIGTPYKFTLG
    HGKSVMLSKLPIGINYYLSEDEANKDGYTTTATLKEQGKEKSSDFTLSTQ
    NQKTDESADEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRK
    KA
  • Orf 81 is thought to to be a SrtC2 type sortase. An example of the nucleotide sequence encoding the SrtC2 sortase (SEQ ID NO: 96) and a SrtC2 sortase amino acid sequence (SEQ ID NO: 97) are set forth below.
    SEQ ID NO: 96
    GTGATTAGTCAAAGAATGATGATGACAATTGTACAGGTTATCAATAAAGC
    CATTGATACTCTCATTCTTATCTTTTGTTTAGTCGTACTATTTTTAGCTG
    GTTTTGGTTTGTGGGATTCTTATCATCTCTATCAACAAGCAGACGCTTCT
    AATTTCAAAAAATTTAAAACAGCTCAACAACAGCCTAAATTTGAAGACTT
    GTTAGCTTTGAATGAGGATGTCATTGGTTGGTTAAATATCCCAGGGACTC
    ATATTGATTATCCTCTAGTTCAGGGAAAAACGAATTTAGAGTATATTAAT
    AAAGCAGTTGATGGCAGTGTTGCCATGTCTGGTAGTTTATTTTTAGATAC
    ACGGAATCATAATGATTTTACGGACGATTACTCTCTGATTTATGGCCATC
    ATATGGCAGGTAATGCCATGTTTGGCGAAATTCCAAAATTTTTAAAAAAG
    GATTTTTTCAACAAACATAATAAAGCTATCATTGAAACAAAAGAGAGAAA
    AAAACTAACCGTCACTATTTTTGCTTGTCTCAAGACAGATGCCTTTGACC
    AGTTAGTTTTTAATCCTAATGCTATTACCAATCAAGACCAACAAAAGCAG
    CTCGTTGATTATATCAGTAAAAGATCAAAACAATTTAAACCTGTTAAATT
    GAAGCATCATACAAAGTTCGTTGCTTTTTCAACGTGTGAAAATTTTTCTA
    CTGACAATCGTGTTATCGTTGTCGGTACTATTCAAGAA
    SEQ ID NO: 97
    MISQRMMMTIVQVINKAIDTLILIFCLVVLFLAGFGLWDSYHLYQQADAS
    NFKKFKTAQQQPKFEDLLALNEDVIGWLNIPGTHIDYPLVQGKTNLEYIN
    KAVDGSVAMSGSLFLDTRNHNDFTDDYSLIYGHHMAGNAMFGEIPKFLKK
    DFFNKHNKAIIETKERKKLTVTIFACLKTDAFDQLVFNPNAITNQDQQKQ
    LVDYISKRSKQFKPVKLKHHTKFVAFSTCENFSTDNRVIVVGTIQE
  • Orf 82 is referred to as a hypothetical protein. It contains a sortase substrate motif LPXAG shown in italics in SEQ ID NO: 99. An example of the nucleotide sequence encoding the hypothetical protein (SEQ ID NO: 98) and a hypothetical protein amino acid sequence (SEQ ID NO: 99) are set forth below.
    SEQ ID NO: 98
    TTGCTTTTTCAACGTGTGAAAATTTTTCTACTGACAATCGTGTTATCGTT
    GTCGGTACTATTCAAGAATAACGAAAGGAGGAGACTTTTGAGAAAATATT
    GGAAAATGTTATTTTCTGTCGTAATGATATTAACCATGCTGGCCTTTAAT
    CAGACTGTTTTAGCAAAAGACAGCACTGTTCAAACTAGCATTAGTGTCGA
    AAATGTCTTAGAGAGAGCAGGCGATAGTACCCCATTTTCGGTTGCATTAG
    AATCAATTGATGCGATGAAAACAATAGACGAAATAACAATTGCTGGTTCT
    GGAAAAGCAAGCTTTTCCCCTCTGACCTTCACAACAGTTGGGCAATATAC
    TTATCGTGTTTATCAGAAGCCTTCACAAAATAAAGATTATCAAGCAGATA
    CTACTGTATTTGACGTTCTTGTCTATGTGACCTATGATGAAGATGGGACT
    CTAGTCGCAAAAGTTATTTCTCGAAGGGCTGGAGACGAAGAAAAATCAGC
    GATTACTTTTAAGCCCAAACGGTTAGTAAAACCAATACCGCCTAGACAAC
    CTAACATCCCTAAAACCCCATTACCATTAGCTGGTGAAGTAAAAAGTTTA
    TTGGGTATCTTAAGTATCGTATTACTGGGGTTACTAGTTCTTCTTTATGT
    TAAAAAACTGAAGAGTAGGCTA
    SEQ ID NO: 99
    MLFQRVKIFLLTIVLSLSVLFKNNERRRLLRKYWKMLFSVVMILTMLAFN
    QTVLAKDSTVQTSISVENVLEPAGDSTPFSVALESIDAMKTIDEITIAGS
    GKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTTVFDVLVYVTYDEDGT
    LVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPNIPKTPLPLAGEVKSL
    LGILSTVLLGLLVLLYVKKLKSRL
  • Orf 82 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 185 LPLAG (shown in italics in SEQ ID NO: 99, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Orf 82 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in Orf 82. The pilin motif sequence is underlined in SEQ ID NO: 99, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 173 and 188. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of Orf 82 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 99
    MLFQRVKIFLLTIVLSLSVLFKNNERRRLLRKYWKMLFSVVMILTMLAFN
    QTVLAKDSTVQTSISVENVLERAGDSTPFSVALESIDAMKTIDEITTAGS
    GKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTTVFDVLVYVTYDEDGT
    LVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPNIPKTPLPLAGEVKSL
    LGILSIVLLGLLVLLYVKKLKSRL
  • An E box containing a conserved glutamic residue has been identified in Orf 82. The E-box motif is underlined in SEQ ID NO: 99, below. The conserved glutamic acid (E), at amino acid residue 163, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of Orf 82. Preferred fragments of Orf 82 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 99
    MLFQRVKIFLLTIVLSLSVLFKNNERRRLLRKYWKMLFSVVMILTMLAFN
    QTVLAKDSTVQTSISVENVLEPAGDSTPFSVALESIDAMKTIDEITIAGS
    GKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTTVFDVLVYVTYDEDGT
    LVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPNIPKTPLPLAGEVKSL
    LGILSTVLLGLLVLLYVKKLKSRL
  • Orf 83 is thought to to be a multiple sugar metabolism regulator protein. An example of a nucleotide sequence encoding the sugar metabolism regulator protein (SEQ ID NO: 100) and a sugar metabolism regulator protein amino acid sequence (SEQ ID NO: 101) are set forth below.
    SEQ ID NO: 100
    ATGATACAACTAAGGATGGGGGCAATCTATCAAATGGTTATATTCGATTT
    AAAACATGTGCAAACATTACACAGCTTGTCTCAATTACCTATTTCAGTGA
    TGTCACAAGATAAGGCACTTATTCAAGTATATGGTAATGACGACTATTTA
    TTATGTTACTATCAATTTTTAAAGCATCTAGCTATTCCTCAAGCTGCACA
    AGATGTTATTTTTTATGAGGGTTTATTTGAAGAGTCCTTTATGATTTTTC
    CTCTTTGTCACTACATTATTGCCATTGGACCTTTCTATCCTTATTCACTT
    AATAAAGACTATCAGGAACAATTAGCTAATAATTTTTTAAAACATTCTTC
    TCATCGTAGCAAAGAAGAGCTCTTGTCCTATATGGCACTTGTCCCACATT
    TTCCAATTAATAATGTGCGGAACCTTTTGATAGCTATTGACGCTTTTTTT
    GACACACAATTTGAGACGACTTGCCAACAAACGATTCATCAATTGTTGCA
    GCATTCAAAACAGATGACTGCTGATCCTGATATCATTCATCGCCTTAAGC
    ATATTAGCAAAGCATCTAGCCAATTACCGCCTGTTTTAGAGCACCTAAAT
    CATATTATGGATCTGGTAAAGCTAGGCAATCCACAATTGCTCAAGCAAGA
    AATCAATCGCATCCCCTTATCAAGTATCACCTCATCTTCTATTTCTGCTC
    TAAGGGCGGAAAGAACCTCACTGTTATCTATTTCAACTAGGTTACTGGAA
    TTCAGTTTTGTAGAAAATACTGACGTAGCAAAGCATTATAGCCTTGTCAA
    ATACTACATGGCCTTAAATGAAGAAGCGAGTGACTTGCTCAAAGTTTTGA
    GAATTCGCTGTGCAGCTATCATCCATTTTTCCGAATCATTAACCAATAAA
    AGTATTTCTGATAAACGTCAAATGTACAATAGTGTGCTTCATTATGTCGA
    TAGTCACCTGTATTCCAAATTAAAGGTATCTGATATCGCTAAGCGCCTAT
    ATGTTTCCGAATCTCACTTACGTTCAGTCTTTAAAAAATACTCAAATGTT
    TCCTTACAACATTATATTCTAAGTACAAAAATCAAAGAAGCTCAACTACT
    CTTAAAACGAGGAATTCCTGTTGGAGAAGTGGCTAAAAGCTTATATTTTT
    ATGACACTACCCATTTTCATAAAATCTTTAAAAAATACACGGGTATTTCT
    TCAAAAGACTATCTTGCTAAATACCGAGATAATATT
    SEQ ID NO: 101
    MIQLRMGAIYQMVIFDLKHVQTLHSLSQLPISVMSQDKALIQVYGNDDYL
    LCYYQFLKHLAIPQAAQDVIFYEGLFEESFMIFPLCHYIIAIGPFYPYSL
    NKDYQEQLANNFLKHSSHRSKEELLSYMALVPHFPINNVRNLLIAIDAFF
    DTQFETTCQQTIHQLLQHSKQMTADPDIIHRLKHISKASSQLPPVLEHLN
    HIMDLVKLGNPQLLKQEINRIPLSSITSSSISALRAEKNLTVIYLTRLLE
    FSFVENTDVAKHYSLVKYYMALNEEASDLLKVLRIRCAAIIHFSESLTNK
    SISDKRQMYNSVLHYVDSHLYSKLKVSDIAKRLYVSESHLRSVFKKYSNV
    SLQHYILSTKIKEAQLLLKRGIPVGEVAKSLYFYDTTHFHKIFKKYTGTS
    SKDYLAKYRDNI
  • Orf 84 is thought to to be a F2-like fibronectin-binding protein. An example of a nucleotide sequence encoding the F2-like fibronectin-binding protein (SEQ ID NO: 102) and a F2-like fibronectin-binding protein amino acid sequence (SEQ ID NO: 103) are set forth below.
    SEQ ID NO: 102
    ATGACACAAAAAAATAGCTATAAGTTAAGCTTCCTGTTATCCCTAACAGG
    ATTTATTTTAGGTTTATTATTGGTTTTTATAGGATTGTCCGGAGTATCAG
    TAGGACATGCGGAAACAAGAAATGGAGCAAACAAACAAGGAGCTTTTGAA
    ATCAAGAAAAATAAAAGTCAAGAAGAATATAATTATGAAGTTTATGATAA
    CAGAAACATACTTCAGGATGGGGAACATAAACTTGAAATAAAAAGAGTTG
    ATGGGACAGGTAAAACTTATCAAGGTTTTTGCTTTCAGTTAACGAAAAAT
    TTTCCCACTGCTCAAGGTGTAAGTAAAAAGCTGTATAAAAAATTGAGTAG
    TAGTGATGAAGAAACACTAAAGCAATATGCCTCTAAGTATACAAGTAATA
    GGAGAGGAGATACTAGTGGTAATCTTAAAAAGCAAATTGCTAAGGTTCTG
    ACAGAAGGTTACCCAACTAACAAAAGTGATTGGTTAAATGGATTGACTGA
    AAACGAAAAAATAGAAGTAACCCAGGATGCAATTTGGTATTTTACAGAAA
    CGACAGTTCCGGCTGATAGAAGTTATACGAATCGCAACGTAAATAGTCAA
    AAAATGAAAGAAGTGTATCAAAAGCTAATTGATACAACAGATATAGATAA
    ATATGAAGATGTACAATTTGATTTATTTGTGCCACAAGATACAAACTTAC
    AGGCAGTAATTAGTGTAGAGCCTGTTATCGAAAGCCTTCCTTGGACATCG
    TTGAAGCCAATAGCCCAGAAGGATATCACTGCCAAAAAAATCTGGGTAGA
    TGCACCTAAAGAAAAACCAATTATTTATTTTAAGCTATATAGACAGCTGC
    CTGGAGAAAAGGAAGTAGCAGTGGATGACGCTGAGCTAAAACAGATAAAT
    AGTGAAGGTCAACAAGAAATATCAGTAACTTGGACAAATCAACTTGTTAC
    AGATGAAAAAGGAATGGCTTACATTTATTCTGTAAAAGAAGTAGATAAAA
    ATGGCGAGTTACTTGAGCCAAAAGATTATATCAAGAAGGAAGATGGACTT
    ACAGTTACTAATACTTATGTAAAGCCAACTAGTGGGCACTATGATATAGA
    AGTGACATTTGGAAATGGACATATTGATATTACAGAAGATACTACACCAG
    ATATTGTTTCAGGTGAAAACCAAATGAAGCAAATAGAGGGAGAAGATAGT
    AAGCCTATTGATGAAGTAACGGAAAATAATTTAATTGAATTTGGTAAAAA
    CACGATGCCAGGTGAAGAAGATGGCACAAATTCTAATAAGTATGAAGAAG
    TCGAAGACTCACGCCCAGTTGATACCTTGTCAGGTTTATCAAGTGAGCAA
    GGTCAGTCCGGTGATATGACAATTGAAGAAGATAGTGCTACCCATATTAA
    ATTCTCAAAACGTGATATTGACGGCAAAGAGTTAGCTGGTGCAACTATGG
    AGTTGCGTGATTCATCTGGTAAAACTATTAGTACATGGATTTCAGATGGA
    CAAGTGAAAGATTTCTACCTGATGCCAGGAAAATATACATTTGTCGAAAC
    CGCAGCACCAGACGGTTATGAGATAGCAACTGCTATTACCTTTACAGTTA
    ATGAGCAAGGTCAGGTTACTGTAAATGGCAAAGCAACTAAAGGTGACGCT
    CATATTGTCATGGTTGATGCTTACAAGCCAACTAAGGGTTCAGGTCAGGT
    TATTGATATTGAAGAAAAGCTTCCAGACGAGCAGGGCCATTCTGGCTCAA
    CTACTGAAATAGAAGATAGCAAGTCTTCAGACGTTATCATTGGTGGTCAG
    GGGCAGATTGTCGAGACAACAGAGGATACCCAAACTGGCATGCACGGGGA
    TTCTGGTTGTAAAACGGAAGTCGAAGATACTAAACTAGTACAATCCTTCC
    ACTTTGATAACAAGGAATCAGAAAGTAACTCTGAGATTCCTAAAAAAGAT
    AAGCCAAAGAGTAATACTAGTTTACCAGCAACTGGTGAGAAGCAACATAA
    TATGTTCTTTTGGATGGTTACTTCTTGCTCACTTATTAGTAGTGTTTTTG
    TAATATCACTAAAAACTAAAAAACGCCTATCATCATGT
    SEQ ID NO: 103
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEFGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFFWMVTSCSLISSVFVISLKTKKRLSSC
  • Orf 84 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 181 LPATG (shown in italics in SEQ ID NO: 103, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant Orf 84 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in Orf 84. The pilin motif sequence is underlined in SEQ ID NO: 103, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 270. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of Orf 84 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 103
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVTSVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEPGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFEWMVTSCSLISSVFVISLKTKKRLSSC
  • An E box containing a conserved glutamic residue has been identified in Orf 84. The E-box motif is underlined in SEQ ID NO: 103, below. The conserved glutamic acid (E), at amino acid residue 516, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of Orf 84. Preferred fragments of Orf 84 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 103
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVTSVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEPGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFEWMVTSCSLISSVFVISLKTKKRLSSC
  • Examples of GAS AI-3 sequences from M18 strain isolate MGAS 8232 are set forth below.
  • SpyM180125 is a negative transcriptional regulator (Nra). An example of SpyM180125 is set forth in SEQ ID NO: 72.
    SEQ ID NO: 72
    MPYVKKKKDSFLVETYLEQSIRDKSELVLLLFKSPTIIFSHVAKQTGLTA
    VQLKYYCKELDDFFGNNLDITIKKGKIICCFVKPVKEFYLHQLYDTSTIL
    KLLVFFIKNGTTSQPLIKFSKKYFLSSSSAYRLRESLIKLLREFGLRVSK
    NTIVGEEYRIRYLIAMLYSKFGIVIYPLDHLDNQIIYRFLSQSATNLRTS
    PWLEEPFSFYNMLLALS
  • SpyM180126 is thought to be a collagen binding protein (CBP). An example of SpyM180126 is set forth in SEQ ID NO: 73.
    SEQ ID NO: 73
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    TETKKTSVIIRKYAEGDYSKLLEGATLKLAQIEGSGFQEQSFESSTSGQK
    LQLSDGTYILTETKSPQGYEIAEPITFKVTAGKVFIKGKDGQFVENQNKE
    VAEPYSVTAYNDFDDSGFINPKTFTPYGKFYYAKNANGTSQVVYCFNVDL
    HSPPDSLDKGETIDPDFNEGKEIKYTHILGADLFSYANNPRASTNDELLS
    QVKKVLEKGYRDDSTTYANLTSVEFRAATQLAIYYFTDSVDLDNLADYHG
    FGALTTEALNATKEIVAYAEDRANLPNISNLDFYVPNSNKYQSLIGTQYH
    PESLVDIIRMEDKQAPIIPITHKLTISKTVTGTIADKKKEFNFEIHLKSS
    DGQAISGTYPTNSGELTVTDGKATFTLKDGESLIVEGLPSGYSYEITETG
    ASDYEVSVNGKNAPDGKATKASVKEDETITFENRKDLVPPTGLTTDGAIY
    LWLLLLVLLGLWVWLIGRKGLKND
  • SpyM180126 contains an amino acid motif indicative of a cell wall anchor: SEQ II ID NO: 184 VPPTG (shown in italics in SEQ ID NO: 73, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM180126 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM180126. The pilin motif sequence is underlined in SEQ ID NO: 73, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 172 and 179. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM180126 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 73
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    TETKKTSVIIRKYAEGDYSKLLEGATLKLAQIEGSGFQEQSFESSTSGQK
    LQLSDGTYILTETKSPQGYEIAEPITFKVTAGKVFIKGKDGQFVENQNKE
    VAEPYSVTAYNDFDDSGFINPKTFTPYGK FYYAKNANGTSQVVYCFNVDL
    HSPPDSLDKGETIDPDFNEGKEIKYTHILGADLFSYANNPRASTNDELLS
    QVKKVLEKGYRDDSTTYANLTSVEFRAATQLAIYYFTDSVDLDNLADYHG
    FGALTTEALNATKEIVAYAEDRANLPNISNLDFYVPNSNKYQSLIGTQYH
    PESLVDIIRMEDKQAPIIPITHKLTISKTVTGTIADKKKEFNFEIHLKSS
    DGQAISGTYPTNSGELTVTDGKATFTLKDGESLIVEGLPSGYSYEITETG
    ASDYEVSVNGKNAPDGKATKASVKEDETITFENRKDLVPPTGLTTDGAIY
    LWLLLLVLLGLWVWLIGRKGLKND
  • Three E boxes containing conserved glutamic residues have been identified in SpyM180126. The E-box motifs are underlined in SEQ ID NO: 73, below. The conserved glutamic acid (E) residues, at amino acid residues 112, 257, and 415, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyM180126. Preferred fragments of SpyM180126 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 73
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    TETKKTSVIIRKYAEGDYSKLLEGATLKLAQIEGSGFQEQSFESSTSGQK
    LQLSDGTYILTETKSPQGYEIAEPITFKVTAGKVFIKGKDGQFVENQNKE
    VAEPYSVTAYNDFDDSGFINPKTFTPYGKFYYAKNANGTSQVVYCFNVDL
    HSPPDSLDKGETIDPDFNEGKEIKYTHILGADLFSYANNPRASTNDELLS
    QVKKVLEKGYRDDSTTYANLTSVEFRAATQLAIYYFTDSVDLDNLADYHG
    FGALTTEALNATKEIVAYAEDRANLPNISNLDFYVPNSNKYQSLIGTQYH
    PESLVDIIRMEDKQAPIIPITHKLTISKTVTGTIADKKKEFNFEIHLKSS
    DGQAISGTYPTNSGELTVTDGKATFTLKDGESLIVEGLPSGYSYEITETG
    ASDYEVSVNGKNAPDGKATKASVKEDETITFENRKDLVPPTGLTTDGAIY
    LWLLLLVLLGLWVWLIGRKGLKND
  • SpyM180127 is a LepA protein. An example of SpyM180127 is shown in SEQ ID NO: 74.
    SEQ ID NO: 74
    MTNYLNRLNENPLEKAFIRLVLKISIIGFLGYILFQYIFGVMIINTNVMS
    PALSAGDGILYYRLTDRYHINDVVVYEVDNTLKVGRIVAQAGDEVSFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKVPTGTYFILNDYREERLDSR
    YYGALPINQIKGKISTLLRVRGI
  • SpyM18 0128 is thought to be a fimbrial protein. An example of SypM18 0128 is shown in SEQ ID NO: 75.
    SEQ ID NO: 75
    MKKNKLLLATAILATALGTASLNQNVKAETAGVIDGSTLVVKKTFPSYTD
    DKVLMPKADYTFKVEADDNAKGKTKDGLDIKPGVTDGLENTKTIHYGNSD
    KTTAKEKSVNFDFANVKFPGVGVYRYTVSEVNGNKAGIAYDSQQWTVDVY
    VVNREDGGFEAKYIVSTEGGQSDKKPVLFKNFFDTTSLKVTKKVTGNTGE
    HQRSFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKVTEEDVTKDGYKTSATLKDGDVTDGYNLGDSKTTDKST
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVATGGVIYITKRKKA
  • SpyM18 0128 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 140 QVPTG (shown in italics in SEQ ID NO: 75, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM18 0128 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM18 0128. The pilin motif sequence is underlined in SEQ ID NO: 75, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 57. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM18 0128 include the conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 75
    MKKNKLLLATAILATALGTASLNQNVKAETAGVIDGSTLVVKKTFPSYTD
    DKVLMPKADYTFKVEADDNAKGKTKDGLDIKPGVIDGLENTKTIHYGNSD
    KTTAKEKSVNFDFANVKFPGVGVYRYTVSEVNGNKAGIAYDSQQWTVDVY
    VVNREDGGFEAKYIVSTEGGQSDKKPVLFKNFFDTTSLKVTKKVTGNTGE
    HQRSFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKVTEEDVTKDGYKTSATLKDGDVTDGYNLGDSKTTDKST
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • An E box containing a conserved glutamic residue has been identified in SpyM18 0128. The E-box motif is underlined in SEQ ID NO: 75, below. The conserved glutamic acid (E), at amino acid residue 266, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyM18 0128. Preferred fragments of SpyM18 0128 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 75
    MKKNKLLLATAILATALGTASLNQNVKAETAGVIDGSTLVVKKTFPSYTD
    DKVLMPKADYTFKVEADDNAKGKTKDGLDIKPGVIDGLENTKTIHYGNSD
    KTTAKEKSVNFDFANVKFPGVGVYRYTVSEVNGNKAGIAYDSQQWTVDVY
    VVNREDGGFEAKYIVSTEGGQSDKKPVLFKNFFDTTSLKVTKKVTGNTGE
    HQRSFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKVTEEDVTKDGYKTSATLKDGDVTDGYNLGDSKTTDKST
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • SpyM180129 is a SrtC2 type sortase. An example of SpyM180129 is shown in SEQ ID NO: 76
    SEQ ID NO: 76
    MISQRMMMTIVQVINKAIDTLILIFCLVVLFLAGFGLWDSYHLYQQADAS
    NFKKFKTAQQQPKFEDLLALNEDVIGWLNIPGTHMDYPLVQGKTNLEYIN
    KAVDGSVAMSGSLFLDTRNHNDFTDDYSLIYGHHMAGNAMFGEIPKFLKK
    DFFNKHNKAIIETKERKKLTVTIFACLKTDAFDQLVFNPNAITNQDQQRQ
    LVDYISKRSKQFKPVKLKHHTKFVAFSTCENFSTDNRVIVVGTIQE
  • SpyM180130 is referred to as a hypothetical protein. An example of SpyM180130 is shown in SEQ ID NO: 77.
    SEQ ID NO: 77
    MRKYWKMLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTSF
    SVALESIDAMKTIDEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKD
    YQADTTVFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPI
    PPRQPDIPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • SpyM180130 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 185 LPLAG (shown in italics in SEQ ID NO: 77, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM180130 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM180130. The pilin motif sequence is underlined in SEQ ID NO: 77, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 144, 159, and 169. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM180130 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 77
    MRKYWKMLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTSF
    SVALESIDAMKTIDEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKD
    YQADTTVFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPI
    PPRQPDIPKTPLPLAGEVK SLLGTLSIVLLGLLVLLYVKKLKSRL
  • An E box containing a conserved glutamic residue has been identified in SpyM180130. The E-box motif is underlined in SEQ ID NO: 77, below. The conserved glutamic acid (E), at amino acid residue 134, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyM180130. Preferred fragments of SpyM180130 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 77
    MRKYWKMLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTSF
    SVALESIDAMKTIDEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKD
    YQADTTVFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPI
    PPRQPDIPKTPLPLAGEVKSLLGTLSIVLLGLLVLLYVKKLKSRL
  • SpyM180131 is referred to as a putative multiple sugar metabolism regulator. An example of SpyM180131 is set forth in SEQ ID NO: 78.
    SEQ ID NO: 78
    MAIFDLKHVQTLHSLSQLPISVMSQDKALIQVYGNDDYLLCYYQFLKHLA
    IPQAAQDVIFYEGLFEESFMIFPLCHYIIAIGPFYPYSLNKDYQEQLANN
    CLKHSSHRSKEELLSYMALVPHFPINNVRNLLIAIDAFFDTQFETTCQQT
    IHQLLQHSKQMTADPDIIHRLKHISKASSQLPPVLEHLNHIMDLVKLGNP
    QLLKQEINRIPLSSITSSSISALRAEKNLTVIYLTRLLEFSFVENTDVAK
    HYSLVKYYMALNEEASDLLKVLRIRCAAIIHFSESLTNKSISDKRQMYNS
    VLHYVDSHLYSKLKVSDTAKRLYVSESHLRSVFKKYSNVSLQHYTLSTKT
    KEAQLLLKRGIPVGEVAKSLYFYDTTHFHKIFKKYTGISSKDYLAKYRDN
    I
  • SpyM180132 is a F2 like fibronectic-binding protein. An example of SpyM180132 is set forth in SEQ ID NO: 79.
    SEQ ID NO: 79
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEPGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFFWMVTSCSLISSVPVISLKTKKRLSSC
  • SpyM180132 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 180 LPATG (shown in italics in SEQ ID NO: 79, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyM180132 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyM180132. The pilin motif sequence is underlined in SEQ ID NO: 79, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 270. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyM180132 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 79
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEPGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFFWMVTSCSLISSVPVISLKTKKRLSSC
  • An E box containing a conserved glutamic residue has been identified in SpyM180132. The E-box motif is underlined in SEQ ID NO: 79, below. The conserved glutamic acid (E), at amino acid residue 516, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyM180132. Preferred fragments of SpyM180132 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 79
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGAFE
    IKKNKSQEEYNYEVYDNRNILQDGEHKLEIKRVDGTGKTYQGFCFQLTKN
    FPTAQGVSKKLYKKLSSSDEETLKQYASKYTSNRRGDTSGNLKKQIAKVL
    TEGYPTNKSDWLNGLTENEKIEVTQDAIWYFTETTVPADRSYTNRNVNSQ
    KMKEVYQKLIDTTDIDKYEDVQFDLFVPQDTNLQAVISVEPVIESLPWTS
    LKPIAQKDITAKKIWVDAPKEKPIIYFKLYRQLPGEKEVAVDDAELKQIN
    SEGQQEISVTWTNQLVTDEKGMAYIYSVKEVDKNGELLEPKDYIKKEDGL
    TVTNTYVKPTSGHYDIEVTFGNGHIDITEDTTPDIVSGENQMKQIEGEDS
    KPIDEVTENNLIEPGKNTMPGEEDGTNSNKYEEVEDSRPVDTLSGLSSEQ
    GQSGDMTIEEDSATHIKFSKRDIDGKELAGATMELRDSSGKTISTWISDG
    QVKDFYLMPGKYTFVETAAPDGYEIATAITFTVNEQGQVTVNGKATKGDA
    HIVMVDAYKPTKGSGQVIDIEEKLPDEQGHSGSTTEIEDSKSSDVIIGGQ
    GQIVETTEDTQTGMHGDSGCKTEVEDTKLVQSFHFDNKESESNSEIPKKD
    KPKSNTSLPATGEKQHNMFFWMVTSCSLISSVPVISLKTKKRLSSC
  • Examples of GAS AI-3 sequences from M49 strain isolate 591 are set forth below.
  • SpyoM01000156 is a negative transcriptional regulator (Nra). An example of SpyoM01000156 is set forth in SEQ ID NO: 243.
    SEQ ID NO: 243
    MPYVKKKKDSFLVETYLEQSIRDKSELVLLLFKSPTIIFSHVAKQTGLTA
    VQLKYYCKELDDEFGNNLDITIKKGKIIGCFVKPVKEFYLHQLYDTSTIL
    KLLVFFIKNGTSSQPLIKFSKKYFLSSSSAYRLRESLIKLLREFGLRVSK
    NTIVGEEYRIRYLIAMLYSKFGIVIYPLDHLDNQIIYRFLSQSATNLRTS
    PWLEEPFSFYNMLLALSWKRHQFAVSIPQTRIFRQLKKLFIYDCLTRSSR
    QVIENAFSLTFSQGDLDYLELIYITTNNSEASLQWTPQHIETCCHIFEKN
    DTERLLLEPILKRLPQLNHSKQDLIKALMYFSKSFLFNLQHEVIEIPSFS
    LPTYTGNSNLYKALKNIVNQWLAQLPGKRHLNEKHLQLFCSHIEQILKNK
    QPALTVVLISSNFINAKLLTDTIPRYFSDKGIHFYSFYLLRDDTYQIPSL
    KPDLVITHSRLTPFVKNDLVKGVTVAEFSFDNPDYSIASIQNLIYQLKDK
    KYQDFLNEQLQ
  • SpyoM01000155 is thought to be a collagen binding protein (CPA). An example of SpyoM01000155 is set forth in SEQ ID NO: 244.
    SEQ ID NO: 244
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGESIRAFGAEEQS
    VPNRQSSIQDYPWYGYDSYPKGYPDYSPLKTYHNLKVNLEGSKDYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNNRNGIMKGIDPLNAILVTQNAIWYYTDSAQINPDESFKTEARSN
    GINDQQLGLMRKALKELIDPNLGSKYSNKTPSGYRLNVFESHDKTFQNLL
    SAEYVPDTPPKPGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLSQIE
    GSGFQEKDFQSNSLGETVELPNGTYTLTETSSPDGYKIAEPIKFRVENKK
    VETVQKDGSQVENPNKEVAEPYSVEAYNDFMDEEVLSGFTPYGKFYYAKN
    KDKSSQVVYCPNADLHSPPDSYDSGETTNPDTSTMKEVKYTHTAGSDLFK
    YALRPRDTNPEDFLKHIKKVIEKGYKKKGDSYNGLTETQFRAATQLAIYY
    FTDSADLKTLKTYNNGKGYHGFESMDEKTLAVTKELITYAQNGSAPQLTN
    LDFFVPNNSKYQSLIGTEYHPDDLVDVTRMEDKKQEVIPVTHSLTVKKTV
    VGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVAKDGKYSFNLKHG
    DTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQSVGKDITEDKKVT
    FENRKDLVPPTGLTTDGAIYLWLLLLVPLGLLVWLFGRKGLKND
  • SpyoM01000155 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 184 VPPTG (shown in italics in SEQ ID NO: 244, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyoM1000155 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, discussed above, containing conserved lysine (K) residues have also been identified in SpyoM01000155. The pilin motif sequence is underlined in SEQ ID NO: 244, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 71 and 261. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyoM01000155 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 244
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGFSIRAFGAEEQS
    VPNRQSSIQDYPWYGYDSYPK GYPDYSPLKTYHNLKVNLEGSKDYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNNRNGIMKGIDPLNAILVTQNAIWYYTDSAQINPDESFKTEARSN
    GINDQQLGLMRKALKELIDPNLGSKYSNKTPSGYRLNVFESHDKTFQNLL
    SAEYVPDTPPK PGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLSQIE
    GSGFQEKDFQSNSLGETVELPNGTYTLTETSSPDGYKIAEPIKFRVENKK
    VFIVQKDGSQVENPNKEVAEPYSVEAYNDFMDEEVLSGFTPYGKFYYAKN
    KDKSSQVVYCFNADLHSPPDSYDSGETINPDTSTMKEVKYTHTAGSDLFK
    YALRPRDTNPEDFLKHIKKVIEKGYKKKGDSYNGLTETQFRAATQLAIYY
    FTDSADLKTLKTYNNGKGYHGFESMDEKTLAVTKELITYAQNGSAPQLTN
    LDFFVPNNSKYQSLIGTEYHPDDLVDVIRMEDKKQEVIPVTHSLTVKKTV
    VGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVAKDGKYSFNLKHG
    DTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQSVGKDITEDKKVT
    FENRKDLVPPTGLTTDGAIYLWLLLLVPLGLLVWLFGRKGLKND
  • Two E boxes containing conserved glutamic residues have been identified in SpyoM1000155. The E-box motifs are underlined in SEQ ID NO: 244, below. The conserved glutamic acid (E) residues, at amino acid residues 329 and 668, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyoM01000155. Preferred fragments of SpyoM01000155 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 244
    MQKRDKTNYGSANNKRRQTTIGLLKVFLTFVALIGIVGESIRAFGAEEQS
    VPNRQSSIQDYPWYGYDSYPKGYPDYSPLKTYHNLKVNLEGSKDYQAYCF
    NLTKHFPSKSDSVRSQWYKKLEGTNENFIKLADKPRIEDGQLQQNILRIL
    YNGYPNNRNGIMKGIDPLNAILVTQNAIWYYTDSAQINPDESFKTEARSN
    GINDQQLGLMRKALKELIDPNLGSKYSNKTPSGYRLNVFESHDKTFQNLL
    SAEYVPDTPPKPGEEPPAKTEKTSVIIRKYAEGDYSKLLEGATLKLSQIE
    GSGFQEKDFQSNSLGETVELPNGTYTLTETSSPDGYKIAEPIKFRVENKK
    VETVQKDGSQVENPNKEVAEPYSVEAYNDFMDEEVLSGFTPYGKFYYAKN
    KDKSSQVVYCPNADLHSPPDSYDSGETTNPDTSTMKEVKYTHTAGSDLFK
    YALRPRDTNPEDFLKHIKKVIEKGYKKKGDSYNGLTETQFRAATQLAIYY
    FTDSADLKTLKTYNNGKGYHGFESMDEKTLAVTKELITYAQNGSAPQLTN
    LDFFVPNNSKYQSLIGTEYHPDDLVDVTRMEDKKQEVIPVTHSLTVKKTV
    VGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVAKDGKYSFNLKHG
    DTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQSVGKDITEDKKVT
    FENRKDLVPPTGLTTDGAIYLWLLLLVPLGLLVWLFGRKGLKND
  • SpyoM01000154 is a LepA protein. An example of SpyoM01000154 is shown in SEQ ID NO: 245.
    SEQ ID NO: 245
    MTNYLNRLNENSLFKAFIRLVLKISTIGFLGYILFQYVFGVMIINTNDMS
    PALSAGDGVLYYRLADRSHINDVVVYEVDNTLKVGRIAAQAGDEVNFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKVPTGTYFILNDYREERLDSR
    YYGALPINQIKGKISTLLRVRGI
  • SpyoM01000153 is thought to be a fimbrial protein. An example of SpyoM01000153 is shown in SEQ ID NO: 246.
    SEQ ID NO: 246
    MKKNKLLLATAILATALGMASMSQNIKAETAGVIDGSTLVVKKTFPSYTD
    DNVLMPKADYSFKVEADDNAKGKTKDGLDIKPGVIDGLENTKTIRYSNSD
    KITAKEKSVNFEFANVKFPGVGVYRYTVAEVNGNKAGITYDSQQWTVDVY
    VVNKEGGGFEVKYIVSTEVGQSEKKPVLFKNSFDTTSLKIEKQVTGNTGE
    HQRLFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKLTEEDVTKDGYKTSATLKDGEQSSTYELGKDHKTDKSA
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • SpyoM01000153 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 140 QVPTG (shown in italics in SEQ ID NO: 246, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyoM01000153 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyoM01000153. The pilin motif sequence is underlined in SEQ ID NO: 246, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 57. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyoM01000153 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 246
    MKKNKLLLATAILATALGMASMSQNIKAETAGVIDGSTLVVKKTFPSYTD
    DNVLMPK ADYSFKVEADDNAKGKTKDGLDIKPGVIDGLENTKTIRYSNSD
    KITAKEKSVNFEFANVKFPGVGVYRYTVAEVNGNKAGITYDSQQWTVDVY
    VVNKEGGGEEVKYIVSTEVGQSEKKPVLFKNSFDTTSLKIEKQVTGNTGE
    HQRLFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKLTEEDVTKDGYKTSATLKDGEQSSTYELGKDHKTDKSA
    DEIVVTNKRDTQVPTGVVGTLAPEAVLSIVAIGGVIYITKRKKA
  • An E box containing a conserved glutamic residue has been identified in SpyoM01000153. The E-box motif is underlined in SEQ ID NO: 246, below. The conserved glutamic acid (E), at amino acid residue 265, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of SpyoM01000153. Preferred fragments of SpyoM01000153 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 246
    MKKNKLLLATAILATALGMASMSQNIKAETAGVIDGSTLVVKKTFPSYTD
    DNVLMPKADYSFKVEADDNAKGKTKDGLDIKPGVIDGLENTKTIRYSNSD
    KITAKEKSVNFEFANVKFPGVGVYRYTVAEVNGNKAGITYDSQQWTVDVY
    VVNKEGGGFEVKYIVSTEVGQSEKKPVLFKNSFDTTSLKIEKQVTGNTGE
    HQRLFSFTLLLTPNECFEKGQVVNILQGGETKKVVIGEEYSFTLKDKESV
    TLSQLPVGIEYKLTEEDVTKDGYKTSATLKDGEQSSTYELGKDHKTDKSA
    DEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • SpyoM01000152 is a SrtC2 type sortase. An example of SpyoM01000152 is shown in SEQ ID NO: 247
    SEQ ID NO: 247
    MMMTIVQVINKAIDTLILIFCLVVLFLAGFGLWDSYHLYQQADASNFKKF
    KTAQQQPKFEDLLALNEDVIGWLNIPGTHIDYPLVQGKTNLEYINKAVDG
    SVAMSGSLFLDTRNHNDFTDDYSLIYGHHMAGNAMFGEIPKFLKKNFFNK
    HNKAIIETKERKKLTVTIFACLKTDAFDQLVFNPNAITNQDQQRQLVDYI
    SKRSKQFKPVKLKHHTKFVAPSTCENFSTDNRVTVVGTIQE
  • SpyoM01000151 is referred to as a hypothetical protein. An example of SpyoM01000151 is shown in SEQ ID NO: 248.
    SEQ ID NO: 248
    MLFSVVMMLTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPD
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • SpyoM01000151 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 185 LPLAG (shown in italics in SEQ ID NO: 248, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyoM01000151 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in SpyoM01000151. The pilin motif sequence is underlined in SEQ ID NO: 248, below. Conserved lysine (K) residues are also marked in bold, at amino acid residue 138. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyoM01000151 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 248
    MLFSVVMMLTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPK RLVKPIPPRQPD
    IPKTPLPLAGEVKSLLGILSTVLLGLLVLLYVKKLKSRL
  • Two E boxes containing conserved glutamic residues have been identified in SpyoM01000151. The E-box motifs are underlined in SEQ ID NO: 248, below. The conserved glutamic acid (E) residues, at amino acid residues 58 and 128, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyoM01000151. Preferred fragments of SpyoM01000151 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 248
    MLFSVVMMLTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPD
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSRL
  • SpyoM01000150 is referred to as a putative MsmRL. An example of SpyoM01000150 is set forth in SEQ ID NO: 249.
    SEQ ID NO: 249
    MVIFDLKHVQTLHSLSQLPISVMSQDKALIQVYGNDDYLLCYYQFLKHLA
    IPQAAQDVIFYEGLFEESFMIFPLCHYIIAIGPFYPYSLNKDYQEQLANN
    FLKHSSHRSKEELLSYMALVPHFPINNVRNLLIAIDAFFDTQFETTCQQT
    THQLLQHSKQMTADPDIIHRLKHISKASSQLPPVLEHLNHIMDLVKLGNP
    QLLKQEINRIPLSSITSSSISALRAEKNLTVIYLTRLLEFSFVENTDVAK
    HYSLVKYYMALNEEASDLLKVLRIRCAAIIHFSESLTNKSISDKRQMYNS
    VLHYVDSHLYSKLKVSDIAKRLYVSESHLRSVFKKYSNVSLQHYILSTKI
    KEAQLLLKRGIPVGEVAKSLYFYDTTHFHKIFKKYTGISSKDYLAKYRDN
    I
  • SpyoM01000149 is a F2 like fibronectin-binding protein. An example of SpyoM01000149 is set forth in SEQ ID NO: 250.
    SEQ ID NO: 250
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGYFE
    IKKVDQNNKPLSGATFSLTPKDGKGKPVQTFTSSEEGIIDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSKYGEQEKTSNSADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNPKGISQDIPKIIYDSENSPLAIGKYDAKTHQLTYTE
    TNYIAGLDKVQLSAELSLFLENKEVLENTNISDFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNIPY
    AVLNLWGFAKRTAQGENDNSSVSSAQLTGYDIYEVPHNYRLPTSYGVDIS
    RLNLRKDLEAKLPQGSTQGANKRLRIDFGENLQGKAFVVKVTGKADQSGK
    ELIVQSHLSSPNNWGSYKTLRPNSHVSETNEIALSPSKGSGSGTSEETKP
    AITVANLKRVAQLRFKKVSTDNVPLPEAAFELRSSNGNSQKLEASSNTQG
    EIHPKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEQMVKWEKP
    HSFVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNGQ
    KMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVKVPDGYKVSYL
    GNDIFNTRETEFVFEQNNENLEFGNAEIKGQSGSKIIDEDTLTSFKGKKI
    WKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYNG
    TGNDIIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPNLEIEETLPL
    ESGASGGTTTVEDSRSVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRDI
    DGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDGY
    EIATAITFTVNEQGQVTVNGKATKGDAHIVMVDAYKPTKGSGQVIDIEEK
    LPDEQGHSGSTTEIEDSKPSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTE
    IEDSKSSDVIIGGQGQVVETTEDTQTGMHGDSGCKTEVEDTKLVQFFHFD
    NKEPESNSEIPKKDKPKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVIS
    LKSKKRLLSC
  • SpyoM01000149 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 180 LPATG (shown in italics in SEQ ID NO: 250, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant SpyoM01000149 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, discussed above, containing conserved lysine (K) residues have also been identified in SpyoM01000149. The pilin motif sequences are underlined in SEQ ID NO: 250, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 157 and 163, and 216 and 224. The pilin sequences, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of SpyoM01000149 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 250
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGYFE
    IKKVDQNNKPLSGATFSLTPKDGKGKPVQTFTSSEEGIIDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSK YGEQEKTSNSADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNP KGISQDIPK IIYDSENSPLAIGKYDAKTHQLTYTE
    TNYIAGLDKVQLSAELSLFLENKEVLENTNISDFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNIPY
    AVLNLWGFAKRTAQGENDNSSVSSAQLTGYDIYEVPHNYRLPTSYGVDIS
    RLNLRKDLEAKLPQGSTQGANKRLRIDFGENLQGKAFVVKVTGKADQSGK
    ELIVQSHLSSPNNWGSYKTLRPNSHVSETNEIALSPSKGSGSGTSEETKP
    AITVANLKRVAQLRFKKVSTDNVPLPEAAFELRSSNGNSQKLEASSNTQG
    EIHPKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEQMVKWEKP
    HSFVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNGQ
    KMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVKVPDGYKVSYL
    GNDIFNTRETEFVFEQNNENLEFGNAEIKGQSGSKIIDEDTLTSFKGKKI
    WKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYNG
    TGNDIIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPNLEIEETLPL
    ESGASGGTTTVEDSRSVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRDI
    DGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDGY
    EIATAITFTVNEQGQVTVNGKATKGDAHIVMVDAYKPTKGSGQVIDIEEK
    LPDEQGHSGSTTEIEDSKPSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTE
    IEDSKSSDVIIGGQGQVVETTEDTQTGMHGDSGCKTEVEDTKLVQFFHFD
    NKEPESNSEIPKKDKPKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVIS
    LKSKKRLLSC
  • Two E boxes containing conserved glutamic residues have been identified in SpyoM01000149. The E-box motifs are underlined in SEQ ID NO: 250, below. The conserved glutamic acid (E) residues, at amino acid residues 329 and 668, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of SpyoM01000149. Preferred fragments of SpyoM01000149 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 250
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGYFE
    IKKVDQNNKPLSGATFSLTPKDGKGKPVQTFTSSEEGIIDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSKYGEQEKTSNSADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNPKGISQDIPKIIYDSENSPLAIGKYDAKTHQLTYTE
    TNYIAGLDKVQLSAELSLFLENKEVLENTNISDFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNIPY
    AVLNLWGFAKRTAQGENDNSSVSSAQLTGYDIYEVPHNYRLPTSYGVDIS
    RLNLRKDLEAKLPQGSTQGANKRLRIDFGENLQGKAFVVKVTGKADQSGK
    ELIVQSHLSSPNNWGSYKTLRPNSHVSETNEIALSPSKGSGSGTSEETKP
    AITVANLKRVAQLRFKKVSTDNVPLPEAAFELRSSNGNSQKLEASSNTQG
    EIHPKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEQMVKWEKP
    HSFVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNGQ
    KMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVKVPDGYKVSYL
    GNDIFNTRETEFVFEQNNENLEFGNAEIKGQSGSKIIDEDTLTSFKGKKI
    WKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYNG
    TGNDIIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPNLEIEETLPL
    ESGASGGTTTVEDSRSVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRDI
    DGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDGY
    EIATAITFTVNEQGQVTVNGKATKGDAHIVMVDAYKPTKGSGQVIDIEEK
    LPDEQGHSGSTTEIEDSKPSDVIIGGQGEVVDTTEDTQSGMTGHSGSTTE
    IEDSKSSDVIIGGQGQVVETTEDTQTGMHGDSGCKTEVEDTKLVQFFHFD
    NKEPESNSEIPKKDKPKSNTSLPATGEKQHNKFFWMVTSCSLISSVFVIS
    LKSKKRLLSC
  • As discussed above, applicants have also determined the nucleotide and encoded amino acid sequence of fimbrial structural subunits in several other GAS AI-3 strains of bacteria. Examples of sequences of these fimbrial structural subunits are set forth below.
  • M3 strain isolate ISS 3040 is a GAS AI-3 strain of bacteria. ISS3040_fimbrial is thought to be a fimbrial structural subunit of M3 strain isolate ISS 3040. An example of a nucleotide sequence encoding the ISS3040_fimbrial protein (SEQ ID NO: 263) and an ISS3040_fimbrial protein amino acid sequence (SEQ ID NO: 264) are set forth below.
    SEQ ID NO: 263
    gagacggcaggagtgtccgaaaatgcaaaattaatagtaaaaaagacatt
    tgactcttatacagacaatgaagttttaatgccaaaagctgattatactt
    ttaaagtagaggcagatagtacagctagtggcaaaacgaaagacggttta
    gagattaagccaggtattgttaatggtttaacagaacagattaccagcta
    tactaatactgataaaccagatagtaaagttaaaagtacagagtttgatt
    tttcaaaagtagtattccctggtattggtgtttaccgctatactgtttca
    gaaaaacaaggtgatgttgaaggaattacctacgatactaagaagtggac
    agtagatgtttatgttggaaacaaagaaggtggtggttttgaacctaagt
    ttattgtatctaaggaacaaggaacagacgtcaaaaaaccagttaatttt
    aacaactcgtttgcaactacttcgttaaaagttaagaagaatgtatcggg
    gaatactggagaattgcaaaaagaatttgactttacattgacgcttaatg
    aaagcacgaattttaaaaaagatcaaattgtttctttacaaaaaggaaac
    gagaaatttgaagttaagattggtactccctacaagtttaaactcaaaaa
    tggggaatctattcaactagacaagttaccagttggtattacttataaag
    tcaatgaaatggaagctaataaagatgggtataaaacaacagcatccttg
    aaagagggagatggtcaatctaaaatgtatcaattggatatggaacaaaa
    aacagacgaatctgctgacgaaatcgttgtcacaaataagcgtgacactc
    aagttccaactggtgttgtaggcacccttgctccatttgcagttcttagc
    SEQ ID NO: 264
    ETAGVSENAKLIVKKTFDSYTDNEVLMPKADYTFKVEADSTASGKTKDGL
    EIKPGIVNGLTEQIISYTNTDKPDSKVKSTEFDFSKVVFPGIGVYRTVSE
    KQGDVEGITYDTKKWTVDVYVGNKEGGGFEPKFIVSKEQGTDVKKPVNFN
    NSFATTSLKVKKNVSGNTGELQKEFDFTLTLNESTNFKKDQIVSLQKGNE
    KFEVKIGTPYKFKLKNGESIQLDKLPVGITYKVNEMEANKDGYKTTASLK
    EGDGQSKMYQLDMEQKTDESADEIVVTNKRDTQVPTGVVGTLAPFAVLS
  • M44 strain isolate ISS 3776 is a GAS AI-3 strain of bacteria. ISS3776_fimbrial is thought to be a fimbrial structural subunit of M44 isolate ISS 3776. An example of a nucleotide sequence encoding the ISS3776_fimbrial protein (SEQ ID NO: 253) and an ISS3776_fimbrial protein amino acid sequence (SEQ ID NO: 254) are set forth below.
    SEQ ID NO: 253
    ttggagagagaaaaaatgaaaaaaaacaaattattacttgctactgcaat
    cttagcaactgctttaggaacagcttctttaaatcaaaacgtaaaagctg
    agacggcaggggttgtaacaggaaaatcactacaagttacaaagacaatg
    acttatgatgatgaagaggtgttaatgcccgaaaccgcctttacttttac
    tatagagcctgatatgactgcaagtggaaaagaaggcagcctagatatta
    aaaatggaattgtagaaggcttagacaaacaagtaacagtaaaatataag
    aatacagataaaacatctcaaaaaactaaaatagcacaatttgatttttc
    taaggttaaatttccagctataggtgtttaccgctatatggtttcagaga
    aaaacgataaaaaagacggaattacgtacgatgataaaaagtggactgta
    gatgtttatgttgggaataaggccaataacgaagaaggtttcgaagttct
    atatattgtatcaaaagaaggtacttctagtactaaaaaaccaattgaat
    ttacaaactctattaaaactacttccttaaaaattgaaaaacaaataact
    ggcaatgcaggagatcgtaaaaaatcattcaacttcacattaacattaca
    accaagtgaatattataaaactggatcagttgtgaaaatcgaacaggatg
    gaagtaaaaaagatgtgacgataggaacgccttacaaatttactttggga
    cacggtaagagtgtcatgttatcgaaattaccaattggtatcaattacta
    tcttagtgaagacgaagcgaataaagacggctacactacaacggcaacat
    taaaagaacaaggcaaagaaaagagttccgatttcactttgagtactcaa
    aaccagaaaacagacgaatctgctgacgaaatcgttgtcacaaataagcg
    tgacactcaagttccaactggtgttgtagggacccttgctccatttgcag
    ttcttagcattgtggctattggtggagttatctatattacaaaacgtaaa
    aaagcttaa
    SEQ ID NO: 254
    MEREKMKKNKLLLATAILATALGTASLNQNVKAETAGVVTGKSLQVTKTM
    TYDDEEVLMPETAFTFTIEPDMTASGKEGSLDIKNGIVEGLDKQVTVKYK
    NTDKTSQKTKIAQFDFSKVKFPAIGVYRYMVSEKNDKKDGTTYDDKKWTV
    DVYVGNKANNEEGFEVLYIVSKEGTSSTKKPIEFTNSIKTTSLKIEKQIT
    GNAGDRKKSFNFTLTLQPSEYYKTGSVVKIEQDGSKKDVTIGTPYKFTLG
    HGKSVMLSKLPIGINYYLSEDEANKDGYTTTATLKEQGKEKSSDFTLSTQ
    NQKTDESADEIVVTNKRDTQVPTGVVGTLAPFAVLSIVAIGGVIYTTKRK
    KA
  • M77 strain isolate ISS4959 is a GAS AI-3 strain of bacteria. ISS4959_fimbrial is thought to be a fimbrial structural subunit of M77 strain ISS 4959. An example of a nucleotide sequence encoding the ISS4959_fimbrial protein (SEQ ID NO: 271) and an ISS4959_fimbrial protein amino acid sequence (SEQ ID NO: 272) are set forth below.
    SEQ ID NO: 271
    gtaacagtaaaatataagaatacagataaaacatctcaaaaaactaaaat
    agcacaatttgatttttctaaggttaaatttccagctataggtgtttacc
    gctatatggtttcagagaaaaacgataaaaaagacggaattacgtacgat
    gataaaaagtggacngtagatgtttatgttgggaataaggccaataacga
    agaaggtttcgaagttctatatattgtatcaaaagaaggtacttctagtn
    ctaaaaaaccaattgaatttacaaactctattaaaactacttccttaaaa
    attgaaaaacaaataactggcaatgcaggagatcgtaaaaaatcattcaa
    cttcacattnacattacanccaagtgaatattataaaactggatcagttg
    tgaaaatcgaacaggatggaagtaaaaaagatgtgacgataggaacgcct
    tacaaatttactttgggacacggtaagagtgtcatgttatcgaaattncc
    aattggtatcaattactatcttagtgaagacgaagcgaataaagacggnt
    acactacancggcaacattaaaagaacaaggcaaagaaaagagttccgat
    ttcactttgagtactcaaaaccagaaaacagacgaatctgctg
    SEQ ID NO: 272
    VTVKYKNTDKTSQKTKIAQFDFSKVKFPAIGVYRYMVSEKNDKKDGITYD
    DKKWTVDVYVGNKANNEEGREVLYIVSKEGTSSXKKPIEFTNSIKTTSLK
    IEKQITGNAGDRKKSFNFTXTLXPSEYYKTGSVVKIEQDGSKKDVTIGTP
    YKFTLGHGKSVNLSKXPIGINYYLSEDEANKDGYTTXATLKEQGKEKSSD
    FTLSTQNQKTDESA
  • Examples of GAS AI-4 sequences from M12 strain isolate A735 are set forth below.
  • 19224133 is thought to be a RofA regulatory protein. An example of a nucleotide sequence encoding the RofA regulatory protein (SEQ ID NO: 104) and a RofA regulatory protein amino acid sequence (SEQ ID NO: 105) are set forth below.
    SEQ ID NO: 104
    ATGACCATCCAAAAAAGGATGATATCTTGCCAATTTACACATCCTTCTAA
    AGAAACTTATCTTTACCAACTCTATGCATCATCTAATGTCTTACAATTAC
    TAGCGTTTTTAATAAAAAATGGTTCCCACTCTCGTCCCCTTACGGATTTT
    GCAAGAAGTCATTTTTTATCAAACTCCTCAGCTTATCGGATGCGCGAAGC
    ATTGATTCCTTTATTAAGAAACTTTGAATTAAAACTCTCTAAGAACAAGA
    TTGTCGGTGAGGAATATCGTATCCGTTACCTCATCGCTCTGCTATATAGT
    AAGTTTGGCATTAAAGTTTATGACTTGACGCAGCAAGACAAAAACATTAT
    TCATAGCTTTTTATCCCATAGTTCCACCCACCTTAAAACTTCTCCTTGGT
    TATCGGAATCGTTTTCTTTCTATGACATTTTATTAGCTTTATCGTGGAAG
    CGGCATCAATTTTCGGTAACTATTCCCCAAACCAGAATTTTTCAACAATT
    AAAAAAACTTTTTGTCTACGATTCTTTGAAAAAAAGTAGCCGTGATATTA
    TCGAAACTTACTGCCAACTAAACTTTTCAGCAGGAGATTTGGACTACCTC
    TATTTAATTTATATCACCGCTAATAATTCTTTTGCGAGCTTACAATGGAC
    ACCTGAGCATATCAGACAATGTTGTCAACTTTTTGAAGAAAATGATACTT
    TTCGCCTGCTTTTAAATCCTATCATCACTCTTTTACCTAACCTAAAAGAG
    CAAAAGGCTAGTTTAGTAAAAGCTCTTATGTTTTTTTCAAAATCATTCTT
    GTTTAATCTGCAACATTTTATTCCTACAGATTCTTTCCCAAGGTATTTCT
    CGGATAAAAGCATTGATTTTCATTCCTATTATCTATTGCAAGATAATGTT
    TATCAAATTCCTGATTTAAAGCCAGATTTGGTCATCACTCACAGTCAACT
    GATTCCTTTTGTTCACCATGAACTTACAAAAGGAATTGCTGTTGCTGAAA
    TATCTTTTGATGAATCGATTCTGTCTATCCAAGAATTGATGTATCAAGTT
    AAAGAGGAAAAATTCCAAGCTGATTTAACCAAACAATTAACATAA
    SEQ ID NO: 105
    MTIQKRMISCQFTHPSKETYLYQLYASSNVLQLLAFLIKNGSHSRPLTDF
    ARSHFLSNSSAYRMREALIPLLRNFELKLSKNKIVGEEYRIRYLIALLYS
    KFGIKVYDLTQQDKNIIHSFLSHSSTHLKTSPWLSESFSFYDILLALSWK
    RHQFSVTIPQTRIFQQLKKLFVYDSLKKSSRDIIETYCQLNFSAGDLDYL
    YLIYITANNSFASLQWTPEHIRQCCQLFEENDTFRLLLNPIITLLPNLKE
    QKASLVKALMFFSKSFLFNLQHFIPETNLFVSPYYKGNQKLYTSLKLIVE
    EWMAKLPGKRYLNHKHFHLFCHYVEQILRNIQPPLVVVFVASNFINAHLL
    TDSFPRYFSDKSIDFHSYYLLQDNVYQIPDLKPDLVITHSQLIPFVHHEL
    TKGTAVAEISFDESTLSIQELMYQVKEEKFQADLTKQLT
  • 19224134 is thought to be a protein F fibronectin binding protein. An example of a nucleotide sequence encoding the protein F fibronectin binding protein (SEQ ID NO: 106) and a protein F fibronectin binding protein amino acid sequence (SEQ ID NO: 107) are set forth below.
    SEQ ID NO: 106
    ATGGTAAGCTCATATATGTTTGCGAGAGGAGAGAAAATGAATAACAAAAT
    GTTTTTGAACAAAGAAGCCGGTTTTTTGGTACACACAAAAAGAAAAAGGC
    GATTTGCTGTCACTTTAGTGGGAGTCTTTTTTCTGCTTTTGGCATGTGCG
    GGTGCTATCGGTTTTGGTCAAGTAGCCTATGCTGCGGATGAGAAGACTGT
    GCCGAATTTTAAAAGCCCAGATCCAGATTATCCCTGGTATGGTTATGATT
    CGTATAGAGGAATATTTGCAAGATATCACAATTTAAAAGTAAATCTAAAA
    GGAAGTAAGGAGTATCAAGCGTATTGTTTTAACCTAACAAAATACTTTCC
    TCGCCCCACTTATAGTACTACAAATAATTTTTACAAGAAAATTGATGGGA
    GTGGATCAGCGTTCAAATCTTATGCAGCGAATCCTAGGGTTTTAGATGAG
    AATTTAGATAAATTAGAAAAAAATATACTGAATGTAATTTATAATGGATA
    TAAAAGTAATGCAAATGGTTTTATGAATGGTATAGAAGATCTTAATGCTA
    TACTAGTAACTCAAAACGCTATTTGGTACTATTCAGATAGTGCTCCATTA
    AATGATGTTAATAAAATGTGGGAAAGAGAGGTTCGGAATGGGGAGATTAG
    TGAGTCACAAGTTACTTTAATGCGTGAGGCATTGAAAAAACTAATTGATC
    CCAATTTAGAAGCTACTGCAGCTAATAAAATCCCATCAGGATATCGTTTA
    AATATCTTTAAGTCTGAAAATGAAGATTACCAAAATCTTTTAAGTGCTGA
    ATATGTACCTGATGATCCCCCTAAACCTGGTGATACGTCAGAACATAATC
    CTAAAACTCCCGAGTTGGATGGCACTCCAATTCCCGAGGACCCAAAACGT
    CCAGATGAGAGTTCAGAACCTGCGCTTCCCCCATTAATGCCAGAGCTAGA
    TGGTGAAGAAGTCCCAGAAGTTCCAAGCGAGAGCTTAGAACCTGCGCTTC
    CCCCATTGATGCCAGAGCTAGATGGTGAAGAAGTCCCAGAAGTTCCAAGC
    GAGAGCTTAGAACCTGCGCTTCCCCCATTGATGCCAGAGCTAGATGGTGA
    AGAAGTCCCAGAAGTTCCAAGCGAGAGCTTAGAACCTGCGCTTCCCCCAT
    TAATGCCAGAGCTAGATGGTGAAGAAGTCCCAGAAGTTCCAAGCGAGAGC
    TTAGAACCTGCGCTTCCCCCATTGATGCCAGAGTTAGATGGTGAAGAAGT
    CCCTGAAAAACCTAGTGTTGACTTACCTATTGAAGTTCCTCGTTATGAGT
    TTAACAATAAAGACCAGTCACCTCTAGCGGGTGAGTCTGGTGAGACGGAG
    TATATTACCGAAGTCTATGGAAATCAACAGAACCCTGTTGATATTGATAA
    AAAACTTCCGAATGAAACAGGTTTTTCAGGAAATATGGTTGAGACAGAAG
    ATACGAAAGAGCCAGAAGTGTTGATGGGAGGTCAAAGTGAGTCTGTTGAA
    TTTACTAAAGACACTCAAACAGGCATGAGTGGTCAAACAACTCCTCAGGT
    TGAGACAGAAGATACGAAAGAGCCAGAAGTGTTGATGGGAGGTCAAAGTG
    AGTCTGTTGAATTTACTAAAGACACTCAAACAGGCATGAGTGGTCAAACA
    ACTCCTCAGGTTGAGACAGAAGATACGAAAGAGCCAGGAGTGTTGATGGG
    AGGCCAAAGTGAGTCTGTTGAATTTACTAAAGACACTCAAACAGGCATGA
    GTGGTCAAACAACTCCTCAGGTTGAGACAGAAGACACGAAAGAGCCAGGA
    AATCGGGAAAAGCCTACAAAAAATATAACACCTATCCTTCCTGCAACAGG
    AGATATTGAGAATGTTTTGGCCTTTCTTGGAATCCTTATTTTGTCAGTAC
    TTTCTATTTTTAGCCTTTTAAAAACAAACAAAACAATAAAGTCTGA
    SEQ ID NO: 107
    MVSSYMFARGEKMNNKMFLNKEAGFLVHTKRKRRFAVTLVGVFFLLLACA
    GAIGFGQVAYAADEKTVPNFKSPDPDYPWYGYDSYRGIFARYHNLKVNLK
    GSKEYQAYCFNLTKYFPRPTYSTTNNFYKKIDGSGSAFKSYAANPRVLDE
    NLDKLEKNILNVIYNGYKSNANGFMNGIEDLNAILVTQNAIWYYSDSAPL
    NDVNKMWEREVRNGEISESQVTLMREALKKLTDPNLEATAANKIPSGYRL
    NIFKSENEDYQNLLSAEYVPDDPPKPGDTSEHNPKTPELDGTPIPEDPKR
    PDESSEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPS
    ESLEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPSES
    LEPALPPLMPELDGEEVPEKPSVDLPIEVPRYEFNNKDQSPLAGESGETE
    YITEVYGNQQNPVDIDKKLPNETGFSGNMVETEDTKEPEVLMGGQSESVE
    FTKDTQTGMSGQTTPQVETEDTKEPEVLMGGQSESVEETKDTQTGMSGQT
    TPQVETEDTKEPGVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDTKEPG
    VLMGGQSESVEFTKDTQTGMSFFSETVTIVEDTRPKLVFHFDNNEPKVEE
    NREKPTKNITPILPATGDIENVLAFLGILILSVLSIFSLLKNKQNNKV
  • 19224134 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 181 LPATG (shown in italics in SEQ ID NO: 107, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 19224134 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in 19224134. The pilin motif sequence is underlined in SEQ ID NO: 107, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 275, 285, and 299. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of 19224134 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 107
    MVSSYMFARGEKMNNKMFLNKEAGFLVHTKRKRRFAVTLVGVFFLLLACA
    GAIGFGQVAYAADEKTVPNFKSPDPDYPWYGYDSYRGIFARYHNLKVNLK
    GSKEYQAYCFNLTKYFPRPTYSTTNNFYKKIDGSGSAFKSYAANPRVLDE
    NLDKLEKNTLNVIYNGYKSNANGFMNGIEDLNAILVTQNAIWYYSDSAPL
    NDVNKMWEREVRNGEISESQVTLMREALKKLIDPNLEATAANKIPSGYRL
    NIFKSENEDYQNLLSAEYVPDDPPKPGDTSEHNPKTPELDGTPIPEDPK R
    PDESSEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPS
    ESLEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPSES
    LEPALPPLMPELDGEEVPEKPSVDLPIEVPRYEFNNKDQSPLAGESGETE
    YITEVYGNQQNPVDIDKKLPNETGFSGNMVETEDTKEPEVLMGGQSESVE
    FTKDTQTGMSGQTTPQVETEDTKEPEVLMGGQSESVEFTKDTQTGMSGQT
    TPQVETEDTKEPGVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDTKEPG
    VLMGGQSESVEFTKDTQTGMSGFSETVTIVEDTRPKLVEHFDNNEPKVEE
    NREKPTKNITPILPATGDIENVLAFLGILILSVLSIPSLLKNKQNNKV
  • Two E boxes containing conserved glutamic residues have been identified in 19224134. The E-box motifs are underlined in SEQ ID NO: 107, below. The conserved glutamic acid (E) residues, at amino acid residues 487 and 524, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of 19224134. Preferred fragments of 19224134 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 107
    MVSSYMFARGEKMNNKMFLNKEAGFLVHTKRKRRFAVTLVGVFFLLLACA
    GAIGFGQVAYAADEKTVPNFKSPDPDYPWYGYDSYRGIFARYHNLKVNLK
    GSKEYQAYCFNLTKYFPRPTYSTTNNFYKKIDGSGSAFKSYAANPRVLDE
    NLDKLEKNTLNVIYNGYKSNANGFMNGIEDLNAILVTQNAIWYYSDSAPL
    NDVNKMWEREVRNGEISESQVTLMREALKKLIDPNLEATAANKIPSGYRL
    NIFKSENEDYQNLLSAEYVPDDPPKPGDTSEHNPKTPELDGTPIPEDPKR
    PDESSEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPS
    ESLEPALPPLMPELDGEEVPEVPSESLEPALPPLMPELDGEEVPEVPSES
    LEPALPPLMPELDGEEVPEKPSVDLPIEVPRYEFNNKDQSPLAGESGETE
    YITEVYGNQQNPVDIDKKLPNETGFSGNMVETEDTKEPEVLMGGQSESVE
    FTKDTQTGMSGQTTPQVETEDTKEPEVLMGGQSESVEFTKDTQTGMSGQT
    TPQVETEDTKEPGVLMGGQSESVEFTKDTQTGMSGQTTPQVETEDTKEPG
    VLMGGQSESVEFTKDTQTGMSGFSETVTIVEDTRPKLVEHFDNNEPKVEE
    NREKPTKNITPILPATGDIENVLAFLGILILSVLSIPSLLKNKQNNKV
  • 19224135 is thought to be a capsular polysaccharide adhesin (Cpa) protein. An example of a nucleotide sequence encoding the Cpa protein (SEQ ID NO: 108) and a Cpa protein amino acid sequence (SEQ ID NO: 109) are set forth below.
    SEQ ID NO: 108
    ATGAATAACAAAAAATTGCAAAAGAAGCAAGATGCTCCTCGGGTATCAAA
    CAGAAAGCCAAAACAATTAACTGTCACTTTAGTGGGAGTATTTTTAATGT
    TTTTGACCTTGGTAAGTTCCATGAGAGGTGCTCAAAGCATATTTGGAGAG
    GAAAAGAGAATTGAAGAAGTCAGTGTTCCTAAAATAAAAAGTCCAGATGA
    TGCCTACCCTTGGTATGGCTATGATTCATATGACTCTAGTCATCCTTACT
    ATGAACGTTTTAAAGTAGCACATGATTTAAGGGTTAATTTAAATGGAAGT
    AAGAGCTACCAAGTATATTGCTTTAATATCAATTCTCATTATCCGAATAG
    AAAAAATGCTTTTTCTAAACAATGGTTTAAGAGAGTTGATGGGACAGGTG
    ATGTGTTCACAAATTATGCTCAGACACCTAAGATTCGTGGAGAATCATTG
    AATAATAAACTTTTAAGTATTATGTACAACGCTTATCCTAAAAATGCTAA
    TGGCTATATGGATAAGATAGAACCATTAAATGCTATTTTAGTAACTCAAC
    AAGCTGTTTGGTACTATTCTGACAGTTCTTATGGTAATATAAAAACGTTA
    TGGGCATCTGAGCTTAAAGACGGAAAAATAGATTTTGAACAAGTAAAATT
    AATGCGTGAAGCTTACTCAAAACTAATTAGTGATGATTTAGAAGAAACAT
    CTAAAAATAAGCTACCTCAAGGATCTAAACTGAATATTTTTGTTCCGCAA
    GATAAATCTGTTCAAAATTTATTAAGTGCAGAGTACGTGCCTGAATCCCC
    TCCGGCACCAGGTCAGTCTCCAGAACCGCCAGTGCAAACAAAAAAAACAT
    CAGTCATTATCAGAAAATATGCGGAAGGTGACTACTCTAAACTTCTAGAG
    GGAGCAACTTTGCGTTTAACAGGGGAAGATATCCTAGATTTTCAAGAAAA
    AGTCTTCCAAAGTAATGGAACAGGAGAAAAGATTGAATTATCAAATGGGA
    CTTATACCTTAACAGAAACATCATCTCCAGATGGATATAAAATTGCGGAG
    CCGATTAAGTTTAGAGTAGTGAATAAAAAAGTATTTATCGTCCAAAAAGA
    TGGTTCTCAAGTGGAAAATCCAAACAAAGAAGTAGCAGAGCCATACTCAG
    TGGAAGCGTACAGCGATATGCAAGATAGTAACTATATTAATCCAGAAACG
    TTCACTCCTTATGGGAAATTTTATTACGCTAAAAATAAGGATAAAAGTTC
    ACAAGTTGTCTACTGTTTTAATGCTGATTTACACTCTCCACCTGAATCAG
    AGGATGGGGGAGGAACTATAGATCCTGATATTAGTACGATGAAAGAAGTC
    AAGTACACACATACGGCAGGTAGTGATTTGTTTAAATACGCGCTAAGACC
    GAGAGATACAAATCCAGAAGACTTCTTAAAGCACATTAAAAAAGTAATTG
    AAAAAGGCTACAATAAAAAAGGTGATAGCTATAATGGATTAACAGAAACA
    CAGTTTCGCGCGGCTACTCAGCTTGCTATCTATTACTTTACAGACAGCAC
    TGACTTAAAAACCTTAAAAACTTATAACAATGGGAAAGGTTACCATGGAT
    TTGAATCTATGGATGAAAAAACCCTAGCTGTAACAAAAGAATTAATTAAT
    TACGCTCAAGATAATAGTGCCCCTCAACTAACAAATCTTGATTTCTTCGT
    ACCTAATAATAGCAAATACCAATCTCTTATTGGGACAGAATACCATCCAG
    ATGATTTGGTTGACGTGATTCGTATGGAAGATAAAAAGCAAGAAGTTATT
    CCAGTAACTCACAGTTTGACAGTGAAAAAAACAGTAGTCGGTGAGTTGGG
    AGATAAAACTAAAGGCTTCCAATTTGAACTTGAGTTGAAAGATAAAACTG
    GACAGCCTATTGTTAACACTCTAAAAACTAATAATCAAGATTTAGTAGCT
    AAAGATGGGAAATATTCATTTAATCTAAAGCATGGTGACACCATAAGAAT
    AGAAGGATTACCGACGGGATATTCTTATACTCTGAAAGAGACTGAAGCTA
    AGGATTATATAGTAACCGTTGATAACAAAGTTAGTCAAGAAGCTCAATCA
    GCAAGTGAGAATGTCACAGCAGACAAAGAAGTCACTTTTGAAAACCGTAA
    AGATCTTGTCCCACCAACTGGTTTTATTACTGATGGTGGAACCTATGTGT
    GGTTATTATTGCTTGTCGCATTTGGTTTGTTAGTGTGGTTCTTTGGTGGT
    AAAGGACTAAAAAATGACTAA
    SEQ ID NO: 109
    MNNKKLQKKQDAPRVSNRKPKQLTVTLVGVFLMELTLVSSMRGAQSIFGE
    EKRIEEVSVPKIKSPDDAYPWYGYDSYDSSHPYYERFKVAHDLRVNLNGS
    KSYQVYCFNINSHYPNRKNAFSKQWFKRVDGTGDVFTNYAQTPKIRGESL
    NNKLLSIMYNAYPKNANGYMDKIEPLNAILVTQQAVWYYSDSSYGNIKTL
    WASELKDGKIDFEQVKLMREAYSKLISDDLEETSKNKLPQGSKLNIFVPQ
    DKSVQNLLSAEYVPESPPAPGQSPEPPVQTKKTSVIIRKYAEGDYSKLLE
    GATLRLTGEDILDFQEKVFQSNGTGEKIELSNGTYTLTETSSPDGYKIAE
    PIKFRVVNKKVFIVQKDGSQVENPNKEVAEPYSVEAYSDMQDSNYINPET
    FTPYGKFYYAKNKDKSSQVVYCFNADLHSPPESEDGGGTIDPDISTMKEV
    KYTHTAGSDLFKYALRPRDTNPEDFLKHIKKVTEKGYNKKGDSYNGLTET
    QFRAATQLAIYYFTDSTDLKTLKTYNNGKGYHGFESMDEKTLAVTKELIN
    YAQDNSAPQLTNLDFFVPNNSKYQSLIGTEYHPDDLVDVIRMEDKKQEVI
    PVTHSLTVKKTVVGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVA
    KDGKYSFNLKHGDTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQS
    ASENVTADKEVTFENRKDLVPPTGFITDGGTYLWLLLLVPFGLLVWFFGR
    KGLKND
  • 19224135 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 184 VPPTG (shown in italics in SEQ ID NO: 109, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 19224135 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in 19224135. The pilin motif sequence is underlined in SEQ ID NO: 109, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 164 and 172. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of 19224135 include at least one conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 109
    MNNKKLQKKQDAPRVSNRKPKQLTVTLVGVFLMELTLVSSMRGAQSIFGE
    EKRIEEVSVPKIKSPDDAYPWYGYDSYDSSHPYYERFKVAHDLRVNLNGS
    KSYQVYCFNINSHYPNRKNAFSKQWFKRVDGTGDVFTNYAQTPKIRGESL
    NNKLLSIMYNAYPKNANGYMDK IEPLNAILVTQQAVWYYSDSSYGNIKTL
    WASELKDGKIDFEQVKLMREAYSKLISDDLEETSKNKLPQGSKLNIFVPQ
    DKSVQNLLSAEYVPESPPAPGQSPEPPVQTKKTSVIIRKYAEGDYSKLLE
    GATLRLTGEDILDFQEKVFQSNGTGEKIELSNGTYTLTETSSPDGYKIAE
    PIKFRVVNKKVFIVQKDGSQVENPNKEVAEPYSVEAYSDMQDSNYINPET
    FTPYGKFYYAKNKDKSSQVVYCFNADLHSPPESEDGGGTIDPDISTMKEV
    KYTHTAGSDLFKYALRPRDTNPEDFLKHIKKVTEKGYNKKGDSYNGLTET
    QFRAATQLAIYYFTDSTDLKTLKTYNNGKGYHGFESMDEKTLAVTKELIN
    YAQDNSAPQLTNLDFFVPNNSKYQSLIGTEYHPDDLVDVIRMEDKKQEVI
    PVTHSLTVKKTVVGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVA
    KDGKYSFNLKHGDTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQS
    ASENVTADKEVTFENRKDLVPPTGFITDGGTYLWLLLLVPFGLLVWFFGR
    KGLKND
  • An E box containing a conserved glutamic residue has been identified in 19224135. The E-box motif is underlined in SEQ ID NO: 109, below. The conserved glutamic acid (E), at amino acid residue 339, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of 19224135. Preferred fragments of 19224135 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 109
    MNNKKLQKKQDAPRVSNRKPKQLTVTLVGVFLMELTLVSSMRGAQSIFGE
    EKRIEEVSVPKIKSPDDAYPWYGYDSYDSSHPYYERFKVAHDLRVNLNGS
    KSYQVYCFNINSHYPNRKNAFSKQWFKRVDGTGDVFTNYAQTPKIRGESL
    NNKLLSIMYNAYPKNANGYMDKIEPLNAILVTQQAVWYYSDSSYGNIKTL
    WASELKDGKIDFEQVKLMREAYSKLISDDLEETSKNKLPQGSKLNIFVPQ
    DKSVQNLLSAEYVPESPPAPGQSPEPPVQTKKTSVIIRKYAEGDYSKLLE
    GATLRLTGEDILDFQEKVFQSNGTGEKIELSNGTYTLTETSSPDGYKIAE
    PIKFRVVNKKVFIVQKDGSQVENPNKEVAEPYSVEAYSDMQDSNYINPET
    FTPYGKFYYAKNKDKSSQVVYCFNADLHSPPESEDGGGTIDPDISTMKEV
    KYTHTAGSDLFKYALRPRDTNPEDFLKHIKKVTEKGYNKKGDSYNGLTET
    QFRAATQLAIYYFTDSTDLKTLKTYNNGKGYHGFESMDEKTLAVTKELIN
    YAQDNSAPQLTNLDFFVPNNSKYQSLIGTEYHPDDLVDVIRMEDKKQEVI
    PVTHSLTVKKTVVGELGDKTKGFQFELELKDKTGQPIVNTLKTNNQDLVA
    KDGKYSFNLKHGDTIRIEGLPTGYSYTLKETEAKDYIVTVDNKVSQEAQS
    ASENVTADKEVTFENRKDLVPPTGFITDGGTYLWLLLLVPFGLLVWFFGR
    KGLKND
  • 19224136 is thought to be a LepA protein. An example of a nucleotide sequence encoding the LepA protein (SEQ ID NO: 110) and a LepA protein amino acid sequence (SEQ ID NO: 111) are set forth below.
    SEQ ID NO: 110
    ATGACTAATTACCTAAATCGCTTAAATGAGAATCCACTATTTAAAGCTTT
    CATACGGTTAGTACTTAAGATTTCTATTATTGGATTTCTAGGTTACATTC
    TATTTCAGTATGTTTTTGGCGTCATGATTGTTAACACAAATCAGATGAGT
    CCTGCTGTAAGTGCTGGTGATGGAGTCTTATATTATCGTTTGACTGATCG
    CTATCATATTAATGATGTGGTGGTCTATGAGGTTGATAACACTTTGAAAG
    TTGGTCGAATTGCCGCTCAAGCTGGCGATGAGGTTAGTTTTACGCAAGAA
    GGAGGACTGTTGATTAATGGGCATCCACCAGAAAAAGAGGTCCCTTACCT
    GACGTATCCTCACTCAAGTGGTCCAAACTTTCCCTATAAAGTTCCTACGG
    GTACGTATTTCATATTGAATGATTATCGTGAAGAACGTTTGGACAGTCGT
    TATTATGGGGCGTTACCCATCAATCAAATCAAAGGGAAAATCTCAACTCT
    ATTAAGAGTGAGAGGAATTTAA
    SEQ ID NO: 111
    MTNYLNRLNENPLFKAFIRLVLKISIIGFLGYILFQYVFGVMIVNTNQMS
    PAVSAGDGVLYYRLTDRYHINDVVVYEVDNTLKVGRIAAQAGDEVSFTQE
    GGLLINGHPPEKEVPYLTYPHSSGPNFPYKVPTGTYFILNDYREERLDSR
    YYGALPINQIKGKISTLLRVRGI
  • 19224137 is thought to be a fimbrial protein. An example of a nucleotide sequence encoding the fimbrial protein (SEQ ID NO: 112) and a fimbrial protein amino acid sequence (SEQ ID NO: 113) are set forth below.
    SEQ ID NO: 112
    ATGAAAAAAAATAAATTATTACTTGCTACTGCAATCTTAGCAACTGCTTT
    AGGAACAGCTTCTTTAAATCAAAACGTAAAAGCTGAGACGGCAGGGGTTG
    TTAGCAGTGGTCAATTAACAATAAAAAAATCAATTACAAATTTTAATGAT
    GATACACTTTTGATGCCTAAGACAGACTATACTTTTAGCGTTAATCCGGA
    TAGTGCGGCTACAGGTACTGAAAGTAATTTACCAATTAAACCAGGTATTG
    CTGTTAACAATCAAGATATTAAGGTTTCTTATTCTAATACTGATAAGACA
    TCAGGTAAAGAAAAACAAGTTGTTGTTGACTTTATGAAAGTTACTTTTCC
    TAGCGTTGGTATTTACCGTTATGTTGTTACCGAGAATAAAGGGACAGCAG
    AAGGAGTTACATATGATGATACAAAATGGTTAGTTGACGTCTATGTTGGT
    AATAATGAAAAGGGAGGTCTTGAACCAAAGTATATTGTATCTAAAAAAGG
    AGATTCTGCTACTAAAGAACCAATCCAGTTTAATAATTCATTCGAAACAA
    CGTCATTAAAAATTGAAAAGGAAGTTACTGGTAATACAGGAGATCATAAA
    AAAGCATTTACCTTTACATTAACATTGCAACCAAATGAATACTATGAGGC
    AAGTTCGGTTGTGAAAATTGAAGAGAACGGACAAACGAAAGATGTGAAAA
    TTGGGGAGGCATATAAGTTTACTTGAACGAATAGTCAGAGTGTGATATTG
    TCTAAATTACCAGTTGGTATTAATTATAAAGTTGAAGAAGCAGAAGCTAA
    TCAAGGTGGATATACTACAACAGCAACTTTAAAAGATGGAGAAAAGTTAT
    CTACTTATAACTTAGGTCAGGAACATAAAACAGACAAGACTGCTGATGAA
    ATCGTTGTCACAAATAACCGTGACACTCAAGTTCCAACTGGTGTTGTAGG
    CACCCTTGCTCCATTTGCAGTTCTTAGCATTGTGGCTATTGGTGGAGTTA
    TCTATATTACAAAACGTAAAAAAGCTTAA
    SEQ ID NO: 113
    MKKNKLLLATAILATALGTASLNQNVKAETAGVVSSGQLTIKKSITNFND
    DTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIAVNNQDIKVSYSNTDKT
    SGKEKQVVVDFMKVTFPSVGIYRYVVTENKGTAEGVTYDDTKWLVDVYVG
    NNEKGGLEPKYIVSKKGDSATKEPIQFNNSFETTSLKIEKEVTGNTGDHK
    KAFTFTLTLQPNEYYEASSVVKIEENGQTKDVKIGEAYKFTLNDSQSVIL
    SKLPVGINYKVEEAEANQGGYTTTATLKDGEKLSTYNLGQEHKTDKTADE
    IVVTNNRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • 19224137 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 140 QVPTG (shown in italics in SEQ ID NO: 113, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 19224137 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in 19224137. The pilin motif sequence is underlined in SEQ ID NO: 113, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 160. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of 19224137 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 113
    MKKNKLLLATAILATALGTASLNQNVKAETAGVVSSGQLTIKKSITNFND
    DTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIAVNNQDIKVSYSNTDKT
    SGKEKQVVVDFMKVTFPSVGIYRYVVTENKGTAEGVTYDDTKWLVDVYVG
    NNEKGGLEPKYIVSKKGDSATKEPIQFNNSFETTSLKIEKEVTGNTGDHK
    KAFTFTLTLQPNEYYEASSVVKIEENGQTKDVKIGEAYKFTLNDSQSVIL
    SKLPVGINYKVEEAEANQGGYTTTATLKDGEKLSTYNLGQEHKTDKTADE
    IVVTNNRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • An E box containing a conserved glutamic residue has been identified in 19224137. The E-box motif is underlined in SEQ ID NO: 113, below. The conserved glutamic acid (E), at amino acid residue 263, is marked in bold. The E box motif, in particular the conserved glutamic acid residue, is thought to be important for the formation of oligomeric pilus-like structures of 19224137. Preferred fragments of 19224137 include the conserved glutamic acid residue. Preferably, fragments include the E box motif.
    SEQ ID NO: 113
    MKKNKLLLATAILATALGTASLNQNVKAETAGVVSSGQLTIKKSITNFND
    DTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIAVNNQDIKVSYSNTDKT
    SGKEKQVVVDFMKVTFPSVGIYRYVVTENKGTAEGVTYDDTKWLVDVYVG
    NNEKGGLEPKYIVSKKGDSATKEPIQFNNSFETTSLKIEKEVTGNTGDHK
    KAFTFTLTLQPNEYYEASSVVKIEENGQTKDVKIGEAYKFTLNDSQSVIL
    SKLPVGINYKVEEAEANQGGYTTTATLKDGEKLSTYNLGQEHKTDKTADE
    IVVTNNRDTQVPTGVVGTLAPFAVLSIVAIGGVIYITKRKKA
  • 19224138 is thought to be a SrtC2-type sortase. An example of a nucleotide sequence encoding the SrtC2 sortase (SEQ ID NO: 114) and a SrtC2 sortase amino acid sequence (SEQ ID NO: 115) are set forth below.
    SEQ ID NO: 114
    ATGATGATGACAATTGTACAGGTTATCAATAAAGCCATTGATACTCTCAT
    TCTTATCTTTTGTTTAGTCGTACTATTTTTAGCTGGTTTTGGTTTGTGGG
    ATTCTTATCATCTCTATCAACAAGCAGACGCTTCTAATTTCAAAAAATTT
    AAAACAGCTCAACAACAGCCTAAATTTGAAGACTTGTTAGCTTTGAATGA
    GGATGTCATTGGTTGGTTAAATATCCCGGGGACTCATATTGATTATCCTC
    TAGTTCAGGGAAAAACGAATTTAGAGTATATTAATAAAGCAGTTGATGGC
    AGTGTTGCCATGTCTGGTAGTTTATTTTTAGATACACGGAATCATAATGA
    TTTTACGGACGATTACTCTCTGATTTATGGCCATCATATGGCAGGTAATG
    CCATGTTTGGCGAAATTCCAAAATTTTTAAAAAAGGATTTTTTCAACAAA
    CATAATAAAGCTATCATTGAAACAAAAGAGAGAAAAAAACTAACCGTCAC
    TATTTTTGCTTGTCTCAAGACAGATGCCTTTGACCAGTTAGTTTTTAATC
    CTAATGCTATTACCAATCAAGACCAACAAAGGCAGCTCGTTGATTATATC
    AGTAAAAGATCAAAACAATTTAAACCTGTTAAATTGAAGCATCATACAAA
    GTTCGTTGCTTTTTCAACGTGTGAAAATTTTTCTACTGACAATCGTGTTA
    TCGTTGTCGGTACTATTCAAGAATAA
    SEQ ID NO: 115
    MMMTIVQVINKAIDTLILIFCLVVLFLAGFGLWDSYHLYQQADASNFKKF
    KTAQQQPKFEDLLALNEDVIGWLNIPGTHIDYPLVQGKTNLEYINKAVDG
    SVAMSGSLFLDTRNHNDFTDDYSLIYGHHMAGNAMFGEIPKFLKKDFFNK
    HNKAIIETKERKKLTVTIFACLKTDAFDQLVFNPNAITNQDQQRQLVDYI
    SKRSKQFKPVKLKHHTKFVAFSTCENFSTDNRVIVVGTIQE
  • 19224139 is an open reading frame that encodes a sortase substrate motif LPXAG shown in italics in SEQ ID NO: 117. An example of a nucleotide sequence of the open reading frame (SEQ ID NO: 116) and the amino acid sequence encoded by the open reading frame (SEQ ID NO: 117) are set forth below.
    SEQ ID NO: 116
    ATGTTATTTTCTGTCGTAATGATATTAACCATGCTGGCCTTTAATCAGAC
    TGTTTTAGCAAAAGACAGCACTGTTCAAACTAGCATTAGTGTCGAAAATG
    TCTTAGAGAGAGCAGGCGATAGTACCCCATTTTCGATTGCATTAGAATCA
    ATTGATGCGATGAAAACAATAGAAGAAATAACAATTGCTGGTTCTGGAAA
    AGCAAGCTTTTCCCCTCTGACCTTCACAACAGTTGGGCAATATACTTATC
    GTGTTTATCAGAAGCCTTCACAAAATAAAGATTATCAAGCAGATACTACT
    GTATTTGACGTTCTTGTCTATGTGACCTATGATGAAGATGGGACTCTAGT
    CGCAAAAGTTATTTCTCGAAGGGCTGGAGACGAAGAAAAATCAGCGATTA
    CTTTTAAGCCCAAACGGTTAGTAAAACCAATACCGCCTAGACAACCTAAC
    ATCCCTAAAACCCCATTACCATTAGCTGGTGAAGTAAAAAGTTTATTGGG
    TATCTTAAGTATCGTATTACTGGGGTTACTAGTTCTTCTTTATGTTAAAA
    AACTGAAGAG
    SEQ ID NO: 117
    MLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPN
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSKL
  • 19224139 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 185 LPLAG (shown in italics in SEQ ID NO: 117, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 19224139 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • A pilin motif, discussed above, containing a conserved lysine (K) residue has also been identified in 19224139. The pilin motif sequence is underlined in SEQ ID NO: 117, below. A conserved lysine (K) residue is also marked in bold, at amino acid residue 138. The pilin sequence, in particular the conserved lysine residue, is thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of 19224139 include the conserved lysine residue. Preferably, fragments include the pilin sequence.
    SEQ ID NO: 117
    MLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPN
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSKL
  • Two E boxes containing conserved glutamic residues have been identified in 19224139. The E-box motifs are underlined in SEQ ID NO: 117, below. The conserved glutamic acid (E) residues, at amino acid residues 58 and 128, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of 19224139. Preferred fragments of 19224139 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 117
    MLFSVVMILTMLAFNQTVLAKDSTVQTSISVENVLERAGDSTPFSIALES
    IDAMKTIEEITIAGSGKASFSPLTFTTVGQYTYRVYQKPSQNKDYQADTT
    VFDVLVYVTYDEDGTLVAKVISRRAGDEEKSAITFKPKRLVKPIPPRQPN
    IPKTPLPLAGEVKSLLGILSIVLLGLLVLLYVKKLKSKL
  • 19224140 is thought to be a MsmRL protein. An example of a nucleotide sequence encoding the MsmRL protein (SEQ ID NO: 118) and a MsmRL protein amino acid sequence (SEQ ID NO: 119) are set forth below.
    SEQ ID NO: 118
    ATGGTTATATTCGATTTAAAACATGTGCAAACATTACACAGCTTGTCTCA
    ATTACCTATTTCAGTGATGTCACAAGATAAGGCACTTATTCAAGTATATG
    GTAATGACGACTATTTATTATGTTACTATCAATTTTTAAAGCATCTAGCT
    ATTCCTCAAGCTGCACAAGATGTTATTTTTTATGAGGGTTTATTTGAAGA
    GTCCTTTATGATTTTTCCTCTTTGTCACTACATTATTGCCATTGGACCTT
    TCTACCCTTATTCACTTAATAAAGACTATCAGGAACAATTAGCTAATAAT
    TTTTTAAAACATTCTTCTCATCGTAGCAAAGAAGAGCTCTTATCCTATAT
    GGCACTTGTCCCACATTTTCCAATTAATAATGTGCGGAACCTTTTGATAG
    CTATTGACGCTTTTTTTGACACACAATTTGAGACGACTTGCCAACAAACA
    ATTCATCAATTGTTGCAGCATTCAAAACAGATGACTGCTGATCCTGATAT
    CATTCATCGCCTTAAGCATATTAGCAAAGCATCTAGCCAACTACCGCCTG
    TTTTAGAGCACCTAAATCATATTATGGATCTGGTAAAGCTAGGCAATCCA
    CAATTGCTCAAGCAAGAAATCAATCGCATCCCCTTATCAAGTATCACCTC
    ATCTTCTATTTCTGCTCTAAGGGCGGAAAAGAACCTCACTGTTATCTATT
    TAACTAGGTTACTGGAATTCAGTTTTGTAGAAAATACTGACGTAGCAAAG
    CATTATAGCCTTGTCAAATACTACATGGCCTTAAATGAAGAAGCGAGTGA
    CTTGCTCAAAGTTTTGAGAATTCGCTGTGCAGCCATCATCCATTTTTCCG
    AATCATTAACCAATAAAAGTATTTCTGATAAACGTCAAATGTACAATAGT
    GTGCTTCATTATGTCGATAGTCACCTGTATTCCAAATTAAAGGTATCTGA
    TATCGCTAAGCGCCTATATGTTTCCGAATCTCACTTACGTTCAGTCTTTA
    AAAAATACTCAAATGTTTCCTTACAACATTATATTCTAAGTACAAAAATC
    AAAGAAGCTCAACTACTCTTAAAACGAGGAATTCCTGTTGGAGAAGTGGC
    TAAAAGCTTATATTTTTATGACACTACCCATTTTCATAAAATCTTTAAAA
    AATACACGGGTATTTCTTCAAAAGACTATCTTGCTAAATACCGAGATAAT
    ATTTAA
    SEQ ID NO: 119
    MVIFDLKHVQTLHSLSQLPISVMSQDKALIQVYGNDDYLLCYYQFLKHLA
    TPQAAQDVIFYEGLFEESFMIFPLCHYIIAIGPFYPYSLNKDYQEQLANN
    FLKHSSHRSKEELLSYMALVPHFPINNVRNLLIAIDAFFDTQFETTCQQT
    IHQLLQHSKQMTADPDIIHRLKHISKASSQLPPVLEHLNHIMDLVKLGNP
    QLLKQEINRIPLSSITSSSISALRAEKNLTVIYLTRLLEFSFVENTDVAK
    HYSLVKYYMALNEEASDLLKVLRIRCAAIIHFSESLTNKSISDKRQMYNS
    VLHYVDSHLYSKLKVSDIAKRLYVSESHLRSVFKKYSNVSLQHYILSTKI
    KEAQLLLKRGIPVGEVAKSLYFYDTTHFHKIFKKYTGISSKDYLAKYRDN
    I
  • 19224141 is thought to be a protein F2 fibronectin binding protein. An example of a nucleotide sequence encoding the protein F2 fibronectin binding protein (SEQ ID NO: 120) and a protein F2 fibronectin binding protein amino acid sequence (SEQ ID NO: 121) are set forth below.
    SEQ ID NO: 120
    ATGACACAAAAAAATAGCTATAAGTTAAGCTTCCTGTTATCCCTAACAGG
    ATTTATTTTAGGTTTATTATTGGTTTTTATAGGATTGTCCGGAGTATCAG
    TAGGACATGCGGAAACAAGAAATGGAGCAAACAAACAAGGATCTTTTGAA
    ATCAAGAAAGTCGAGCAAAAGAATAAGGCTTTACCGGGAGCAAGTTTTTC
    AGTGACATCAAAGGATGGCAAGGGAACATGTGTTCAAAGGTTGACTTGAA
    ATGATAAAGGTATTGTAGATGGTCAAAATCTCGAACCAGGGACTTATAGC
    TTAAAAGAAGAAACAGCACCAGATGGTTATGATAAAACCAGCCGGAGTTG
    GACAGTGACTGTTTATGAGAACGGCTATAGCAAGTTGGTTGAAAATCCCT
    ATAATGGGGAAATCATCAGTAAAGCAGGGTCAAAAGATGTTAGTAGTTCT
    TTACAGTTGGAAAATCCGAAAATGTCAGTTGTTTCTAAATATGGGAAAAC
    AGAGGTTAGTAGTGGCGCAGCGGATTTCTAGCGGAAGGATGCCGCCTATT
    TTAAAATGTGTTTTGAGTTGAAACAAAAGGATAAATCTGAAACAATCAAC
    CCAGGTGATACCTTTGTGTTACAGCTGGATAGACGTCTGAATCCTAAAGG
    TATCAGTCAAGATATCCCTAAAATCATTTACGACAGTGGAAATAGTCGGG
    TTGCGATTGGAAAATACCATGGTGAGAACCATCAACTTATCTATACTTTC
    ACAGATTATATTGCGGGTTTAGATAAAGTCCAGTTGTCTGCAGAATTGAG
    CTTATTCCTAGAGAATAAGGAAGTGTTGGAAAATACTAGTATGTCAAATT
    TTAAGAGTAGCATAGGTGGGCAGGAGATCAGCTATAAAGGAACGGTTAAT
    GTTCTTTATGGAAATGAGAGCACTAAAGAAAGCAATTATATTAGTAATGG
    ATTGAGGAATGTGGGTGGGAGTATTGAAAGCTACAACACCGAAACGGGAG
    AATTTGTCTGGTATGTTTATGTCAATCCAAACCGTACCAATATTCCTTAT
    GCGACGATGAATTTATGGGGATTTGGAAGGGCTCGTTCAAATACAAGCGA
    CTTAGAAAACGACGCTAATACAAGTAGTGCTGAGCTTGGAGAGATTCAGG
    TCTATGAAGTACCTGAAGGAGAAAAATTACCATCAAGTTATGGGGTTGAT
    GTTAGAAAAGTTACTTTAAGAACGGATATCACAGCAGGCCTAGGAAATGG
    TTTTCAAATGAGCAAACGTCAGCGAATTGACTTTGGAAATAATATCCAAA
    ATAAAGCATTTATCATCAAAGTAACAGGGAAAACAGACCAATCTGGTAAG
    CCATTGGTTGTTCAATCCAATTTGGCAAGTTTTCGTGGTGCTTCTGAATA
    TGCTGCTTTTACTCCAGTTGGAGGAAATGTCTACTTCCAAAACGAAATTG
    CCTTGTCTCCTTCTAAGGGTAGTGGTTCTGGGAAAAGTGAATTTACTAAG
    CCCTCTATTACAGTAGCAAATCTAAAACGAGTGGCTCAGCTTCGCTTTAA
    GAAAATGTCAACTGAGAATGTGCCATTGCCAGAAGCGGCTTTTGAGCTGC
    GTTCATCAAATGGTAATAGTCAGAAATTAGAAGCCAGTTCAAACACACAA
    GGAGAGGTTCACTTTAAGGACCTGACCTCGGGCACATATGACCTGTATGA
    AACAAAAGCGCCAAAAGGTTATCAGCAGGTGACAGAGAAATTGGCGACCG
    TTACTGTTGATACTACCAAACCTGCTGAGGAAATGGTCACTTGGGGAAGC
    CCACATTCGTCTGTAAAAGTAGAAGCTAACAAAGAAGTCACGATTGTCAA
    CCATAAAGAAACCCTTACGTTTTCAGGGAAGAAAATTTGGGAGAATGACA
    GACCAGATCAACGCCCAGCAAAGATTCAAGTGCAACTGTTGCAAAATGGT
    CAAAAGATGCCTAACCAGATTCAAGAAGTAACGAAGGATAACGATTGGTC
    TTATCACTTCAAAGACTTGCCTAAGTACGATGCCAAGAATCAGGAGTATA
    AGTACTCAGTTGAAGAAGTAAATGTTCCAGACGGCTACAAGGTGTCGTAT
    TTAGGAAATGATATATTTAACACCAGAGAAACAGAATTTGTGTTTGAACA
    GAATAACTTTAACCTTGAATTTGGAAATGCTGAAATAAAAGGTCAATCTG
    GGTCAAAAATCATTGATGAAGACACGCTAACGTCTTTCAAAGGTAAGAAA
    ATTTGGAAAAATGATACGGCAGAAAATCGTCCCCAAGCCATTCAAGTGCA
    GCTTTATGCTGATGGAGTGGCTGTGGAAGGTCAAACCAAATTTATTTCTG
    GCTCAGGTAATGAGTGGTCATTTGAGTTTAAAAACTTGAAGAAGTATAAT
    GGAACAGGTAATGACATCATTTACTCAGTTAAAGAAGTAACTGTTCCAAC
    AGGTTATGATGTGACTTACTCAGCTAATGATATTATTAATACCAAACGTG
    AGGTTATTACACAACAAGGACCGAAACTAGAGATTGAAGAAACGCTTCCG
    CTAGAATCAGGTGCTTCAGGCGGTACCACTACTGTCGAAGACTCACGCCC
    AGTTGATACCTTATCAGGTTTATCAAGTGAGCAAGGTCAGTCCGGTGATA
    TGACAATTGAAGAAGATAGTGCTACCCATATTAAATTCTCAAAACGTGAT
    ATTGACGGCAAAGAGTTAGCTGGTGCAACTATGGAGTTGCGTGATTCATC
    TGGTAAAACTATTAGTACATGGATTTCAGATGGACAAGTGAAAGATTTCT
    ACCTGATGCCAGGAAAATATACATTTGTCGAAACCGCAGCACCAGACGGT
    TATGAGATAGCAACTGCTATTACCTTTACAGTTAATGAGCAAGGTCAGGT
    TACTGTAAATGGCAAAGCAACTAAAGGTGACACTCATATTGTCATGGTTG
    ATGCTTACAAGCCAACTAAGGGTTCAGGTCAGGTTATTGATATTGAAGAA
    AAGCTTCCAGACGAGCAAGGTCATTCTGGTTCAACTACTGAAATAGAAGA
    CAGTAAATCTTCAGACCTTATCATTGGCGGTCAAGGTGAAGTTGTTGACA
    CAACAGAAGACACACAAAGTGGTATGACGGGCCATTCTGGCTCAACTACT
    GAAATAGAAGATAGCAAGTCTTCAGACCTTATCATTGGTGGTCAGGGGCA
    GGTTGTCGAGACAACAGAGGATACCCAAACTGGCATGTACGGGGATTCTG
    GTTGTAAAACGGAAGTCGAAGATACTAAACTAGTACAATCCTTCCACTTT
    GATAACAAGGAACCAGAAAGTAACTCTGAGATTCCTAAAAAAGATAAGCC
    AAAGAGTAATAGTAGTTTACCAGCAACTGGTGAGAAGCAACATAATATGT
    TCTTTTGGATGGTTACTTCTTGCTCACTTATTAGTAGTGTTTTTGTAATA
    TCACTAAAATCCAAAAAACGCCTATCATCATGTTAA
    SEQ ID NO: 121
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGSFE
    IKKVDQNNKPLPGATFSLTSKDGKGTSVQTFTSNDKGIVDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSKYGKTEVSSGAADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNPKGISQDIPKIIYDSANSPLAIGKYHAENHQLIYTF
    TDYIAGLDKVQLSAELSLFLENKEVLENTSISNFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNTPY
    ATMNLWGFGRARSNTSDLENDANTSSAELGEIQVYEVPEGEKLPSSYGVD
    VTKLTLRTDITAGLGNGFQMTKRQRIDFGNNIQNKAFIIKVTGKTDQSGK
    PLVVQSNLASERGASEYAAFTPVGGNVYFQNEIALSPSKGSGSGKSEFTK
    PSITVANLKRVAQLRFKKMSTDNVPLPEAAFELRSSNGNSQKLEASSNTQ
    GEVHFKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEEMVTWGS
    PHSSVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNG
    QKMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVNVPDGYKVSY
    LGNDIFNTRETEFVFEQNNFNLEFGNAETKGQSGSKIIDEDTLTSFKGKK
    IWKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYN
    GTGNDTIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPKLEIEETLP
    LESGASGGTTTVEDSRPVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRD
    IDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDG
    YEIATAITFTVNEQGQVTVNGKATKGDTHTVMVDAYKPTKGSGQVIDIEE
    KLPDEQGHSGSTTEIEDSKSSDLIIGGQGEVVDTTEDTQSGMTGHSGSTT
    EIEDSKSSDVIIGGQGQVVETTEDTQTGMYGDSGCKTEVEDTKLVQSFHF
    DNKEPESNSEIPKKDKPKSNTSLPATGEKQHNMFFWMVTSCSLISSVFVI
    SLKSKKRLSSC
  • 19224141 contains an amino acid motif indicative of a cell wall anchor: SEQ ID NO: 181 LPATG (shown in italics in SEQ ID NO: 121, above). In some recombinant host cell systems, it may be preferable to remove this motif to facilitate secretion of a recombinant 19224141 protein from the host cell. Alternatively, in other recombinant host cell systems, it may be preferable to use the cell wall anchor motif to anchor the recombinantly expressed protein to the cell wall. The extracellular domain of the expressed protein may be cleaved during purification or the recombinant protein may be left attached to either inactivated host cells or cell membranes in the final composition.
  • Two pilin motifs, discussed above, containing conserved lysine (K) residues have also been identified in 19224141. The pilin motif sequences are underlined in SEQ ID NO: 121, below. Conserved lysine (K) residues are also marked in bold, at amino acid residues 157 and 163 and at amino acid residues 216, 224, and 238. The pilin sequence, in particular the conserved lysine residues, are thought to be important for the formation of oligomeric, pilus-like structures. Preferred fragments of 19224141 include at least one conserved lysine residue. Preferably, fragments include at least one pilin sequence.
    SEQ ID NO: 121
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGSFE
    IKKVDQNNKPLPGATFSLTSKDGKGTSVQTFTSNDKGIVDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSK YGKTEVSSGAADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNPKGISQDIPKIIYDSANSPLAIGK YHAENHQLIYTF
    TDYIAGLDKVQLSAELSLFLENKEVLENTSISNFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNTPY
    ATMNLWGFGRARSNTSDLENDANTSSAELGEIQVYEVPEGEKLPSSYGVD
    VTKLTLRTDITAGLGNGFQMTKRQRIDFGNNIQNKAFIIKVTGKTDQSGK
    PLVVQSNLASERGASEYAAFTPVGGNVYFQNEIALSPSKGSGSGKSEFTK
    PSITVANLKRVAQLRFKKMSTDNVPLPEAAFELRSSNGNSQKLEASSNTQ
    GEVHFKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEEMVTWGS
    PHSSVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNG
    QKMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVNVPDGYKVSY
    LGNDIFNTRETEFVFEQNNFNLEFGNAETKGQSGSKIIDEDTLTSFKGKK
    IWKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYN
    GTGNDTIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPKLEIEETLP
    LESGASGGTTTVEDSRPVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRD
    IDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDG
    YEIATAITFTVNEQGQVTVNGKATKGDTHTVMVDAYKPTKGSGQVIDIEE
    KLPDEQGHSGSTTEIEDSKSSDLIIGGQGEVVDTTEDTQSGMTGHSGSTT
    EIEDSKSSDVIIGGQGQVVETTEDTQTGMYGDSGCKTEVEDTKLVQSFHF
    DNKEPESNSEIPKKDKPKSNTSLPATGEKQHNMFFWMVTSCSLISSVFVI
    SLKSKKRLSSC
  • Two E boxes containing conserved glutamic residues have been identified in 19224141. The E-box motifs are underlined in SEQ ID NO: 121, below. The conserved glutamic acid (E) residues, at amino acid residues 567 and 944, are marked in bold. The E box motifs, in particular the conserved glutamic acid residues, are thought to be important for the formation of oligomeric pilus-like structures of 19224141. Preferred fragments of 19224141 include at least one conserved glutamic acid residue. Preferably, fragments include at least one E box motif.
    SEQ ID NO: 121
    MTQKNSYKLSFLLSLTGFILGLLLVFIGLSGVSVGHAETRNGANKQGSFE
    IKKVDQNNKPLPGATFSLTSKDGKGTSVQTFTSNDKGIVDAQNLQPGTYT
    LKEETAPDGYDKTSRTWTVTVYENGYTKLVENPYNGEIISKAGSKDVSSS
    LQLENPKMSVVSKYGKTEVSSGAADFYRNHAAYFKMSFELKQKDKSETIN
    PGDTFVLQLDRRLNPKGISQDIPKIIYDSANSPLAIGKYHAENHQLIYTF
    TDYIAGLDKVQLSAELSLFLENKEVLENTSISNFKSTIGGQEITYKGTVN
    VLYGNESTKESNYITNGLSNVGGSIESYNTETGEFVWYVYVNPNRTNTPY
    ATMNLWGFGRARSNTSDLENDANTSSAELGEIQVYEVPEGEKLPSSYGVD
    VTKLTLRTDITAGLGNGFQMTKRQRIDFGNNIQNKAFIIKVTGKTDQSGK
    PLVVQSNLASERGASEYAAFTPVGGNVYFQNEIALSPSKGSGSGKSEFTK
    PSITVANLKRVAQLRFKKMSTDNVPLPEAAFELRSSNGNSQKLEASSNTQ
    GEVHFKDLTSGTYDLYETKAPKGYQQVTEKLATVTVDTTKPAEEMVTWGS
    PHSSVKVEANKEVTIVNHKETLTFSGKKIWENDRPDQRPAKIQVQLLQNG
    QKMPNQIQEVTKDNDWSYHFKDLPKYDAKNQEYKYSVEEVNVPDGYKVSY
    LGNDIFNTRETEFVFEQNNFNLEFGNAETKGQSGSKIIDEDTLTSFKGKK
    IWKNDTAENRPQAIQVQLYADGVAVEGQTKFISGSGNEWSFEFKNLKKYN
    GTGNDTIYSVKEVTVPTGYDVTYSANDIINTKREVITQQGPKLEIEETLP
    LESGASGGTTTVEDSRPVDTLSGLSSEQGQSGDMTIEEDSATHIKFSKRD
    IDGKELAGATMELRDSSGKTISTWISDGQVKDFYLMPGKYTFVETAAPDG
    YEIATAITFTVNEQGQVTVNGKATKGDTHTVMVDAYKPTKGSGQVIDIEE
    KLPDEQGHSGSTTEIEDSKSSDLIIGGQGEVVDTTEDTQSGMTGHSGSTT
    EIEDSKSSDVIIGGQGQVVETTEDTQTGMYGDSGCKTEVEDTKLVQSFHF
    DNKEPESNSEIPKKDKPKSNTSLPATGEKQHNMFFWMVTSCSLISSVFVI
    SLKSKKRLSSC
  • As discussed above, applicants have also determined the nucleotide and encoded amino acid sequence of fimbrial structural subunits in several other GAS AI-4 strains of bacteria. Examples of sequences of these fimbrial structural subunits are set forth below.
  • M12 strain isolate 20010296 is a GAS AI-4 strain of bacteria. 20010296_fimbrial is thought to be a fimbrial structural subunit of M12 strain isolate 20010296. An example of a nucleotide sequence encoding the 20010296_fimbrial protein (SEQ ID NO: 257) and a 20010296_fimbrial protein amino acid sequence (SEQ ID NO: 258) are set forth below.
    SEQ ID NO: 257
    agcagtggtcaattaacaataaaaaaatcaattacaaattttaatgatga
    tacacttttgatgcctaagacagactatacttttagcgttaatccggata
    gtgcggctacaggtactgaaagtaatttaccaattaaaccaggtattgct
    gttaacaatcaagatattaaggtttcttattctaatactgataagacatc
    aggtaaagaaaaacaagttgttgttgactttatgaaagttacttttccta
    gcgttggtatttaccgttatgttgttaccgagaataaagggacagcagaa
    ggagttacatatgatgatacaaaatggttagttgacgtctatgttggtaa
    taatgaaaagggaggtcttgaaccaaagtatattgtatctaaaaaaggag
    attctgctactaaagaaccaatccagtttaataattcattcgaaacaacg
    tcattaaaaattgaaaaggaagttactggtaatacaggagatcataaaaa
    agcatttaactttacattaacattgcaaccaaatgaatactatgaggcaa
    gttcggttgtgaaaattgaagagaacggacaaacgaaagatgtgaaaatt
    ggggaggcatataagtttactttgaacgatagtcagagtgtgatattgtc
    taaattaccagttggtattaattataaagttgaagaagcagaagctaatc
    aaggtggatatactacaacagcaactttaaaagatggagaaaagttatct
    acttataacttaggtcaggaacataaaacagacaagactgctgatgaaat
    cgt
    SEQ ID NO: 258
    SSGQLTIKKSITNFNDDTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIA
    VNNQDIKVSYSNTDKTSGKEKQVVVDFMKVTFPSVGIYRYVVTENKGTAE
    GVTYDDTKWLVDVYVGNNEKGGLEPKYIVSKKGDSATKEPIQFNNSFETT
    SLKIEKEVTGNTGDHKKAFNFTLTLQPNEYYEASSVVKIEENGQTKDVKI
    GEAYKFTLNDSQSVILSKLPVGINYKVEEAEANQGGYTTTATLKDGEKLS
    TYNLGQEHKTDKTADEIV
  • M12 strain isolate 20020069 is a GAS AI-4 strain of bacteria. 20020069_fimbrial is thought to be a fimbrial structural subunit of M12 strain isolate 20020069. An example of a nucleotide sequence encoding the 20020069_fimbrial protein (SEQ ID NO: 259) and a 20020069_fimbrial protein amino acid sequence (SEQ ID NO: 260) are set forth below.
    SEQ ID NO: 259
    agcagtggtcaattaacaataaaaaaatcaattacaaattttaatgatga
    tacacttttgatgcctaagacagactatacttttagcgttaatccggata
    gtgcggctacaggtactgaaagtaatttaccaattaaaccaggtattgct
    gttaacaatcaagatattaaggtttcttattctaatactgataagacatc
    aggtaaagaaaaacaagttgttgttgactttatgaaagttacttttccta
    gcgttggtatttaccgttatgttgttaccgagaataaagggacagcagaa
    ggagttacatatgatgatacaaaatggttagttgacgtctatgttggtaa
    taatgaaaagggaggtcttgaaccaaagtatattgtatctaaaaaaggag
    attctgctactaaagaaccaatccagtttaataattcattcgaaacaacg
    tcattaaaaattgaaaaggaagttactggtaatacaggagatcataaaaa
    agcatttaactttacattaacattgcaaccaaatgaatactatgaggcaa
    gttcggttgtgaaaattgaagagaacggacaaacgaaagatgtgaaaatt
    ggggaggcatataagtttactttgaacgatagtcagagtgtgatattgtc
    taaattaccagttggtattaattataaagttgaagaagcagaagctaatc
    aaggtggatatactacaacagcaactttaaaagatggagaaaagttatct
    acttataacttaggtcaggaacataaaacagacaagactgctgatgaaat
    cgt
    SEQ ID NO: 260
    SSGQLTIKKSITNFNDDTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIA
    VNNQDIKVSYSNTDKTSGKEKQVVVDEMKVTFPSVGIYRYVVTENKGTAE
    GVTYDDTKWLVDVYVGNNEKGGLEPKYIVSKKGDSATKEPIQFNNSEETT
    SLKIEKEVTGNTGDHKKAFNFTLTLQPNEYYEASSVVKIEENGQTKDVKI
    GEAYKFTLNDSQSVILSKLPVGINYKVEEAEANQGGYTTTATLKDGEKLS
    TYNLGQEHKTDKTADEIV
  • M12 strain isolate CDC SS 635 is a GAS AI-4 strain of bacteria. CDC SS 635_fimbrial is thought to be a fimbrial structural subunit of M12 strain isolate CDC SS 635. An example of a nucleotide sequence encoding the CDC SS 635_fimbrial protein (SEQ ID NO: 261) and a CDC SS 635_fimbrial protein amino acid sequence (SEQ ID NO: 262) are set forth below.
    SEQ ID NO: 261
    gagacggcaggggttgttagcagtggtcaattaacaataaaaaaatcaat
    tacaaattttaatgatgatacacttttgatgcctaagacagactatactt
    ttagcgttaatccggatagtgcggctacaggtactgaaagtaatttacca
    attaaaccaggtattgctgttaacaatcaagatattaaggtttcttattc
    taatactgataagacatcaggtaaagaaaaacaagttgttgttgacttta
    tgaaagttacttttcctagcgttggtatttaccgttatgttgttaccgag
    aataaagggacagcagaaggagttacatatgatgatacaaaatggttagt
    tgacgtctatgttggtaataatgaaaagggaggtcttgaaccaaagtata
    ttgtatctaaaaaaggagattctgctactaaagaaccaatccagtttaat
    aattcattcgaaacaacgtcattaaaaattgaaaaggaagttactggtaa
    tacaggagatcataaaaaagcatttaactttacattaacattgcaaccaa
    atgaatactatgaggcaagttcggttgtgaaaattgaagagaacggacaa
    acgaaagatgtgaaaattggggaggcatataagtttactttgaacgatag
    tcagagtgtgatattgtctaaattaccagttggtattaattataaagttg
    aagaagcagaagctaatcaaggtggatatactacaacagcaactttaaaa
    gatggagaaaagttatctacttataacttaggtcaggaacataaaacaga
    caagactgctgatgaaatcgttgtcacaaataaccgtgacact
    SEQ ID NO: 262
    ETAGVVSSGQLTTKKSITNFNDDTLLMPKTDYTFSVNPDSAATGTESNLP
    IKPGIAVNNQDIKVSYSNTDKTSGKEKQVVVDPMKVTFPSVGIYRYVVTE
    NKGTAEGVTYDDTKWLVDVYVGNNEKGGLEPKYIVSKKGDSATKEPIQFN
    NSFETTSLKIEKEVTGNTGDHKKAFNFTLTLQPNEYYEASSVVKIEENGQ
    TKDVKIGEAYKFTLNDSQSVILSKLPVGINYKVEEAEANQGGYTTTATLK
    DGEKLSTYNLGQEHKTDKTADEIVVTNNRDT
  • M5 strain isolate ISS 4883 is a GAS AI-4 strain of bacteria. ISS4883_fimbrial is thought to be a fimbrial structural subunit of M5 strain isolate ISS 4883. An example of a nucleotide sequence encoding the ISS4883_fimbrial protein (SEQ ID NO: 265) and an ISS4883_fimbrial protein amino acid sequence (SEQ ID NO: 266) are set forth below.
    SEQ ID NO: 265
    gagacggcaggggttgtaacaggaaaatcactacaagttacaaagacaat
    gacttatgatgatgaagaggtgttaatgcccgaaaccgcctttactttta
    ctatagagcctgatatgactgcaagtggaaaagaaggcgacctagatatt
    aaaaatggaattgtagaaggcttagacaaacaagtaacagtaaaatataa
    gaatacagataaaacatctcaaaaaactaaaatagcacaatttgattttt
    ctaaggttaaatttccagctataggtgtttaccgctatatggtttcagag
    aaaaacgataaaaaagacggaattaggtacgatgataaaaagtggactgt
    agatgtttatgttgggaataaggccaataacgaagaaggtttcgaagttc
    tatatattgtatcaaaagaaggtacttctagtactaaaaaaccaattgaa
    tttacaaactctattaaaactacttccttaaaaattgaaaaacaaataac
    tggcaatgcaggagatcgtaaaaaatcattcaacttcacattaacattac
    aaccaagtgaatattataaaaccggatcagttgtgaaaatcgaacaggat
    ggaagtaaaaaagatgtgacgataggaacgccttacaaatttactttggg
    acacggtaagagtgtcatgttatcgaaattaccaattggtatcaattact
    atcttagtgaagacgaagcgaataaagacggttacactacaacggcaaca
    ttaaaagaacaaggcaaagaaaagagttccgatttcactttgagtactca
    aaaccagaaaacagacgaatctgctgacgaaatcgttgtcacaaataagc
    gtgacactctcgag
    SEQ ID NO: 266
    ETAGVVTGKSLQVTKTMTYDDEEVLMPETAFTFTIEPDMTASGKEGDLDI
    KNGIVEGLDKQVTVKYKNTDKTSQKTKIAQFDFSKVKFPAIGVYRYMVSE
    KNDKKDGIRYDDKKWTVDVYVGNKANNEEGFEVLYIVSKEGTSSTKKPIE
    FTNSIKTTSLKIEKQITGNAGDRKKSFNFTLTLQPSEYYKTGSVVKIEQD
    GSKKDVTIGTPYKFTLGHGKSVMLSKLPIGINYYLSEDEANKDGYTTTAT
    LKEQGKEKSSDFTLSTQNQKTDESADEIVVTNKRDTLE
  • M50 strain isolate ISS4538 is a GAS AI-4 strain of bacteria. ISS4538_fimbrial is thought to be a fimbrial structural subunit of M50 strain ISS 4538. An example of a nucleotide sequence encoding the ISS4538_fimbrial protein (SEQ ID NO: 255) and an ISS4538_fimbrial protein amino acid sequence (SEQ ID NO: 256) are set forth below.
    SEQ ID NO: 255
    atgaaaaaaaataaattattacttgctactgcaatcttagcaactgcttt
    aggaacagcttctttaaatcaaaacgtaaaagctgagacggcaggggttg
    ttagcagtggtcaattaacaataaaaaaatcaattacaaattttaatgat
    gatacacttttgatgcctaagacagactatacttttagcgttaatccgga
    tagtgcggctacaggtactgaaagtaatttaccaattaaaccaggtattg
    ctgttaacaatcaagatattaaggtttcttattctaatactgataagaca
    tcaggtaaagaaaaacaagttgttgttgactttatgaaagttacttttcc
    tagcgttggtatttaccgttatgttgttaccgagaataaagggacagcag
    aaggagttacatatgatgatacaaaatggttagttgacgtctatgttggt
    aataatgaaaagggaggtcttgaaccaaagtatattgtatctaaaaaagg
    agattctgctactaaagaaccaatccagtttaataattcattcgaaacaa
    cgtcattaaaaattgaaaagaaagttactggtaatacaggagatcataaa
    aaagcatttaactttacattaacattgcaaccaaatgaatactatgaggc
    aagttcggttgtgaaaattgaagagaacggacaaacgaaagatgtgaaaa
    ttggggaggcatataagtttactttgaacgatagtcagagtgtgatattg
    tctaaattaccagttggtattaattataaagttgaagaagcagaagctaa
    tcaaggtggatatactacaacagcaactttaaaagatggagaaaagttat
    ctacttataacttaggtcaggaacataaaacagacaagactgctgatgaa
    atcgttgtcacaaataancgngacactcnagttccaacnggtgtngtagg
    caccccncctccattcncagttcttancattgnggctantggtggngtna
    tntatnttacaaaacgnaaaaaagnataa
    SEQ ID NO: 256
    MKKNKLLLATAILATALGTASLNQNVKAETAGVVSSGQLTIKKSITNFND
    DTLLMPKTDYTFSVNPDSAATGTESNLPIKPGIAVNNQDIKVSYSNTDKT
    SGKEKQVVVDFMKVTFPSVGIYRYVVTENKGTAEGVTYDDTKWLVDVYVG
    NNEKGGLEPKYIVSKKGDSATKEPIQFNNSFETTSLKIEKKVTGNTGDHK
    KAFNFTLTLQPNEYYEASSVVKTEENGQTKDVKIGEAYKFTLNDSQSVIL
    SKLPVGINYKVEEAEANQGGYTTTATLKDGEKLSTYNLGQEHKTDKTADE
    IVVTNXRDTXVPTGVVGTPPPFXVLXIXAXGGVXYXTKRKKX
  • There may be an upper limit to the number of GAS proteins which will be in the compositions of the invention. Preferably, the number of GAS proteins in a composition of the invention is less than 20, less than 19, less than 18, less than 17, less than 16, less than 15, less than 14, less than 13, less than 12, less than 11, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, or less than 3. Still more preferably, the number of GAS proteins in a composition of the invention is less than 6, less than 5, or less than 4. Still more preferably, the number of GAS proteins in a composition of the invention is 3.
  • The GAS proteins and polynucleotides used in the invention are preferably isolated, i.e., separate and discrete, from the whole organism with which the molecule is found in nature or, when the polynucleotide or polypeptide is not found in nature, is sufficiently free of other biological macromolecules so that the polynucleotide or polypeptide can be used for its intended purpose.
  • Examples Other Gram Positive Bacterial Adhesin Island Sequences
  • The Gram positive bacteria AI polypeptides of the invention can, of course, be prepared by various means (e.g. recombinant expression, purification from a gram positive bacteria, chemical synthesis etc.) and in various forms (e.g. native, fusions, glycosylated, non-glycosylated etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other streptococcal or host cell proteins) or substantially isolated form.
  • The Gram positive bacteria AI proteins of the invention may include polypeptide sequences having sequence identity to the identified Gram positive bacteria proteins. The degree of sequence identity may vary depending on the amino acid sequence (a) in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more). Polypeptides having sequence identity include homologs, orthologs, allelic variants and mutants of the identified Gram positive bacteria proteins. Typically, 50% identity or more between two proteins is considered to be an indication of functional equivalence. Identity between proteins is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affinity gap search with parameters gap open penalty=12 and gap extension penalty=1.
  • The Gram positive bacteria adhesin island polynucleotide sequences may include polynucleotide sequences having sequence identity to the identified Gram positive bacteria adhesin island polynucleotide sequences. The degree of sequence identity may vary depending on the polynucleotide sequence in question, but is preferably greater than 50% (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more).
  • The Gram positive bacteria adhesin island polynucleotide sequences of the invention may include polynucleotide fragments of the identified adhesin island sequences. The length of the fragment may vary depending on the polynucleotide sequence of the specific adhesin island sequence, but the fragment is preferably at least 10 consecutive polynucleotides, (e.g. at least 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more).
  • The Gram positive bacteria adhesin island amino acid sequences of the invention may include polypeptide fragments of the identified Gram positive bacteria proteins. The length of the fragment may vary depending on the amino acid sequence of the specific Gram positive bacteria antigen, but the fragment is preferably at least 7 consecutive amino acids, (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200 or more). Preferably the fragment comprises one or more epitopes from the sequence. The fragment may comprise at least one T-cell or, preferably, a B-cell epitope of the sequence. T- and B-cell epitopes can be identified empirically (e.g., using PEPSCAN [Geysen et al. (1984) PNAS USA 81:39984002; Carter (1994) Methods Mol. Biol. 36:207-223, or similar methods], or they can be predicted (e.g., using the Jameson-Wolf antigenic index [Jameson, B A et al. 1988, CABIOS 4 (1):1818-186], matrix-based approaches [Raddrizzani and Hammer (2000) Brief Bioinform. 1(2):179-189], TEPITOPE [De Lalla et al. (199) J. Immunol. 163:1725-1729], neural networks [Brusic et al. (1998) Bioinformatics 14(2):121-130], OptiMer & EpiMer [Meister et al. (1995) Vaccine 13(6):581-591; Roberts et al. (1996) AIDS Res. Hum. Retroviruses 12(7):593-610], ADEPT [Maksyutov & Zagrebelnaya (1993) Comput. Appl. Biosci. 9(3):291-297], Tsites [Feller & de la Cruz (1991) Nature 349(6311):720-721], hydrophilicity [Hopp (1993) Peptide Research 6:183-190], antigenic index [Welling et al. (1985) FEBS Lett. 188:215-218] or the methods disclosed in Davenport et al. (1995) Immunogenetics 42:392-297, etc. Other preferred fragments include (1) the N-terminal signal peptides of each identified Gram positive bacteria protein, (2) the identified Gram positive bacteria protein without their N-terminal signal peptides, (3) each identified Gram positive bacteria protein wherein up to 10 amino acid residues (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) are deleted from the N-terminus and/or the C-terminus e.g. the N-terminal amino acid residue may be deleted. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain), and (4) the polypeptides, but without their N-terminal amino acid residue.
  • As indicated in the above text, nucleic acids and polypeptides of the invention may include sequences that:
      • (a) are identical (i.e., 100% identical) to the sequences disclosed in the sequence listing;
      • (b) share sequence identity with the sequences disclosed in the sequence listing;
      • (c) have 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 single nucleotide or amino acid alterations (deletions, insertions, substitutions), which may be at separate locations or may be contiguous, as compared to the sequences of (a) or (b);
      • (d) when aligned with a particular sequence from the sequence listing using a pairwise alignment algorithm, a moving window of x monomers (amino acids or nucleotides) moving from start (N-terminus or 5′) to end (C-terminus or 3′), such that for an alignment that extends top monomers (where p>x) there are p−x+1 such windows, each window has at least x·y identical aligned monomers, where: x is slected from 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150, 200; y is selected from 0.50, 0.60, 0.70, 0.75, 0.80, 0.85, 0.90, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99; and if x·y is not an integer then it is rounded up to the nearest integer. The preferred pairwise alignment algorithm is the Needleman-Wunsch global alignment algorithm [Needlman &Wunsch (1970) J. Mol. Biol. 48, 443-453], using default parameters (e.g., with Gap opening penalty=10.0, and with Gap extension penalty=0.5, using the EBLOSUM62 scoring matrix). This algorithm is conveniently implemented in the needle tool in the EMBOSS package [Rice et al. (2000) Trends Genet. 16:276-277].
  • The nucleic acids and polypeptides of the inention may additionally have further sequences to the N-terminus/5′ and/or C-terminus/3′ of these sequences (a) to (d).
  • All of the Gram positive bacterial sequences referenced herein are publicly available through PubMed on GenBank.
  • Streptococcus pneumoniae Adhesin Island Sequences
  • As discussed above, a S. pneumoniae AI sequence is present in the TIGR4 S. pneumoniae genome. Examples of S. pneumoniae AI sequences are set forth below.
  • SrtD (Sp0468) is a sortase. An example of an amino acid sequence of SrtD is set forth in SEQ ID NO: 80.
    SEQ ID NO: 80
    MSRTKLRALLGYLLMLVACLIPIYCFGQMVLQSLGQVKGHATFVKSMTTE
    MYQEQQNHSLAYNQRLASQNRTVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLGMGLAHVDGTPLPLDGTGIRSVIAGHRAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • SrtC (Sp0467) is a sortase. An example of an amino acid sequence of SrtC is set forth in SEQ ID NO: 81.
    SEQ ID NO: 81
    MSRYYYRIESNEVIKEFDETVSQMDKAELEERWRLAQAENATLKPSEILD
    PFTEQEKKKGVSEYANMLKVHERIGYVEIPAIDQEIPMYVGTSEDILQKG
    AGLLEGASLPVGGENTHTVITAHRGLPTAELFSQLDKMKKGDIFYLHVLD
    QVLAYQVDQIVTVEPNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGK
    RIPYTAPIAERNRAVRERGQFWLWLLLGAMAVILLLLYRVYRNRRIVKGL
    EKQLEGRHVKD
  • SrtB (SP0466) is a sortase. An example of an amino acid sequence of SrtB is set forth in SEQ ID NO: 82.
    SEQ ID NO: 82
    MAVMAYPLVSRLYYRVESNQQIADFDKEKATLDEADIDERMKLAQAFNDS
    LNNVVSGDPWSEEMKKKGRAEYARMLEIHERMGHVEIPVIDVDLPVYAGT
    AEEVLQQGAGHLEGTSLPIGGNSTHAVITAHTGLPTAKMFTDLTKLKVGD
    KFYVHNIKEVMAYQVDQVKVIEPTNFDDLLIVPGHDYVTLLTCTPYMINT
    HRLLVRGHRIPYVAEVEEEFIAANKLSHLYRYLFYVAVCLIVTLLWITRR
    LRKKKKQPEKALKALKAARKEVKVEDGQQ
  • Sp0465 is a hypothetical protein. An example of an amino acid sequence of Sp0465 is set forth in SEQ ID NO: 83.
    SEQ ID NO: 83
    MFLPFLSASLYLQTHHFIAFPNRQSYLLRETRKSHFFLIHHPF
  • RrgC (SP0464) is a cell wall surface anchor family protein. RrgC contains a sortase substrate motif VPXTG (SEQ ID NO: 137), shown in italics in SEQ ID NO: 84.
    SEQ ID NO: 84
    MISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVSQLPSRDGH
    RLQVWKLDDSYSYDDRVQIVRDLHSWDENKLSSFKKTSFEMTFLENQIEV
    SHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAKKTDTMTTK
    VKLIKVDQDHNRLEGVGFKLVSVARDVSEKEVPLIGEYRYSSSGQVGRTL
    YTDKNGEIFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVDHQLVTITV
    VNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEESGHYTPVLQNGKEVVV
    TSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGKDTRKELVT
    VVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKPNN
  • RrgB (Sp0463) is a cell wall surface anchor protein. RrgB contains a sortase substrate motif IPXTG (SEQ ID NO: 133), shown in italics in SEQ ID NO: 85.
    SEQ ID NO: 85
    MKSINKFLTMLAALLLTASSLFSAATVFAAGTTTTSVTVHKLLATDGDMD
    KIANELETGNYAGNKVGVLPANAKEIAGVMFVWTNTNNEIIDENGQTLGV
    NIDPQTFKLSGAMPATAMKKLTEAEGAKFNTANLPAAKYKIYEIHSLSTY
    VGEDGATLTGSKAVPIEIELPLNDVVDAHVYPKNTEAKPKIDKDFKGKAN
    PDTPRVDKDTPVNHQVGDVVEYEIVTKIPALANYATANWSDRMTEGLAFN
    KGTVKVTVDDVALEAGDYALTEVATGFDLKLTDAGLAKVNDQNAEKTVKI
    TYSATLNDKAIVEVPESNDVTFNYGNNPDHGNTPKPNKPNENGDLTLTKT
    WVDATGAPIPAGAEATFDLVNAQTGKVVQTVTLTTDKNTVTVNGLDKNTE
    YKFVERSIKGYSADYQEITTAGEIAVKNWKDENPKPLDPTEPKVVTYGKK
    FVKVNDKDNRLAGAEFVIANADNAGQYLARKADKVSQEEKQLVVTTKDAL
    DRAVAAYNALTAQQQTQQEKEKVDKAQAAYNAAVIAANNAFEWVADKDNE
    NVVKLVSDAQGRFEITGLLAGTYYLEETKQPAGYALLTSRQKFEVTATSY
    SATGQGIEYTAGSGKDDATKVVNKKITIPQTGGIGTIIFAVAGAAIMGIA
    VYAYVKNNKDEDQLA
  • RrgA (Sp0462) is a cell wall surface anchor protein. RrgA contains a sortase substrate motif YPXTG (SEQ ID NO: 186), indicated in italics in SEQ ID NO: 86.
    SEQ ID NO: 86
    MLNRETHMKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKV
    VIKETGEGGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYT
    LTEAQPPVGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGT
    YPDVQTPYQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLD
    DNQYGIELTVSGKTVYEQKDKSVPLDVVILLDNSNSMSNIRNKNARRAER
    AGEATRSLIDKITSDSENRVALVTYASTIFDGTEFTVEKGVADKNGKRLN
    DSLFWNYDQTSFTTNTKDYSYLKLTNDKNDIVELKNKVPTEAEDHDGNRL
    MYQFGATFTQKALMKADEILTQQARQNSQKVIFHITDGVPTMSYPINFNH
    ATFAPSYQNQLNAFFSKSPNKDGILLSDFITQATSGEHTIVRGDGQSYQM
    FTDKTVYEKGAPAAFPVKPEKYSEMKAAGYAVIGDPINGGYIWLNWRESI
    LAYPFNSNTAKITNHGDPTRWYYNGNIAPDGYDVFTVGIGINGDPGTDEA
    TATSFMQSISSKPENYTNVTDTTKILEQLNRYFHTIVTEKKSIENGTITD
    PMGELIDLQLGTDGRFDPADYTLTANDGSRLENGQAVGGPQNDGGLLKNA
    KVLYDTTEKRIRVTGLYLGTDEKVTLTYNVRLNDEFVSNKFYDTNGRTTL
    HPKEVEQNTVRDFPIPKIRDVRKYPEITISKEKKLGDIEFIDVNDNDKKP
    LRGAVFSLQKQHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYR
    LFENSEPAGYKPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKH
    YITNEPIPPKREYPRTGGIGMLPFYLIGCMMMGGVLLYTRKHP
  • RlrA (Sp0461) is a transcriptional regulator. An example of an amino acid sequence for RlrA is set forth in SEQ ID NO: 87.
    SEQ ID NO: 87
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGTEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKE
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLESLYLTETIFSSLPAIPIKIIL
    NNQADVNLTKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • As discussed above, a S. pneumoniae AI sequence is present in the S. pneumoniae strain 670 genome. Examples of S. pneumoniae AI sequences are set forth below.
  • Orf1 670 is a transposase. An example of an amino acid sequence of orf1 670 is set forth in SEQ ID NO: 171.
    SEQ ID NO: 171
    MEHINHTTLLIGIKDKNITLNKAIQHDTHIEVFATLDYHPPKCKHCKGKQ
    IKYDFQKPSKIPFIEIGGFPSLIHLKKRRFQCKSCRKVTVAETTLVQKNC
    QISEMVRQKIAQLLLNREALTHIASKLAISTSTSTVYRKLKQFHFQEDYT
    TLPEILSWDEFSYQKGKLAFIAQDFNTKKIMTILDNRRQTTIRNHFFKYS
    KEARKKVKVVTVDMSGSYIPLIKKLFPNAKIVLDRFHIVQHMSRALNQTR
    INIMKQFDDKSLEYRALKYYWKFILKDSRKLSLKPFYARTFRETLTPREC
    LKKTFTLVPELKDYYDLYQLLLFHLQEKNTDQFWGLIQDTLPHLNRTFKT
    TLSTFICYKNYITNAIELPYSNAKLEATNKLIKDIKRNAFGFRNFENFKK
    RIFIALNTKKERTKFVLSRA
  • Orf2 670 is a transcriptional regulator. An example of an amino acid sequence of Orf2 670 is set forth in SEQ ID NO: 172.
    SEQ ID NO: 172
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNETDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • Orf3 670 is a cell wall surface anchor family proten. An example of an amino acid sequence of Orf3 670 is set forth in SEQ ID NO: 173.
    SEQ ID NO: 173
    MLNRETHMKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKV
    VIKETGEGGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYT
    LTEAQPPVGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGT
    YPDVQTPYQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLD
    DNQYGIELTVSGKTTVETKEASTPLDVVILLDNSNSMSNIRHNHAHRAEK
    AGEATPALVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKILN
    DSALWTEDRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKL
    MYQFGATFTQKALMTADDILTKQARPNSKKVIFHITDGVPTMSYPINFKY
    TGTTQSYRTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQM
    FTKKPVTDQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAY
    PFNSSTDWITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEATAT
    RFMQSISSSPDNYTNVADPSQILQELNRYFYTIVNEKKSIENGTITDPMG
    ELIDFQLGADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVF
    YDTTEKRIRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPK
    EVEKNTVRDFPIPKIRDVRKYPEITIPKEKKLGEIEFIKINKNDKKPLRD
    AVFSLQKQHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFE
    NSEPAGYKPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYIT
    NEPIPPKREYPRTGGIGMLPFYLIGCMMMGGVLLYTRKHP
  • Orf4 670 is a cell wall surface anchor family protein. An example of an amino acid sequence of orf4 670 is set forth in SEQ ID NO: 174.
    SEQ ID NO: 174
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRIREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVIDAHVFPKNSYNKPVVDKRIADTL
    NYNDQNGLSIGTKIPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGNNGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDITYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINPEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKFGYVEVAGKDEAMVLTSNTDGQFQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIFAVAGAAIMGIAVYAYVKNNKDEDQLA
  • Orf5 670 is a cell wall surface anchor family protein. An example of an amino acid sequence of orf5 670 is set forth in SEQ ID NO: 175.
    SEQ ID NO: 175
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDDRVQIVRDLHSWDENKSLLFKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KTDTMTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVD
    HQLVTTTVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEESGHYTPVL
    QNGKEVVVTSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKPN
    N
  • Orf6 670 is a sortase. An example of an amino acid sequence of orf6 670 is set forth in SEQ ID NO: 176.
    SEQ ID NO: 176
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPVIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • Orf7 670 is a sortase. An example of an amino acid sequence of orf7 670 is set forth in SEQ ID NO: 177.
    SEQ ID NO: 177
    VSRYYYRIESNEVIKEFDETVSQMDKAELEERWRLAQAFNATLKPSEILD
    PFTEQEKKKGVSEYANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKG
    AGLLEGASLPVGGENTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLD
    QVLAYQVDQILTVEPNDEEPVLIQHGEDYATLLTCTPYMINSHRLLVRGK
    RIPYTAPTAERNRAVRERGQFWLWLLLAALVMILVLSYGVYRHRRIVKGL
    EKQLEEHHVKG
  • Orf8 670 is a sortase. An example of an amino acid sequence of orf8 670 is set forth in SEQ ID NO: 178.
    SEQ ID NO: 178
    MSKAKLQKLLGYLLMLVALVIPVYCEGQMVLQSLGQVKGHEIFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGIRSVIAGHRAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 19A Hungary 6 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 19A Hungary 6 are set forth below.
  • ORF219AH is a transcriptional regulator. An example of an amino acid sequence of ORF219AH is set forth in SEQ ID NO: 187.
    SEQ ID NO: 187
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVTDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSTILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF319AH is a cell wall surface protein. An example of an amino acid sequence of ORF319AH is set forth in SEQ ID NO: 188.
    SEQ ID NO: 188
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTTVETKEASTPLDVVILLDNSNSMSNTRHNHAHRAEKAGEATRA
    LVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKILNDSALWTF
    DRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKLMYQFGAT
    FTQKALMTADDILTKQARPNSKKVIFHITDGVPTMSYPINFKYTGTTQSY
    RTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQMFTKKPVT
    DQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAYPFNSSTD
    WITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEATATRFMQSIS
    SSPDNYTNVADPSQILQELNRYFYTTVNEKKSIENGTITDPMGELIDFQL
    GADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVFYDTTEKR
    IRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPKEVEKNTV
    RDFPIPKIRDVRKYPEITIPKEKKLGETEFIKINKNDKKPLRDAVFSLQK
    QHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEPAGY
    KPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPIPPK
    REYPRTGGIGMLPFYLIGCMMMGGVLLYTRKNP
  • ORF419AH is a cell wall surface protein. An example of an amino acid sequence of ORF419AH is set forth in SEQ ID NO: 189.
    SEQ ID NO: 189
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRIREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVTDAHVFPKNSYNKPVVDKRIADTL
    NYNDQNGLSIGTKTPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGXNGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDITYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINPEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKEGYVEVAGKDEAMVLTSNTDGQFQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIFAVAGAAIMGIAVYAYVKNNKDEDQLA
  • ORF519AH is a cell wall surface protein. An example of an amino acid sequence of ORF519AH is set forth in SEQ ID NO: 190.
    SEQ ID NO: 190
    MTMQKMQKMISRIFFVMALCPSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDDRVQIVRDLHSWDENKLSSEKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEELFEMTDQTVEPLVIVAK
    KTDTMTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVD
    HQLVTITVVNQKLPRGNVDEMKVDGRTNTSLQGAMFKVMKEESGHYTPVL
    QNGKEVVVTSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSETIGK
    DTRKELVTVVKNNKRPRTDVPDTGEETLYILMLVAILLFGSGYYLTKKPN
    N
  • ORF619AH is a putative sortase. An example of an amino acid sequence of ORF619AH is set forth in SEQ ID NO: 191.
    SEQ ID NO: 191
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQTADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPVIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • ORF719AH is a putative sortase. An example of an amino acid sequence of ORF719AH is set forth in SEQ ID NO: 192.
    SEQ ID NO: 192
    MDNSRRSRKKGTKKKKHPLILLLIFLVGFAVAIYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLDQVLAYQVDQILTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLAALVMILVLSYGVYRHRRIVKGLEKQLEEHHVKG
  • ORF819AH is a putative sortase. An example of an amino acid sequence of ORF819AH is set forth in SEQ ID NO: 193.
    SEQ ID NO: 193
    MSKAKLQKLLGYLLMLVALVIPVYCFGQMVLQSLGQVKGHEIFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGIRSVIAGHPAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFMGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 6B Finland 12 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 6B Finland 12 are set forth below.
  • ORF26BF is a transcriptional regulator. An example of an amino acid sequence of ORF26BF is set forth in SEQ ID NO: 194.
    SEQ ID NO: 194
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSENEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHGQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF36BF is a cell wall surface protein. An example of an amino acid sequence of ORF36BF is set forth in SEQ ID NO: 195.
    SEQ ID NO: 195
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRTYQVNNLDDNQYGIE
    LTVSGKTTVETKEASTPLDVVILLDNSNSMSNIRHNHAHRAEKAGEATRA
    LVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKILNDSALWTF
    DRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKLMYQFGAT
    FTQKALMTADDILTKQARPNSKKVIFHITDGVPTMSYPINFKYTGTTQSY
    RTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQMFTKKPVT
    DQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAYPFNSSTD
    WITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEATATRFMQSIS
    SSPDNYTNVADPSQILQELNRYFYTIVNEKKSIENGTITDPMGELIDFQL
    GADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVFYDTTEKR
    IRVTGLYLGTGEKVTLTYNVRLNDQFVSNKEYDTNGRTTLHPKEVEKNTV
    RDFPIPKIRDVRKYPEITTPKEKKLGEIEFIKINKNDKKPLRDAVFSLQK
    QHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEPAGY
    KPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPIPPK
    REYPRTGGIGMLPEYLIGCMMMGGVLLYTRKHP
  • ORF46BF is a cell wall surface protein. An example of an amino acid sequence of ORF46BF is set forth in SEQ ID NO: 196.
    SEQ ID NO: 196
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRIREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVIDAHVFPKNSYNKPVVDKRIADTL
    NYNDQNGLSIGTKIPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGNNGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDITYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINPEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKFGYVEVAGKDEAMVLTSNTDGQFQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIEAVAGAAIMGIAVYAYVKNNKDEDQLA
  • ORF56BF is a cell wall surface protein. An example of an amino acid sequence of ORF56BF is set forth in SEQ ID NO: 197.
    SEQ ID NO: 197
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDDRVQIVRDLHSWDENKLSSFKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KTDTMTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEESGHYTPVL
    QNGKEVVVTSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKPN
    N
  • ORF66BF is a putative sortase. An example of an amino acid sequence of ORF66BF is set forth in SEQ ID NO: 198.
    SEQ ID NO: 198
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPVIDVDLPVYAGTAEEVLQQGAGHLEGTSLPTGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • ORF76BF is a putative sortase. An example of an amino acid sequence of ORF76BF is set forth in SEQ ID NO: 199.
    SEQ ID NO: 199
    MDNSRRSRKKGTKKKKHPLILLLIFLVGFAVAIYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLDQVLAYQVDQILTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLAALVMILVLSYGVYRHRRIVKGLEKQLEEHHVKG
  • ORF86BF is a putative sortase. An example of an amino acid sequence of ORF86BF is set forth in SEQ ID NO: 200.
    SEQ ID NO: 200
    MSKAKLQKLLGYLLMLVALVTPVYCFGQMVLQSLGQVKGHEIFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLETMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGIRSVIAGHRAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAPTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 6B Spain 2 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 6B Spain 2 are set forth below.
  • ORF26BSP is a transcriptional regulator. An example of an amino acid sequence of ORF26BSP is set forth in SEQ ID NO: 201.
    SEQ ID NO: 201
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSTLQELQ
    ETFEEELTFNLDTQQVQLTEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSPLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFITL
    NNQADVNLTKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDTRKEAFDKRVAM
    IAKKAHYLL
  • ORF36BSP is a cell wall surface protein. An example of an amino acid sequence of ORF36BSP is set forth in SEQ ID NO: 202.
    SEQ ID NO: 202
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTTVETKEASTPLDVVILLDNSNSMSNIRHNHAHRAEKAGEATRA
    LVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKILNDSALWTF
    DRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKLMYQFGAT
    FTQKALMTAKKILTKQARPNSKKVIFHITDGVPTMSYPINFKYTGTTQSY
    RTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQMFTKKPVT
    DQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAYPFNSSTD
    WITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEATATRFMQSIS
    SSPDNYTNVADPSQILQELNRYEYTIVNEKKSIENGTITDPMGELIDFQL
    GADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVFYDTTEKR
    TRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPKEVEKNTV
    RDFPIPKIRDVRKYPEITIPKEKKLGEIEFIKINKNDKKPLRDAVFSLQK
    QHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEPAGY
    KPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPIPPK
    REYPRTGGIGMLPFYLIGCMMMGGVLLYTRKHP
  • ORF46BSP is a cell wall surface protein. An example of an amino acid sequence of ORF46BSP is set forth in SEQ ID NO: 203.
    SEQ ID NO: 203
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRTREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVTDAHVFPKNSYNKPVVDKRIADTL
    NYNDQNGLSIGTKIPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGNNGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDTTYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINPEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKPGYVEVAGKDEAMVLTSNTDGQEQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIFAVAGAAIMGIAVYAYVKNNKDEDQLA
  • ORF56BSP is a cell wall surface protein. An example of an amino acid sequence of ORF56BSP is set forth in SEQ ID NO: 204.
    SEQ ID NO: 204
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDDRVQIVRDLHSWDENKLSSFKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KTDTMTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEESGHYTPVL
    QNGKEVVVTSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKPN
    N
  • ORF66BSP is a putative sortase. An example of an amino acid sequence of ORF66BSP is set forth in SEQ ID NO: 205.
    SEQ ID NO: 205
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVETPVIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFTAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • ORF76BSP is a putative sortase. An example of an amino acid sequence of ORF76BSP is set forth in SEQ ID NO: 206.
    SEQ ID NO: 206
    MDNSRRSRKKGTKKKKHPLILLLIFLVGFAVAIYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLDQVLAYQVDQILTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLAALVMILVLSYGVYRHRRIVKGLEKQLEEHHVKG
  • ORF86BSP is a putative sortase. An example of an amino acid sequence of ORF86BSP is set forth in SEQ ID NO: 207.
    SEQ ID NO: 207
    MSKAKLQKLLGYLLMLVALVIPVYCFGQMVLQSLGQVKGHEIFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLETMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGIRSVIAGHRAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPTPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 9V Spain 3 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 9V Spain 3 are set forth below.
  • ORF29VSP is a transcriptional regulator. An example of an amino acid sequence of ORF29VSP is set forth in SEQ ID NO: 208.
    SEQ ID NO: 208
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILREELLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMTVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILILSPPPSEEHLTEPLIITTK
    EYLPYVKKQYPKGKHHFLTTALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF39VSP is a cell wall surface protein. An example of an amino acid sequence of ORF39VSP is set forth in SEQ ID NO: 209.
    SEQ ID NO: 209
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTNGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQRTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTVYERKDKSVPLDVVILLDNSNSMSNIRNKNARRAERAGEATRS
    LIDKITSDPENRVALVTYASTIFDGTEFTVEKGVADKNGKRLNDSLFWNY
    DQTSFTTNTKDYSYLKLTNDKNDIVELKNKVPTEAEDHDGNRLMYQFGAT
    FTQKALMKADEILTQQARQNSQKVIFHITDGVPTMSYPINFNHATFAPSY
    QNQLNAFFSKSPNKDGILLSDFITQATSGEHTIVRGDGQSYQMFTDKTVY
    EKGAPAAFPVKPEKYSEMKAVGYAVIGDPINGGYIWLNWRESILAYPFNS
    NTAKITNHGDPTRWYYNGNIAPDGYDVFTVGIGINGDPGTDEATATSFMQ
    SISSKPENYTNVTDTTKILEQLNRYFHTIVTEKKSIENGTITDPMGELID
    LQLGTDGRFDPADYTLTANDGSRLENGQAVGGPQNDGGLLKNAKVFYDTT
    EKRIRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPKEVEK
    NTVRDFPIPKIRDVRKYPAITIAKEKKLGEIEFIKINKNDKKPLRDAVFS
    LQKQHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEP
    AGYKPVQNKPIVAFQIVNGEVRDVISTVPQDIPAGYEFTNDKHYITNEPI
    PPKREYPRTGGIGMLLFYLIGCMMMGGVLLYTRKHP
  • ORF49VSP is a cell wall surface protein. An example of an amino acid sequence of ORF49VSP is set forth in SEQ ID NO: 210.
    SEQ ID NO: 210
    MKSINKFLTMLAALLLTASSLFSAATVFAAGTTTTSVTVHKLLATDGDMD
    KIANELETGNYAGNKVGVLPANAKEIAGVMFVWTNTNNEIIDENGQTLGV
    NIDPQTFKLSGAMPATAMKKLTEAEGAKFNTANLPAAKYKIYEIHSLSTY
    VGEDGATLTGSKAVPIEIELPLNDVVDAHVYPKNTEAKPKIDKDFKGKAN
    PDTPRVDKDTPVNHQVGDVVEYEIVTKIPALANYATANWSDRMTEGLAFN
    KGTVKVTVDDVALEAGDYALTEVATGFDLKLTDAGLAKVNDQNAEKTVKI
    TYSATLNDKAIVEVPESNDVTFNYGNNPDHGNTPKPNKPNENGDLTLTKT
    WVDATGAPIPAGAEATFDLVNAQTGDVVQTVTLTTDKNTVTVNGLDKNTE
    YKFVERSIKGYSADYQEITTAGEIAVKNWKDENPKPLDPTEPKVVTYGKK
    FVKVNDKDNRLAGAEFVIANADNAGQYLARKADKVSQEEKQLVVTTKDAL
    DRAVAAYNALTAQQQTQQEKEKVDKAQAAYNAAVTAANNAFEWVADKDNE
    NVVKLVSDAQGRFEITGLLAGTYYLEETKQPAGYALLTSRQKFEVTATSY
    SATGQGIEYTAGSGKDDATKVVNKKITIPQTGGIGTIIFAVAGAVIMGIA
    VYAYVKNNKDEDQLA
  • ORF59VSP is a cell wall surface protein. An example of an amino acid sequence of ORF59VSP is set forth in SEQ ID NO: 211.
    SEQ ID NO: 211
    MTMQKMQKMQKMQKMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTL
    VLQLENYQEVVSQLPSRDGHRLQVWKLDDSYSYDNRVQTVRDLHSWDENK
    LSSFKKTSFEMTFLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMT
    DQTVEPLVIVAKKADTVTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEK
    EVPLIGEYRYSSSGQVGRTLYTDKNGEIVVTNLPLGTYRFKEVEPLAGYT
    VTTMDTDVQLVDHQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKV
    MKEENGHYTPVLQNGKEVVVASGKDGRFRVEGLEYGTYYLWELQAPTGYV
    QLTSPVSFTIGKDTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILL
    FGSGYYLTKKTNN
  • ORF69VSP is a putative sortase. An example of an amino acid sequence of ORF69VSP is set forth in SEQ ID NO: 212.
    SEQ ID NO: 212
    MLIKMAKTKKQKRNNLLLGVVFFIGIAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPAIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKRQSERALKALKEATKEVKVE
    DE
  • ORF79VSP is a putative sortase. An example of an amino acid sequence of ORF79VSP is set forth in SEQ ID NO: 213.
    SEQ ID NO: 213
    MSKSRYSRKKSVKKKKNPFILLLIFLVGLAVANYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERTGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDIEYLHVLDQVLAYQVDQIVTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLGAMAVILLLLYRVYRNRRIVKGLEKQLEGRHVKD
  • ORF89VSP is a putative sortase. An example of an amino acid sequence of ORF89VSP is set forth in SEQ ID NO: 214.
    SEQ ID NO: 214
    MSRTKLRALLGYLLMLVACLIPIYGFGQMVLQSLGQVKGHATFVKSMTTE
    MYQEQQNHSLAYNQRLASQNRIVDPELAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLGMGLAHVDGTPLPLDGTGIRSVIAGHPAEPSH
    VFTRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNEERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 14 CSR 10 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 14 CSR 10 are set forth below.
  • ORF214CSR is a transcriptional regulator. An example of an amino acid sequence of ORF214CSR is set forth in SEQ ID NO: 215.
    SEQ ID NO: 215
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKTLRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTTQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHTSKATVQEWMTEQKIEGVTDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF314CSR is a cell wall surface protein. An example of an amino acid sequence of ORF314CSR is set forth in SEQ ID NO: 216.
    SEQ ID NO: 216
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTTVETKEASTPLDVVTLLDNSNSMSNIRHNHAHRAEKAGEATRA
    LVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKILNDSALWTF
    DRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKLMYQFGAT
    FTQKALMTADDILTKQARPNSKKVIFHITDGVPTMSYPINFKYTGTTQSY
    RTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQMFTKKPVT
    DQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAYPFNSSTD
    WITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEARARTFMQSIS
    SSPDNYTNVADPSQILQELNRYFYTIVNEKKSIENGTITDPMGELIDFQL
    GADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVFYDTTEKR
    IRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPKEVEKNTV
    RDFPIPKIRDVRKYPEITIPKEKKLGEIEFIKINKNDKKPLRDAVPSLQK
    QHPDYPDIYGAIDQNGTYQNVRTGEDGKLTEKNLSDGKYRLFENSEPAGY
    KPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPTPPK
    REYPRTGGIGMLPFYLTGCMMMGGVLLYTRKHP
  • ORF414CSR is a cell wall surface protein. An example of an amino acid sequence of ORF414CSR is set forth in SEQ ID NO: 217.
    SEQ ID NO: 217
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRIREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVIDAHVFPKNSYNKPVVDKRTADTL
    NYNDQNGLSIGTKIPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGNNGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDITYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINPEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKFGYVEVAGKDEAMVLTSNTDGQFQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIFAVAGAAIMGIAVYAYVKNNKDEDQLA
  • ORF514CSR is a cell wall surface protein. An example of an amino acid sequence of ORF514CSR is set forth in SEQ ID NO: 218.
    SEQ ID NO: 218
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDDRVQIVRDLHSWDENKLSSFKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KTDTMTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGETFVTNLPLGNYRFKEVEPLAGYAVTTLDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEESGHYTPVL
    QNGKEVVVTSGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKPN
    N
  • ORF614CSR is a putative sortase. An example of an amino acid sequence of ORF614CSR is set forth in SEQ ID NO: 219.
    SEQ ID NO: 219
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPVIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • ORF714CSR is a putative sortase. An example of an amino acid sequence of ORF714CSR is set forth in SEQ ID NO: 220.
    SEQ ID NO: 220
    MDNSRRSRKKGTKKKKHPLILLLIFLVGFAVAIYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLDQVLAYQVDQILTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQEWLWLLLAALVMILVLSYGVYRHRRIVKGLEKQLEEHHVKG
  • ORF814CSR is a putative sortase. An example of an amino acid sequence of ORF814CSR is set forth in SEQ ID NO: 221.
    SEQ ID NO: 221
    MSKAKLQKLLGYLLMLVALVIPVYCFGQMVLQSLGQVKGHEIFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGIRSVIAGHPAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTPNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGTLFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 19F Taiwan 14 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 19F Taiwan 14 are set forth below.
  • ORF219FTW is a transcriptional regulator. An example of an amino acid sequence of ORF219FTW is set forth in SEQ ID NO: 222.
    SEQ ID NO: 222
    MLNKYIEKRITDKITILNILLDIRSTELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKTLRFFLLQG
    NQSENEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LTALLQFHFGTEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHE
    STLVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILTSPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF319FTW is a cell wall surface protein. An example of an amino acid sequence of ORF319FTW is set forth in SEQ ID NO: 223.
    SEQ ID NO: 223
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIESNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVTPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTVYERKDKSVPLDVVILLDNSNSMSNIRNKNARRAERAGEATRS
    LIDKITSDPENRVALVTYASTIPDGTEFTVEKGVADKNGKRLNDSLFWNY
    DQTSFTTNTKDYSYLKLTNDKNDIVELKNKVPTEAEDHDGNRLMYQFGAT
    FTQKALMKADEILTQQARQNSQKVIFHITDGVPTMSYPINFNHATFAPSY
    QNQLNAFFSKSPNKDGILLSDFITQATSGEHTIVRGDGQSYQMFTDKTVY
    EKGAPAAFPVKPEKYSEMKAVGYAVIGDPINGGYIWLNWRESILAYPFNS
    NTAKITNHGAPTRWYYNGNIAPDGYDVFTVGIGINGDPGTDEATATSFMQ
    SISSKPENYTNVTDTTKILEQLNRYFHTIVTEKKSIENGTITDPMGELID
    LQLGTDGRFDPADYTLTANDGSRLENGQAVGGPQNDGGLLKNAKVFYDTT
    EKRIRVTGLYLGTGEKVTLTYNVRLNDQEVSNKFYDTNGRTTLHPKEVEK
    NTVRDFPIPKIRDVRKYPAITIAKEKKLGEIEFIKINKNDKKPLRDAVFS
    LQKQHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEP
    AGYKPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPI
    PPKREYPRTGGIGMLPFYLIGCMMMGGVLLYTRKHP
  • ORF419FTW is a cell wall surface protein. An example of an amino acid sequence of ORF419FTW is set forth in SEQ ID NO: 224.
    SEQ ID NO: 224
    MKSINKFLTMLAALLLTASSLFSAATVFAAGTTTTSVTVHKLLATDGDMD
    KIANELETGNYAGNKVGVLPANAKEIAGVMFVWTNTNNEIIDENGQTLGV
    NIDPQTFKLSGAMPATAMKKLTEAEGAKFNTANLPAAKYKIYEIHSLSTY
    VGEDGATLTGSKAVPIEIELPLNDVVDAHVYPKNTEAKPKIDKDFKGKAN
    PDTPRVDKDTPVNHQVGDVVEYEIVTKIPALANYATANWSDRMTEGLAFN
    KGTVKVTVDDVALEAGDYALTEVATGFDLKLTDAGLAKVNDQNAEKTVKI
    TYSATLNDKAIVEVPESNDVTFNYGNNPDHGNTPKPNKPNENGDLTLTKT
    WVDATGAPIPAGAEATFDLVNAQTGKVVQTVTLTTDKNTVTVNGLDKNTE
    YKFVERSIKGYSADYQEITTAGEIAVKNWKDENPKPLDPTEPKVVTYGKK
    FVKVNDKDNRLAGAEFVTANADNAGQYLARKADKVSQEEKQLVVTTKDAL
    DRAVAAYNALTAQQQTQQEKEKVDKAQAAYNAAVIAANNAFEWVADKDNE
    NVVKLVSDAQGRFEITGLLAGTYYLEETKQPAGYALLTSRQKFEVTATSY
    SATGQGIEYTAGSGKDDATKVVNKKITIPQTGGIGTIIFAVAGAVIMGIA
    VYAYVKNNKDEDQLA
  • ORF519FTW is a cell wall surface protein. An example of an amino acid sequence of ORF519FTW is set forth in SEQ ID NO: 225.
    SEQ ID NO: 225
    MTMQKMQKMTSRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDNRVQIVRDLHSWDENKLSSFKKTSFEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KADTVTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIVVTNLPLGTYRFKEVEPLAGYTVTTMDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEENGHYTPVL
    QNGKEVVVASGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKTN
    N
  • ORF619FTW is a putative sortase. An example of an amino acid sequence of ORF619FTW is set forth in SEQ ID NO: 226.
    SEQ ID NO: 226
    MLIKMAKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADTDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPAIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKRQSERALKALKEATKEVKVE
    DE
  • ORF719FTW is a putative sortase. An example of an amino acid sequence of ORF719FTW is set forth in SEQ ID NO: 227.
    SEQ ID NO: 227
    MSKSRYSRKKSVKKKKNPFILLLIFLVGLAVAMYPLVSRYYYRIESNEVT
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTDQEKKQGVSEY
    ANMLKVHERIGYVEIPAIEQEIPMYVGTSEDILQKGAGLLEGASLPVGGE
    NTHTVITAHRGLPTAELFSQLDKMKKGDIFYLHVLDQVLAYQVDQIVTVE
    PNDFEPVLIQHGQDYATLLTCTPYMLNSHRLLVRGKRTPYTAPIAERNRA
    VRERGQFWLWLLLGAMAVILLLLYRVYRNRRTVKGLEKQLEGRHVKD
  • ORF819FTW is a putative sortase. An example of an amino acid sequence of ORF819FTW is set forth in SEQ ID NO: 228.
    SEQ ID NO: 228
    MSRTKLRALLGYLLMLVACLIPIYCFGQMVLQSLGQVKGHATFVKSMTTE
    MYQEQQNHSLAYNQRLASQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLGMGLAHVDGTPLPLDGTGIRSVIAGHRAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 23F Taiwan 15 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 23F Taiwan 15 are set forth below.
  • ORF223FTW is a transcriptional regulator. An example of an amino acid sequence of ORF223FTW is set forth in SEQ ID NO: 229.
    SEQ ID NO: 229
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTPNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLETTPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKAIVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLITITTK
    EYLPYVKKQYPKGKHHFLTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAM
    IAKKAHYLL
  • ORF323FTW is a cell wall surface protein. An example of an amino acid sequence of ORF323FTW is set forth in SEQ ID NO: 230.
    SEQ ID NO: 230
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTVYEQKDKSVPLDVVILLDNSNSMSNIRNKNARRAERAGEATRS
    LIDKITSDPENRVALVTYASTIFDGTEFTVEKGVADKNGKRLNDSLFWNY
    DQTSFTTNTKDYSYLKLTNDKNDIVELKNKVPTFAEDHDGNRLMYQFGAT
    FTQKALMKADEILTQQARQNSQKVIFHITDGVPTMSYPINFNHATFAPSY
    QNQLNAETSKSPNKDGILLSDFITQATSGEHTIVRGDGQSYQMFTDKTVY
    EKGAPAAFPVKPEKYSEMKAAGYAVIGDPINGGYIWLNWRESILAYPFNS
    NTAKITNHGDPTRWYYNGNIAPDGYDVFTVGIGINGDPGTDEATATSFMQ
    SISSKPENYTNVTDTTKILEQLNRYFHTIVTEKKSIENGTITDPMGELID
    LWLGTDGRFDPADYTLTANDGSRLENGQAVGGPQNDGGLLKNAKVLYDTT
    EKRIRVTGLYLGTKEKVTLTYNVRLNDEFVSNKFYDTNGRTTLHPKEVEQ
    NTVRDFPIPKIRDVRKYPEITISKEKKLGDIEFIKVNKNDKKPLRDAVFS
    LQKQHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEP
    AGYKPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEFTNDKHYITNEPI
    PPKREYPRTGGIGMLPFYLIGCMMMGGVLLYTRKHP
  • ORF423FTW is a cell wall surface protein. An example of an amino acid sequence of ORF423FTW is set forth in SEQ ID NO: 231.
    SEQ ID NO: 231
    MKSINKFLTILAALLLTVSSLFSAATVFAAEQKTKTLTVHKLLMTDQELD
    AWNSDAITTAGYDGSQNFEQFKQLQGVPQGVTEISGVAFELQSYTGPQGK
    EQENLTNDAVWTAVNKGVTTETGVKFDTEVLQGTYRLVEVRKESTYVGPN
    GKVLTGMKAVPALITLPLVNQNGVVENAHVYPKNSEDKPTATKTFDTAAG
    FVDPGEKGLAIGTKVPYIVTTTIPKNSTLATAPWSDEMTEGLDYNGDVVV
    NYNGQPLDNSHYTLEAGHNGFILKLNEKGLEAINGKDAEATITLKYTATL
    NALAVADVPEANDVTFHYGNNPGHGNTPKPNKPKNGELTITKTWADAKDA
    PIAGVEVTFDLVNAQTGEVVKVPGHETGIVLNQTNNWTFTATGLDNNTEY
    KFVERTIKGYSADYQTITETGKIAVKNWKDENPEPINPEEPRVKTYGKKF
    VKVDQKDERLKEAQFVVKNEQGKYLALKSAAQQAVNEKAAAEAKQALDAA
    IAAYTNAADKNAAQAVVDAAQKTYNDNYRAARFGYVEVERKEDALVLTSN
    TDGQFQISGLAAGSYTLEETKAPEGFAKLGDVKFEVGAGSWNQGDFNYLK
    DVQKNDATKVVNKKITIPQTGGIGTIIFAVAGAVIMGIAVYAYVKNNKDE
    DQLA
  • ORF523FTW is a cell wall surface protein. An example of an amino acid sequence of ORF523FTW is set forth in SEQ ID NO: 232.
    SEQ ID NO: 232
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDNRVQIVRDLHSWDENKLSSFKKTSEEMT
    FLENQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KADTVTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIVVTNLPLGTYRFKEVEPLAGYTVTTMDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEENGHYTPVL
    QNGKEVVVASGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRIDVPDTGEETLYILMLVAILLFGSGYYLTKKTN
    N
  • ORF623FTW is a putative sortase. An example of an amino acid sequence of ORF623FTW is set forth in SEQ ID NO: 233.
    SEQ ID NO: 233
    MLIKMVKTKKQKRNNLLLGVVFFIGMAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVEIPVIDVDLPVYAGTAEEVLQQGAGQLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKKQPEKALKALKAARKEVKVE
    DGQQ
  • ORF723FTW is a putative sortase. An example of an amino acid sequence of ORF723FTW is set forth in SEQ ID NO: 234.
    SEQ ID NO: 234
    MDNSRRSRKKGTKKKKHPLILLLIFLVGFAVAIYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFThQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEEILQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDVFYLHVLDQVLAYQVDQILTVE
    PNDFEPVLIQHGKDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLAALVMILVLSYGVYRHRRIVKGLEKQLEEHHVKG
  • ORF823FTW is a putative sortase. An example of an amino acid sequence of ORF823FTW is set forth in SEQ ID NO: 235.
    SEQ ID NO: 235
    MSKAKLQKLLGYLLMLVALVIPVYCFGQMVLQSLGQVKGHETFSESVTAD
    SYQEQLQRSLDYNQRLDSQNRIVDPFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLAMGLAHVDGTPLPVEGKGTRSVIAGHPAEPSH
    VFFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • As discussed above, a S. pneumoniae AI sequence is present in the 23F Poland 16 S. pneumoniae genome. Examples of S. pneumoniae AI sequences from 23F Poland 16 are set forth below.
  • ORF223FP is a transcriptional regulator. An example of an amino acid sequence of ORF223FP is set forth in SEQ ID NO: 236.
    +TR, SEQ ID NO: 236
    MLNKYIEKRITDKITILNILLDIRSIELDELSTLTSLQSKSLLSILQELQ
    ETFEEELTFNLDTQQVQLIEHHSHQTNYYFHQLYNQSTILKILRFFLLQG
    NQSFNEFTQKEYISIATGYRVRQKCGLLLRSVGLDLVKNQVVGPEYRIRF
    LIALLQFHFGIEIYDLNDGSMDWVTHMIVQSNSQLSHELLEITPDEYVHF
    SILVALTWKRREFPLEFPESKEFEKLKNLFMYPILMEHCQTYLEPHANMT
    FTQEELDYIFLVYCSANSSFSKDKWNQEKKTHTIQLILQHTRGKHLLSKF
    KNILGNDISNSLSFLTALTFLTRTFLFGLQNLVPYYNYYEHYGIESDKPL
    YHISKATVQEWMTEQKIEGVIDQHRLYLFSLYLTETIFSSLPAIPIFIIL
    NNQADVNLIKSIILRNFTDKVASVTGYNILISPPPSEEHLTEPLIIITTK
    EYLPYVKKQYPKGKHHELTIALDLHVSQQRLIYQTIVDIRKEAFDKRVAN
    IAKKAHYLL
  • ORF323FP is a cell wall surface protein. An example of an amino acid sequence of ORF323FP is set forth in SEQ ID NO: 237.
    SEQ ID NO: 237
    MKKVRKIFQKAVAGLCCISQLTAFSSIVALAETPETSPAIGKVVIKETGE
    GGALLGDAVFELKNNTDGTTVSQRTEAQTGEAIFSNIKPGTYTLTEAQPP
    VGYKPSTKQWTVEVEKNGRTTVQGEQVENREEALSDQYPQTGTYPDVQTP
    YQIIKVDGSEKNGQHKALNPNPYERVIPEGTLSKRIYQVNNLDDNQYGIE
    LTVSGKTTVETKEASTPLDVVTLLDNSNSMSNIRHNHAHRAEKAGEATRA
    LVDKITSNPDNRVALVTYGSTIFDGSEATVEKGVADANGKTLNDSALWTE
    DRTTFTAKTYNYSFLNLTSDPTDIQTIKDRIPSDAEELNKDKLMYQFGAT
    FTQKALMTADDILTKQARPNSKKVIFHITDGVPTMSYPINFKYTGTTQSY
    RTQLNNFKAKTPNSSGILLEDFVTWSADGEHKIVRGDGESYQMFTKKPVT
    DQYGVHQILSITSMEQRAKLVSAGYRFYGTDLYLYWRDSILAYPFNSSTD
    WITNHGDPTTWYYNGNMAQDGYDVFTVGVGVNGDPGTDEATATRFMQSIS
    SSPDNYTNVADPSQILQELNRYFYTIVNEKKSIENGTITDPMGELIDFQL
    GADGRFDPADYTLTANDGSSLVNNVPTGGPQNDGGLLKNAKVFYDTTEKR
    IRVTGLYLGTGEKVTLTYNVRLNDQFVSNKFYDTNGRTTLHPKEVEKNTV
    RDFPIPKIRDVRKYPEITIPKEKKLGEIEFIKINKNDKKPLRDAVFSLQK
    QHPDYPDIYGAIDQNGTYQNVRTGEDGKLTFKNLSDGKYRLFENSEPAGY
    KPVQNKPIVAFQIVNGEVRDVTSIVPQDIPAGYEETNDKHYITNEPIPPK
    REYPRTGGIGMLPFYLIGCMMMGGVLLYTRKNP
  • ORF423FP is a cell wall surface protein. An example of an amino acid sequence of ORF423FP is set forth in SEQ ID NO: 238.
    SEQ ID NO: 238
    MKSINKFLTMLAALLLTASSLFSAATVFAADNVSTAPDAVTKTLTIHKLL
    LSEDDLKTWDTNGPKGYDGTQSSLKDLTGVVAEEIPNVYFELQKYNLTDG
    KEKENLKDDSKWTTVHGGLTTKDGLKIETSTLKGVYRIREDRTKTTYVGP
    NGQVLTGSKAVPALVTLPLVNNNGTVIDAHVEPKNSYNKPVVDKRIADTL
    NYNDQNGLSIGTKIPYVVNTTIPSNATFATSFWSDEMTEGLTYNEDVTIT
    LNNVAMDQADYEVTKGINGFNLKLTEAGLAKINGKDADQKIQITYSATLN
    SLAVADIPESNDITYHYGNHQDHGNTPKPTKPNNGQITVTKTWDSQPAPE
    GVKATVQLVNAKTGEKVGAPVELSENNWTYTWSGLDNSIEYKVEEEYNGY
    SAEYTVESKGKLGVKNWKDNNPAPINLEEPRVKTYGKKFVKVDQKDTRLE
    NAQFVVKKADSNKYIAFKSTAQQAADEKAAATAKQKLDAAVAAYTNAADK
    QAAQALVDQAQQEYNVAYKEAKFGYVEVAGKDEAMVLTSNTDGQFQISGL
    AAGTYKLEEIKAPEGFAKIDDVEFVVGAGSWNQGEFNYLKDVQKNDATKV
    VNKKITIPQTGGIGTIIFAVAGAVIMGIAVYAYVKNNKDEDQLA
  • ORF523FP is a cell wall surface protein. An example of an amino acid sequence of ORF523FP is set forth in SEQ ID NO: 239.
    SEQ ID NO: 239
    MTMQKMQKMISRIFFVMALCFSLVWGAHAVQAQEDHTLVLQLENYQEVVS
    QLPSRDGHRLQVWKLDDSYSYDNRVQIVRDLHSWDENKLSSFKKTSFEMT
    FLSNQIEVSHIPNGLYYVRSIIQTDAVSYPAEFLFEMTDQTVEPLVIVAK
    KADTVTTKVKLIKVDQDHNRLEGVGFKLVSVARDGSEKEVPLIGEYRYSS
    SGQVGRTLYTDKNGEIVVTNLPLGTYRFKEVEPLAGYAVTTMDTDVQLVD
    HQLVTITVVNQKLPRGNVDFMKVDGRTNTSLQGAMFKVMKEENGHYTPVL
    QNGKEVVVASGKDGRFRVEGLEYGTYYLWELQAPTGYVQLTSPVSFTIGK
    DTRKELVTVVKNNKRPRTDVPDTGEETLYILMLVAILLFGSGYYLTKKTN
    N
  • ORF623FP is a putative sortase. An example of an amino acid sequence of ORF623FP is set forth in SEQ ID NO: 240.
    SEQ ID NO: 240
    MLIKMAKTKKQKRNNLLLGVVFFIGIAVMAYPLVSRLYYRVESNQQIADF
    DKEKATLDEADIDERMKLAQAFNDSLNNVVSGDPWSEEMKKKGRAEYARM
    LEIHERMGHVETPAIDVDLPVYAGTAEEVLQQGAGHLEGTSLPIGGNSTH
    AVITAHTGLPTAKMFTDLTKLKVGDKFYVHNIKEVMAYQVDQVKVIEPTN
    FDDLLIVPGHDYVTLLTCTPYMINTHRLLVRGHRIPYVAEVEEEFIAANK
    LSHLYRYLFYVAVGLIVILLWIIRRLRKKKRQSERALKALKEATKEVKVE
    DE
  • ORF723FP is a putative sortase. An example of an amino acid sequence of ORF723FP is set forth in SEQ ID NO: 241.
    SEQ ID NO: 241
    MSKSRYSRKKSVKKKKNPFILLLIFLVGLAVAMYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFNATLKPSEILDPFTEQEKKKGVSEY
    ANMLKVHERIGYVEIPAIDQEIPMYVGTSEETLQKGAGLLEGASLPVGGE
    NTHTVVTAHRGLPTAELFSQLDKMKKGDIFYLHVLDQVLAYQVDQIVTVE
    PNDFEPVLIQHGEDYATLLTCTPYMINSHRLLVRGKRIPYTAPIAERNRA
    VRERGQFWLWLLLGAMAVILLLLYRVYRNRRIVKGLEKQLEGRHVKD
  • ORF823FP is a putative sortase. An example of an amino acid sequence of ORF823FP is set forth in SEQ ID NO: 242.
    SEQ ID NO: 242
    MSKSRYSRKKSVKKKKNPFILLLIFLVGLAVAMYPLVSRYYYRIESNEVI
    KEFDETVSQMDKAELEERWRLAQAFFLAEGYEVNYQVSDDPDAVYGYLSI
    PSLEIMEPVYLGADYHHLGMGLAHVDGTPLPLDGTGIRSVIAGHPAEPSH
    VEFRHLDQLKVGDALYYDNGQEIVEYQMMDTEIILPSEWEKLESVSSKNI
    MTLITCDPIPTFNKRLLVNFERVAVYQKSDPQTAAVARVAFTKEGQSVSR
    VATSQWLYRGLVVLAFLGILFVLWKLARLLRGK
  • Immunogenic compositions of the invention comprising AI antigens may further comprise one or more antigenic agents. Preferred antigens include those listed below. Additionally, the compositions of the present invention may be used to treat or prevent infections caused by any of the below-listed microbes. Antigens for use in the immunogenic compositions include, but are not limited to, one or more of the following set forth below, or antigens derived from one or more of the following set forth below:
  • Bacterial Antigens
  • N. meningitides: a protein antigen from N. meningitides serogroup A, C, W135, Y, and/or B (1-7); an outer-membrane vesicle (OMV) preparation from N. meningitides serogroup B. (8, 9, 10, 11); a saccharide antigen, including LPS, from N. meningitides serogroup A, B, C W135 and/or Y, such as the oligosaccharide from serogroup C (see PCT/US99/09346; PCT IB98/01665; and PCT IB99/00103);
  • Streptococcus pneumoniae: a saccharide or protein antigen, particularly a saccharide from Streptooccus pneumoniae;
  • Streptococcus agalactiae: particularly, Group B streptococcus antigens;
  • Streptococcus pyogenes: particularly, Group A streptococcus antigens;
  • Enterococcus faecalis or Enterococcus faecium: Particularly a trisaccharide repeat or other Enterococcus derived antigens provided in U.S. Pat. No. 6,756,361;
  • Helicobacter pylori. including: Cag, Vac, Nap, HopX, HopY and/or urease antigen;
  • Bordetella pertussis: such as petussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B. pertussis, optionally also combination with pertactin and/or agglutinogens 2 and 3 antigen;
  • Staphylococcus aureus: including S. aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa exotoxin A, such as StaphVAX™, or antigens derived from surface proteins, invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit phagocytic engulfment (capsule, Protein A), carotenoids, catalase production, Protein A, coagulase, clotting factor, and/or membrane-damaging toxins (optionally detoxified) that lyse eukaryotic cell membranes (hemolysins, leukotoxin, leukocidin);
  • Staphylococcus epidermis: particularly, S. epidermidis slime-associated antigen (SAA);
  • Staphylococcus saprophyticus: (causing urinary tract infections) particularly the 160 kDa hemagglutinin of S. saprophyticus antigen;
  • Pseudomonas aeruginosa. particularly, endotoxin A, Wzz protein, P. aeruginosa LPS, more particularly LPS isolated from PAO1 (O5 serotype), and/or Outer Membrane Proteins, including Outer Membrane Proteins F (OprF) (Infect Immun. 2001 May; 69(5): 3510-3515);
  • Bacillus anthracis (anthrax): such as B. anthracis antigens (optionally detoxified) from A-components (lethal factor (LF) and edema factor (EF)), both of which can share a common B-component known as protective antigen (PA);
  • Moraxella catarrhalis. (respiratory) including outer membrane protein antigens (HMW-OMP), C-antigen, and/or LPS;
  • Yersinia pestis (plague): such as F1 capsular antigen (Infect Immun. 2003 January; 71(1)): 374-383, LPS (Infect Immun. 1999 October; 67(10): 5395), Yersinia pestis V antigen (Infect Immun. 1997 November; 65(11): 4476-4482);
  • Yersinia enterocolitica (gastrointestinal pathogen): particularly LPS (Infect Immun. 2002 August; 70(8): 4414);
  • Yersinia pseudotuberculosis: gastrointestinal pathogen antigens;
  • Mycobacterium tuberculosis: such as lipoproteins, LPS, BCG antigens, a fusion protein of antigen 85B (Ag85B) and/or ESAT-6 optionally formulated in cationic lipid vesicles (Infect Immun. 2004 October; 72(10): 6148), Mycobacterium tuberculosis (Mtb) isocitrate dehydrogenase associated antigens (Proc Natl Acad Sci USA. 2004 Aug. 24; 101(34): 12652), and/or MPT51 antigens (Infect Immun. 2004 July; 72(7): 3829);
  • Legionella pneumophila (Legionnairs' Disease): L. pneumophila antigens—optionally derived from cell lines with disrupted asd genes (Infect Immun. 1998 May; 66(5): 1898);
  • Rickettsia: including outer membrane proteins, including the outer membrane protein A and/or B (OmpB) (Biochim Biophys Acta. 2004 Nov. 1; 1702(2): 145), LPS, and surface protein antigen (SPA) (J Autoimmun. 1989 June; 2 Suppl:81);
  • E. coli: including antigens from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), and/or enterohemorrhagic E. coli (EHEC);
  • Vibrio cholerae: including proteinase antigens, LPS, particularly lipopolysaccharides of Vibrio cholerae II, O1 Inaba O-specific polysaccharides, V. cholera O139, antigens of IEM108 vaccine (Infect Immun. 2003 October; 71(10):5498-504), and/or Zonula occludens toxin (Zot);
  • Salmonella typhi (typhoid fever): including capsular polysaccharides preferably conjugates (Vi, i.e. vax-TyVi);
  • Salmonella typhimurium (gastroenteritis): antigens derived therefrom are contemplated for microbial and cancer therapies, including angiogenesis inhibition and modulation of flk;
  • Listeria monocytogenes (sytemic infections in immunocompromised or elderly people, infections of fetus): antigens derived from L. monocytogenes are preferably used as carriers/vectors for intracytoplasmic delivery of conjugates/associated compositions of the present invention;
  • Porphyromonas gingivalis: particularly, P. gingivalis outer membrane protein (OMP);
  • Tetanus: such as tetanus toxoid (TT) antigens, preferably used as a carrier protein in conjunction/conjugated with the compositions of the present invention;
  • Diphtheria: such as a diphtheria toxoid, preferably CRM197, additionally antigens capable of modulating, inhibiting or associated with ADP ribosylation are contemplated for combination/co-administration/conjugation with the compositions of the present invention, the diphtheria toxoids are preferably used as carrier proteins;
  • Borrelia burgdorferi (Lyme disease): such as antigens associated with P39 and P13 (an integral membrane protein, Infect Immun. 2001 May; 69(5): 3323-3334), VlsE Antigenic Variation Protein (J Clin Microbiol. 1999 December; 37(12): 3997);
  • Haemophilis influenzae B: such as a saccharide antigen therefrom;
  • Klebsiella: such as an OMP, including OMP A, or a polysaccharide optionally conjugated to tetanus toxoid;
  • Neiserria gonorrhoeae: including, a Por (or porin) protein, such as PorB (see Zhu et al., Vaccine (2004) 22:660-669), a transferring binding protein, such as ThpA and TbpB (See Price et al., Infection and Immunity (2004) 71(1):277-283), a opacity protein (such as Opa), a reduction-modifiable protein (Rmp), and outer membrane vesicle (OMV) preparations (see Plante et al., J Infectious Disease (2000) 182:848-855), also see e.g. WO99/24578, WO99/36544, WO99/57280, WO02/079243);
  • Chlamydia pneumoniae. particularly C. pneumoniae protein antigens;
  • Chlamydia trachomatis: including antigens derived from serotypes A, B, Ba and C are (agents of trachoma, a cause of blindness), serotypes L1, L2 & L3 (associated with Lymphogranuloma venereum), and serotypes, D-K;
  • Treponema pallidum (Syphilis): particularly a TmpA antigen; and
  • Haemophilus ducreyi (causing chancroid): including outer membrane protein (DsrA).
  • Where not specifically referenced, further bacterial antigens of the invention may be capsular antigens, polysaccharide antigens or protein antigens of any of the above. Further bacterial antigens may also include an outer membrane vesicle (OMV) preparation. Additionally, antigens include live, attenuated, split, and/or purified versions of any of the aforementioned bacteria. The bacterial or microbial derived antigens of the present invention may be gram-negative or gram-positive and aerobic or anaerobic.
  • Additionally, any of the above bacterial-derived saccharides (polysaccharides, LPS, LOS or oligosaccharides) can be conjugated to another agent or antigen, such as a carrier protein (for example CRM197). Such conjugation may be direct conjugation effected by reductive amination of carbonyl moieties on the saccharide to amino groups on the protein, as provided in U.S. Pat. No. 5,360,897 and Can J Biochem Cell Biol. 1984 May;62(5):270-5. Alternatively, the saccharides can be conjugated through a linker, such as, with succinamide or other linkages provided in Bioconjugate Techniques, 1996 and CRC, Chemistry of Protein Conjugation and Cross-Linking, 1993.
  • Viral Antigens
  • Influenza: including whole viral particles (attenuated), split, or subunit comprising hemagglutinin (HA) and/or neuraminidase (NA) surface proteins, the influenza antigens may be derived from chicken embryos or propogated on cell culture, and/or the influenza antigens are derived from influenza type A, B, and/or C, among others;
  • Respiratory syncytial virus (RSV): including the F protein of the A2 strain of RSV (J Gen Virol. 2004 November; 85(Pt 11):3229) and/or G glycoprotein;
  • Parainfluenza virus (PIV): including PIV type 1, 2, and 3, preferably containing hemagglutinin, neuraminidase and/or fusion glycoproteins;
  • Poliovirus: including antigens from a family of picornaviridae, preferably poliovirus antigens such as OPV or, preferably IPV;
  • Measles: including split measles virus (MV) antigen optionally combined with the Protollin and or antigens present in MMR vaccine;
  • Mumps: including antigens present in MMR vaccine;
  • Rubella: including antigens present in MMR vaccine as well as other antigens from Togaviridae, including dengue virus;
  • Rabies: such as lyophilized inactivated virus (RabAvert™);
  • Flaviviridae viruses: such as (and antigens derived therefrom) yelow fever virus, Japanese encephalitis virus, dengue virus ( types 1, 2, 3, or 4), tick borne encephalitis virus, and West Nile virus;
  • Caliciviridae; antigens therefrom;
  • HIV: including HIV-1 or HIV-2 strain antigens, such as gag (p24gag and p55gag), env (gp160 and gp41), pol, tat, nef, rev vpu, miniproteins, (preferably p55 gag and gp140v delete) and antigens from the isolates HIVIIb, HIVSF2, HIVLAV, HIVLA1, HUVMN, HIV-1CM235, HIV-1US4, HIV-2; simian immunodeficiency virus (SIV) among others;
  • Rotavirus: including VP4, VP5, VP6, VP7, VP8 proteins (Protein Expr Purif. 2004 December; 38(2):205) and/or NSP4;
  • Pestivirus: such as antigens from classical porcine fever virus, bovine viral diarrhoea virus, and/or border disease virus;
  • Parvovirus: such as parvovirus B19;
  • Coronavirus: including SARS virus antigens, particularly spike protein or proteases therefrom, as well as antigens included in WO 04/92360;
  • Hepatitis A virus: such as inactivated virus;
  • Hepatitis B virus: such as the surface and/or core antigens (sAg), as well as the presurface sequences, pre-S1 and pre-S2 (formerly called pre-S), as well as combinations of the above, such as sAg/pre-S1, sAg/pre-S2, sAg/pre-S1/pre-S2, and pre-S1/pre-S2, (see, e.g., AHBV Vaccines—Human Vaccines and Vaccination, pp. 159-176; and U.S. Pat. Nos. 4,722,840, 5,098,704, 5,324,513; Beames et al., J. Virol. (1995) 69:6833-6838, Birnbaum et al., J. Virol. (1990) 64:3319-3330; and Zhou et al., J. Virol. (1991) 65:5457-5464);
  • Hepatitis C virus: such as E1, E2, E1/E2 (see, Houghton et al., Hepatology (1991) 14:381), NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions (International Publication Nos. WO 89/04669; WO 90/11089; and WO 90/14436);
  • Delta hepatitis virus (HDV): antigens derived therefrom, particularly δ-antigen from HDV (see, e.g., U.S. Pat. No. 5,378,814);
  • Hepatitis E virus (HEV); antigens derived therefrom;
  • Hepatitis G virus (HGV), antigens derived therefrom;
  • Varcicella zoster virus: antigens derived from varicella zoster virus (VZV) (J. Gen. Virol. (1986) 67:1759);
  • Epstein-Barr virus: antigens derived from EBV (Baer et al., Nature (1984) 310:207);
  • Cytomegalovirus: CMV antigens, including gB and gH (Cytomegaloviruses (J. K. McDougall, ed., Springer-Verlag 1990) pp. 125-169);
  • Herpes simplex virus: including antigens from HSV-1 or HSV-2 strains and glycoproteins gB, gD and gH (McGeoch et al., J. Gen. Virol. (1988)69:1531 and U.S. Pat. No. 5,171,568);
  • Human Herpes Virus: antigens derived from other human herpesviruses such as HHV6 and HHV7; and
  • HPV: including antigens associated with or derived from human papillomavirus (HPV), for example, one or more of E1-E7, L1, L2, and fusions thereof, particularly the compositions of the invention may include a virus-like particle (VLP) comprising the L1 major capsid protein, more particular still, the HPV antigens are protective against one or more of HPV serotypes 6, 11, 16 and/or 18.
  • Further provided are antigens, compostions, methods, and microbes included in Vaccines, 4th Edition (Plotkin and Orenstein ed. 2004); Medical Microbiology 4th Edition (Murray et al. ed. 2002); Virology, 3rd Edition (W. K. Joklik ed. 1988); Fundamental Virology, 2nd Edition (B. N. Fields and D. M. Knipe, eds. 1991), which are contemplated in conjunction with the compositions of the present invention.
  • Additionally, antigens include live, attenuated, split, and/or purified versions of any of the aforementioned viruses.
  • Fungal Antigens
  • Fungal antigens for use herein, associated with vaccines include those described in: U.S. Pat. Nos. 4,229,434 and 4,368,191 for prophylaxis and treatment of trichopytosis caused by Trichophyton mentagrophytes; U.S. Pat. Nos. 5,277,904 and 5,284,652 for a broad spectrum dermatophyte vaccine for the prophylaxis of dermatophyte infection in animals, such as guinea pigs, cats, rabbits, horses and lambs, these antigens comprises a suspension of killed T. equinum, T. mentagrophytes (var. granulare), M. canis and/or M gypseum in an effective amount optionally combined with an adjuvant; U.S. Pat. Nos. 5,453,273 and 6,132,733 for a ringworm vaccine comprising an effective amount of a homogenized, formaldehyde-killed fungi, i.e., Microsporum canis culture in a carrier; U.S. Pat. No. 5,948,413 involving extracellular and intracellular proteins for pythiosis. Additional antigens identified within antifungal vaccines include Ringvac bovis LTF-130 and Bioveta.
  • Further, fungal antigens for use herein may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var. discoides, var. ochraceum, Trichophyton violaceum, and/or Trichophyton faviforme.
  • Fungal pathogens for use as antigens or in derivation of antigens in conjunction with the compositions of the present invention comprise Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida parapsilosis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladosporium carrionii, Coccidioides immitis, Blastomyces dermatidis, Cryptococcus neoformans, Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Paracoccidioides brasiliensis, Pneumocystis carinii, Pythiumn insidiosum, Pityrosporum ovale, Sacharomyces cerevisae, Saccharomyces boulardii, Saccharomyces pombe, Scedosporium apiosperum, Sporothrix schenckii, Trichosporon beigelii, Toxoplasma gondii, Penicillium marneffei, Malassezia spp., Fonsecaea spp., Wangiella spp., Sporothrix spp., Basidiobolus spp., Conidiobolus spp., Rhizopus spp, Mucor spp, Absidia spp, Mortierella spp, Cunninghamella spp, and Saksenaea spp.
  • Other fungi from which antigens are derived include Alternaria spp, Curvularia spp, Helminthosporium spp, Fusarium spp, Aspergillus spp, Penicillium spp, Monolinia spp, Rhizoctonia spp, Paecilomyces spp, Pithomyces spp, and Cladosporium spp.
  • Processes for producing a fungal antigens are well known in the art (see U.S. Pat. No. 6,333,164). In a preferred method a solubilized fraction extracted and separated from an insoluble fraction obtainable from fungal cells of which cell wall has been substantially removed or at least partially removed, characterized in that the process comprises the steps of: obtaining living fungal cells; obtaining fungal cells of which cell wall has been substantially removed or at least partially removed; bursting the fungal cells of which cell wall has been substantially removed or at least partially removed; obtaining an insoluble fraction; and extracting and separating a solubilized fraction from the insoluble fraction.
  • STD Antigens
  • In particular embodiments, microbes (bacteria, viruses and/or fungi) against which the present compositions and methods can be implement include those that cause sexually transmitted diseases (STDs) and/or those that display on their surface an antigen that can be the target or antigen composition of the invention. In a preferred embodiment of the invention, compositions are combined with antigens derived from a viral or bacterial STD. Antigens derived from bacteria or viruses can be administered in conjunction with the compositions of the present invention to provide protection against at least one of the following STDs, among others: chlamydia, genital herpes, hepatitis (particularly HCV), genital warts, gonorrhoea, syphilis and/or chancroid (See, WO00/15255).
  • In another embodiment the compositions of the present invention are co-administered with an antigen for the prevention or treatment of an STD.
  • Antigens derived from the following viruses associated with STDs, which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: hepatitis (particularly HCV), HPV, HIV, or HSV.
  • Additionally, antigens derived from the following bacteria associated with STDs, which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: Neiserria gonorrhoeae, Chlamydia pneumoniae, Chlamydia trachomatis, Treponema pallidum, or Haemophilus ducreyi.
  • Respiratory Antigens
  • The antigen may be a respiratory antigen and could further be used in an immunogenic composition for methods of preventing and/or treating infection by a respiratory pathogen, including a virus, bacteria, or fungi such as respiratory syncytial virus (RSV), PIV, SARS virus, influenza, Bacillus anthracis, particularly by reducing or preventing infection and/or one or more symptoms of respiratory virus infection. A composition comprising an antigen described herein, such as one derived from a respiratory virus, bacteria or fungus is administered in conjunction with the compositions of the present invention to an individual which is at risk of being exposed to that particular respiratory microbe, has been exposed to a respiratory microbe or is infected with a respiratory virus, bacteria or fungus. The composition(s) of the present invention is/are preferably co-administered at the same time or in the same formulation with an antigen of the respiratory pathogen. Administration of the composition results in reduced incidence and/or severity of one or more symptoms of respiratory infection.
  • Pediatric/Geriatric Antigens
  • In one embodiment the compositions of the present invention are used in conjunction with an antigen for treatment of a pediatric population, as in a pediatric antigen. In a more particular embodiment the pediatric population is less than about 3 years old, or less than about 2 years, or less than about 1 years old. In another embodiment the pediatric antigen (in conjunction with the composition of the present invention) is administered multiple times over at least 1, 2, or 3 years.
  • In another embodiment the compositions of the present invention are used in conjunction with an antigen for treatment of a geriatric population, as in a geriatric antigen.
  • Other Antigens
  • Other antigens for use in conjunction with the compositions of the present include hospital acquired (nosocomial) associated antigens.
  • In another embodiment, parasitic antigens are contemplated in conjunction with the compositions of the present invention. Examples of parasitic antigens include those derived from organisms causing malaria and/or Lyme disease.
  • In another embodiment, the antigens in conjunction with the compositions of the present invention are associated with or effective against a mosquito bom illness. In another embodiment, the antigens in conjunction with the compositions of the present invention are associated with or effective against encephalitis. In another embodiment the antigens in conjunction with the compositions of the present invention are associated with or effective against an infection of the nervous system.
  • In another embodiment, the antigens in conjunction with the compositions of the present invention are antigens transmissible through blood or body fluids.
  • Antigen Formulations
  • In other aspects of the invention, methods of producing microparticles having adsorbed antigens are provided. The methods comprise: (a) providing an emulsion by dispersing a mixture comprising (i) water, (ii) a detergent, (iii) an organic solvent, and (iv) a biodegradable polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate. The polymer is typically present in the mixture at a concentration of about 1% to about 30% relative to the organic solvent, while the detergent is typically present in the mixture at a weight-to-weight detergent-to-polymer ratio of from about 0.00001:1 to about 0.1:1 (more typically about 0.0001:1 to about 0.1:1, about 0.001:1 to about 0.1:1, or about 0.005:1 to about 0.1:1); (b) removing the organic solvent from the emulsion; and (c) adsorbing an antigen on the surface of the microparticles. In certain embodiments, the biodegradable polymer is present at a concentration of about 3% to about 10% relative to the organic solvent.
  • Microparticles for use herein will be formed from materials that are sterilizable, non-toxic and biodegradable. Such materials include, without limitation, poly(α-hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate. Preferably, microparticles for use with the present invention are derived from a poly(α-hydroxy acid), in particular, from a poly(lactide) (“PLA”) or a copolymer of D,L-lactide and glycolide or glycolic acid, such as a poly(D,L-lactide-co-glycolide) (“PLG” or “PLGA”), or a copolymer of D,L-lactide and caprolactone. The microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of which will be largely a matter of choice, depending in part on the coadministered macromolecule. These parameters are discussed more fully below.
  • Further antigens may also include an outer membrane vesicle (OMV) preparation.
  • Additional formulation methods and antigens (especially tumor antigens) are provided in U.S. patent Ser. No. 09/581,772.
  • Antigen References
  • The following references include antigens useful in conjunction with the compositions of the present invention:
    • 1 International patent application WO99/24578
    • 2 International patent application WO99/36544.
    • 3 International patent application WO99/57280.
    • 4 International patent application WO00/22430.
    • 5 Tettelin et al. (2000) Science 287:1809-1815.
    • 6 International patent application WO96/29412.
    • 7 Pizza et al. (2000) Science 287:1816-1820.
    • 8 PCT WO 01/52885.
    • 9 Bjune et al. (1991) Lancet 338(8775).
    • 10 Fuskasawa et al. (1999) Vaccine 17:2951-2958.
    • 11 Rosenqist et al. (1998) Dev. Biol. Strand 92:323-333.
    • 12 Constantino et al. (1992) Vaccine 10:691-698.
    • 13 Constantino et al. (1999) Vaccine 17:1251-1263.
    • 14 Watson (2000) Pediatr Infect Dis J 19:331-332.
    • 15 Rubin (20000) Pediatr Clin North Am 47:269-285, v.
    • 16 Jedrzejas (2001) Microbiol Mol Biol Rev 65:187-207.
    • 17 International patent application filed on 3rd Jul. 2001 claiming priority from GB-0016363.4; WO 02/02606; PCT IB/01/00166.
    • 18 Kalman et al. (1999) Nature Genetics 21:385-389.
    • 19 Read et al. (2000) Nucleic Acids Res 28:1397-406.
    • 20 Shirai et al. (2000) J. Infect. Dis 181(Suppl 3):S524-S527.
    • 21 International patent application WO99/27105.
    • 22 International patent application WO00/27994.
    • 23 International patent application WO00/37494.
    • 24 International patent application WO99/28475.
    • 25 Bell (2000) Pediatr Infect Dis J 19:1187-1188.
    • 26 Iwarson (1995) APMIS 103:321-326.
    • 27 Gerlich et al. (1990) Vaccine 8 Suppl:S63-68 & 79-80.
    • 28 Hsu et al. (1999) Clin Liver Dis 3:901-915.
    • 29 Gastofsson et al. (1996) N. Engl. J. Med. 334-:349-355.
    • 30 Rappuoli et al. (1991) TIBTECH 9:232-238.
    • 31 Vaccines (1988) eds. Plotkin & Mortimer. ISBN 0-7216-1946-0.
    • 32 Del Guidice et al. (1998) Molecular Aspects of Medicine 19:1-70.
    • 33 International patent application WO93/018150.
    • 34 International patent application WO99/53310.
    • 35 International patent application WO98/04702.
    • 36 Ross et al. (2001) Vaccine 19:135-142.
    • 37 Sutter et al. (2000) Pediatr Clin North Am 47:287-308.
    • 38 Zimmerman & Spann (1999) Am Fan Physician 59:113-118, 125-126.
    • 39 Dreensen (1997) Vaccine 15 Suppl″ S2-6.
    • 40 MMWR Morb Mortal Wkly rep 1998 Jan. 16: 47(1):12, 9.
    • 41 McMichael (2000) Vaccine 19 Suppl 1: S101-107.
    • 42 Schuchat (1999) Lancer 353(9146):51-6.
    • 43 GB patent applications 0026333.5, 0028727.6 & 0105640.7.
    • 44 Dale (1999) Infect Disclin North Am 13:227-43, viii.
    • 45 Ferretti et al. (2001) PNAS USA 98: 4658-4663.
    • 46 Kuroda et al. (2001) Lancet 357(9264):1225-1240; see also pages 1218-1219.
    • 47 Ramsay et al. (2001) Lancet 357(9251): 195-196.
    • 48 Lindberg (1999) Vaccine 17 Suppl 2:S28-36.
    • 49 Buttery & Moxon (2000) J R Coil Physicians Long 34:163-168.
    • 50 Ahmad & Chapnick (1999) Infect Dis Clin North Am 13:113-133, vii.
    • 51 Goldblatt (1998) J. Med. Microbiol. 47:663-567.
    • 52 European patent 0 477 508.
    • 53 U.S. Pat. No. 5,306,492.
    • 54 International patent application WO98/42721.
    • 55 Conjugate Vaccines (eds. Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114.
    • 56 Hermanson (1996) Bioconjugate Techniques ISBN: 012323368 & 012342335X.
    • 57 European patent application 0372501.
    • 58 European patent application 0378881.
    • 59 European patent application 0427347.
    • 60 International patent application WO93/17712.
    • 61 International patent application WO98/58668.
    • 62 European patent application 0471177.
    • 63 International patent application WO00/56360.
    • 64 International patent application WO00/67161.
  • The contents of all of the above cited patents, patent applications and journal articles are incorporated by reference as if set forth fully herein.
  • There may be an upper limit to the number of Gram positive bacterial proteins which will be in the compositions of the invention. Preferably, the number of Gram positive bacterial proteins in a composition of the invention is less than 20, less than 19, less than 18, less than 17, less than 16, less than 15, less than 14, less than 13, less than 12, less than 11, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, or less than 3. Still more preferably, the number of Gram positive bacterial proteins in a composition of the invention is less than 6, less than 5, or less than 4. Still more preferably, the number of Gram positive bacterial proteins in a composition of the invention is 3.
  • The Gram positive bacterial proteins and polynucleotides used in the invention are preferably isolated, i.e., separate and discrete, from the whole organism with which the molecule is found in nature or, when the polynucleotide or polypeptide is not found in nature, is sufficiently free of other biological macromolecules so that the polynucleotide or polypeptide can be used for its intended purpose.
  • Fusion Proteins: GBS AI Sequences
  • The GBS AI proteins used in the invention may be present in the composition as individual separate polypeptides, but it is preferred that at least two (i.e. 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18) of the antigens are expressed as a single polypeptide chain (a “hybrid” or “fusion” polypeptide). Such fusion polypeptides offer two principal advantages: first, a polypeptide that may be unstable or poorly expressed on its own can be assisted by adding a suitable fusion partner that overcomes the problem; second, commercial manufacture is simplified as only one expression and purification need be employed in order to produce two polypeptides which are both antigenically useful.
  • The fusion polypeptide may comprise one or more AI polypeptide sequences. Preferably, the fusion comprises an AI surface protein sequence. Preferably, the fusion polypeptide includes one or more of GBS 80, GBS 104, and GBS 67. Most preferably, the fusion peptide includes a polypeptide sequence from GBS 80. Accordingly, the invention includes a fusion peptide comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a GBS AI surface protein or a fragment thereof. Preferably, the first and second amino acid sequences in the fusion polypeptide comprise different epitopes.
  • Hybrids (or fusions) consisting of amino acid sequences from two, three, four, five, six, seven, eight, nine, or ten GBS antigens are preferred. In particular, hybrids consisting of amino acid sequences from two, three, four, or five GBS antigens are preferred.
  • Different hybrid polypeptides may be mixed together in a single formulation. Within such combinations, a GBS antigen may be present in more than one hybrid polypeptide and/or as a non-hybrid polypeptide. It is preferred, however, that an antigen is present either as a hybrid or as a non-hybrid, but not as both.
  • Hybrid polypeptides can be represented by the formula NH2-A-{-X-L-}n-B—COOH, wherein: X is an amino acid sequence of a GBS AI protein or a fragment thereof; L is an optional linker amino acid sequence; A is an optional N-terminal amino acid sequence; B is an optional C-terminal amino acid sequence; and n is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • If a —X— moiety has a leader peptide sequence in its wild-type form, this may be included or omitted in the hybrid protein. In some embodiments, the leader peptides will be deleted except for that of the —X— moiety located at the N-terminus of the hybrid protein i.e. the leader peptide of X1 will be retained, but the leader peptides of X2 . . . Xn will be omitted. This is equivalent to deleting all leader peptides and using the leader peptide of X1 as moiety -A-.
  • For each n instances of {-X-L-}, linker amino acid sequence -L- may be present or absent. For instance, when n=2 the hybrid may be NH2—X1-L1-X2-L2-COOH, NH2—X1—X2—COOH, NH2—X1-L1-X2—COOH, NH2—X1—X2-L2-COOH, etc. Linker amino acid sequence(s) -L- will typically be short (e.g. 20 or fewer amino acids i.e. 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples comprise short peptide sequences which facilitate cloning, poly-glycine linkers (i.e. comprising Glyn where n=2, 3, 4, 5, 6, 7, 8, 9, 10 or more), and histidine tags (i.e. Hisn where n=3, 4, 5, 6, 7, 8, 9, 10 or more). Other suitable linker amino acid sequences will be apparent to those skilled in the art. A useful linker is GSGGGG, with the Gly-Ser dipeptide being formed from a BamHI restriction site, thus aiding cloning and manipulation, and the (Gly)4 tetrapeptide being a typical poly-glycine linker.
  • -A- is an optional N-terminal amino acid sequence. This will typically be short (e.g. 40 or fewer amino acids i.e. 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples include leader sequences to direct protein trafficking, or short peptide sequences which facilitate cloning or purification (e.g. histidine tags i.e. Hisn where n=3, 4, 5, 6, 7, 8, 9, 10 or more). Other suitable N-terminal amino acid sequences will be apparent to those skilled in the art. If X1 lacks its own N-terminus methionine, -A- is preferably an oligopeptide (e.g. with 1, 2, 3, 4, 5, 6, 7 or 8 amino acids) which provides a N-terminus methionine.
  • —B— is an optional C-terminal amino acid sequence. This will typically be short (e.g. 40 or fewer amino acids i.e. 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples include sequences to direct protein trafficking, short peptide sequences which facilitate cloning or purification (e.g. comprising histidine tags i.e. Hisn, where n=3, 4, 5, 6, 7, 8, 9, 10 or more), or sequences which enhance protein stability. Other suitable C-terminal amino acid sequences will be apparent to those skilled in the art.
  • Most preferably, n is 2 or 3.
  • Fusion Proteins: Gram Positive Bacteria AI Sequences
  • The Gram positive bacteria AI proteins used in the invention may be present in the composition as individual separate polypeptides, but it is preferred that at least two (i.e. 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18) of the antigens are expressed as a single polypeptide chain (a “hybrid” or “fusion” polypeptide). Such fusion polypeptides offer two principal advantages: first, a polypeptide that may be unstable or poorly expressed on its own can be assisted by adding a suitable fusion partner that overcomes the problem; second, commercial manufacture is simplified as only one expression and purification need be employed in order to produce two polypeptides which are both antigenically useful.
  • The fusion polypeptide may comprise one or more AI polypeptide sequences. Preferably, the fusion comprises an AI surface protein sequence. Accordingly, the invention includes a fusion peptide comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a Gram positive bacteria AI protein or a fragment thereof. Preferably, the first and second amino acid sequences in the fusion polypeptide comprise different epitopes.
  • Hybrids (or fusions) consisting of amino acid sequences from two, three, four, five, six, seven, eight, nine, or ten Gram positive bacteria antigens are preferred. In particular, hybrids consisting of amino acid sequences from two, three, four, or five Gram positive bacteria antigens are preferred.
  • Different hybrid polypeptides may be mixed together in a single formulation. Within such combinations, a Gram positive bacteria AI sequence may be present in more than one hybrid polypeptide and/or as a non-hybrid polypeptide. It is preferred, however, that an antigen is present either as a hybrid or as a non-hybrid, but not as both.
  • Hybrid polypeptides can be represented by the formula NH2-A-{-X-L-}n-B—COOH, wherein: X is an amino acid sequence of a Gram positive bacteria AI sequence or a fragment thereof; L is an optional linker amino acid sequence; A is an optional N-terminal amino acid sequence; B is an optional C-terminal amino acid sequence; and n is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • If a —X— moiety has a leader peptide sequence in its wild-type form, this may be included or omitted in the hybrid protein. In some embodiments, the leader peptides will be deleted except for that of the —X— moiety located at the N-terminus of the hybrid protein i.e. the leader peptide of X1 will be retained, but the leader peptides of X2 . . . Xn will be omitted. This is equivalent to deleting all leader peptides and using the leader peptide of X1 as moiety -A-.
  • For each n instances of {—X-L-}, linker amino acid sequence -L- may be present or absent. For instance, when n=2 the hybrid may be NH2—X1-L1-X2-L2-COOH, NH2—X1—X2—COOH, NH2—X1-L1-X2—COOH, NH2—X1—X2-L2-COOH, etc. Linker amino acid sequence(s) -L- will typically be short (e.g. 20 or fewer amino acids i.e. 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples comprise short peptide sequences which facilitate cloning, poly-glycine linkers (i.e. comprising Glyn, where n=2, 3, 4, 5, 6, 7, 8, 9, 10 or more), and histidine tags (i.e. Hisn where n=3, 4, 5, 6, 7, 8, 9, 10 or more). Other suitable linker amino acid sequences will be apparent to those skilled in the art. A useful linker is GSGGGG, with the Gly-Ser dipeptide being formed from a BamHI restriction site, thus aiding cloning and manipulation, and the (Gly)4 tetrapeptide being a typical poly-glycine linker.
  • -A- is an optional N-terminal amino acid sequence. This will typically be short (e.g. 40 or fewer amino acids i.e. 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples include leader sequences to direct protein trafficking, or short peptide sequences which facilitate cloning or purification (e.g. histidine tags i.e. His, where n=3, 4, 5, 6, 7, 8, 9, 10 or more). Other suitable N-terminal amino acid sequences will be apparent to those skilled in the art. If X1 lacks its own N-terminus methionine, -A- is preferably an oligopeptide (e.g. with 1, 2, 3, 4, 5, 6, 7 or 8 amino acids) which provides a N-terminus methionine.
  • —B— is an optional C-terminal amino acid sequence. This will typically be short (e.g. 40 or fewer amino acids i.e. 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1). Examples include sequences to direct protein trafficking, short peptide sequences which facilitate cloning or purification (e.g. comprising histidine tags i.e. Hisn, where n=3, 4, 5, 6, 7, 8, 9, 10 or more), or sequences which enhance protein stability. Other suitable C-terminal amino acid sequences will be apparent to those skilled in the art.
  • Most preferably, n is 2 or 3.
  • Antibodies: GBS AI Sequences
  • The GBS AI proteins of the invention may also be used to prepare antibodies specific to the GBS AI proteins. The antibodies are preferably specific to the an oligomeric or hyper-oligomeric form of an AI protein. The invention also includes combinations of antibodies specific to GBS AI proteins selected to provide protection against an increased range of GBS serotypes and strain isolates. For example, a combination may comprise a first and second antibody, wherein said first antibody is specific to a first GBS AI protein and said second antibody is specific to a second GBS AI protein. Preferably, the nucleic acid sequence encoding said first GBS AI protein is not present in a GBS genome comprising a polynucleotide sequence encoding for said second GBS AI protein. Preferably, the nucleic acid sequence encoding said first and second GBS AI proteins are present in the genomes of multiple GBS serotypes and strain isolates.
  • The GBS specific antibodies of the invention include one or more biological moieties that, through chemical or physical means, can bind to or associate with an epitope of a GBS polypeptide. The antibodies of the invention include antibodies which specifically bind to a GBS AI protein. The invention includes antibodies obtained from both polyclonal and monoclonal preparations, as well as the following: hybrid (chimeric) antibody molecules (see, for example, Winter et al. (1991) Nature 349: 293-299; and U.S. Pat. No. 4,816,567; F(ab′)2 and F(ab) fragments; Fv molecules (non-covalent heterodimers, see, for example, Inbar et al. (1972) Proc Natl Acad Sci USA 69:2659-2662; and Ehrlich et al. (1980) Biochem 19:4091-4096); single-chain Fv molecules (sFv) (see, for example, Huston et al. (1988) Proc Natl Acad Sci USA 85:5897-5883); dimeric and trimeric antibody fragment constructs; minibodies (see, e.g., Pack et al. (1992) Biochem 31:1579-1584; Cumber et al. (1992) J Immunology 149B: 120-126); humanized antibody molecules (see, for example, Riechmann et al. (1988) Nature 332:323-327; Verhoeyan et al. (1988) Science 239:1534-1536; and U.K. Patent Publication No. GB 2,276,169, published 21 Sep. 1994); and, any functional fragments obtained from such molecules, wherein such fragments retain immunological binding properties of the parent antibody molecule. The invention further includes antibodies obtained through non-conventional processes, such as phage display.
  • Preferably, the GBS specific antibodies of the invention are monoclonal antibodies. Monoclonal antibodies of the invention include an antibody composition having a homogeneous antibody population. Monoclonal antibodies of the invention may be obtained from murine hybridomas, as well as human monoclonal antibodies obtained using human rather than murine hybridomas. See, e.g., Cote, et al. Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, 1985, p 77.
  • The antibodies of the invention may be used in diagnostic applications, for example, to detect the presence or absence of GBS in a biological sample. The antibodies of the invention may also be used in the prophylactic or therapeutic treatment of GBS infection.
  • Antibodies: Gram Positive Bacteria AI Sequences
  • The Gram positive bacteria AI proteins of the invention may also be used to prepare antibodies specific to the Gram positive bacteria AI proteins. The antibodies are preferably specific to the an oligomeric or hyper-oligomeric form of an AI protein. The invention also includes combinations of antibodies specific to Gram positive bacteria AI proteins selected to provide protection against an increased range of Gram positive bacteria genus, species, serotypes and strain isolates.
  • For example, a combination may comprise a first and second antibody, wherein said first antibody is specific to a first Gram positive bacteria AI protein and said second antibody is specific to a second Gram positive bacteria AI protein. Preferably, the nucleic acid sequence encoding said first Gram positive bacteria AI protein is not present in a Gram positive bacterial genome comprising a polynucleotide sequence encoding for said second Gram positive bacteria AI protein. Preferably, the nucleic acid sequence encoding said first and second Gram positive bacteria AI proteins are present in the genomes of multiple Gram positive bacteria genus, species, serotypes or strain isolates.
  • As an example of an instance where the combination of antibodies provides protection against an increased range of bacteria serotypes, the first antibody may be specific to a first GAS AI protein and the second antibody may be specific to a second GAS AI protein. The first GAS AI protein may comprise a GAS AI-1 surface protein, while the second GAS AI protein may comprise a GAS AI-2 or AI-3 surface protein.
  • As an example of an instance where the combination of antibodies provides protection against an increased range of bacterial species, the first antibody may be specific to a GBS AI protein and the second antibody may be specific to a GAS AI protein. Alternatively, the first antibody may be specific to a GAS AI protein and the second antibody may be specific to a S. pneumoniae AI protein.
  • The Gram positive specific antibodies of the invention include one or more biological moieties that, through chemical or physical means, can bind to or associate with an epitope of a Gram positive bacteria AI polypeptide. The antibodies of the invention include antibodies which specifically bind to a Gram positive bacteria AI protein. The invention includes antibodies obtained from both polyclonal and monoclonal preparations, as well as the following: hybrid (chimeric) antibody molecules (see, for example, Winter et al. (1991) Nature 349: 293-299; and U.S. Pat. No. 4,816,567; F(ab′)2 and F(ab) fragments; Fv molecules (non-covalent heterodimers, see, for example, Inbar et al. (1972) Proc Natl Acad Sci USA 69:2659-2662; and Ehrlich et al. (1980) Biochem 19:40914096); single-chain Fv molecules (sFv) (see, for example, Huston et al. (1988) Proc Natl Acad Sci USA 85:5897-5883); dimeric and trimeric antibody fragment constructs; minibodies (see, e.g., Pack et al. (1992) Biochem 31:1579-1584; Cumber et al. (1992) J Immunology 149B: 120-126); humanized antibody molecules (see, for example, Riechmann et al. (1988) Nature 332:323-327; Verhoeyan et al. (1988) Science 239:1534-1536; and U.K. Patent Publication No. GB 2,276,169, published 21 Sep. 1994); and, any functional fragments obtained from such molecules, wherein such fragments retain immunological binding properties of the parent antibody molecule. The invention further includes antibodies obtained through non-conventional processes, such as phage display.
  • Preferably, the Gram positive specific antibodies of the invention are monoclonal antibodies. Monoclonal antibodies of the invention include an antibody composition having a homogeneous antibody population. Monoclonal antibodies of the invention may be obtained from murine hybridomas, as well as human monoclonal antibodies obtained using human rather than murine hybridomas. See, e.g., Cote, et al. Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, 1985, p 77.
  • The antibodies of the invention may be used in diagnostic applications, for example, to detect the presence or absence of Gram positive bacteria in a biological sample. The antibodies of the invention may also be used in the prophylactic or therapeutic treatment of Gram positive bacteria infection.
  • Nucleic Acids
  • The invention provides nucleic acids encoding the Gram positive bacteria sequences and/or the hybrid fusion polypeptides of the invention. The invention also provides nucleic acid encoding the GBS antigens and/or the hybrid fusion polypeptides of the invention. Furthermore, the invention provides nucleic acid which can hybridise to these nucleic acids, preferably under “high stringency” conditions (e.g. 65° C. in a 0.1×SSC, 0.5% SDS solution).
  • Polypeptides of the invention can be prepared by various means (e.g. recombinant expression, purification from cell culture, chemical synthesis, etc.) and in various forms (e.g. native, fusions, non-glycosylated, lipidated, etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other GAS or host cell proteins).
  • Nucleic acid according to the invention can be prepared in many ways (e.g. by chemical synthesis, from genomic or cDNA libraries, from the organism itself, etc.) and can take various forms (e.g. single stranded, double stranded, vectors, probes, etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other GBS or host cell nucleic acids).
  • The term “nucleic acid” includes DNA and RNA, and also their analogues, such as those containing modified backbones (e.g. phosphorothioates, etc.), and also peptide nucleic acids (PNA), etc. The invention includes nucleic acid comprising sequences complementary to those described above (e.g. for antisense or probing purposes).
  • The invention also provides a process for producing a polypeptide of the invention, comprising the step of culturing a host cell transformed with nucleic acid of the invention under conditions which induce polypeptide expression.
  • The invention provides a process for producing a polypeptide of the invention, comprising the step of synthesising at least part of the polypeptide by chemical means.
  • The invention provides a process for producing nucleic acid of the invention, comprising the step of amplifying nucleic acid using a primer-based amplification method (e.g. PCR).
  • The invention provides a process for producing nucleic acid of the invention, comprising the step of synthesising at least part of the nucleic acid by chemical means.
  • Purification and Recombinant Expression
  • The Gram positive bacteria AI proteins of the invention may be isolated from the native Gram positive bacteria, or they may be recombinantly produced, for instance in a heterologous host. For example, the GAS, GBS, and S. pneumoniae antigens of the invention may be isolated from Streptococcus agalactiae, S. pyogenes, S. pneumoniae, or they may be recombinantly produced, for instance, in a heterologous host. Preferably, the GBS antigens are prepared using a heterologous host.
  • The heterologous host may be prokaryotic (e.g. a bacterium) or eukaryotic. It is preferably E. coli, but other suitable hosts include Bacillus subtilis, Vibrio cholerae, Salmonella typhi, Salmonella typhimurium, Neisseria lactamica, Neisseria cinerea, Mycobacteria (e.g. M. tuberculosis), S. gordonii, L. lactis, yeasts, etc.
  • Recombinant production of polypeptides is facilitated by adding a tag protein to the Gram positive bacteria AI sequence to be expressed as a fusion protein comprising the tag protein and the Gram positive bacteria antigen. For example, recombinant production of polypeptides is facilitated by adding a tag protein to the GBS antigen to be expressed as a fusion protein comprising the tag protein and the GBS antigen. Such tag proteins can facilitate purification, detection and stability of the expressed protein. Tag proteins suitable for use in the invention include a polyarginine tag (Arg-tag), polyhistidine tag (His-tag), FLAG-tag, Strep-tag, c-myc-tag, S-tag, calmodulin-binding peptide, cellulose-binding domain, SBP-tag, chitin-binding domain, glutathione S-transferase-tag (GST), maltose-binding protein, transcription termination anti-terminiantion factor (NusA), E. coli thioredoxin (TrxA) and protein disulfide isomerase I (DsbA). Preferred tag proteins include His-tag and GST. A full discussion on the use of tag proteins can be found at Terpe et al., “Overview of tag protein fusions: from molecular and biochemical fundamentals to commercial systems”, Appl Microbiol Biotechnol (2003) 60:523-533.
  • After purification, the tag proteins may optionally be removed from the expressed fusion protein, i.e., by specifically tailored enzymatic treatments known in the art. Commonly used proteases include enterokinase, tobacco etch virus (TEV), thrombin, and factor Xa.
  • GBS Polysaccharides
  • The compositions of the invention may be further improved by including GBS polysaccharides. Preferably, the GBS antigen and the saccharide each contribute to the immunological response in a recipient. The combination is particularly advantageous where the saccharide and polypeptide provide protection from different GBS serotypes.
  • The combined antigens may be present as a simple combination where separate saccharide and polypeptide antigens are administered together, or they may be present as a conjugated combination, where the saccharide and polypeptide antigens are covalently linked to each other.
  • Thus the invention provides an immunogenic composition comprising (i) one or more GBS AI proteins and (ii) one or more GBS saccharide antigens. The polypeptide and the polysaccharide may advantageously be covalently linked to each other to form a conjugate.
  • Between them, the combined polypeptide and saccharide antigens preferably cover (or provide protection from) two or more GBS serotypes (e.g. 2, 3, 4, 5, 6, 7, 8 or more serotypes). The serotypes of the polypeptide and saccharide antigens may or may not overlap. For example, the polypeptide might protect against serogroup II or V, while the saccharide protects against either serogroups Ia, Ib, or III. Preferred combinations protect against the following groups of serotypes: (1) serotypes Ia and Ib, (2) serotypes Ia and H, (3) serotypes Ia and III, (4) serotypes Ia and IV, (5) serotypes Ia and V, (6) serotypes Ia and VI, (7) serotypes Ia and VII, (8) serotypes Ia and VIII, (9) serotypes Ib and II, (10) serotypes Ib and III, (11) serotypes Ib and IV, (12) serotypes Ib and V, (13) serotypes Ib and VI, (14) serotypes Ib and VII, (15) serotypes Ib and VIII, 16) serotypes II and III, (17) serotypes II and IV, (18) serotypes II and V, (19) serotypes II and VI, (20) serotypes H and VII, (21) serotypes II and VII, (22) serotypes III and IV, (23) serotypes III and V, (24) serotypes III and VI, (25) serotypes III and VII, (26) serotypes III and VIII, (27) serotypes IV and V, (28) serotypes IV and VI, (29) serotypes IV and VII, (30) serotypes IV and VIII, (31) serotypes V and VI, (32) serotypes V and VII, (33) serotypes V and VIII, (34) serotypes VI and VII, (35) serotypes VI and VIII, and (36) serotypes VII and VIII.
  • Still more preferably, the combinations protect against the following groups of serotypes: (1) serotypes Ia and II, (2) serotypes Ia and V; (3) serotypes Ib and II, (4) serotypes Ib and V, (5) serotypes III and II, and (6) serotypes III and V. Most preferably, the combinations protect against serotypes III and V.
  • Protection against serotypes II and V is preferably provided by polypeptide antigens. Protection against serotypes Ia, Ib and/or III may be polypeptide or saccharide antigens.
  • Immunogenic Compositions and Medicaments
  • Compositions of the invention are preferably immunogenic compositions, and are more preferably vaccine compositions. The pH of the composition is preferably between 6 and 8, preferably about 7. The pH may be maintained by the use of a buffer. The composition may be sterile and/or pyrogen-free. The composition may be isotonic with respect to humans.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat infection), but will typically be prophylactic. Accordingly, the invention includes a method for the therapeutic or prophylactic treatment of a Gram positive bacteria infection in an animal susceptible to such gram positive bacterial infection comprising administering to said animal a therapeutic or prophylactic amount of the immunogenic composition of the invention. For example, the invention includes a method for the therapeutic or prophylactic treatment of a Streptococcus agalactiae, S. pyogenes, or S. pneumoniae infection in an animal susceptible to streptococcal infection comprising administering to said animal a therapeutic or prophylactic amount of the immunogenic compositions of the invention.
  • The invention also provides a composition of the invention for use of the compositions described herein as a medicament. The medicament is preferably able to raise an immune response in a mammal (i.e. it is an immunogenic composition) and is more preferably a vaccine.
  • The invention also provides the use of the compositions of the invention in the manufacture of a medicament for raising an immune response in a mammal. The medicament is preferably a vaccine.
  • The invention also provides kits comprising one or more containers of compositions of the invention. Compositions can be in liquid form or can be lyophilized, as can individual antigens. Suitable containers for the compositions include, for example, bottles, vials, syringes, and test tubes. Containers can be formed from a variety of materials, including glass or plastic. A container may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The composition may comprise a first component comprising one or more Gram positive bacteria AI proteins. Preferably, the AI proteins are surface AI proteins. Preferably, the AI surface proteins are in an oligomeric or hyperoligomeric form. For example, the first component comprises a combination of GBS antigens or GAS antigens, or S. pneumoniae antigens. Preferably said combination includes GBS 80. Preferably GBS 80 is present in an oligomeric or hyperoligomeric form.
  • The kit can further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, or dextrose solution. It can also contain other materials useful to the end-user, including other buffers, diluents, filters, needles, and syringes. The kit can also comprise a second or third container with another active agent, for example an antibiotic.
  • The kit can also comprise a package insert containing written instructions for methods of inducing immunity against S agalactiae and/or S. pyogenes and/or S pneumoniae or for treating S agalactiae and/or S. pyogenes and/or S pneumoniae infections. The package insert can be an unapproved draft package insert or can be a package insert approved by the Food and Drug Administration (FDA) or other regulatory body.
  • The invention also provides a delivery device pre-filled with the immunogenic compositions of the invention.
  • The invention also provides a method for raising an immune response in a mammal comprising the step of administering an effective amount of a composition of the invention. The immune response is preferably protective and preferably involves antibodies and/or cell-mediated immunity. This immune response will preferably induce long lasting (e.g., neutralising) antibodies and a cell mediated immunity that can quickly respond upon exposure to one or more GBS and/or GAS and/or S. pneumoniae antigens. The method may raise a booster response.
  • The invention provides a method of neutralizing GBS, GAS, or S. pneumoniae infection in a mammal comprising the step of administering to the mammal an effective amount of the immunogenic compositions of the invention, a vaccine of the invention, or antibodies which recognize an immunogenic composition of the invention.
  • The mammal is preferably a human. Where the vaccine is for prophylactic use, the human is preferably a female (either of child bearing age or a teenager). Alternatively, the human may be elderly (e.g., over the age of 50, 55, 60, 65, 70 or 75) and may have an underlying disease such as diabetes or cancer. Where the vaccine is for therapeutic use, the human is preferably a pregnant female or an elderly adult.
  • These uses and methods are preferably for the prevention and/or treatment of a disease caused by Streptococcus agalactiae, or S. pyogenes, or S. pneumoniae. The compositions may also be effective against other streptococcal bacteria. The compositions may also be effective against other Gram positive bacteria.
  • One way of checking efficacy of therapeutic treatment involves monitoring Gram positive bacterial infection after administration of the composition of the invention. One way of checking efficacy of prophylactic treatment involves monitoring immune responses against the Gram positive bacterial antigens in the compositions of the invention after administration of the composition.
  • One way of checking efficacy of therapeutic treatment involves monitoring GBS infection after administration of the composition of the invention. One way of checking efficacy of prophylactic treatment involves monitoring immune responses against the GBS antigens in the compositions of the invention after administration of the composition.
  • A way of assessing the immunogenicity of the component proteins of the immunogenic compositions of the present invention is to express the proteins recombinantly and to screen patient sera or mucosal secretions by immunoblot. A positive reaction between the protein and the patient serum indicates that the patient has previously mounted an immune response to the protein in question—that is, the protein is an immunogen. This method may also be used to identify immunodominant proteins and/or epitopes.
  • Another way of checking efficacy of therapeutic treatment involves monitoring GBS or GAS or S pneumoniae infection after administration of the compositions of the invention. One way of checking efficacy of prophylactic treatment involves monitoring immune responses both systemically (such as monitoring the level of IgG1 and IgG2a production) and mucosally (such as monitoring the level of IgA production) against the GBS and/or GAS and/or S pneumoniae antigens in the compositions of the invention after administration of the composition. Typically, GBS and/or GAS and/or S pneumoniae serum specific antibody responses are determined post-immunization but pre-challenge whereas mucosal GBS and/or GAS and/or S pneumoniae specific antibody body responses are determined post-immunization and post-challenge.
  • The vaccine compositions of the present invention can be evaluated in in vitro and in vivo animal models prior to host, e.g., human, administration.
  • The efficacy of immunogenic compositions of the invention can also be determined in vivo by challenging animal models of GBS and/or GAS and/or S pneumoniae infection, e.g., guinea pigs or mice, with the immunogenic compositions. The immunogenic compositions may or may not be derived from the same serotypes as the challenge serotypes. Preferably the immunnogenic compositions are derivable from the same serotypes as the challenge serotypes. More preferably, the immunogenic composition and/or the challenge serotypes are derivable from the group of GBS and/or GAS and/or S pneumoniae serotypes.
  • In vivo efficacy models include but are not limited to: (i) A murine infection model using human GBS and/or GAS and/or S pneumoniae serotypes; (ii) a murine disease model which is a murine model using a mouse-adapted GBS and/or GAS and/or S pneumoniae strain, such as those strains outlined above which is particularly virulent in mice and (iii) a primate model using human GBS or GAS or S pneumoniae isolates.
  • The immune response may be one or both of a TH1 immune response and a TH2 response.
  • The immune response may be an improved or an enhanced or an altered immune response.
  • The immune response may be one or both of a systemic and a mucosal immune response.
  • Preferably the immune response is an enhanced system and/or mucosal response.
  • An enhanced systemic and/or mucosal immunity is reflected in an enhanced TH1 and/or TH2 immune response. Preferably, the enhanced immune response includes an increase in the production of IgG1 and/or IgG2a and/or IgA Preferably the mucosal immune response is a TH2 immune response. Preferably, the mucosal immune response includes an increase in the production of IgA.
  • Activated TH2 cells enhance antibody production and are therefore of value in responding to extracellular infections. Activated TH2 cells may secrete one or more of IL-4, IL-5, IL-6, and IL-10. A TH2 immune response may result in the production of IgG1, IgE, IgA and memory B cells for future protection.
  • A TH2 immune response may include one or more of an increase in one or more of the cytokines associated with a TH2 immune response (such as IL-4, IL-5, IL-6 and IL-10), or an increase in the production of IgG1, IgE, IgA and memory B cells. Preferably, the enhanced TH2 immune resonse will include an increase in IgG1 production.
  • A TH1 immune response may include one or more of an increase in CTLs, an increase in one or more of the cytokines associated with a TH1 immune response (such as IL-2, IFNγ, and TNFβ, an increase in activated macrophages, an increase in NK activity, or an increase in the production of IgG2a. Preferably, the enhanced TH1 immune response will include an increase in IgG2a production.
  • Immunogenic compositions of the invention, in particular, immunogenic composition comprising one or more GAS antigens of the present invention may be used either alone or in combination with other GAS antigens optionally with an immunoregulatory agent capable of eliciting a Th1 and/or Th2 response.
  • Compositions of the invention will generally be administered directly to a patient. Certain routes may be favored for certain compositons, as resulting in the generation of a more effective immune response, preferably a CMI response, or as being less likely to induce side effects, or as being easier for administration. Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intradermally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (e.g. see WO 99/27961) or transcutaneous (e.g. see WO 02/074244 and WO 02/064162), intranasal (e.g. see WO03/028760), ocular, aural, pulmonary or other mucosal administration.
  • The invention may be used to elicit systemic and/or mucosal immunity.
  • In one particularly preferred embodiment, the immunogenic composition comprises one or more GBS or GAS or S pneumoniae antigen(s) which elicits a neutralising antibody response and one or more GBS or GAS or S pneumoniae antigen(s) which elicit a cell mediated immune response. In this way, the neutralising antibody response prevents or inhibits an initial GBS or GAS or S pneumoniae infection while the cell-mediated immune response capable of eliciting an enhanced Th1 cellular response prevents further spreading of the GBS or GAS or S pneumoniae infection. Preferably, the immunogenic composition comprises one or more GBS or GAS or S pneumoniae surface antigens and one or more GBS or GAS or S pneumoniae cytoplasmic antigens. Preferably the immunogenic composition comprises one or more GBS or GAS or S pneumoniae surface antigens or the like and one or other antigens, such as a cytoplasmic antigen capable of eliciting a Th1 cellular response.
  • Dosage treatment can be a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. In a multiple dose schedule the various doses may be given by the same or different routes e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc.
  • The compositions of the invention may be prepared in various forms. For example, the compositions may be 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 (e.g. a lyophilised composition). The composition may be prepared for topical administration e.g. as an ointment, cream or powder. The composition may be prepared for oral administration e.g. as a tablet or capsule, as a spray, or as a syrup (optionally flavoured). The composition may be prepared for pulmonary administration e.g. as an inhaler, using a fine powder or a spray. The composition may be prepared as a suppository or pessary. The composition may be prepared for nasal, aural or ocular administration e.g. as drops. The composition may be in kit form, designed such that a combined composition is reconstituted just prior to administration to a patient. Such kits may comprise one or more antigens in liquid form and one or more lyophilised antigens.
  • Immunogenic compositions used as vaccines comprise an immunologically effective amount of antigen(s), as well as any other components, such as antibiotics, as needed. By ‘immunologically effective amount’, it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention, or increases a measurable immune response or prevents or reduces a clinical symptom. This amount varies depending upon the health and physical condition of the individual to be treated, age, the taxonomic group of individual to be treated (e.g. non-human primate, primate, etc.), the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • Further Components of the Composition
  • The composition of the invention will typically, in addition to the components mentioned above, comprise one or more ‘pharmaceutically acceptable carriers’, which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Suitable carriers are typically large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes). Such carriers are well known to those of ordinary skill in the art. The vaccines may also contain diluents, such as water, saline, glycerol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present. A thorough discussion of pharmaceutically acceptable excipients is available in Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th ed., ISBN: 0683306472.
  • Adjuvants
  • Vaccines of the invention may be administered in conjunction with other immunoregulatory agents. In particular, compositions will usually include an adjuvant. Adjuvants for use with the invention include, but are not limited to, one or more of the following set forth below:
  • A. Mineral Containing Compositions
  • Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminum salts and calcium salts. The invention includes mineral salts such as hydroxides (e.g. oxyhydroxides), phosphates (e.g. hydroxyphosphates, orthophosphates), sulfates, etc. (e.g. see chapters 8 & 9 of Vaccine Design . . . (1995) eds. Powell & Newman. ISBN: 030644867X. Plenum.), or mixtures of different mineral compounds (e.g. a mixture of a phosphate and a hydroxide adjuvant, optionally with an excess of the phosphate), with the compounds taking any suitable form (e.g. gel, crystalline, amorphous, etc.), and with adsorption to the salt(s) being preferred. The mineral containing compositions may also be formulated as a particle of metal salt (WO 00/23105).
  • Aluminum salts may be included in vaccines of the invention such that the dose of Al3+ is between 0.2 and 1.0 mg per dose.
  • B. Oil-Emulsions
  • Oil-emulsion compositions suitable for use as adjuvants in the invention include squalene-water emulsions, such as MF59 (5% Squalene, 0.5% Tween 80, and 0.5% Span 85, formulated into submicron particles using a microfluidizer). See WO90/14837. See also, Podda, “The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine”, Vaccine (2001) 19: 2673-2680; Frey et al., “Comparison of the safety, tolerability, and immunogenicity of a MF59-adjuvanted influenza vaccine and a non-adjuvanted influenza vaccine in non-elderly adults”, Vaccine (2003) 21:42344237. MF59 is used as the adjuvant in the FLUAD™ influenza virus trivalent subunit vaccine.
  • Particularly preferred adjuvants for use in the compositions are submicron oil-in-water emulsions. Preferred submicron oil-in-water emulsions for use herein are squalene/water emulsions optionally containing varying amounts of MTP-PE, such as a submicron oil-in-water emulsion containing 4-5% w/v squalene, 0.25-1.0% w/v Tween 80™ (polyoxyelthylenesorbitan monooleate), and/or 0.25-1.0% Span85™ (sorbitan trioleate), and, optionally, N-acetylmuramyl-L-alanyl-D-isogluatminyl-L-alanine-2-(1′-2′-dipalmitoyl-sn-glycero-3-huydroxyphosphophoryloxy)-ethylamine (MTP-PE), for example, the submicron oil-in-water emulsion known as “MF59” (International Publication No. WO 90/14837; U.S. Pat. Nos. 6,299,884 and 6,451,325, incorporated herein by reference in their entireties; and Ott et al., “MF59—Design and Evaluation of a Safe and Potent Adjuvant for Human Vaccines” in Vaccine Design: The Subunit and Adjuvant Approach (Powell, M. F. and Newman, M. J. eds.) Plenum Press, New York, 1995, pp. 277-296). MF59 contains 4-5% w/v Squalene (e.g. 4.3%), 0.25-0.5% w/v Tween 80™, and 0.5% w/v Span 85™ and optionally contains various amounts of MTP-PE, formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton, Mass.). For example, MTP-PE may be present in an amount of about 0-500 μg/dose, more preferably 0-250 μg/dose and most preferably, 0-100 μg/dose. As used herein, the term “MF59-0” refers to the above submicron oil-in-water emulsion lacking MTP-PE, while the term MF59-MTP denotes a formulation that contains MTP-PE. For instance, “MF59-100” contains 100 μg MTP-PE per dose, and so on. MF69, another submicron oil-in-water emulsion for use herein, contains 4.3% w/v squalene, 0.25% w/v Tween 80™, and 0.75% w/v Span 85™ and optionally MTP-PE. Yet another submicron oil-in-water emulsion is MF75, also known as SAF, containing 10% squalene, 0.4% Tween 80™, 5% pluronic-blocked polymer L121, and thr-MDP, also microfluidized into a submicron emulsion. MF75-MTP denotes an MF75 formulation that includes MTP, such as from 100-400 μg MTP-PE per dose.
  • Submicron oil-in-water emulsions, methods of making the same and immunostimulating agents, such as muramyl peptides, for use in the compositions, are described in detail in International Publication No. WO 90/14837 and U.S. Pat. Nos. 6,299,884 and 6,451,325, incorporated herein by reference in their entireties.
  • Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IFA) may also be used as adjuvants in the invention.
  • C. Saponin Formulations
  • Saponin formulations, may also be used as adjuvants in the invention. Saponins are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponin from the bark of the Quillaia saponaria Molina tree have been widely studied as adjuvants. Saponin can also be commercially obtained from Smilax ornata (sarsaprilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root). Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
  • Saponin compositions have been purified using High Performance Thin Layer Chromatography (HP-LC) and Reversed Phase High Performance Liquid Chromatography (RP-HPLC). Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C. Preferably, the saponin is QS21. A method of production of QS21 is disclosed in U.S. Pat. No. 5,057,540. Saponin formulations may also comprise a sterol, such as cholesterol (see WO96/33739).
  • Combinations of saponins and cholesterols can be used to form unique particles called Immunostimulating Complexs (ISCOMs). ISCOMs typically also include a phospholipid such as phosphatidylethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs. Preferably, the ISCOM includes one or more of Quil A, QHA and QHC. ISCOMs are further described in EP0109942, WO 96/11711 and WO 96/33739. Optionally, the ISCOMS may be devoid of additional detergent. See WO 00/07621.
  • A review of the development of saponin based adjuvants can be found at Barr, et al., “ISCOMs and other saponin based adjuvants”, Advanced Drug Delivery Reviews (1998) 32:247-271. See also Sjolander, et al., “Uptake and adjuvant activity of orally delivered saponin and ISCOM vaccines”, Advanced Drug Delivery Reviews (1998) 32:321-338.
  • D. Virosomes and Virus Like Particles (VLPs)
  • Virosomes and Virus Like Particles (VLPs) can also be used as adjuvants in the invention. These structures generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally non-pathogenic, non-replicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses. These viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA), Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and-Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA-phages, Qβ-phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein p1). VLPs are discussed further in WO 03/024480, WO 03/024481, and Niikura et al., “Chimeric Recombinant Hepatitis E Virus-Like Particles as an Oral Vaccine Vehicle Presenting Foreign Epitopes”, Virology (2002) 293:273-280; Lenz et al., “Papillomarivurs-Like Particles Induce Acute Activation of Dendritic Cells”, Journal of Immunology (2001) 5246-5355; Pinto, et al., “Cellular Immune Responses to Human Papillomavirus (HPV)-16 L1 Healthy Volunteers Immunized with Recombinant HPV-16 L1 Virus-Like Particles”, Journal of Infectious Diseases (2003) 188:327-338; and Gerber et al., “Human Papillomavrisu Virus-Like Particles Are Efficient Oral Immunogens when Coadministered with Escherichia coli Heat-Labile Entertoxin Mutant R192G or CpG”, Journal of Virology (2001) 75(10):4752-4760. Virosomes are discussed further in, for example, Gluck et al., “New Technology Platforms in the Development of Vaccines for the Future”, Vaccine (2002) 20:B10-B16. Immunopotentiating reconstituted influenza virosomes (IRIV) are used as the subunit antigen delivery system in the intranasal trivalent IFLEXAL™ product {Mischler & Metcalfe (2002) Vaccine 20 Suppl 5:B17-23} and the INFLUVAC PLUS™ product.
  • E. Bacterial or Microbial Derivatives
  • Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as:
  • (1) Non-Toxic Derivatives of Enterobacterial Lipopolysaccharide (LPS)
  • Such derivatives include Monophosphoryl lipid A (MPL) and 3-O-deacylated MPL (3dMPL). 3dMPL is a mixture of 3 De-O-acylated monophosphoryl lipid A with 4, 5 or 6 acylated chains. A preferred “small particle” form of 3 De-O-acylated monophosphoryl lipid A is disclosed in EP 0 689 454. Such “small particles” of 3dMPL are small enough to be sterile filtered through a 0.22 micron membrane (see EP 0 689 454). Other non-toxic LPS derivatives include monophosphoryl lipid A mimics, such as aminoalkyl glucosaminide phosphate derivatives e.g. RC-529. See Johnson et al. (1999) Bioorg Med Chem Lett 9:2273-2278.
  • (2) Lipid A Derivatives
  • Lipid A derivatives include derivatives of lipid A from Escherichia coli such as OM-174. OM-174 is described for example in Meraldi et al., “OM-174, a New Adjuvant with a Potential for Human Use, Induces a Protective Response with Administered with the Synthetic C-Terminal Fragment 242-310 from the circumsporozoite protein of Plasmodium berghei”, Vaccine (2003) 21:2485-2491; and Pajak, et al., “The Adjuvant OM-174 induces both the migration and maturation of murine dendritic cells in vivo”, Vaccine (2003) 21:836-842.
  • (3) Immunostimulatory Oligonucleotides
  • Immunostimulatory oligonucleotides suitable for use as adjuvants in the invention include nucleotide sequences containing a CpG motif (a sequence containing an unmethylated cytosine followed by guanosine and linked by a phosphate bond). Bacterial double stranded RNA or oligonucleotides containing palindromic or poly(dG) sequences have also been shown to be immunostimulatory.
  • The CpG's can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or single-stranded. Optionally, the guanosine may be replaced with an analog such as 2′-deoxy-7-deazaguanosine. See Kandimalla, et al., “Divergent synthetic nucleotide motif recognition pattern: design and development of potent immunomodulatory oligodeoxyribonucleotide agents with distinct cytokine induction profiles”, Nucleic Acids Research (2003) 31(9): 2393-2400; WO02/26757 and WO99/62923 for examples of possible analog substitutions. The adjuvant effect of CpG oligonucleotides is further discussed in Krieg, “CpG motifs: the active ingredient in bacterial extracts?”, Nature Medicine (2003) 9(7): 831-835; McCluskie, et al., “Parenteral and mucosal prime-boost immunization strategies in mice with hepatitis B surface antigen and CpG DNA”, FEMS Immunology and Medical Microbiology (2002) 32:179-185; WO98/40100; U.S. Pat. No. 6,207,646; U.S. Pat. No. 6,239,116 and U.S. Pat. No. 6,429,199.
  • The CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT. See Kandimalla, et al., “Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic CpG DNAs”, Biochemical Society Transactions (2003) 31 (part 3): 654-658. The CpG sequence may be specific for inducing a Th1 immune response, such as a CpG-A ODN, or it may be more specific for inducing a B cell response, such a CpG-B ODN. CpG-A and CpG-B ODNs are discussed in Blackwell, et al., “CpG-A-Induced Monocyte IFN-gamma-Inducible Protein-10 Production is Regulated by Plasmacytoid Dendritic Cell Derived IFN-alpha”, J. Immunol. (2003) 170(8):4061-4068; Krieg, “From A to Z on CpG”, TRENDS in Immunology (2002) 23(2): 64-65 and WO01/95935. Preferably, the CpG is a CpG-A ODN.
  • Preferably, the CpG oligonucleotide is constructed so that the 5′ end is accessible for receptor recognition. Optionally, two CpG oligonucleotide sequences may be attached at their 3′ ends to form “immunomers”. See, for example, Kandimalla, et al., “Secondary structures in CpG oligonucleotides affect immunostimulatory activity”, BBRC (2003) 306:948-953; Kandimalla, et al., “Toll-like receptor 9: modulation of recognition and cytokine induction by novel synthetic GpG DNAs”, Biochemical Society Transactions (2003) 31(part 3):664-658; Bhagat et al., “CpG penta- and hexadeoxyribonucleotides as potent immunomodulatory agents” BBRC (2003) 300:853-861 and WO 03/035836.
  • (4) ADP-Ribosylating Toxins and Detoxified Derivatives Thereof.
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention. Preferably, the protein is derived from E. coli (i.e., E. coli heat labile enterotoxin “LT), cholera (“CT”), or pertussis (“PT”). The use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in WO95/17211 and as parenteral adjuvants in WO98/42375. Preferably, the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LTR192G. The use of ADP-ribosylating toxins and detoxified derivaties thereof, particularly LT-K63 and LT-R72, as adjuvants can be found in the following references, each of which is specifically incorporated by reference herein in their entirety: Beignon, et al., “The LTR72 Mutant of Heat-Labile Enterotoxin of Escherichia coli Enahnces the Ability of Peptide Antigens to Elicit CD4+ T Cells and Secrete Gamma Interferon after Coapplication onto Bare Skin”, Infection and Immunity (2002) 70(6):3012-3019; Pizza, et al., “Mucosal vaccines: non toxic derivatives of LT and CT as mucosal adjuvants”, Vaccine (2001) 19:2534-2541; Pizza, et al., “LTK63 and LTR72, two mucosal adjuvants ready for clinical trials” Int. J. Med. Microbiol (2000) 290(4-5):455-461; Scharton-Kersten et al., “Transcutaneous Immunization with Bacterial ADP-Ribosylating Exotoxins, Subunits and Unrelated Adjuvants”, Infection and Immunity (2000) 68(9):5306-5313; Ryan et al., “Mutants of Escherichia coli Heat-Labile Toxin Act as Effective Mucosal Adjuvants for Nasal Delivery of an Acellular Pertussis Vaccine: Differential Effects of the Nontoxic AB Complex and Enzyme Activity on Th1 and Th2 Cells” Infection and Immunity (1999) 67(12):6270-6280; Partidos et al., “Heat-labile enterotoxin of Escherichia coli and its site-directed mutant LTK63 enhance the proliferative and cytotoxic T-cell responses to intranasally co-immunized synthetic peptides”, Immunol. Lett. (1999) 67(3):209-216; Peppoloni et al., “Mutants of the Escherichia coli heat-labile enterotoxin as safe and strong adjuvants for intranasal delivery of vaccines”, Vaccines (2003) 2(2):285-293; and Pine et al., (2002) “Intranasal immunization with influenza vaccine and a detoxified mutant of heat labile enterotoxin from Escherichia coli (LTK63)” J. Control Release (2002) 85(1-3):263-270. Numerical reference for amino acid substitutions is preferably based on the alignments of the A and B subunits of ADP-ribosylating toxins set forth in Domenighini et al., Mol. Microbiol (1995) 15(6):1165-1167, specifically incorporated herein by reference in its entirety.
  • F. Bioadhesives and Mucoadhesives
  • Bioadhesives and mucoadhesives may also be used as adjuvants in the invention. Suitable bioadhesives include esterified hyaluronic acid microspheres (Singh et al. (2001) J. Cont. Rele. 70:267-276) or mucoadhesives such as cross-linked derivatives of poly(acrylic acid), polyvinyl alcohol, polyvinyl pyrollidone, polysaccharides and carboxymethylcellulose. Chitosan and derivatives thereof may also be used as adjuvants in the invention. E.g. WO99/27960.
  • G. Microparticles
  • Microparticles may also be used as adjuvants in the invention. Microparticles (i.e. a particle of ˜100 nm to ˜150 μm in diameter, more preferably ˜200 nm to ˜30 μm in diameter, and most preferably ˜500 nm to ˜10 μm in diameter) formed from materials that are biodegradable and non-toxic (e.g. a poly(α-hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.), with poly(lactide-co-glycolide) are preferred, optionally treated to have a negatively-charged surface (e.g. with SDS) or a positively-charged surface (e.g. with a cationic detergent, such as CTAB).
  • H. Liposomes
  • Examples of liposome formulations suitable for use as adjuvants are described in U.S. Pat. No. 6,090,406, U.S. Pat. No. 5,916,588, and EP 0 626 169.
  • I. Polyoxyethylene Ether and Polyoxyethylene Ester Formulations
  • Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters. WO99/52549. Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol (WO01/21207) as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol (WO 01/21152).
  • Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether (laureth 9), polyoxyethylene-9-steoryl ether, polyoxytheylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
  • J. Polyphosphazene (PCPP)
  • PCPP formulations are described, for example, in Andrianov et al., “Preparation of hydrogel microspheres by coacervation of aqueous polyphophazene solutions”, Biomaterials (1998) 19(1-3):109-115 and Payne et al., “Protein Release from Polyphosphazene Matrices”, Adv. Drug. Delivery Review (1998) 31(3):185-196.
  • K. Muramyl Peptides
  • Examples of muramyl peptides suitable for use as adjuvants in the invention include N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-1-alanyl-d-isoglutamine (nor-MDP), and N-acetylmuramyl-1-alanyl-d-isoglutaminyl-1-alanine-2-(1′-2′-dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-ethylamine MTP-PE).
  • L. Imidazoquinolone Compounds.
  • Examples of imidazoquinolone compounds suitable for use adjuvants in the invention include Imiquamod and its homologues, described further in Stanley, “Imiquimod and the imidazoquinolones: mechanism of action and therapeutic potential” Clin Exp Dermatol (2002) 27(7):571-577 and Jones, “Resiquimod 3M”, Curr Opin Investig Drugs (2003) 4(2):214-218.
  • The invention may also comprise combinations of aspects of one or more of the adjuvants identified above. For example, the following adjuvant compositions may be used in the invention:
  • (1) a saponin and an oil-in-water emulsion (WO 99/11241);
  • (2) a saponin (e.g., QS21)+a non-toxic LPS derivative (e.g. 3dMPL) (see WO 94/00153);
  • (3) a saponin (e.g., QS21)+a non-toxic LPS derivative (e.g. 3dMPL)+a cholesterol;
  • (4) a saponin (e.g. QS21)+3dMPL+IL-12 (optionally+a sterol) (WO 98/57659);
  • (5) combinations of 3dMPL with, for example, QS21 and/or oil-in-water emulsions (See European patent applications 0835318, 0735898 and 0761231);
  • (6) SAF, containing 10% Squalane, 0.4 % Tween 80, 5% pluronic-block polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion.
  • (7) Ribi™ adjuvant system (RAS), (Ribi Immunochem) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL+CWS (Detox™);
  • (8) one or more mineral salts (such as an aluminum salt)+a non-toxic derivative of LPS (such as 3dPML).
  • (9) one or more mineral salts (such as an aluminum salt)+an immunostimulatory oligonucleotide (such as a nucleotide sequence including a CpG motif). Combination No. (9) is a preferred adjuvant combination.
  • M. Human Immunomodulators
  • Human immunomodulators suitable for use as adjuvants in the invention include cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, 1L-6, IL-7, IL-12, etc.), interferons (e.g. interferon-γ), macrophage colony stimulating factor, and tumor necrosis factor.
  • Aluminum salts and MF59 are preferred adjuvants for use with injectable influenza vaccines. Bacterial toxins and bioadhesives are preferred adjuvants for use with mucosally-delivered vaccines, such as nasal vaccines.
  • The immunogenic compositions of the present invention may be administed in combination with an antibiotic treatment regime. In one embodiment, the antibiotic is administered prior to administration of the antigen of the invention or the composition comprising the one or more of the antigens of the invention.
  • In another embodiment, the antibiotic is administered subsequent to the adminstration of the one or more antigens of the invention or the composition comprising the one or more antigens of the invention. Examples of antibiotics suitable for use in the treatment of the Steptococcal infections of the invention include but are not limited to penicillin or a derivative thereof or clindamycin or the like.
  • Further Antigens
  • The compositions of the invention may further comprise one or more additional Gram positive bacterial antigens which are not associated with an AI. Preferably, the Gram positive bacterial antigens that are not associated with an AI can provide protection across more than one serotype or strain isolate. For example, a first non-AI antigen, in which the first non-AI antigen is at least 90% (i.e., at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%) homologous to the amino acid sequence of a second non-AI antigen, wherein the first and the second non-AI antigen are derived from the genomes of different serotypes of a Gram positive bacteria, may be further included in the compositions. The first non-AI antigen may also be homologous to the amino acid sequence of a third non-AI antigen, such that the first non-AI antigen, the second non-AI antigen, and the third non-AI antigen are derived from the genomes of different serotypes of a Gram positive bacteria. The first non-AI antigen may also be homologous to the amino acid sequence of a fourth non-AI antigen, such that the first non-AI antigen, the second non-AI antigen, the third non-AI antigen, and the fourth non-AI antigen are derived from the genomes of different serotypes of a Gram positive bacteria.
  • The first non-AI antigen may be GBS 322. The amino acid sequence of GBS 322 across GBS strains from serotypes Ia, Ib, II, III, V, and VIII is greater than 90%. Alternatively, the first non-AI antigen may be GBS 276. The amino acid sequence of GBS 276 across GBS strain from serotypes Ia, Ib, II, III, V, and VIII is greater than 90%. Table 13 provides the percent amino acid sequence identity of GBS 322 and GBS 276 across different GBS strains and serotypes.
    TABLE 13
    Conservation of GBS 322 and GBS 276 amino acid sequences
    GBS
    322 GBS 276
    Serotype Strains cGH % AA identity cGH % AA identity
    Ia
    090 + 98.60 + 97.90
    A909 + 98.30 + 97.90
    515 + 98.80 + 97.50
    DK1 + +
    DK8 + +
    Davis + +
    Ib 7357b + +
    H36B + 98.30 + 97.80
    II 18RS21 + 100.00 + 99.90
    DK21 + +
    III NEM316 + 100.00 + 97.00
    COH31 + +
    D136 + +
    M732 + 98.00 + 100.00
    COH1 + 98.30 + 100.00
    M781 + 98.30 + 99.60
    No type CJB110 + 98.60 + 97.90
    1169NT + 97.40 + 97.90
    V CJB111 + 100.00 +
    2603 + 100.00 + 100.00
    VIII JM130013 + 100.00 + 97.90
    SMU014 + +
    total 22/22 98.28 +/− 0.4 22/22 98.44 +/− 1.094
  • As an example, inclusion of a non-AI protein, GBS 322, in combination with AI antigens GBS 67, GBS 80, and GBS 104 provided protection to newborn mice in an active maternal immunization assay.
    TABLE 14
    Active maternal immunization assay for a combination of fragments from
    GBS 322, GBS 80, GBS 104, and GBS 67
    MIX = 322 +
    FACS (Δ Mean) 80 + 104 + 67 PBS
    GBS strains Type GBS 80 GBS 67 GBS 322 alive/treated % protection alive/treated % protection
    515 Ia 0 409 227 39/40 97  6/40 15
    7357b- Ib 91 316 102 19/30 63  1/30 3
    DK21 II 0 331 416 25/34 73 17/48 35
    5401 II 170 618 135 35/40 87  3/37 8
    3050 II 43 460 188 48/48 100  1/30 3
    COH1 III 305 0 130 36/36 100  7/40 17
    M781 III 65 0 224 30/40 75  4/39 10
    2603 V 125 105 313 27/33 82 10/35 28
    CJB111 V 370 481 63 25/28 89  4/46 9
    JM9130013 VIII 597 83 143 37/39 95  5/40 12
    JMU071 VIII 556 79 170 44/50 88 18/50 36
    NT1169 NT 0 443 213 12/32 37 11/35 31
  • In fact, the non-AI GBS 322 antigen may itself provide protection to newborn mice in an active maternal immunization assay.
    TABLE 16
    Active maternal immunization assay for each of
    GBS 80 and GBS 322 antigens
    GBS
    80 GBS 322
    Protection Protection
    FACS (% survival) FACS (% survival)
    GBS strains Type Δ Mean antigen ctrl- Δ Mean antigen ctrl-
    CJB111 V 370 72% 40% 63 57% 40%
    COH1 III
    305 76% 10% 130 3% 10%
    2603 V 82 22% 34% 313 83% 34%
    7357b- Ib 91 36% 34% 102 43% 34%
    18RS21 II
    0 15% 24% 268 84% 24%
    DK21 II 0 10% 21% 416 67% 25%
    A909 Ia
    0 0% 14%
    090 Ia 0 0% 0%
    H36B Ib
    105 34% 32%

    Thus, inclusion of a non-AI protein in an immunogenic composition of the invention may provide increased protection a mammal.
  • The immunogenic compositions comprising S. pneumonaie AI polypeptides may further secondary SP protein antigens which include (a) any of the SP protein antigens disclosed in WO 02/077021 or U.S. provisional application ______, filed Apr. 20, 2005 (Attorney Docket Number 002441.00154), (2) immunogenic portions of the antigens comprising at least 7 contiguous amino acids, (3) proteins comprising amino acid sequences which retain immunogenicity and which are at least 95% identical to these SP protein antigens (e.g., 95%, 96%, 97%, 98%, 99%, or 99.5% identical), and (4) fusion proteins, including hybrid SP protein antigens, comprising (1)-(3).
  • Alternatively, the invention may include an immunogenic composition comprising a first and a second Gram positive bacteria non-AI protein, wherein the polynucleotide sequence encoding the sequence of the first non-AI protein is less than 90% (i.e., less than 90, 88, 86, 84, 82, 81, 78, 76, 74, 72, 70, 65, 60, 55, 50, 45, 40, 35, or 30 percent) homologous than the corresponding sequence in the genome of the second non-AI protein.
  • The compositions of the invention may further comprise one or more additional non-Gram positive bacterial antigens, including additional bacterial, viral or parasitic antigens. The compositions of the invention may further comprise one or more additional non-GBS antigens, including additional bacterial, viral or parasitic antigens.
  • In another embodiment, the GBS antigen combinations of the invention are combined with one or more additional, non-GBS antigens suitable for use in a vaccine designed to protect elderly or immunocomprised individuals. For example, the GBS antigen combinations may be combined with an antigen derived from the group consisting of Enterococcus faecalis, Staphylococcus aureus, Staphylococcus epidermis, Pseudomonas aeruginosa, Legionella pneumophila, Listeria monocytogenes, Neisseria meningitides, influenza, and Parainfluenza virus (‘PIV’).
  • Where a saccharide or carbohydrate antigen is used, it is preferably conjugated to a carrier protein in order to enhance immunogenicity {e.g. Ramsay et al. (2001) Lancet 357(9251):195-196; Lindberg (1999) Vaccine 17 Suppl 2:S28-36; Buttery & Moxon (2000) J R Coll Physicians Lond 34:163-168; Ahmad & Chapnick (1999) Infect Dis Clin North Am 13:113-133, vii.; Goldblatt (1998) J. Med. Microbiol. 47:563-567; European patent 0 477 508; U.S. Pat. No. 5,306,492; International patent application WO98/42721; Conjugate Vaccines (eds. Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114; and Hermanson (1996) Bioconjugate Techniques ISBN: 0123423368 or 012342335X}. Preferred carrier proteins are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids. The CRM197 diphtheria toxoid is particularly preferred {Research Disclosure, 453077 (January 2002)}. Other carrier polypeptides include the N. meningitidis outer membrane protein (EP-A-0372501), synthetic peptides (EP-A-0378881; EP-A-0427347), heat shock proteins (WO 93/17712; WO 94/03208), pertussis proteins (WO 98/58668; EP A 0471177), protein D from H. influenzae (WO 00/56360), cytokines (WO 91/01146), lymphokines, hormones, growth factors, toxin A or B from C. difficile (WO00/61761), iron-uptake proteins (WO01/72337), etc. Where a mixture comprises capsular saccharides from both serogroups A and C, it may be preferred that the ratio (w/w) of MenA saccharide:MenC saccharide is greater than 1 (e.g. 2:1, 3:1, 4:1, 5:1, 10:1 or higher). Different saccharides can be conjugated to the same or different type of carrier protein. Any suitable conjugation reaction can be used, with any suitable linker where necessary.
  • Toxic protein antigens may be detoxified where necessary e.g. detoxification of pertussis toxin by chemical and/or genetic means.
  • Where a diphtheria antigen is included in the composition it is preferred also to include tetanus antigen and pertussis antigens. Similarly, where a tetanus antigen is included it is preferred also to include diphtheria and pertussis antigens. Similarly, where a pertussis antigen is included it is preferred also to include diphtheria and tetanus antigens.
  • Antigens in the composition will typically be present at a concentration of at least 1 μg/ml each. In general, the concentration of any given antigen will be sufficient to elicit an immune response against that antigen.
  • As an alternative to using protein antigens in the composition of the invention, nucleic acid encoding the antigen may be used {e.g. refs. Robinson & Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al. (1997) Annu Rev Immunol 15:617-648; Scott-Taylor & Dalgleish (2000) Expert Opin Investig Drugs 9:471480; Apostolopoulos & Plebanski (2000) Curr Opin Mol Ther 2:441-447; Ilan (I 999) Curr Opin Mol Ther 1:116-120; Dubensky et al. (2000) Mol Med 6:723-732; Robinson & Pertmer (2000) Adv Virus Res 55:1-74; Donnelly et al. (2000) Am J Respir Crit Care Med 162(4 Pt 2):S 190-193; and Davis (1999) Mt. Sinai J. Med. 66:84-90}. Protein components of the compositions of the invention may thus be replaced by nucleic acid (preferably DNA e.g. in the form of a plasmid) that encodes the protein.
  • Definitions
  • The term “comprising” means “including” as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X+Y.
  • The term “about” in relation to a numerical value x means, for example, x±10%.
  • References to a percentage sequence identity between two amino acid sequences means that, when aligned, that percentage of amino acids are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987) Supplement 30. A preferred alignment is determined by the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The Smith-Waterman homology search algorithm is disclosed in Smith & Waterman (1981) Adv. Appl. Math. 2: 482-489.
  • The invention is further illustrated, without limitation, by the following examples.
  • EXAMPLE 1 Binding of an Adhesin Island Surface Protein, GBS 80, to Fibrinogen and Fibronectin
  • This example demonstrates that an Adhesin Island surface protein, GBS 80 can bind to fibrinogen and fibronectin.
  • An enzyme-linked immunosorbent assay (ELISA) was used to analyse the in vitro binding ability of recombinant GBS 80 to immobilized extra-cellular matrix (ECM) proteins but not to bovine serum albumin (BSA). Microtiter plates were coated with ECM proteins (fibrinogen, fibronectin, laminin, collagen type IV) and binding assessed by adding varying concentrations of a recombinant form of GBS 80, over-expressed and purified from E. coli (FIG. 5A). Plates were then incubated sequentially with a) mouse anti-GBS 80 primary antibody; b) rabbit anti-mouse AP-conjugated secondary antibody; c) pNPP colorimetric substrate. Relative binding was measured by monitoring absorbance at 405 nm, using 595 nm as a reference wavelength. FIG. 5 b shows binding of recombinant GBS 80 to immobilized ECM proteins (1 μg) as a function of concentration of GBS 80. BSA was used as a negative control. Data points represent the means of OD405 values±standard deviation for 3 wells.
  • Binding of GBS 80 to the tested ECM proteins was found to be concentration dependent and exhibited saturation kinetics. As is also evident from FIG. 5, binding of GBS 80 to fibronectin and fibrinogen was greater than binding to laminin and collagen type IV at all the concentrations tested.
  • EXAMPLE 2 GBS 80 is Required for Surface Localization of GBS 104
  • This example demonstrates that co-expression of GBS 80 is required for surface localization of GBS 104.
  • The polycistronic nature of the Adhesin Island I mRNA was investigated through reverse transcriptase-PCR (RT-PCR) analysis employing primers designed to detect transcripts arising from contiguous genes. Total RNA was isolated from GBS cultures grown to an optical density at 600 nm (OD600) of 0.3 in THB (Todd-Hewitt broth) by the RNeasy Total RNA isolation method (Qiagen) according to the manufacturer's instructions. The absence of contaminating chromosomal DNA was confirmed by failure of the gene amplification reactions to generate a product detectable by agarose gel electrophoresis, in the absence of reverse transcriptase. RT-PCR analysis was performed with the Access RT-PCR system (Promega) according to the manufacturer's instructions, employing PCR cycling temperatures of 60° C. for annealing and 70° C. for extension. Amplification products were visualized alongside 100-bp DNA markers in 2% agarose gels after ethidium bromide staining.
  • FIG. 5 shows that all the genes are co-transcribed as an operon. A schematic of the AI-1 operon is shown above the agarose gel analysis of the RT-PCR products. Large rectangular arrows indicate the predicted transcript direction. Primer pairs were selected such as “1-4” cross the 3′finish-5′start of successive genes and overlap each gene by at least 200 bp. Additionally, “1” crosses a putative rho-independent transcriptional terminator. “5” is an internal GBS 80 control and “6” is an unrelated control from a highly expressed gene. Lanes: “a”: RNA plus RTase enzyme; “b” RNA without RTase; “c”: genomic DNA control.
  • In the effort to elucidate the functions of the AI-1 proteins, in frame deletions of all of the genes within the operon have been constructed and the resulting mutants characterized with respect to surface exposure of the encoded antigens (see FIG. 8).
  • Each in-frame deletion mutation was constructed by splice overlap extension PCR(SOE-PCR) essentially as decribed by Horton et al. [Horton R. M., Z. L. Cai, S. N. Ho, L. R. Pease (1990) Biotechniques 8:528-35] using suitable primers and cloned into the temperature sensitive shuttle vector pJRS233 to replace the wild type copy by allelic exchange [Perez-Casal, J., J. A. Price, et al. (1993) Mol Microbiol 8(5): 809-19.]. All plasmid constructions utilized standard molecular biology techniques, and the identities of DNA fragments generated by PCR were verified by sequencing. Following SOE-PCR, the resulting mutant DNA fragments were digested with XhoI and EcoRI, and ligated into a similarly digested pJRS233. The resuting vectors were introduced by electroporation into the chromosome of 2603 and COH1 GBS strains in a three-step process, essentially as described in Framson et al. [Framson, P. E., A. Nittayajarn, J. Merry, P. Youngman, and C. E. Rubens. (1997) Appl. Environ. Microbiol. 63(9):353947]. Briefly, the vector pJRS233 contains an erm gene encoding erythromycin resistance and a temperature-sensitive gram-positive replicon that is active at 30° C. but not at 37° C. Initially, the constructs are electroporated into GBS electro-competent cells prepared as described by Frameson et al., and transformants containing free plasmid are selected by their ability to grow at 30° C. on Todd-Hewitt Broth (THB) agar plates containing 1 μg/ml erythromycin. The second step includes a selection step for strains in which the plasmid has integrated into the chromosome via a single recombination event over the homologous plasmid insert and chromosome sequence by their ability to grow at 37° C. on THB agar medium containing 1 mg/ml erythromycin. In the third step, GBS cells containing the plasmid integrated within the chromosome (integrants) are serially passed in broth culture in the absence of antibiotics at 30° C. Plasmid excision from the chromosome via a second recombination event over the duplicated target gene sequence either completed the allelic exchange or reconstituted the wild-type genotype. Subsequent loss of the plasmid in the absence of antibiotic selection pressure resulted in an erythromycin-sensitive phenotype. In order to assess gene replacement a screening of erythromycin-sensitive colonies was performed by analysis of the target gene PCR amplicons.
  • FIG. 7 reports a schematic of the IS-1 operon for each knock-out strain generated, along with the deletion position within the amino acidic sequence. Most data presented here concern the COH1 deletion strains, in which the expression of each of the antigens is higher by DNA microarray analysis (data not shown) as well as detectable by FACS analysis (see FIG. 8). The double mutant in 2603 Δ80, Δ104 double mutant was constructed by sequential allelic exchanges of the shown alleles.
  • Immunization Protocol
  • Immune sera for FACS experiments were obtained as follows.
  • Groups of 4 CD-1 outbred female mice 6-7 weeks old (Charles River Laboratories, Calco Italy) were immunized with the selected GBS antigens, (20 μg of each recombinant GBS antigen), suspended in 100 μl of PBS. Each group received 3 doses at days 0, 21 and 35. Immunization was performed through intra-peritoneal injection of the protein with an equal volume of Complete Freund's Adjuvant (CFA) for the first dose and Incomplete Freund's Adjuvant (IFA) for the following two doses. In each immunization scheme negative and positive control groups are used. Immune response was monitored by using serum samples taken on day 0 and 49.
  • FACS Analysis
  • Preparation of paraformaldehyde treated GBS cells and their FACS analysis were carried out as follows.
  • GBS serotype COH1 strain cells were grown in Todd Hewitt Broth (THB; Difco Laboratories, Detroit, Mich.) to OD600 nm=0.5. The culture was centrifuged for 20 minutes at 5000 rpm and bacteria were washed once with PBS, resuspended in PBS containing 0.05% paraformaldehyde, and incubated for 1 hours at 37° C. and then overnight at 4° C. 50 μl of fixed bacteria (OD600 0.1) were washed once with PBS, resuspended in 20 μl of Newborn Calf Serum, (Sigma) and incubated for 20 min. at room temperature. The cells were then incubated for 1 hour at 4° C. in 100 μl of preimmune or immune sera, diluted 1:200 in dilution buffer (PBS, 20% Newborn Calf Serum, 0.1% BSA). After centrifugation and washing with 200 μl of washing buffer (0.1% BSA in PBS), samples were incubated for 1 hour at 4° C. with 50 μl of R-Phicoerytrin conjugated F(ab)2 goat anti-mouse IgG (Jackson ImmunoResearch Laboratories; Inc.), diluted 1:100 in dilution buffer. Cells were washed with 200 μl of washing buffer and resuspended in 200 μl of PBS. Samples were analysed using a FACS Calibur apparatus (Becton Dickinson, Mountain View, Calif.) and data were analyzed using the Cell Quest Software (Becton Dickinson). A shift in mean fluorescence intensity of >75 channels compared to preimmune sera from the same mice was considered positive. This cutoff was determined from the mean plus two standard deviations of shifts obtained with control sera raised against mock purified recombinant proteins from cultures of E. coli carrying the empty expression vector and included in every experiment. Artifacts due to bacterial lysis were excluded using antisera raised against 6 different known cytoplasmic proteins all of which were negative
  • FACS data on COH1 single KO mutants for GBS 104 and GBS 80 indicated that GBS 80 is required for surface localization of GBS 104.
  • As shown in FIG. 8, GBS 104 is not surface exposed in the Δ80 strain (second column, bottom), but is present in the whole protein extracts (see FIG. 10). Mean shift values suggest that GBS 104 is partially responsible for GBS 80 surface exposure (Mean shift of GBS 80 is reduced to ˜60% wild-type levels in Δ104), and that GBS 80 is over-expressed in the complemented strain (mean shift value ˜200% wild-type level). The Δ80/pGBS 80 strain contains the GBS 80 orf cloned in the shuttle-vector pAM401 (Wirth, R., F. Y. An, et al. (1986). J Bacteriol 165(3): 831-6). The vector alone does not alter the secretion pattern of GBS 104 (right column). FACS was performed on mid-log fixed bacteria with mouse polyclonal antibodies as indicated at left. Black peak is pre-immune sera, colored peaks are sera from immunized animals.
  • EXAMPLE 3 Deletion of GBS 80 Causes Attenuation In Vivo
  • This example demonstrates that deletion of GBS 80 causes attenuation in vivo, suggesting that this protein contributes to bacterial virulence.
  • By using a mouse animal model, we studied the role of GBS 80 and GBS 104 in the virulence of S. agalactiae.
  • Groups of ten outbred female mice 5-6 week weeks old (Charles River Laboratories, Calco Italy) were inoculated intraperitoneally with different dilutions of the mutant strains and LD50 (lethal dose 50) were calculated according to the method of Reed and Muench [Reed, L. J. and H. Muench (1938). The American Journal of Hygiene 27(3): 493-7]. As presented in the table below the number of colony forming units (cfu) counted for both the Δ80 and the Δ80, Δ104 double mutants is about 10 fold higher when compared to the wild type strain suggesting that inactivation of GBS 80 but not GBS 104 is responsible for an attenuation in virulence. This finding indicates that GBS 80 gene in the AI-1 might contribute to virulence.
    TABLE
    Lethal dose
    50% analysis of AI-1 mutants in the 2603 strain background.
    LD50s were performed by IP injection of female CD1 mice at an age
    of 5-6 weeks. LD50s were calculated by the
    method of Reed and Muench (8).
    GBS strain LD50, cfu Number of Experiments
    Wild Type 2603  2 × 108 4
    Δ104 mutant ˜2 × 108 1
    Δ80 mutant 2.6 × 109  3
    Δ80, Δ104 double mutant ˜2 × 109 1
  • EXAMPLE 4 Effect of Adhesin Island Sortase Deletions on Surface Antigen Presentation
  • This example demonstrates the effect of adhesin island sortase deletions on surface antigen presentation.
  • FACS analysis results set forth in FIG. 9 show that a deletion in sortase SAG0648 prevented GBS 104 from reaching the surface and slightly reduced the surface exposure of GBS 80 (fourth panel; mean shift value ˜60% wild-type COH1). In the double sortase knock-out strain, neither antigen was surface exposed (far right panel). Either sortase alone was sufficient for GBS 80 to arrive at the bacterial surface (third and fourth columns, top). No effect was seen on surface exposure of antigens GBS 80 or GBS 104 in the ΔGBS 52 strain. Antibodies derived from purified GBS 52 were either non-specific or were FACS negative for GBS 52 (data not shown). FACS analysis was performed as described above (see EXAMPLE 2).
  • As shown in FIG. 10, inactivation of GBS 80 has no effect on GBS 104 expression as much as GBS 104 knock out doesn't change the total amount GBS 80 expressed. The Western blot of whole protein extracts (strains noted above lanes) probed with anti-GBS 80 antisera is shown in panel A. Arrow indicates expected size of GBS 80 (60 kDa). GBS 80 antibodies recognize a doublet, the lower band is not present in ΔGBS 80 strains. Panel B shows a Western blot of whole protein extracts probed with anti-GBS 104 antisera. Arrow indicates expected size of GBS 104 (99.4 kDa). Protein extracts were prepared from the same bacterial cultures used for FACS (FIGS. 8 and 9). In conclusion, although GBS 104 does not arrive at the surface in the Δ80 strain by FACS (FIG. 8, second column), it is present at approximately wild-type levels in the whole protein preps (B, second lane). Approximately 20 μg of each protein extract was loaded per lane.
  • Western-Blot Analysis
  • Aliquots of total protein extract mixed with SDS loading buffer (1×:60 mM TRIS-HCl pH 6.8, 5% w/v SDS, 10% v/v glycerin, 0.1% Bromophenol Blue, 100 mM DTT) and boiled 5 minutes at 95° C., were loaded on a 12.5% SDS-PAGE precast gel (Biorad). The gel is run using a SDS-PAGE running buffer containing 250 mM TRIS, 2.5 mM Glycine and 0.1% SDS. The gel is electroblotted onto nitrocellulose membrane at 200 mA for 60 minutes. The membrane is blocked for 60 minutes with PBS/0.05% Tween-20 (Sigma), 10% skimmed milk powder and incubated O/N at 4° C. with PBS/0.05 % Tween 20, 1% skimmed milk powder, with the appropriate dilution of the sera. After washing twice with PBS/0.05% Tween, the membrane is incubated for 2 hours with peroxidase-conjugated secondary anti-mouse antibody (Amersham) diluted 1:4000. The nitrocellulose is washed three times for 10 minutes with PBS/0.05% Tween and once with PBS and thereafter developed by Opti-4CN Substrate Kit (Biorad).
  • EXAMPLE 5 Binding of Adhesin Island Proteins to Epithelial Cells and Effect of Adhesin Island Proteins on Capacity of GBS to Adhere to Epithelial Cells
  • This example illustrates the binding of AI proteins to epithelial cells and the effect of AI proteins on the capacity of GBS to adhere to epithelial cells.
  • Applicants analysed whether recombinant AI surface proteins GBS 80 or GBS 104 would demonstrate binding to various epithelial cells in a FACS analysis. Applicants also analysed whether deletion of AI surface proteins GBS 80 or GBS 104 would effect the capacity of GBS to adhere to and invade ME180 cervical epithelial cells.
  • As shown in FIG. 28, deletion of GBS 80 sequence from GBS strain isolate 2603 (serotype V) did not affect the capacity of the mutated GBS to adhere to and invade ME180 cervical epithelial cells. Here ME180 cervical carcinoma epithelial cells were infected with wild type GBS 2603 or GBS 2603 Δ80 isogenic mutant. After two hours of infection, non-adherent bacteria were washed off and infection prolonged for a further two hours and four hours. In invasion experiments, after each time point, was followed by a two hour antibiotic treatment. Cells were then lysed with 1% saponin and lysates platedon TSA plates. As shown in FIG. 28, there was little difference between the percent invasion or percent adhesion of wild type and mutant strains up to the four hour time point.
  • FIG. 30 repeats this experiment with both Δ104 and Δ80 mutants from a different strain isolate. Here, ME180 cervical carcinoma epithelial cells were infected with GBS strain isolate COH (serotype III) wild type or COH1 ΔGBS 104 or COH1 Δ80 isogenic mutant. After one hour of infection, non-adherent bacteria were washed off and the cells were lysed with 1% saponin. The lysates were plated on TSA plates. As shown in FIG. 30, while there was little difference in the percent invasion, there was a significant decrease in the percent association of the Δ104 mutant compared to both the wild type and Δ80 mutant.
  • The affect of AI surface proteins on the ability of GBS to translocate through an epithelial monolayer was also analysed. As shown in FIG. 31, a GBS 80 knockout mutant strain partially loses the ability to translocate through an epithelial monolayer. Here epithelial monolayers were inoculated with wildtype or knockout mutant in the apical chamber of a transwell system for two hours and then non-adherent bacteria were washed off. Infection was prolonged for a further two and four hours. Samples were taken from the media of the basolateral side and the number of colony forming unties measured. Transepithelial electrical resistance measured prior to and after infection gave comparable values, indicating the maintenance of the integrity of the monolayer. By the six hour time point, the Δ80 mutants demonstrated a reduced percent transcytosis.
  • A similar experiment was conducted with GBS 104 knock out mutants. Here, as shown in FIG. 22, the Δ104 mutants also demonstrated a reduced percent transcytosis, indicating that the mutant strains translocate through an epithelial monolayer less efficiently than their isogenic wild type counterparts.
  • Applicants also studied the effect of AI proteins on the capacity of a GBS strain to invade J774 macrophage-like cells. Here, J774 cells were infected with GBS COH1 wild type or COH1 ΔGBS 104 or COH1 ΔGBS80 isogenic mutants. After one hour of infection, non-adherent bacteria were washed off and intracellular bacteria were recovered at two, four and six hours post antibiotic treatment. At each time point, cells were lysed with 0.25% Triton X-100 and lysates plated on TSA plates. As shown in FIG. 32, the Δ104 mutant demonstrated a significantly reduced percent invasion compared to both the wild type and Δ80 mutant.
  • EXAMPLE 6 Hyperoligomeric Structures Comprising AI Surface Proteins GBS 80 and GBS 104
  • This example illustrates hyperoligomeric structures comprising AI surface proteins GBS 80 and GBS 104. A GBS isolate COH1 (serotype III) was adapted to increase expression of GBS 80. FIG. 34 presents a regular negative stain electron micrograph of this mutant; no pilus or hyperoligomeric structures are distinguishable on the surface of the bacteria. When the EM stain is based on anti-GBS 80 antibodies labelled with 10 or 20 nm gold particles, the presence of GBS 80 throughout the hyperoligomeric structure is clearly indicated (FIGS. 36, 37 and 38). EM staining against GBS 104 (anti-GBS 104 antibodies labelled with 10 nm gold particles) also reveals the presence of GBS 104 primarily on or near the surface of the bacteria or potentially associated with bacterial peptidoglycans (FIG. 39). Analysis of this same strain (over-expressing GBS 80) with a combination of both anti-GBS 80 (using 20 nm gold particles) and anti-GBS 104 (using 10 nm gold particles) reveals the presence of GBS 104 on the surface and within the hyperoligomeric structures (see FIGS. 40 and 41).
  • EXAMPLE 7 GBS 80 is Necessary for Polymer Formation and GBS 104 and Sortase SAG0648 are Necessary for Efficient Pili Assembly
  • This example demonstrates that GBS 80 is necessary for formation of polymers and that GBS 104 and sortase SAG0648 are necessary for efficient pili assembly. GBS 80 and GBS 104 polymeric assembly was systematically analyzed in Coh1 strain single knock out mutants of each of the relevant coding genes in AI-1 (GBS 80, GBS 104, GBS 52, sag0647, and sag0648). FIG. 41 provides Western blots of total protein extracts (strains noted above lanes) probed with either anti-GBS 80 (left panel) sera or anti-GBS 104 sera (right panel) for each of these Coh1 and Coh1 knock out strains. (Coh1, wild type Coh1; Δ80, Coh1 with GBS 80 knocked out; Δ104, Coh1 with GBS 104 knocked out; Δ52, Coh1 with GBS 52 knocked out; A647, Coh1 with SAG0647 knocked out; A648, Coh1 with SAG0648 knocked out, A647-8, Coh1 with SAG0647 and SAG0648 knocked out; Δ80/pGBS80, Coh1 with GBS 80 knocked out but complemented with a high copy number plasmid expressing GBS 80. Asterisks identify the monomer of GBS 80 and GBS 104.)
  • The smear of immunoreactive material observed in the wild type strain, along with its disappearance in Δ80 and Δ104 mutants, is consistent with the notion that such high molecular weight structures are composed of covalently linked (SDS-resistant) GBS 80 and GBS 104 subunits. The immunoblotting with both anti-GBS 80 (α-GBS 80) and anti-GBS 104 (α-GBS 104) revealed that deletion of sortase SAG0648 also interferes with the assembly of high molecular weight species, whereas the knock out mutant of the second sortase (SAG0647), even if somehow reduced, still maintains the ability to form polymeric structures.
  • Total extracts form GBS were prepared as follows. Bacteria were grown in 50 ml of Todd-Hewitt broth (Difco) to an OD600nm of 0.5-0.6 and successively pelleted. After two washes in PBS the pellet was resuspended and incubated 3 hours at 37° C. with mutanolisin. Cells were then lysed with at least three freezing-thawing cycles in dry ice and a 37° C. bath. The lysate was then centrifuged to eliminate the cellular debris and the supernatant was quantified. Approximately 40 μg of each protein extract was separated on SDS-PAGE. The gel was then subjected to immunoblotting with mice antisera and detected with chemiluminescence.
  • EXAMPLE 8 GBS 80 is Polymerized by an AI-2 Sortase
  • This example illustrates that GBS 80 can be polymerized not only by AI-1 sortases, but also by AI-2 sortases. FIG. 42 shows total cell extract immunoblots of GBS 515 strain, which lacks AI-1. The left panel, where an anti-GBS 67 sera was used, shows that GBS 67 from AI-2 is assembled into high-molecular weight-complexes, suggesting the formation of a second type of pilus. The same high molecular structure is observed when GBS 80 is highly expressed by reintroducing the gene within a plasmid (pGBS 80). By using anti-GBS 80 (right panel) sera on the same extracts, again it is observed that, with GBS 80 over expression (515/pGBS 80), a high-molecular weight structure is assembled. This implies that, in the absence of AI-1 sortases, AI-2 sortases (SAG1405 and SAG1406) can complement the lacking function, still being able to assemble GBS 80 in a pilus structure.
  • EXAMPLE 9 Coh1 Produces a High Molecular Weight Molecule, the GBS 80 Pilin
  • This example illustrates that Coh1 produces a high molecular weight molecule, greater than 1000 kDa, which is the GBS 80 pilin. FIG. 43 provides silver-stained electrophoretic gels that show that Coh1 produces two macromolecules. One of these macromolecules disappears in the Coh1 GBS 80 knock out cells, but does not disappear in the Coh1 GBS 52 knock out mutant cells. The last two lanes on the right were loaded with 15 times the amount loaded in the other lanes. This was done in order to be able to count the bands. By doing this, a conservative size estimate of the top bands was calculated by starting at 240 kDa and considering each of 14 higher bands as the result of consecutive additions of a GBS 80 monomer.
  • Coh1, wild type Coh1; Δ80, Coh1 cells with GBS 80 knocked out; Δ52, Coh1 cells with GBS 52 knocked out; Δ80/pGBS 80, Coh1 cells with GBS 80 knocked out and complemented with a high copy number construct expressing GBS 80.
  • EXAMPLE 10 GBS 52 is a Minor Component of the GBS Pilus
  • This example illustrates that GBS 52 is present in the GBS pilus and is a minor component of the pilus. FIG. 45 shows an immunoblot of total cell extracts from a GBS Coh1 strain and a GBS Coh1 strain knocked out for GBS 52 (Δ52). The total cell extracts were immunoblotted anti-GBS 80 antisera (left) and anti-GBS 52 antisera (right). Immunoblotting was performed using a 3-8% Tris-acetate polyacrylamide gel (Invitrogen) which provided excellent separation of large molecular weight proteins (see FIG. 41). When the gel was incubated with anti-GBS 80 sera, the bands from the Coh1 wild-type strain appeared shifted when compared to the Δ52 mutant. This observation indicated a different size of the pilus polymeric components in the two strains. When the same gel was stripped and incubated with anti-GBS 52 sera the high-molecular subunits in the Coh1 wild-type strain showed similar molecular size of those in the correspondent lane in the left panel. These findings confirmed that GBS 52 is indeed associated with GBS 80 macro-molecular structures but represents a minor component of the GBS pilus.
  • EXAMPLE 11 Pilus Structures are Present in the Supernatant of GBS Bacterial Cultures
  • This example illustrates that the pilus structure assembled in Coh1 GBS is present in the supernatant of a bacterial cell culture. FIG. 46 shows an immunoblot where the protein extract of the supernatant from cultures of different GBS mutant strains (117=Coh1 GBS 80 knockout; 159=Coh1 GBS 104 knockout; 202=Coh1 GBS 52 knockout; 206=Coh1 GBS sag0647 knockout; 208=Coh1 GBS sag0648 knockout; 197=Coh1 GBS sag0647/sag0648 knockout; 179=Coh1 GBS 80 knockout complemented with a high copy plasmid expressing GBS 80). GBS 80 antisera detects the presence of pilus structures in the appropriate Coh1 strains.
  • The protein extract was prepared as follows. Bacteria were grown in THB to an OD600nm of 0.5-0.6 and the supernatant was separated from the cells by centrifugation. The supernatant was then filtered (527 0.2 μm) and 1 ml was added with 60% TCA for protein precipitation.
  • GBS pili were also extracted from the fraction of surface-exposed proteins in Coh1 strain and its GBS 80 knock out mutant as described hereafter. Bacteria were grown to an OD600nm of 0.6 in 50 ml of THB at 37° C. Cells were washed once with PBS and the pellet was then resuspended in 0.1 M KPO4 pH 6.2, 40% sucrose, 10 mM MgCl2, 400 U/ml mutanolysin and incubated 3 hours at 37° C. Protoplasts were separated by centrifugation and the supernatant was recovered and its protein content measured.
  • In order to study the dynamics of pilus production during different growth phases, 1 ml supernatant of a culture at different OD600nm was TCA precipitated and loaded onto a 3-8% SDS-PAGE as described before. FIG. 47 shows the corresponding Western blot with GBS 80 anti-sera. The first group of lanes (left five sample lanes) refer to a Coh1 strain growth (OD600nm are noted above the lanes) whereas the second group of lanes (right five samples) are from a GBS 80 knock out strain over expressing GBS 80. The experiment shows that pilus macromolecular structures can be found in the supernatant in all of the growth phases tested.
  • EXAMPLE 12 In GBS Strain Coh1, only GBS 80 and a Sortase (sag0647 or sag0648) is Required for Polymerization
  • This example describes requirements for pilus formation in Coh1. FIG. 48 shows a Western blot of total protein extracts (prepared as described before) using anti-GBS 80 sera on Coh1 clones. (Coh1, wild type Coh1; Δ104, Coh1 knocked out for GBS 104, Δ647, Coh1 knocked out for sag0647, Δ648, Coh1 knocked for sag0648, Δ647-8, Coh1 knocked out for sag0647 and sag0648; 515, wild type bacterial strain 515, which lacks an AI-1; p80 a high copy number plasmid which expresses GBS 80.) The data show that only the double sortase mutant is unable to polymerize GBS 80 indicating that the ‘conditio sine qua non’ for pilus polymerization is the co-existence of GBS 80 with at least one sortase. This result leads to a reasonable assumption that SAG1405 and SAG1406 are responsible for polymerization in this strain.
  • EXAMPLE 13 GBS 80 can be Expressed in L. lactis Under its Own Promoter and Terminator Sequences
  • This example demonstrates that L. lactis, a non-pathogenic bacterium, can express GBS AI polypeptides such as GBS 80. L. lactis M1363 (J. Bacteriol. 154 (1983):1-9) was transformed with a construct encoding GBS 80. Briefly, the construct was prepared by cloning a DNA fragment containing the gene coding for GBS 80 under its own promoter and terminator sequences into plasmid pAM401 (a shuttle vector for E. coli and other Gram positive bacteria; J. Bacteriol. 163 (1986):831-836). Total extracts of the transformed bacteria in log phase were separated on SDS-PAGE, transferred to membranes, and incubated with antiserum against GBS 80. A polypeptide corresponding to the molecular weight of GBS 80 was detected in the lanes containing total extracts of L. lactis transformed with the GBS 80 construct. See FIGS. 133A and 133B, lanes 6 and 7. This same polypeptide was not detected in the lane containing total extracts of L. lactis not transformed with the GBS 80 construct, lane 9. This example shows that L. lactis can express GBS 80 under its own promoter and terminator.
  • EXAMPLE 14 L. lactis Modified to Express GBS AI-1 Under the GBS 80 Promoter and Terminator Sequences Expresses GBS 80 in Polymeric Structures
  • This example demonstrates the ability of L. lactis to express GBS AI-1 polypeptides and to incorporate at least some of the polypeptides into oligomers. L. lactis was transformed with a construct containing the genes encoding GBS AI-1 polypeptides. Briefly, the construct was prepared by cloning a DNA fragment containing the genes for GBS 80, GBS 52, SAG0647, SAG0648, and GBS 104 under the GBS 80 promoter and terminator sequences into construct pAM401. The construct was transformed into L. lactis M1363. Total extracts of log phase transformed bacteria were separated on reducing SDS-PAGE, transferred to membranes, and incubated with antiserum against GBS 80. A polypeptide with a molecular weight corresponding to the molecular weight of GBS 80 was detected in the lanes containing L. lactis transformed with the GBS AI-1 encoding construct. See FIG. 134, lane 2. In addition, the same lane also showed immunoreactivity of polypeptides having higher molecular weights than the polypeptide having the molecular weight of GBS 80. These higher molecular weight polypeptides are likely oligomers of GBS 80. Oligomers of similar molecular weights were also observed on a Western blot of the culture supernatant of the transformed L. lactis. See lane 4 of FIG. 135. Thus, this example shows that L. lactis transformed to express GBS AI-1 can efficiently polymerize GBS 80 in the form of a pilus. This pilus structure can likely be purified from either the cell culture supernatant or cell extracts.
  • EXAMPLE 15 Cloning and Expression of S. pneumoniae Sp0462
  • This example describes the production of a clone encoding a Sp0462 polypeptide and expression of the clone. To produce a clone encoding Sp0462, the open reading frame encoding Sp0462 was amplified using primers that annealed within the full-length Sp0462 open reading frame sequence. FIG. 150A provides a 893 amino acid sequence of Sp0462. The primers used to produce a clone encoding the Sp0462 polypeptide are shown in FIG. 150B. These primers annealed to the nucleotide sequences encoding the amino acid residues indicated by underlining in FIG. 150A. Amplification of the open reading frame encoding Sp0462 using these primers produced the amplicon shown at lane 2 of the agarose gel provided in FIG. 160. The Sp0462 clone encodes amino acid residues 38-862 of the 893 amino acid residue Sp0462 protein; the italicized residues in FIG. 150A were eliminated. FIG. 151A provides a schematic depiction of the recombinant Sp0462 polypeptide. FIG. 151B shows a schematic depiction of the full-length Sp0462 polypeptide. Both the recombinant Sp0462 encoded by the clone and the full-length Sp0462 protein have two collagen binding protein type B (Cna B) domains and a von Hillebrand factor A (vWA) domain. The cloned recombinant Sp0462 lacks the LPXTG motif present in the full-length Sp0462 protein. Western blot analysis for expression of the Sp0462 clone did not result in detection of polypeptides with serum obtained from S. pneumoniae-infected patients (FIG. 152A) or GBS 80 antiserum (FIG. 152B).
  • EXAMPLE 16 Cloning and Expression of S. pneumoniae Sp0463
  • This example describes the production of a clone encoding a Sp0463 polypeptide and detection of recombinant Sp0463 polypeptide expressed from the clone. To produce a clone encoding Sp0463, the open reading frame encoding Sp0463 was amplified using primers that annealed within the full-length Sp0463 open reading frame sequence. FIG. 153A provides a 665 amino acid sequence of Sp0463. The primers used to produce the clone encoding Sp0463 polypeptide are shown in FIG. 153B. These primers annealed to the nucleotide sequences encoding the amino acid residues indicated by underlining in FIG. 153A. Amplification of the open reading frame encoding Sp0463 using these primers produced the amplicon shown at lane 3 of the agarose gel provided in FIG. 160. The Sp0463 clone encodes amino acid residues 23-627 of the 665 amino acid residue Sp0463 protein; the italicized residues in FIG. 153A were eliminated. FIG. 154A provides a schematic depiction of the recombinant Sp0463 polypeptide. FIG. 154B shows a schematic depiction of the full-length Sp0463 polypeptide. Both the recombinant Sp0463 encoded by the clone and the full-length Sp0463 protein have a Cna B domain and an E box motif. The cloned recombinant Sp0463 lacks the LPXTG motif present in the full-length Sp0463 protein. Expression of the Sp0463 clone resulted in the detection of a 60 kD polypeptide, the expected molecular weight of the recombinant Sp0463 polypeptide, by Western blot analysis. See FIG. 155.
  • EXAMPLE 17 Cloning and Expression of S. pneumoniae Sp0464
  • This example describes the production of a clone encoding a Sp0464 polypeptide and detection of recombinant Sp0464 polypeptide expressed from the clone. To produce a clone encoding Sp0464, the open reading frame encoding Sp0464 was amplified using primers that annealed either within the full-length Sp0464 open reading frame sequence. FIG. 157A provides a 393 amino acid sequence of Sp0464. The primers used to produce a clone encoding the Sp0464 polypeptide are shown in FIG. 157B. These primers annealed to the nucleotide sequences encoding the amino acid residues indicated by underlining in FIG. 157A. Amplification of the open reading frame encoding Sp0464 using these primers produced the amplicon shown at lane 4 of the agarose gel provided in FIG. 160. The Sp0464 clone encodes amino acid residues 19-356 of the 393 amino acid residue Sp0464 protein; the italicized residues in FIG. 157A were eliminated. FIG. 158A provides a schematic depiction of the recombinant Sp0464 polypeptide. FIG. 158B shows a schematic depiction of the full-length Sp0464 polypeptide. Both the recombinant Sp0464 encoded by the clone and the full-length Sp0464 protein have two Cna B domains. The cloned recombinant Sp0464 lacks the LPXTG motif present in the full-length Sp0464 protein. Expression of the Sp0464 clone resulted in the detection of a 38 kD polypeptide, the expected molecular weight of the recombinant Sp0464 polypeptide, by Western blot analysis. See FIG. 159.
  • EXAMPLE 18 Intranasal Immunization of Mice with Recombinant L. lactis Expressing GBS 80 and Subsequent Challenge
  • This example describes a method of intranasally immunizing mice using L. lactis that express GBS 80. Intranasal immunization consisted of 3 doses at days 0, 14 and 28, each dose administered in three consecutive days. Each day, groups of 3 CD-1 outbred female mice 6-7 weeks old (Charles River Laboratories, Calco Italy) were immunized intranasally with 109 or 1010 CFU of the recombinant Lactococcus lactis suspended in 20 μl of PBS. In each immunization scheme negative (wild-type L. lactis) and positive (recombinant GBS80) control groups were used. The immune response of the dams was monitored by using serum samples taken on day 0 and 49. The female mice were bred 2-7 days after the last immunization (at approximately t=36-37), and typically had a gestation period of 21 days. Within 48 hours of birth, the pups were challenged via I.P. with GBS in a dose approximately equal to an amount which would be sufficient to kill 90% of immunized pups (as determined by empirical data gathered from PBS control groups). The GBS challenge dose is preferably administered in 50 ml of THB medium. Preferably, the pup challenge takes place at 56 to 61 days after the first immunization. The challenge inocula were prepared starting from frozen cultures diluted to the appropriate concentration with THB prior to use. Survival of pups was monitored for 5 days after challenge.
  • EXAMPLE 19 Subcutaneous Immunization of Mice with Recombinant L. lactis Expressing GBS 80 and Subsequent Challenge
  • This example describes a method of subcutaneous immunization mice using L. lactis that express GBS 80. Subcutaneous immunization consists of 3 doses at days 0, 14 and 28. Groups of 3 CD-1 outbred female mice 6-7 weeks old (Charles River Laboratories, Calco Italy) were injected subcutaneously with 109 or 1010 CFU of the recombinant Lactococcus lactis suspended in 100 μl of PBS. In each immunization scheme, negative (wild-type L. lactis) and positive (recombinant GBS80) control groups were used. The immune response of the dams was monitored by using serum samples taken on day 0 and 49. The female mice were bred 2-7 days after the last immunization (at approximately t=36-37), and typically had a gestation period of 21 days. Within 48 hours of birth, the pups were challenged via I.P. with GBS in a dose approximately equal to an amount which would be sufficient to kill 90% of immunized pups (as determined by empirical data gathered from PBS control groups). The GBS challenge dose is preferably administered in 50 ml of THB medium. Preferably, the pup challenge takes place at 56 to 61 days after the first immunization. The challenge inocula were prepared starting from frozen cultures diluted to the appropriate concentration with THB prior to use. Survival of pups was monitored for 5 days after challenge.
  • EXAMPLE 20 Immunization of Mice with GAS AI Polypeptides and Subsequent Intranasal Challenge
  • This example describes a method of immunizing mice with GAS AI polypeptides and subsequently intranasally challenging the mice with GAS bacteria. Groups of 10 CD1 female mice aged between 6 and 7 weeks are immunized with a combination of GAS antigens of the invention GAS 15, GAS 16, and GAS 18, (15 μg of each recombinant antigen, derived from M1 strain SF370) or L. lactis expressing the M1 strain SF370 adhesin island, suspended in 100 μl of suitable solution. Each group receives 3 doses at days 0, 21 and 45. Immunization is performed through subcutaneous or intraperitoneal injection for the GAS 15, GAS 16, GAS 18 protein combination. The protein combination is administered with an equal volume of Complete Freund's Adjuvant (CFA) for the first dose and Incomplete Freund's Adjuvant (IFA) for the following two doses. Immunization is performed intranasally for the L. lactis expressing the M1 strain SF370 adhesin island. In each immunization scheme negative and positive control groups are used.
  • The negative control group for the mice immunized with the GAS 15, GAS 16, GAS 18 protein combination included mice immunized with PBS. The negative control group for the mice immunized with L. lactis expressing the M1 strain SF370 adhesin island, included mice immunized with either wildtype L. lactis or L. lactis transformed with the pAM401 expression vector lacking any cloned adhesin island sequence.
  • The positive control groups included mice immunized with purified M1 strain SF370 M protein.
  • Immunized mice are then anaesthetized with Zoletil and challenged intranasally with a 25 μL suspension containing 1.2×106 or 1.2×108 CFU of ISS 3348 in THB. Animals are observed daily and checked for survival.
  • EXAMPLE 21 Active Maternal Immunization Assay
  • As used herein, an Active Maternal Immunization assay refers to an in vivo protection assay where female mice are immunized with the test antigen composition. The female mice are then bred and their pups are challenged with a lethal dose of GBS. Serum titers of the female mice during the immunization schedule are measured as well as the survival time of the pups after challenge.
  • Mouse Immunization
  • Specifically, groups of 4 CD-1 outbred female mice 6-8 weeks old (Charles River Laboratories, Calco Italy) are immunized with one or more GBS antigens, (20 μg of each recombinant GBS antigen), suspended in 100 μl of PBS. Each group receives 3 doses at days 0, 21 and 35. Immunization is performed through intra-peritoneal injection of the protein with an equal volume of Complete Freund's Adjuvant (CFA) for the first dose and Incomplete Freund's Adjuvant (IFA) for the following two doses. In each immunization scheme negative and positive control groups are used.
  • Immune response is monitored by using serum samples taken on day 0 and 49. The sera are analyzed as pools from each group of mice.
  • Active Maternal Immunization
  • A maternal immunization/neonatal pup challenge model of GBS infection was used to verify the protective efficacy of the antigens in mice. The mouse protection study was adapted from Rodewald et al. (Rodewald et al. J. Infect. Diseases 166, 635 (1992)). In brief, CD-1 female mice (6-8 weeks old) were immunized before breeding, as described above. The mice received 20 μg of protein per dose when immunized with a single antigen and 60 μg of protein per dose (15 μg of each antigen) when immunized with the combination of antigens. Mice were bred 2-7 days after the last immunization. Within 48 h of birth, pups were injected intraperitoneally with 50 μl of GBS culture. Challenge inocula were prepared starting from frozen cultures diluted to the appropriate concentration with THB before use. In preliminary experiments (not shown), the challenge doses per pup for each strain tested were determined to cause 90% lethality. Survival of pups was monitored for 2 days after challenge. Protection was calculated as (percentage deadControl minus percentage deadVaccine) divided by percentage deadControl multiplied by 100. Data were evaluated for statistical significance by Fisher's exact test.
  • EMBODIMENTS OF THE INVENTION
  • The invention encompasses, but is not limited to, the embodiments enumerated below.
  • 1. An immunogenic composition comprising a purified Group B Streptococcus (GBS) adhesin island (AI) polypeptide in oligomeric form.
  • 2. The immunogenic composition of embodiment 1 wherein the GBS AI polypeptide is selected from a GBS AI-1.
  • 3. The immunogenic composition of embodiment 1 wherein the GBS AI polypeptide is selected from a GBS AI-2.
  • 1. An immunogenic composition comprising a purified Group B Streptococcus (GBS) adhesin island (AI) polypeptide in oligomeric form.
  • 2. The immunogenic composition of embodiment 1 wherein the GBS AI polypeptide is selected from a GBS AI-1.
  • 3. The immunogenic composition of embodiment 1 wherein the GBS AI polypeptide is selected from a GBS AI-2.
  • 4. The immunogenic composition of any of embodiments 1-3 wherein the GBS AI polypeptide comprises a sortase substrate motif.
  • 5. The immunogenic composition of embodiment 4 wherein the sortase substrate motif is an LPXTG motif.
  • 6. The immunogenic composition of embodiment 5 wherein the LPXTG motif is represented by the amino acid sequence XPXTG, wherein the X at amino acid position 1 is an L, an I, or an F and the X at amino acid position 3 is any amino acid residue.
  • 7. The immunogenic composition of any one of embodiments 1-3 wherein the GBS AI polypeptide affects the ability of GBS bacteria to adhere to epithelial cells.
  • 8. The immunogenic composition of any one of embodiments 1-3 wherein the GBS AI polypeptide affects the ability of GBS bacteria to invade epithelial cells.
  • 9. The immunogenic composition of any one of embodiments 1-3 wherein the GBS AI polypeptide affects the ability of GBS bacteria to translocate through an epithelial cell layer.
  • 10. The immunogenic composition of any one of embodiments 1-3 wherein the GBS AI polypeptide is capable of associating with an epithelial cell surface.
  • 11. The immunogenic composition of embodiment 10 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 12. The immunogenic composition of any of embodiments 1-3 wherein the GBS AI polypeptide is a full-length GBS AI protein.
  • 13. The immunogenic composition of any of embodiments 1-3 wherein the GBS AI polypeptide is a fragment of a full-length GBS AI protein.
  • 14. The immunogenic composition of embodiment 13 wherein the fragment comprises at least 7 contiguous amino acid residues of the GBS AI protein.
  • 15. The immunogenic composition of embodiment 2 wherein the GBS AI polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof.
  • 16. The immunogenic composition of embodiment 3 wherein the GBS AI polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof.
  • 17. The immunogenic composition of embodiment 15 wherein the GBS AI polypeptide is GBS 80.
  • 18. The immunogenic composition of any of embodiments 1-3 or 15-17 wherein the oligomeric form is a hyperoligomer.
  • 19. The immunogenic composition of any of embodiments 1-3, or 15-17 further comprising a Gram positive bacterium antigen not associated with an AI.
  • 20. The immunogenic composition of embodiment 19 wherein the antigen is selected from the group consisting of GBS 322 and GBS 276.
  • 21. The immunogenic composition of embodiment 20 wherein the antigen is GBS 322.
  • 22. An immunogenic composition comprising a purified Gram positive bacteria adhesin island (AI) polypeptide in an oligomeric form.
  • 23. The immunogenic composition of embodiment 22 wherein the Gram positive bacteria is of a genus selected from the group consisting of Streptococcus, Enterococcus, Staphylococcus, or Listeria.
  • 24. The immunogenic composition of embodiment 23 wherein the Gram positive bacteria is of the genus Streptococcus.
  • 25. The immunogenic composition of any of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide comprises a sortase substrate motif.
  • 26. The immunogenic composition of embodiment 25 wherein the sortase substrate motif is an LPXTG motif.
  • 27. The immunogenic composition of any one of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide affects the ability of Gram positive bacteria to adhere to epithelial cells.
  • 28. The immunogenic composition of any one of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide affects the ability of Gram positive bacteria to invade epithelial cells.
  • 29. The immunogenic composition of any one of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide affects the ability of Gram positive bacteria to translocate through an epithelial cell layer.
  • 30. The immunogenic composition of any one of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide is capable of associating with an epithelial cell surface.
  • 31. The immunogenic composition of embodiment 30 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 32. The immunogenic composition of any of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide is a full-length Gram positive bacteria AI protein.
  • 33. The immunogenic composition of any of embodiments 22-24 wherein the Gram positive bacteria AI polypeptide is a fragment of a full-length Gram positive bacteria AI protein.
  • 34. The immunogenic composition of embodiment 33 wherein the fragment comprises at least 7 contiguous amino acid residues of the Gram positive bacteria AI protein.
  • 35. The immunogenic composition of embodiment 24 wherein the genus Streptococcus bacteria is Group A Streptococcus (GAS) bacteria and the Gram positive bacteria AI polypeptide is a GAS AI polypeptide.
  • 36. The immunogenic composition of embodiment 35 wherein the GAS AI polypeptide is selected from a GAS AI-1.
  • 37. The immunogenic composition of embodiment 35 wherein the GAS AI polypeptide is selected from a GAS AI-2.
  • 38. The immunogenic composition of embodiment 35 wherein the GAS AI polypeptide is selected from a GAS AI-3.
  • 39. The immunogenic composition of embodiment 35 wherein the GAS AI polypeptide is selected from a GAS AI-4.
  • 40. The immunogenic composition of any of embodiments 35-39 wherein the GAS AI polypeptide comprises a sortase substrate motif.
  • 41. The immunogenic composition of embodiment 40 wherein the sortase substrate motif is an LPXTG motif.
  • 42. The immunogenic composition of embodiment 41 wherein the LPXTG motif is represented by XXXXG, wherein the X at the first amino acid position is an L, a V, an E, or a Q, wherein the X at the second amino acid position is P if the X at the first amino acid position is an L, the X at the second amino acid position is a V if the X at the first amino acid position is an E or a Q, or the X at the second amino acid position is a V or a P if the X at the first amino acid position is a V, wherein the X at the third amino acid position is any amino acid residue, and wherein the X at the fourth amino acid position is a T if the X at the first amino acid position is a V, an E, or a Q, or the X at the fourth amino acid position is a T, an S, or an A if the X at the first amino acid position is an L.
  • 43. The immunogenic composition of any one of embodiments 35-39 wherein the GAS AI polypeptide affects the ability of GAS bacteria to adhere to epithelial cells.
  • 44. The immunogenic composition of any one of embodiments 35-39 wherein the GAS AI polypeptide affects the ability of GAS bacteria to invade epithelial cells.
  • 45. The immunogenic composition of any one of embodiments 35-39 wherein the GAS AI polypeptide affects the ability of GAS bacteria to translocate through an epithelial cell layer.
  • 46. The immunogenic composition of any one of embodiments 35-39 wherein the GAS AI polypeptide is capable of associating with an epithelial cell surface.
  • 47. The immunogenic composition of embodiment 46 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 48. The immunogenic composition of any of embodiments 35-39 wherein the GAS AI polypeptide is a full-length GAS AI protein.
  • 49. The immunogenic composition of any of embodiments 35-39 wherein the GAS AI polypeptide is a fragment of a full-length GAS AI protein.
  • 50. The immunogenic composition of embodiment 49 wherein the fragment comprises at least 7 contiguous amino acid residues of the GAS AI protein.
  • 51. The immunogenic composition of embodiment 36 wherein the GAS AI-1 polypeptide is selected from the group consisting of M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, DSM2071_fimbrial, and fragments thereof.
  • 52. The immunogenic composition of embodiment 37 wherein the GAS AI-2 polypeptide is selected from the group consisting of GAS 15, GAS 16, GAS 18, and fragments thereof.
  • 53. The immunogenic composition of embodiment 38 wherein the GAS AI-3 polypeptide is selected from the group consisting of SpyM30098, SpyM3 0100, SpyM3 0102, SpyM30104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM180126, spyM18 0128, spyM180130, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, ISS4959_fimbrial, and fragments thereof.
  • 53. The immunogenic composition of embodiment 39 wherein the GAS AI-4 polypeptide is selected from the group consisting of 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial, and fragments thereof.
  • 54. The immunogenic composition of embodiment 24 wherein the Streptococcus bacteria is Streptococcus pneumoniae and the Gram positive bacteria AI polypeptide is a S. pneumoniae AI polypeptide.
  • 55. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide comprises a sortase substrate motif.
  • 56. The immunogenic composition of embodiment 55 wherein the sortase substrate motif is an LPXTG motif.
  • 57. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to adhere to epithelial cells.
  • 58. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to invade epithelial cells.
  • 59. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to translocate through an epithelial cell layer.
  • 60. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide is capable of associating with an epithelial cell surface.
  • 61. The immunogenic composition of embodiment 60 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 62. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide is a full-length S. pneumoniae AI protein.
  • 63. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide is a fragment of a full-length S. pneumoniae AI protein.
  • 64. The immunogenic composition of embodiment 63 wherein the fragment comprises at least 7 contiguous amino acid residues of the S. pneumoniae AI protein.
  • 65. The immunogenic composition of embodiment 54 wherein the S. pneumoniae AI polypeptide is selected from the group consisting of SP0462, SP0463, SP0464, orf3 670, orf4 670, orf5 670, ORF314CSR, ORF414CSR, ORF514CSR, ORF319AH, ORF419AH, ORF519AH, ORF319FTW, ORF419FTW, ORF519FTW, ORF323FP, ORF423FP, ORF523FP, ORF323FTW, ORF423FTW, ORF523FTW, ORF36BF, ORF46BF, ORF56BF, ORF36BSP, ORF46BSP, ORF56BSP, ORF39VSP, ORF49VSP, ORF59VSP, and fragments thereof.
  • 66. The immunogenic composition of any one of embodiments 22-24, 35-39, 51-54, or 65 wherein the oligomeric form is a hyperoligomer.
  • 67. The immunogenic composition of any one of embodiments 22-24, 35-39, 51-54, or 65 further comprising a Gram positive bacteria antigen not associated with an AI.
  • 68. The immunogenic composition of embodiment 67 wherein the antigen is selected from the group consisting of GBS 322 and GBS 276.
  • 69. An immunogenic composition comprising a first and a second Group B Streptococcus (GBS) adhesin island (AI) polypeptide.
  • 70. The immunogenic composition of embodiment 69 wherein a full-length polynucleotide sequence encoding for the first GBS AI polypeptide is not present in a GBS bacteria genome comprising a polynucleotide sequence encoding for the second GBS AI polypeptide.
  • 71. The immunogenic composition of embodiment 69 wherein polynucleotides encoding the first and the second GBS AI polypeptide are each present in genomes of more than one GBS serotype and strain isolate.
  • 72. The immunogenic composition of embodiment 69 wherein the first GBS AI polypeptide is encoded by a GBS AI-1.
  • 73. The immunogenic composition of embodiment 69 wherein the first GBS AI polypeptide is encoded by a GBS AI-2.
  • 74. The immunogenic composition of embodiment 72 wherein the second GBS AI polypeptide is encoded by a GBS AI-2.
  • 75. The immunogenic composition of embodiment 73 wherein the second GBS AI polypeptide is encoded by a GBS AI-2.
  • 76. The immunogenic composition of embodiment 72 wherein the second GBS AI polypeptide is encoded by a GBS AI-1.
  • 77. The immunogenic composition of embodiment 73 wherein the second GBS AI polypeptide is encoded by a GBS AI-1.
  • 78. The immunogenic composition of embodiment 72 wherein the first GBS AI polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof.
  • 79. The immunogenic composition of embodiment 73 wherein the first GBS AI polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof.
  • 80. The immunogenic composition of embodiment 74 or 75 wherein the second GBS AI polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof, and wherein the first and the second GBS AI polypeptide are not the same polypeptide.
  • 81. The immunogenic composition of embodiment 76 or 77 wherein the second GBS AI polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof, and wherein the first and the second GBS AI polypeptide are not the same polypeptide.
  • 82. The immunogenic composition of any one of embodiments 69-77 wherein the first GBS AI polypeptide comprises a sortase substrate motif.
  • 83. The immunogenic composition of embodiment 82 wherein the sortase substrate motif is an LPXTG motif.
  • 84. The immunogenic composition of embodiment 83 wherein the LPXTG motif is represented by the sequence XPXTG, wherein the X at amino acid position 1 is an L, an I, or an F and the X at amino acid position 3 is any amino acid residue.
  • 85. The immunogenic composition of any one of embodiments 69-77 wherein the first GBS AI polypeptide affects the ability of GBS bacteria to adhere to epithelial cells.
  • 86. The immunogenic composition of any one of embodiments 69-77 wherein the first GBS AI polypeptide affects the ability of GBS bacteria to invade epithelial cells.
  • 87. The immunogenic composition of any one of embodiments 69-77 wherein the first GBS AI polypeptide affects the ability of GBS bacteria to translocate through an epithelial cell layer.
  • 88. The immunogenic composition of any one of embodiments 69-77 wherein the first GBS AI polypeptide is capable of associating with an epithelial cell surface.
  • 89. The immunogenic composition of embodiment 88 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 90. The immunogenic composition of any of embodiments 69-77 wherein the first GBS AI polypeptide is a full-length GBS AI protein.
  • 91. The immunogenic composition of any of embodiments 69-77 wherein the first GBS AI polypeptide is a fragment of a full-length GBS AI protein.
  • 92. The immunogenic composition of embodiment 91 wherein the fragment comprises at least 7 contiguous amino acid residues of the first GBS AI protein.
  • 93. The immunogenic composition of any one of embodiments 69-79 wherein the first GBS AI polypeptide is in oligomeric form.
  • 94. The immunogenic composition of any one of embodiments 69-77 wherein the second GBS AI polypeptide is in oligomeric form.
  • 95. The immunogenic composition of any one of embodiments 69-79 wherein the first and the second GBS AI polypeptide are associated in a single oligomeric form.
  • 96. The immunogenic composition of embodiment 95 wherein the first and the second GBS AI polypeptides are chemically associated.
  • 97. The immunogenic composition of embodiment 95 wherein the first and the second GBS AI polypeptides are physically associated.
  • 98. The immunogenic composition of embodiment 93 wherein the oligomeric form is a hyperoligomer.
  • 99. The immunogenic composition of embodiment 94 wherein the oligomeric form is a hyperoligomer.
  • 100. The immunogenic composition of embodiment 76 wherein the first GBS AI polypeptide is GBS 80 and the second GBS AI polypeptide is GBS 104.
  • 101. The immunogenic composition of embodiment 74 wherein the first GBS AI polypeptide is GBS 80 and the second GBS AI polypeptide is GBS 67.
  • 102. The immunogenic composition of any one of embodiments 69-79, 100, or 101 further comprising a GBS polypeptide not associated with an AI.
  • 103. The immunogenic composition of embodiment 102 wherein the GBS polypeptide not associated with an AI is selected from the group consisting of GBS 322 and GBS 276.
  • 104. The immunogenic composition of embodiment 103 wherein the GBS polypeptide not associated with an AI is GBS 322.
  • 105. An immunogenic composition comprising a first and a second Gram positive bacteria adhesin island (AI) polypeptide.
  • 106. The immunogenic composition of embodiment 105 wherein a full length polynucleotide sequence encoding for the first Gram positive bacteria AI polypeptide is not present in a genome of a Gram positive bacteria comprising a full length polynucleotide sequence encoding for the second Gram positive bacteria AI polypeptide.
  • 107. The immunogenic composition of embodiment 105 wherein polynucleotides encoding the first and the second Gram positive bacteria AI polypeptide are each present in genomes of more than one Gram positive bacteria serotype and strain isolate.
  • 108. The immunogenic composition of embodiment 105 wherein the first and the second Gram positive bacteria AI polypeptides are of different Gram positive bacteria species.
  • 109. The immunogenic composition of embodiment 105 wherein the first and the second Gram positive bacteria AI polypeptides are of the same Gram positive bacteria species.
  • 110. The immunogenic composition of embodiment 105 wherein the first and the second Gram positive bacteria AI polypeptides are from different AI subtypes.
  • 111. The immunogenic composition of embodiment 105 wherein the first and the second Gram positive bacteria AI polypeptides are from the same AI subtype.
  • 112. The immunogenic composition of embodiment 105 wherein the first Gram positive bacteria AI polypeptide has detectable surface exposure on a first Gram positive bacteria strain or serotype but not a second Gram positive bacteria strain or subtype and the second Gram positive bacteria AI polypeptide has detectable surface exposure on the second Gram positive bacteria strain or serotype but not the first Gram positive bacteria strain or serotype.
  • 113. The immunogenic composition of embodiment 105 wherein the Gram positive bacteria is S. pneumonaie, S. mutans, E. faecalis, E. faecium, C. difficile, L. monocytogenes, or C. diphtheriae.
  • 114. The immunogenic composition of any of embodiments 105-113 wherein the first and the second Gram positive bacteria AI polypeptides comprise a sortase substrate motif.
  • 115. The immunogenic composition of embodiment 114 wherein the sortase substrate motif is an LPXTG motif.
  • 116. The immunogenic composition of embodiment 115 wherein the LPXTG motif is represented by XXXXG, wherein the X at amino acid position 1 is an L, a V, an E, an I, an F, or a Q, wherein X at amino acid position 2 is a P if X at amino acid position 1 is an L, an I, or an F, wherein X at amino acid position 2 is a V if X at amino acid position 1 is a E or a Q, wherein X at amino acid position 2 is a V or a P if X at amino acid position 1 is a V, wherein X at amino acid position 3 is any amino acid residue, wherein X at amino acid position 4 is a T if X at amino acid position 1 is a V, E, I, F, or Q, and wherein X at amino acid position 4 is a T, S, or A if X at amino acid position 1 is an L.
  • 117. The immunogenic composition of embodiment 105 wherein the first Gram positive bacteria AI polypeptide is a first Group A Streptococcus (GAS) AI polypeptide.
  • 118. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide comprises a sortase substrate motif.
  • 119. The immunogenic composition of embodiment 118 wherein the sortase substrate motif is an LPXTG motif.
  • 120. The immunogenic composition of embodiment 119 wherein the LPXTG motif is represented by XXXXG, wherein the X at the first amino acid position is an L, a V, an E, or a Q, wherein the X at the second amino acid position is P if the X at the first amino acid position is an L, the X at the second amino acid position is a V if the X at the first amino acid position is an E or a Q, or the X at the second amino acid position is a V or a P if the X at the first amino acid position is a V, wherein the X at the third amino acid position is any amino acid residue, and wherein the X at the fourth amino acid position is a T if the X at the first amino acid position is a V, an E, or a Q, or the X at the fourth amino acid position is a T, an S, or an A if the X at the first amino acid position is an L.
  • 121. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide affects the ability of GAS bacteria to adhere to epithelial cells.
  • 122. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide affects the ability of GAS bacteria to invade epithelial cells.
  • 123. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide affects the ability of GAS bacteria to translocate through an epithelial cell layer.
  • 124. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is capable of associating with an epithelial cell surface.
  • 125. The immunogenic composition of embodiment 117 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 126. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a full-length GAS AI protein.
  • 127. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a fragment of a full-length GAS AI protein.
  • 128. The immunogenic composition of embodiment 127 wherein the fragment comprises at least 7 contiguous amino acid residues of the GAS AI protein.
  • 129. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a first GAS AI-1 polypeptide.
  • 130. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a first GAS AI-2 polypeptide.
  • 131. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a first GAS AI-3 polypeptide.
  • 132. The immunogenic composition of embodiment 117 wherein the first GAS AI polypeptide is a first GAS AI-4 polypeptide.
  • 133. The immunogenic composition of any one of embodiments 117 or 129-132 wherein the second Gram positive bacteria AI polypeptide is a second GAS AI polypeptide.
  • 134. The immunogenic composition of embodiment 133 wherein the second GAS AI polypeptide is a second GAS AI-1 polypeptide.
  • 135. The immunogenic composition of embodiment 133 wherein the second GAS AI polypeptide is a second GAS AI-2 polypeptide.
  • 136. The immunogenic composition of embodiment 133 wherein the second GAS AI polypeptide is a second GAS AI-3 polypeptide.
  • 137. The immunogenic composition of embodiment 133 wherein the second GAS AI polypeptide is a second GAS AI-4 polypeptide.
  • 138. The immunogenic composition of embodiment 129 wherein the first GAS AI-1 polypeptide is selected from the group consisting of M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, DSM2071_fimbrial, and fragments thereof.
  • 139. The immunogenic composition of embodiment 130 wherein the first GAS AI-2 polypeptide is selected from the group consisting of GAS 15, GAS 16, GAS 18, and fragments thereof.
  • 140. The immunogenic composition of embodiment 131 wherein the first GAS AI-3 polypeptide is selected from the group consisting of SpyM30098, SpyM3 0100, SpyM3 0102, SpyM30104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM180126, spyM18 0128, spyM180130, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, ISS4959_fimbrial, and fragments thereof.
  • 141. The immunogenic composition of embodiment 132 wherein the first GAS AI-4 polypeptide is selected from the group consisting of 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial, and fragments thereof.
  • 142. The immunogenic composition of embodiment 134 wherein the second GAS AI-1 polypeptide is selected from the group consisting of M6_Spy0157, M6_Spy0159, M6_Spy0160, CDC SS 410_fimbrial, ISS3650_fimbrial, DSM2071_fimbrial, and fragments thereof.
  • 143. The immunogenic composition of embodiment 135 wherein the second GAS AI-2 polypeptide is selected from the group consisting of GAS15, GAS16, GAS18, and fragments thereof.
  • 144. The immunogenic composition of embodiment 136 wherein the second GAS AI-3 polypeptide is selected from the group consisting of SpyM30098, SpyM3 0100, SpyM3 0102, SpyM30104, SPs0100, SPs0102, SPs0104, SPs0106, orf78, orf80, orf82, orf84, spyM180126, spyM18 0128, spyM180130, spyM180132, SpyoM01000156, SpyoM01000155, SpyoM01000154, SpyoM01000153, SpyoM01000152, SpyoM01000151, SpyoM01000150, SpyoM01000149, ISS3040_fimbrial, ISS3776_fimbrial, ISS4959_fimbrial, and fragments thereof.
  • 145. The immunogenic composition of embodiment 137 wherein the second GAS AI-4 polypeptide is selected from the group consisting of 19224134, 19224135, 19224137, 19224139, 19224141, 20010296_fimbrial, 20020069_fimbrial, CDC SS 635_fimbrial, ISS4883_fimbrial, ISS4538_fimbrial, and fragments thereof.
  • 146. The immunogenic composition of any one of embodiments 117-132 or 138-141 wherein the second Gram positive bacteria AI polypeptide is a Group B Streptococcus (GBS) AI polypeptide.
  • 147. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide comprises a sortase substrate motif.
  • 148. The immunogenic composition of embodiment 147 wherein the sortase substrate motif is an LPXTG motif.
  • 149. The immunogenic composition of embodiment 148 wherein the LPXTG motif is represented by the amino acid sequence XPXTG, wherein the X at amino acid position 1 is an L, an I, or an F and the X at amino acid position 3 is any amino acid residue.
  • 150. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide affects the ability of GBS bacteria to adhere to epithelial cells.
  • 151. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide affects the ability of GBS bacteria to invade epithelial cells.
  • 152. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide affects the ability of GBS bacteria to translocate through an epithelial cell layer.
  • 153. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide is capable of associating with an epithelial cell surface.
  • 154. The immunogenic composition of embodiment 146 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 155. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide is a full-length GBS AI protein.
  • 156. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide is a fragment of a full-length GBS AI protein.
  • 157. The immunogenic composition of embodiment 156 wherein the fragment comprises at least 7 contiguous amino acid residues of the GBS AI protein.
  • 158. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide is a GBS AI-1 polypeptide.
  • 159. The immunogenic composition of embodiment 146 wherein the GBS AI polypeptide is a GBS AI-2 polypeptide.
  • 160. The immunogenic composition of embodiment 158 wherein the GBS AI-1 polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof.
  • 161. The immunogenic composition of embodiment 159 wherein the GBS AI-2 polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof.
  • 162. The immunogenic composition of any one of embodiments 117-132 or 138-141 wherein the second Gram positive bacteria AI polypeptide is a Streptococcus pneumoniae AI polypeptide.
  • 163. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide comprises a sortase substrate motif.
  • 164. The immunogenic composition of embodiment 163 wherein the sortase substrate motif is an LPXTG motif.
  • 165. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to adhere to epithelial cells.
  • 166. The immunogenic composition of embodiment 162 S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to invade epithelial cells.
  • 167. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide affects the ability of S. pneumoniae to translocate through an epithelial cell layer.
  • 168. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide is capable of associating with an epithelial cell surface.
  • 169. The immunogenic composition of embodiment 168 wherein the associating with an epithelial cell surface is binding to the epithelial cell surface.
  • 170. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide is a full-length S. pneumoniae AI protein.
  • 171. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide is a fragment of a full-length S. pneumoniae AI protein.
  • 172. The immunogenic composition of embodiment 162 wherein the fragment comprises at least 7 contiguous amino acid residues of the S. pneumoniae AI protein.
  • 173. The immunogenic composition of embodiment 162 wherein the S. pneumoniae AI polypeptide is selected from the group consisting of SP0462, SP0463, SP0464, orf3 670, orf4 670, orf5 670, ORF314CSR, ORF414CSR, ORF514CSR, ORF319AH, ORF419AH, ORF519AH, ORF319FTW, ORF419FTW, ORF519FTW, ORF323FP, ORF423FP, ORF523FP, ORF323FTW, ORF423FTW, ORF523FTW, ORF36BF, ORF46BF, ORF56BF, ORF36BSP, ORF46BSP, ORF56BSP, ORF39VSP, ORF49VSP, ORF59VSP, and fragments thereof.
  • 174. The immunogenic composition of any one of embodiments 105-117 wherein the first Gram positive bacteria AI polypeptide is in oligomeric form.
  • 175. The immunogenic composition of embodiment 174 wherein the oligomeric form is a hyperoligomer.
  • 176. The immunogenic composition of embodiment 174 wherein the second Gram positive bacteria AI polypeptide is in oligomeric form.
  • 177. The immunogenic composition of embodiment 176 wherein the oligomeric form is a hyperoligomer.
  • 178. The immunogenic composition of embodiment 176 wherein the first and the second Gram positive bacteria AI polypeptide are associated in a single oligomeric form.
  • 179. The immunogenic composition of embodiment 178 wherein the first and the second Gram positive bacteria AI polypeptide are chemically associated.
  • 180. The immunogenic composition of embodiment 178 wherein the first and the second Gram positive bacteria AI polypeptide are physically associated.
  • 181. The immunogenic composition of any one of embodiments 105-117 further comprising a Gram positive bacteria polypeptide not associated with an AI.
  • 182. The immunogenic composition of embodiment 181 wherein the Gram positive bacteria polypeptide not associated with an AI is selected from the group consisting of GBS 322 and GBS 276.
  • 183. The immunogenic composition of embodiment 182 wherein the Gram positive bacteria polypeptide not associated with an AI is GBS 322.
  • 184. A modified Gram positive bacterium adapted to produce increased levels of AI surface protein.
  • 185. The modified Gram positive bacterium of embodiment 184 wherein the AI surface protein is in oligomeric form.
  • 186. The modified Gram positive bacterium of embodiment 185 wherein the oligomeric form is a hyperoligomer.
  • 187. The modified Gram positive bacterium of any one of embodiments 184-186 which is a Group B Streptococcus bacterium.
  • 188. The modified Gram positive bacterium of any one of embodiments 184-186 which is a Group A Streptococcus bacterium.
  • 189. The modified Gram positive bacterium of any one of embodiments 184-186 which is a non-pathogenic Gram positive bacterium.
  • 190. The modified Gram positive bacterium of embodiment 189 wherein the non-pathogenic Gram positive bacterium is Streptococus gordonii.
  • 191. The modified Gram positive bacterium of embodiment 189 wherein the non-pathogenic Gram positive bacterium is Lactococcus lactis.
  • 192. The modified Gram positive bacterium of any one of embodiments 184-186 which has been inactivated and wherein the AI surface protein is exposed on the surface of the Gram positive bacterium.
  • 193. The modified Gram positive bacterium of any one of embodiments 184-186 which has been attenuated and wherein the AI surface protein is exposed on the surface of the Gram positive bacterium.
  • 194. The modified GBS bacterium of embodiment 187 which has been inactivated and wherein the AI surface protein is exposed on the surface of the GBS bacterium.
  • 195. The modified GBS bacterium of embodiment 187 which has been attenuated and wherein the AI surface protein is exposed on the surface of the GBS bacterium.
  • 196. The modified GAS bacterium of embodiment 188 which has been inactivated and wherein the AI surface protein is exposed on the surface of the GAS bacterium.
  • 197. The modified GAS bacterium of embodiment 188 which has been attenuated and wherein the AI surface protein is exposed on the surface of the GAS bacterium.
  • 198. The modified non-pathogenic bacterium of embodiment 189 which has been inactivated and wherein the AI surface protein is exposed on the surface of the non-pathogenic Gram positive bacterium.
  • 199. The modified non-pathogenic bacterium of embodiment 189 which has been attenuated and wherein the AI surface protein is exposed on the surface of the non-pathogenic Gram positive bacterium.
  • 200. A method for manufacturing an oligomeric adhesin island (AI) surface antigen comprising:
  • culturing a Gram positive bacterium that expresses an oligomeric AI surface antigen and
  • isolating the expressed oligomeric AI surface antigen.
  • 201. The method of embodiment 200 wherein the step of isolating is performed by collecting said oligomeric AI surface antigen from Gram positive bacterium secretions in the Gram positive bacterium culture.
  • 202. The method of embodiment 200 further comprising a step of purifying.
  • 203. The method of embodiment 202 wherein the oligomeric AI surface antigen is purified from the Gram positive bacterium cell surface.
  • 204. The method of embodiment 200 wherein the Gram positive bacterium is adapted for increased AI protein expression.
  • 205. The method of any one of embodiments 200-204 wherein the Gram positive bacterium is a Group A Streptococcus bacterium.
  • 206. The method of any one of embodiments 200-204 wherein the Gram positive bacterium is a Group B Streptococcus bacterium.
  • 207. The method of any one of embodiments 200-204 wherein the oligomeric AI surface antigen is in hyperoligomeric form.
  • 208. The method of embodiment 200 wherein the Gram positive bacterium expresses the oligomeric AI surface antigen recombinantly.
  • 209. The method of embodiment 208 wherein the Gram positive bacterium further manipulated expresses at least 1 AI sortase.
  • 210. The modified Gram positive bacterium of any one of embodiments 184-186 which is a S. pneumoniae bacterium.
  • 211. The method of any one of embodiments 200-204 wherein the Gram positive bacterium is S. pneumoniae.

Claims (40)

1. An immunogenic composition comprising a purified Group B Streptococcus (GBS) adhesin island (AI) polypeptide in oligomeric form.
2. The immunogenic composition of claim 1 wherein the GBS AI polypeptide is selected from a GBS AI-1.
3. The immunogenic composition of claim 1 wherein the GBS AI polypeptide is selected from a GBS AI-2.
4. The immunogenic composition of claim 2 wherein the GBS AI polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof.
5. The immunogenic composition of claim 3 wherein the GBS AI polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof.
6. The immunogenic composition of claim 4 wherein the GBS AI polypeptide is GBS 80.
7. The immunogenic composition of any of claims 1-6 wherein the oligomeric form is a hyperoligomer.
8 (22). An immunogenic composition comprising a purified Gram positive bacteria adhesin island (AI) polypeptide in an oligomeric form.
9 (23). The immunogenic composition of claim 8 wherein the Gram positive bacteria is of a genus selected from the group consisting of Streptococcus, Enterococcus, Staphylococcus, Clostridium, Corynebacterium, or Listeria.
10 (24). The immunogenic composition of claim 9 wherein the Gram positive bacteria is of the genus Streptococcus.
11 (35). The immunogenic composition of claim 10 wherein the genus Streptococcus bacteria is Group A Streptococcus (GAS) bacteria and the Gram positive bacteria AI polypeptide is a GAS AI polypeptide.
12 (36). The immunogenic composition of claim 11 wherein the GAS AI polypeptide is selected from a GAS AI-1.
13 (37). The immunogenic composition of claim 11 wherein the GAS AI polypeptide is selected from a GAS AI-2.
14 (38). The immunogenic composition of claim 11 wherein the GAS AI polypeptide is selected from a GAS AI-3.
15 (39). The immunogenic composition of claim 11 wherein the GAS AI polypeptide is selected from a GAS AI-4.
16 (66). The immunogenic composition of any one of claims 8-15 wherein the oligomeric form is a hyperoligomer.
17. An immunogenic composition comprising a first and a second Group B Streptococcus (GBS) adhesin island (AI) polypeptide.
18. The immunogenic composition of claim 17 wherein the first GBS AI polypeptide is encoded by a GBS AI-1.
19. The immunogenic composition of claim 18 wherein the second GBS AI polypeptide is encoded by a GBS AI-2.
20. The immunogenic composition of claim 18 wherein the first GBS AI polypeptide is selected from the group consisting of GBS 80, GBS 104, GBS 52, and fragments thereof.
21. The immunogenic composition of claim 19 wherein the second GBS AI polypeptide is selected from the group consisting of GBS 59, GBS 67, GBS 150, 01521, 01523, 01524, and fragments thereof, and wherein the first and the second GBS AI polypeptide are not the same polypeptide.
22. The immunogenic composition of claim 19 wherein the first GBS AI polypeptide is GBS 80 and the second GBS AI polypeptide is GBS 67.
23. An immunogenic composition comprising a first and a second Gram positive bacteria adhesin island (AI) polypeptide.
24. The immunogenic composition of claim 23 wherein the Gram positive bacteria is Streptococcus, Enterococcus, Staphylococcus, Clostridium, Corynebacterium, or Listeria.
25. The immunogenic composition of claim 23 wherein the first Gram positive bacteria AI polypeptide is a first Group A Streptococcus (GAS) AI polypeptide.
26. The immunogenic composition of claim 25 wherein the first GAS AI polypeptide is a first GAS AI-1 polypeptide.
27. The immunogenic composition of claim 25 wherein the first GAS AI polypeptide is a first GAS AI-2 polypeptide.
28. The immunogenic composition of claim 25 wherein the first GAS AI polypeptide is a first GAS AI-3 polypeptide.
29. The immunogenic composition of claim 25 wherein the first GAS AI polypeptide is a first GAS AI-4 polypeptide.
30. The immunogenic composition of any one of claims 25-29 wherein the second Gram positive bacteria AI polypeptide is a second GAS AI polypeptide.
31. The immunogenic composition of claim 30 wherein the second GAS AI polypeptide is a second GAS AI-1 polypeptide.
32. The immunogenic composition of claim 30 wherein the second GAS AI polypeptide is a second GAS AI-2 polypeptide.
33. The immunogenic composition of claim 30 wherein the second GAS AI polypeptide is a second GAS AI-3 polypeptide.
34. The immunogenic composition of claim 30 wherein the second GAS AI polypeptide is a second GAS AI-4 polypeptide.
35. A modified Gram positive bacterium adapted to produce increased levels of AI surface protein.
36. The modified Gram positive bacterium of claim 35 wherein the AI surface protein is in oligomeric form.
37. The modified Gram positive bacterium of claim 36 wherein the oligomeric form is a hyperoligomer.
38. The modified Gram positive bacterium of any one of claims 35-37 which is a non-pathogenic Gram positive bacterium.
39. The modified Gram positive bacterium of claim 38 wherein the non-pathogenic Gram positive bacterium is Lactococcus lactis.
40. A method for manufacturing an oligomeric adhesin island (AI) surface antigen comprising:
culturing a Gram positive bacterium that expresses an oligomeric AI surface antigen and
isolating the expressed oligomeric AI surface antigen.
US11/192,046 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae Abandoned US20060165716A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/192,046 US20060165716A1 (en) 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US12/906,510 US8778358B2 (en) 2004-07-29 2010-10-18 Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US59280504P 2004-07-29 2004-07-29
US60983304P 2004-09-13 2004-09-13
US61683304P 2004-10-08 2004-10-08
US63341804P 2004-12-07 2004-12-07
US64006904P 2004-12-30 2004-12-30
US66032105P 2005-03-11 2005-03-11
US67375405P 2005-04-22 2005-04-22
US69300105P 2005-06-21 2005-06-21
US69545305P 2005-07-01 2005-07-01
US69764305P 2005-07-11 2005-07-11
US11/192,046 US20060165716A1 (en) 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/906,510 Continuation US8778358B2 (en) 2004-07-29 2010-10-18 Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae

Publications (1)

Publication Number Publication Date
US20060165716A1 true US20060165716A1 (en) 2006-07-27

Family

ID=36692677

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/192,046 Abandoned US20060165716A1 (en) 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US11/658,842 Abandoned US20090317420A1 (en) 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US12/906,510 Expired - Fee Related US8778358B2 (en) 2004-07-29 2010-10-18 Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/658,842 Abandoned US20090317420A1 (en) 2004-07-29 2005-07-29 Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US12/906,510 Expired - Fee Related US8778358B2 (en) 2004-07-29 2010-10-18 Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae

Country Status (5)

Country Link
US (3) US20060165716A1 (en)
EP (2) EP2612679A1 (en)
JP (2) JP2008508320A (en)
CA (1) CA2575548A1 (en)
WO (1) WO2006078318A2 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060210581A1 (en) * 2000-10-27 2006-09-21 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A & B
US20060252691A1 (en) * 1997-12-19 2006-11-09 Naomi Balaban Methods and compositions for the treatment and prevention of staphylococcus and other bacterial infections
US20070065464A1 (en) * 2003-07-31 2007-03-22 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
US20080031877A1 (en) * 2006-02-17 2008-02-07 Antonello Covacci Purification of bacterial antigens
US20080152701A1 (en) * 1997-12-19 2008-06-26 Naomi Balaban Methods and Compositions for the Treatment and Prevention of Staphylococcus Infections
US20080219976A1 (en) * 1997-12-19 2008-09-11 Naomi Balaban Methods and compositions for treatment and prevention of staphylococcal infections
US20080220010A1 (en) * 2003-09-15 2008-09-11 Novartis Vaccines And Diagnostics Srl Immunogenic Compositions for Streptococcus Agalactiae
US20090117113A1 (en) * 2004-10-08 2009-05-07 Chiron Corporation Immunogenic And Therapeutic Compositions For Streptococcus Pyogenes
US20090147854A1 (en) * 2007-12-10 2009-06-11 Qualcomm Incorporated Selective display of interpolated or extrapolaed video units
US20090162392A1 (en) * 2007-12-21 2009-06-25 Novartis Ag Mutant forms of streptolysin o
US20090317420A1 (en) * 2004-07-29 2009-12-24 Chiron Corporation Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US20100150943A1 (en) * 2006-07-26 2010-06-17 Novartis Ag Immunogenic compositions for gram positive bacteria
US20100303864A1 (en) * 2002-08-26 2010-12-02 Herve Tettelin Conserved and specific streptococcal genomes
US20110038879A1 (en) * 2006-10-30 2011-02-17 Novartis Ag Immunogenic and therapeutic compositions for streptococcus pyogenes
US20110206692A1 (en) * 2006-06-09 2011-08-25 Novartis Ag Conformers of bacterial adhesins
WO2011104632A1 (en) * 2010-02-26 2011-09-01 Novartis Ag Immunogenic proteins and compositions
US20120076814A1 (en) * 2009-06-01 2012-03-29 Novartis Ag Combinations of pneumococcal rrgb clades
US8287885B2 (en) 2007-09-12 2012-10-16 Novartis Ag GAS57 mutant antigens and GAS57 antibodies
US20140086950A1 (en) * 2012-09-24 2014-03-27 Montana State University Recombinant lactococcus lactis expressing escherichia coli colonization factor antigen i (cfa/i) fimbriae and their methods of use
US9777076B2 (en) 2012-07-16 2017-10-03 Pfizer Inc. Saccharides and uses thereof
US10391161B2 (en) * 2016-12-19 2019-08-27 I-Shou University FSBM recombinant protein
EP3872096A4 (en) * 2018-10-25 2022-11-30 Ajinomoto Co., Inc. Method for secretory production of protein

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7989160B2 (en) 2006-02-13 2011-08-02 Alethia Biotherapeutics Inc. Polynucleotides and polypeptide sequences involved in the process of bone remodeling
US8168181B2 (en) 2006-02-13 2012-05-01 Alethia Biotherapeutics, Inc. Methods of impairing osteoclast differentiation using antibodies that bind siglec-15
US8465751B2 (en) * 2009-01-12 2013-06-18 Novartis Ag Cna—B domain antigens in vaccines against gram positive bacteria
ITMI20090946A1 (en) 2009-05-28 2010-11-29 Novartis Ag EXPRESSION OF RECOMBINANT PROTEINS
US20120237536A1 (en) * 2009-09-10 2012-09-20 Novartis Combination vaccines against respiratory tract diseases
WO2011040133A1 (en) * 2009-10-02 2011-04-07 学校法人 東京女子医科大学 Human serum amyloid-a3 antibody and use thereof
GB201114923D0 (en) * 2011-08-30 2011-10-12 Novartis Ag Immunogenic proteins and compositions
DE102011121069A1 (en) * 2011-12-13 2013-06-13 Martin-Luther-Universität Halle-Wittenberg Vaccination by recombinant yeast by generating a protective humoral immune response against defined antigens
CA2928851A1 (en) 2012-07-19 2014-01-23 Alethia Biotherapeutics Inc. Anti-siglec-15 antibodies
AR101455A1 (en) * 2014-08-05 2016-12-21 Glaxosmithkline Biologicals Sa CARRIER MOLECULE

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070036828A1 (en) * 2002-09-13 2007-02-15 Chiron Corporation Group b streptococcus vaccine

Family Cites Families (172)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU548947A1 (en) 1975-09-05 1980-02-05 Всесоюзный Ордена Ленина Институт Экспериментальной Ветеринарии Васхнил Vaccine for specific prophylaxis and therapy of horse trichophytosis
NO153458C (en) 1978-06-07 1986-03-26 Inst Experimentalnoi Veterinar PROCEDURE FOR THE PREPARATION OF PROPHYLAXY VACCINE AND TREATMENT OF RING WORM OF FUR ANIMALS AND Rabbits CAUSED BY TRICHOPHYTON MENTAGROPHYTES.
US5360897A (en) 1981-08-31 1994-11-01 The University Of Rochester Immunogenic conjugates of streptococcus pneumonial capsular polymer and toxin or in toxiad
US4454121A (en) * 1982-07-27 1984-06-12 The University Of Tennessee Research Corporation Synthetic peptides corresponding to antigenic determinants of the M protein of Streptococcus pyogenes
SE8205892D0 (en) 1982-10-18 1982-10-18 Bror Morein IMMUNOGENT MEMBRANE PROTEIN COMPLEX, SET FOR PREPARATION AND USE THEREOF
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
FR2550094B1 (en) * 1983-08-01 1986-03-21 Anvar VACCINE COMPOSITIONS CONDITIONED FOR THE VACCINATION OF IMMUNITARY NON-NATIVE SUBJECTS AGAINST A DETERMINED PATHOGENIC AGENT AND CONTAINING A HAPTENA WHICH INCLUDES A CHARACTERISTIC ANTIGENIC SITE OF SAID PATHOGENIC AGENT OR AN OLIGOMER OF THIS HAPTEN
US5324513A (en) 1984-03-07 1994-06-28 Institut Pasteur Composition useful for the fabrication of vaccines
US5171568A (en) 1984-04-06 1992-12-15 Chiron Corporation Recombinant herpes simplex gb-gd vaccine
US6090406A (en) 1984-04-12 2000-07-18 The Liposome Company, Inc. Potentiation of immune responses with liposomal adjuvants
US5916588A (en) 1984-04-12 1999-06-29 The Liposome Company, Inc. Peptide-containing liposomes, immunogenic liposomes and methods of preparation and use
US5098704A (en) 1984-06-18 1992-03-24 Chiron Corporation Hepatitis surface antigen particle vaccine
US4722840A (en) 1984-09-12 1988-02-02 Chiron Corporation Hybrid particle immunogens
ES2061500T5 (en) 1986-06-17 2003-05-16 Chiron Corp DIAGNOSIS AND VACCINES OF DELTA HEPATITIS, ITS PREPARATION AND USE.
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
DE318216T1 (en) 1987-11-18 1990-06-13 Chiron Corp., Emeryville, Calif., Us NANBV DIAGNOSTICS AND VACCINE.
US5098827A (en) * 1988-02-26 1992-03-24 The University Of Florida Novel bacterial markers for pathogenic group B streptococci
AU631377B2 (en) 1988-08-25 1992-11-26 Liposome Company, Inc., The Affinity associated vaccine
GB8827038D0 (en) 1988-11-18 1988-12-21 Kehoe M Streptolysin o antigens & uses
US5354846A (en) * 1988-11-18 1994-10-11 Michael Kehoe Streptolysin O antigen derivatives, its production and uses
DE3841091A1 (en) 1988-12-07 1990-06-13 Behringwerke Ag SYNTHETIC ANTIGENS, METHOD FOR THEIR PRODUCTION AND THEIR USE
JP3237842B2 (en) 1988-12-16 2001-12-10 オランダ国 Pneumolysin mutants and pneumococcal vaccines produced therefrom
GB2233977B (en) 1989-01-04 1993-03-31 Michael Kehoe Cytolytic streptolysin o mutants and uses
EP0378881B1 (en) 1989-01-17 1993-06-09 ENIRICERCHE S.p.A. Synthetic peptides and their use as universal carriers for the preparation of immunogenic conjugates suitable for the development of synthetic vaccines
HUT54896A (en) 1989-03-17 1991-04-29 Chiron Corp Process for producing aquous diagnosticum and vaccine of nanbv
CA2011896C (en) 1989-04-21 2001-05-08 Mark Werner Ringworm vaccine
MC2188A1 (en) 1989-05-18 1992-09-16 Chiron Corp NANBV DIAGNOSIS: POLYNUCLEOTIDES FOR SCREENING FOR HEPATITIS C VIRUS
JPH0832638B2 (en) 1989-05-25 1996-03-29 カイロン コーポレイション Adjuvant formulation comprising submicron oil droplet emulsion
CA2063271A1 (en) 1989-07-14 1991-01-15 Subramonia Pillai Cytokine and hormone carriers for conjugate vaccines
IT1237764B (en) 1989-11-10 1993-06-17 Eniricerche Spa SYNTHETIC PEPTIDES USEFUL AS UNIVERSAL CARRIERS FOR THE PREPARATION OF IMMUNOGENIC CONJUGATES AND THEIR USE FOR THE DEVELOPMENT OF SYNTHETIC VACCINES.
US5821088A (en) * 1990-05-11 1998-10-13 Siga Pharmaceuticals, Inc. Use of gram-positive bacteria to express recombinant proteins
US6737521B1 (en) * 1990-05-11 2004-05-18 The Rockefeller University Delivery and expression of a hybrid surface protein on the surface of gram positive bacteria
GB2276169A (en) 1990-07-05 1994-09-21 Celltech Ltd Antibodies specific for carcinoembryonic antigen
ATE128628T1 (en) 1990-08-13 1995-10-15 American Cyanamid Co FIBER HEMAGGLUTININ FROM BORDETELLA PERTUSSIS AS A CARRIER FOR CONJUGATE VACCINE.
US5153312A (en) 1990-09-28 1992-10-06 American Cyanamid Company Oligosaccharide conjugate vaccines
US5277904A (en) 1990-10-26 1994-01-11 Pier Allan C Broad spectrum dermatophyte vaccine
US5284652A (en) 1990-10-26 1994-02-08 Pier Allan C Dermatophyte vaccine
US5378620A (en) 1991-08-30 1995-01-03 Beckman Instruments, Inc. Streptolysin O derivatives
US5391712A (en) * 1991-08-30 1995-02-21 Beckman Instruments, Inc. Non-hemolytic streptolysin O variants
EP0967279B1 (en) 1992-03-02 2008-01-02 Novartis Vaccines and Diagnostics S.r.l. Helicobacter pylori cytotoxin useful for vaccines and diagnostics
IT1262896B (en) 1992-03-06 1996-07-22 CONJUGATE COMPOUNDS FORMED FROM HEAT SHOCK PROTEIN (HSP) AND OLIGO-POLY-SACCHARIDES, THEIR USE FOR THE PRODUCTION OF VACCINES.
HU219808B (en) 1992-06-25 2001-08-28 Smithkline Beecham Biologicals S.A. Vaccine composition containing adjuvant and process for preparation thereof
IL102687A (en) 1992-07-30 1997-06-10 Yeda Res & Dev Conjugates of poorly immunogenic antigens and synthetic pepide carriers and vaccines comprising them
DE4240056A1 (en) * 1992-11-28 1994-06-01 Boehringer Mannheim Gmbh Streptolysin O peptide antigens and method for the determination of streptolysin antibodies
EP0613947B1 (en) 1993-02-01 2004-03-03 Beckman-Coulter, Inc. Mitogenic factor, gene thereof and method of microdetection therefor
DE69405551T3 (en) 1993-03-23 2005-10-20 Smithkline Beecham Biologicals S.A. 3-0-DEAZYLATED MONOPHOSPHORYL LIPID A-CONTAINING VACCINE COMPOSITIONS
CA2141535A1 (en) * 1993-06-23 1995-01-05 Craig W. Adams Recombinant dnase b derived from streptococcus pyogenes
US5585098A (en) * 1993-11-23 1996-12-17 Ovimmune, Inc. Oral administration of chicken yolk immunoglobulins to lower somatic cell count in the milk of lactating ruminants
GB9326174D0 (en) 1993-12-22 1994-02-23 Biocine Sclavo Mucosal adjuvant
GB9326253D0 (en) 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
AU697227B2 (en) * 1994-10-07 1998-10-01 Rockefeller University, The Enzyme for cleavage of the anchor region of surface proteins from gram positive bacteria
AUPM873294A0 (en) 1994-10-12 1994-11-03 Csl Limited Saponin preparations and use thereof in iscoms
AUPM885194A0 (en) * 1994-10-14 1994-11-10 Council Of The Queensland Institute Of Medical Research, The Synthetic peptides and vaccines comprising same
IL117483A (en) 1995-03-17 2008-03-20 Bernard Brodeur Proteinase k resistant surface protein of neisseria meningitidis
UA56132C2 (en) 1995-04-25 2003-05-15 Смітклайн Бічем Байолоджікалс С.А. Vaccine composition (variants), method for stabilizing qs21 providing resistance against hydrolysis (variants), method for manufacturing vaccine
US6284884B1 (en) * 1995-06-07 2001-09-04 North American Vaccine, Inc. Antigenic group B streptococcus type II and type III polysaccharide fragments having a 2,5-anhydro-D-mannose terminal structure and conjugate vaccine thereof
US6936259B2 (en) * 1995-06-08 2005-08-30 University Of Saskatchewan CAMP factor of Streptococcus uberis
GB9513261D0 (en) 1995-06-29 1995-09-06 Smithkline Beecham Biolog Vaccines
US5846547A (en) * 1996-01-22 1998-12-08 Regents Of The University Of Minnesota Streptococcal C5a peptidase vaccine
US6346392B1 (en) 1996-11-27 2002-02-12 Smithkline Beecham Corporation Polynucleotides encoding a novel glutamine transport ATP-binding protein
IES960488A2 (en) 1996-07-03 1997-11-19 Trinity College Dublin Subunit Vaccine for Streptococcus Equi
EP0821070A1 (en) 1996-07-22 1998-01-28 Carelli, Claude Marcel Henri Pit-1 gene polymorphism and trait selection in animals
DE19630390A1 (en) 1996-07-26 1998-01-29 Chiron Behring Gmbh & Co Proteins, in particular membrane proteins from Helicobacter pylori, their production and use
KR100524245B1 (en) 1996-09-04 2005-10-26 다카라 바이오 가부시키가이샤 Fungal antigens and process for producing the same
EP1770164B1 (en) 1996-10-31 2010-09-01 Human Genome Sciences, Inc. Streptococcus pneumoniae antigens and vaccines
WO1998019689A1 (en) 1996-11-01 1998-05-14 Smithkline Beecham Corporation Novel coding sequences
US7033765B1 (en) * 1997-02-20 2006-04-25 Toronto Research Chemicals, Inc. Site-specific drug delivery
ATE441432T1 (en) 1997-03-10 2009-09-15 Ottawa Hospital Res Inst USE OF NON-METHYLATED CPG DINUCLEOTIDE IN COMBINATION WITH ALUMINUM AS ADJUVANTS
US6426074B1 (en) * 1997-03-19 2002-07-30 The Brigham And Women's Hospital Inc. Group B Streptococcus vaccine
US6818222B1 (en) 1997-03-21 2004-11-16 Chiron Corporation Detoxified mutants of bacterial ADP-ribosylating toxins as parenteral adjuvants
US6299881B1 (en) 1997-03-24 2001-10-09 Henry M. Jackson Foundation For The Advancement Of Military Medicine Uronium salts for activating hydroxyls, carboxyls, and polysaccharides, and conjugate vaccines, immunogens, and other useful immunological reagents produced using uronium salts
CA2288944A1 (en) * 1997-05-06 1998-11-12 Human Genome Sciences, Inc. Enterococcus faecalis polynucleotides and polypeptides
US6635623B1 (en) * 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
GB9712347D0 (en) 1997-06-14 1997-08-13 Smithkline Beecham Biolog Vaccine
GB9713156D0 (en) 1997-06-20 1997-08-27 Microbiological Res Authority Vaccines
US5948413A (en) 1997-07-17 1999-09-07 Board Of Trustees Operating Michigan State University Method and vaccine for treatment of pythiosis insidiosi in humans and lower animals
EP1009382B1 (en) 1997-09-05 2003-06-18 GlaxoSmithKline Biologicals S.A. Oil in water emulsions containing saponins
WO1999013084A1 (en) 1997-09-12 1999-03-18 Id Vaccine Group a streptococcal vaccines
US6406883B1 (en) 1997-09-26 2002-06-18 Luetticken Rudolf Lmb gene of Streptococcus agalactiae
AU9507698A (en) 1997-09-26 1999-04-23 Med Immune, Inc. (lmb) gene of (streptococcus agalactiae)
US6756361B1 (en) 1997-10-14 2004-06-29 Nabi Enterococcus antigens and vaccines
NZ503319A (en) * 1997-10-17 2002-04-26 Nestle Sa Streptococcus lactic acid bacteria strains whose total protein profile resembles that of CNCM I-1920
BR9813930A (en) 1997-11-06 2006-12-19 Chiron Spa neisserial antigen
CA2307846A1 (en) 1997-11-21 1999-06-03 Genset S.A. Chlamydia pneumoniae genomic sequence and polypeptides, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
AU748950B2 (en) 1997-11-21 2002-06-13 New Zealand Pastoral Agriculture Research Institute Limited Zoocin A immunity factor
GB9725084D0 (en) 1997-11-28 1998-01-28 Medeva Europ Ltd Vaccine compositions
BR9814912A (en) 1997-11-28 2000-10-03 Genset Sa Genetic sequence and polypeptides of chlamydia trachomatis, fragments thereof and uses thereof, in particular for the diagnosis, prevention and treatment of infection
CN1285753A (en) 1997-12-02 2001-02-28 鲍德杰克特疫苗公司 Transdermal delivery of particulate vaccine compositions
EP1042480A1 (en) * 1997-12-31 2000-10-11 Stressgen Biotechnologies Corporation Streptococcal heat shock proteins of the hsp60 family
EP1047784B2 (en) 1998-01-14 2015-03-18 Novartis Vaccines and Diagnostics S.r.l. Neissera meningitidis antigens
US7041814B1 (en) * 1998-02-18 2006-05-09 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to Enterobacter cloacae for diagnostics and therapeutics
HU228497B1 (en) 1998-02-20 2013-03-28 Id Biomedical Corp Quebec Group b steptococcus antigens
GB9807721D0 (en) 1998-04-08 1998-06-10 Chiron Spa Antigen
WO1999052549A1 (en) 1998-04-09 1999-10-21 Smithkline Beecham Biologicals S.A. Adjuvant compositions
GB9808327D0 (en) 1998-04-20 1998-06-17 Chiron Spa Antidiotypic compounds
NZ532665A (en) 1998-05-01 2005-11-25 Inst Genomic Research Neisseria meningitidis antigens and compositions
US6562798B1 (en) 1998-06-05 2003-05-13 Dynavax Technologies Corp. Immunostimulatory oligonucleotides with modified bases and methods of use thereof
US6660520B2 (en) * 1998-06-05 2003-12-09 Smithkline Beecham Corporation Nrde
US6936252B2 (en) * 1998-07-27 2005-08-30 Microbial Technics Limited Streptococcus pneumoniae proteins and nucleic acid molecules
US7098182B2 (en) * 1998-07-27 2006-08-29 Microbial Technics Limited Nucleic acids and proteins from group B streptococcus
JP2002531054A (en) 1998-07-27 2002-09-24 マイクロビアル テクニクス リミティッド Nucleic acids and proteins from group B streptococci
GB9817052D0 (en) 1998-08-05 1998-09-30 Smithkline Beecham Biolog Vaccine
MXPA01003557A (en) 1998-10-09 2004-04-05 Chiron Corp Neisseria genomic sequences and methods of their use.
DE122007000087I1 (en) 1998-10-16 2008-03-27 Glaxosmithkline Biolog Sa ADJUVANCY SYSTEMS AND VACCINES
AU1722300A (en) 1998-11-12 2000-05-29 Regents Of The University Of California, The Chlamydia pneumoniae genome sequence
GB9828000D0 (en) 1998-12-18 1999-02-10 Chiron Spa Antigens
US7128918B1 (en) * 1998-12-23 2006-10-31 Id Biomedical Corporation Streptococcus antigens
BR0009163A (en) 1999-03-19 2001-12-26 Smithkline Beecham Biolog Vaccine
JP2002541808A (en) 1999-04-09 2002-12-10 テクラブ, インコーポレイテッド Recombinant toxin A protein carrier for polysaccharide conjugate vaccine
US7101692B2 (en) * 1999-04-15 2006-09-05 The Regents Of The University Of California Identification of sortase gene
WO2000067161A2 (en) 1999-05-04 2000-11-09 Grant Lee H Method and apparatus for categorizing and retrieving network pages and sites
US6833356B1 (en) * 1999-08-25 2004-12-21 Medimmune, Inc. Pneumococcal protein homologs and fragments for vaccines
KR20020038770A (en) 1999-09-24 2002-05-23 장 스테판느 Use of combination of polyoxyethylene sorbitan ester and octoxynol as adjuvant and its use in vaccines
CZ20021045A3 (en) 1999-09-24 2002-08-14 Smithkline Beecham Biologicals S. A. Auxiliary preparation
EP1265582A2 (en) 1999-09-29 2002-12-18 Human Genome Sciences, Inc. Colon and colon cancer associated polynucleotides and polypeptides
EP2275129A3 (en) 2000-01-17 2013-11-06 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
AU3108001A (en) 2000-01-20 2001-12-24 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for inducing a th2 immune response
US6400152B1 (en) * 2000-01-31 2002-06-04 General Electric Company Spiral trajectory calculations for MRI
US6777547B1 (en) * 2000-01-31 2004-08-17 Andreas Podbielski Collagen-binding proteins from streptococcus pyogenes
EP1268774A2 (en) * 2000-03-21 2003-01-02 Elitra Pharmaceuticals, Inc. Identification of essential genes in prokaryotes
GB0007432D0 (en) 2000-03-27 2000-05-17 Microbiological Res Authority Proteins for use as carriers in conjugate vaccines
AUPQ801700A0 (en) * 2000-06-07 2000-06-29 Peplin Research Pty Ltd Enzyme and viral activation
DE60139690D1 (en) 2000-07-03 2009-10-08 Novartis Vaccines & Diagnostic IMMUNIZATION AGAINST CHLAMYDIA PNEUMONIAE
EP1297145B1 (en) * 2000-07-06 2008-10-29 ID Biomedical Corporation Streptococcus pyogenes antigen
CA2417357A1 (en) * 2000-08-08 2002-02-14 St. Jude Children's Research Hospital Group b streptococcus polypeptides nucleic acids and therapeutic compositions and vaccines thereof
WO2002026757A2 (en) 2000-09-26 2002-04-04 Hybridon, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
JP2004509972A (en) * 2000-09-29 2004-04-02 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Triterpenes with fungicidal activity against yeast
US7160547B2 (en) * 2000-10-10 2007-01-09 University Of Tennessee Research Corporation Streptococcal streptolysin S vaccines
WO2002057315A2 (en) * 2000-10-10 2002-07-25 University Of Tennessee Research Corporation Streptococcal streptolysin s vaccines
JP2002135314A (en) 2000-10-26 2002-05-10 Sharp Corp Communication control method and communication equipment
SG165981A1 (en) * 2000-10-27 2010-11-29 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
GB0103424D0 (en) 2001-02-12 2001-03-28 Chiron Spa Gonococcus proteins
WO2002064162A2 (en) 2001-02-13 2002-08-22 Government Of The United States, As Represented By The Secretary Of The Army Vaccine for transcutaneous immunization
AU2002252378B2 (en) 2001-03-19 2007-10-18 Intercell Usa, Inc. Transcutaneous immunostimulation
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
US20070128229A1 (en) * 2002-04-12 2007-06-07 Wyeth Surface proteins of Streptococcus pyogenes
IL158328A0 (en) * 2001-04-13 2004-05-12 Wyeth Corp Surface proteins of streptococcus pyogenes
FR2824074A1 (en) * 2001-04-26 2002-10-31 Pasteur Institut SEQUENCE OF THE GENOME STREPTOCOCCUS AGALACTIAE, APPLICATION TO THE DEVELOPMENT OF VACCINES, DIAGNOSIS TOOLS, AND THE IDENTIFICATION OF THERAPEUTIC TARGETS
ATE557041T1 (en) * 2001-05-18 2012-05-15 Us Gov Health & Human Serv PEPTIDE VACCINES AGAINST GROUP A STREPTOCOCCE
GB0118249D0 (en) * 2001-07-26 2001-09-19 Chiron Spa Histidine vaccines
US20060073530A1 (en) * 2001-08-15 2006-04-06 Olaf Schneewind Methods and compositions involving sortase B
DK1450856T3 (en) 2001-09-14 2010-05-31 Cytos Biotechnology Ag Packaging of Immunostimulatory CpG in Virus-Like Particles, Method and Use
WO2003024480A2 (en) 2001-09-14 2003-03-27 Cytos Biotechnology Ag In vivo activation of antigen presenting cells for enhancement of immune responses induced by virus like particles
GB0123580D0 (en) 2001-10-01 2001-11-21 Glaxosmithkline Biolog Sa Vaccine
WO2003035836A2 (en) 2001-10-24 2003-05-01 Hybridon Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5' ends
US20040029129A1 (en) * 2001-10-25 2004-02-12 Liangsu Wang Identification of essential genes in microorganisms
JP2005535287A (en) 2002-02-11 2005-11-24 アイディー バイオメディカル コーポレイション Group B Streptococcus antigen
GB2385274B (en) * 2002-02-13 2004-04-14 Ming-Jeng Shue Vaginal suppository delivery device
US20050181388A1 (en) * 2002-04-02 2005-08-18 Affinium Pharmaceuticals, Inc. Novel purified polypeptides from bacteria
AU2003213949A1 (en) * 2002-04-08 2003-10-27 Affinium Pharmaceuticals, Inc. Purified polypeptides involved in membrane biogenesis
GB0210128D0 (en) * 2002-05-02 2002-06-12 Chiron Spa Nucleic acids and proteins from streptococcus groups A & B
US20070053924A1 (en) * 2002-08-26 2007-03-08 Herve Tettelin Conserved and specific streptococcal genomes
TW566366U (en) * 2002-09-27 2003-12-11 Wus Tech Co Ltd Labor-saving portable battery equipment for power-driven walking assisted scooter
EP2314604A3 (en) * 2002-10-15 2011-05-25 Intercell AG Nucleic acids coding for adhesion factors of group B streptococcus, adhesion factors of group B streptococcus and further uses thereof
CN102512670A (en) 2003-03-04 2012-06-27 英特塞尔股份公司 Streptococcus pyogenes antigens
CA2522379C (en) 2003-04-10 2012-10-23 Chiron Corporation The severe acute respiratory syndrome coronavirus
CA2522986A1 (en) * 2003-05-07 2004-11-18 Intercell Ag Streptococcus agalactiae antigens i + ii
BRPI0411857A (en) 2003-06-26 2006-05-23 Chiron Corp immunogenic compositions for chlamydia trachomatis
US7709009B2 (en) * 2003-07-31 2010-05-04 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for streptococcus pyogenes
AU2003904237A0 (en) * 2003-08-08 2003-08-21 Garvan Institute Of Medical Research Novel translocation assay
US8945589B2 (en) * 2003-09-15 2015-02-03 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for Streptococcus agalactiae
US7424370B2 (en) * 2004-02-06 2008-09-09 Council Of Scientific And Industrial Research Computational method for identifying adhesin and adhesin-like proteins of therapeutic potential
US20060041961A1 (en) * 2004-03-25 2006-02-23 Abad Mark S Genes and uses for pant improvement
WO2005108419A1 (en) 2004-05-07 2005-11-17 Lea-Ann Kirkham Mutant cholesterol-binding cytolysin proteins
JP2008508320A (en) * 2004-07-29 2008-03-21 カイロン コーポレイション Immunogenic composition against gram positive bacteria such as STREPTOCOCCUSAGALACTIAE
EP1817432A2 (en) 2004-10-05 2007-08-15 Wyeth a Corporation of the State of Delaware Probe arrays for detecting multiple strains of different species
EP1770171A1 (en) 2005-09-29 2007-04-04 Universität Zu Köln DNA microarray for rapid identification of Candida albicans in blood cultures.
JP2009542196A (en) 2006-07-07 2009-12-03 インターセル アーゲー Small Streptococcus pyogenes antigens and their use
AT514720B1 (en) 2013-08-30 2016-03-15 Blum Gmbh Julius locking device

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070036828A1 (en) * 2002-09-13 2007-02-15 Chiron Corporation Group b streptococcus vaccine

Cited By (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080152701A1 (en) * 1997-12-19 2008-06-26 Naomi Balaban Methods and Compositions for the Treatment and Prevention of Staphylococcus Infections
US8067015B2 (en) 1997-12-19 2011-11-29 Naomi Balaban Methods and compositions for the treatment and prevention of Staphylococcus and other bacterial infections
US7534857B2 (en) 1997-12-19 2009-05-19 Centegen, Inc. Methods and compositions for the treatment and prevention of staphylococcal infections
US20060252691A1 (en) * 1997-12-19 2006-11-09 Naomi Balaban Methods and compositions for the treatment and prevention of staphylococcus and other bacterial infections
US20080219976A1 (en) * 1997-12-19 2008-09-11 Naomi Balaban Methods and compositions for treatment and prevention of staphylococcal infections
US20070009569A1 (en) * 1997-12-19 2007-01-11 Naomi Balaban Methods and compositions for the treatment and prevention of staphylococcus and other bacterial infections
US7939087B2 (en) 2000-10-27 2011-05-10 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from Streptococcus groups A & B
US7955604B2 (en) * 2000-10-27 2011-06-07 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A and B
US8137673B2 (en) * 2000-10-27 2012-03-20 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from Streptococcus groups A & B
US20060258849A1 (en) * 2000-10-27 2006-11-16 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A & B
US20060210579A1 (en) * 2000-10-27 2006-09-21 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A & B
US10428121B2 (en) 2000-10-27 2019-10-01 Novartis Ag Nucleic acids and proteins from streptococcus groups A and B
US8025890B2 (en) 2000-10-27 2011-09-27 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A and B
US20060210582A1 (en) * 2000-10-27 2006-09-21 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A & B
US9840538B2 (en) 2000-10-27 2017-12-12 Novartis Ag Nucleic acids and proteins from Streptococcus groups A and B
US9738693B2 (en) 2000-10-27 2017-08-22 Novartis Ag Nucleic acids and proteins from streptococcus groups A and B
US8431139B2 (en) 2000-10-27 2013-04-30 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from Streptococcus groups A and B
US20060210581A1 (en) * 2000-10-27 2006-09-21 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups A & B
US20100105865A1 (en) * 2000-10-27 2010-04-29 John Telford Nucleic acids and proteins from streptococcus groups a & b
US20100303864A1 (en) * 2002-08-26 2010-12-02 Herve Tettelin Conserved and specific streptococcal genomes
US7709009B2 (en) 2003-07-31 2010-05-04 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for streptococcus pyogenes
US20070065464A1 (en) * 2003-07-31 2007-03-22 Chiron Corporation Immunogenic compositions for streptococcus pyogenes
US8128936B2 (en) 2003-07-31 2012-03-06 Novartis Vaccines And Diagnostics, S.R.L. Immunogenic compositions for Streptococcus pyogenes
US9056912B2 (en) 2003-07-31 2015-06-16 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for Streptococcus pyogenes
US8529913B2 (en) 2003-07-31 2013-09-10 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for Streptococcus pyogenes
US8945589B2 (en) 2003-09-15 2015-02-03 Novartis Vaccines And Diagnostics, Srl Immunogenic compositions for Streptococcus agalactiae
US20080220010A1 (en) * 2003-09-15 2008-09-11 Novartis Vaccines And Diagnostics Srl Immunogenic Compositions for Streptococcus Agalactiae
US8778358B2 (en) 2004-07-29 2014-07-15 Novartis Vaccines And Diagnostics, Inc. Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae
US20090317420A1 (en) * 2004-07-29 2009-12-24 Chiron Corporation Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
US20090117113A1 (en) * 2004-10-08 2009-05-07 Chiron Corporation Immunogenic And Therapeutic Compositions For Streptococcus Pyogenes
US7838010B2 (en) 2004-10-08 2010-11-23 Novartis Vaccines And Diagnostics S.R.L. Immunogenic and therapeutic compositions for Streptococcus pyogenes
US20100074923A1 (en) * 2006-02-17 2010-03-25 Antonello Covacci Purification of bacterial antigens
US20080031877A1 (en) * 2006-02-17 2008-02-07 Antonello Covacci Purification of bacterial antigens
US20110206692A1 (en) * 2006-06-09 2011-08-25 Novartis Ag Conformers of bacterial adhesins
US20100150943A1 (en) * 2006-07-26 2010-06-17 Novartis Ag Immunogenic compositions for gram positive bacteria
US20110038879A1 (en) * 2006-10-30 2011-02-17 Novartis Ag Immunogenic and therapeutic compositions for streptococcus pyogenes
US8858957B2 (en) 2007-09-12 2014-10-14 Novartis Ag GAS57 mutant antigens and GAS57 antibodies
US8399651B2 (en) 2007-09-12 2013-03-19 Novartis Ag Nucleic acids encoding GAS57 mutant antigens
US9102741B2 (en) 2007-09-12 2015-08-11 Novartis Ag GAS57 mutant antigens and GAS57 antibodies
US8287885B2 (en) 2007-09-12 2012-10-16 Novartis Ag GAS57 mutant antigens and GAS57 antibodies
WO2009054858A1 (en) * 2007-10-25 2009-04-30 Centegen, Inc. Methods and compositions for the treatment and prevention of staphylococcus aureus infections
US8953685B2 (en) 2007-12-10 2015-02-10 Qualcomm Incorporated Resource-adaptive video interpolation or extrapolation with motion level analysis
US9426414B2 (en) 2007-12-10 2016-08-23 Qualcomm Incorporated Reference selection for video interpolation or extrapolation
US20090147853A1 (en) * 2007-12-10 2009-06-11 Qualcomm Incorporated Resource-adaptive video interpolation or extrapolation
US8660175B2 (en) 2007-12-10 2014-02-25 Qualcomm Incorporated Selective display of interpolated or extrapolated video units
US20090147854A1 (en) * 2007-12-10 2009-06-11 Qualcomm Incorporated Selective display of interpolated or extrapolaed video units
US20090148058A1 (en) * 2007-12-10 2009-06-11 Qualcomm Incorporated Reference selection for video interpolation or extrapolation
US8409589B2 (en) 2007-12-21 2013-04-02 Novartis Ag Mutant forms of streptolysin O
US20090162392A1 (en) * 2007-12-21 2009-06-25 Novartis Ag Mutant forms of streptolysin o
US7731978B2 (en) 2007-12-21 2010-06-08 Novartis Ag Mutant forms of streptolysin O
US8039005B2 (en) 2007-12-21 2011-10-18 Novartis Ag Mutant forms of streptolysin O
US20120076814A1 (en) * 2009-06-01 2012-03-29 Novartis Ag Combinations of pneumococcal rrgb clades
AU2011219524B2 (en) * 2010-02-26 2015-05-21 Novartis Ag Immunogenic proteins and compositions
WO2011104632A1 (en) * 2010-02-26 2011-09-01 Novartis Ag Immunogenic proteins and compositions
CN102770444A (en) * 2010-02-26 2012-11-07 诺华有限公司 Immunogenic proteins and compositions
US9777076B2 (en) 2012-07-16 2017-10-03 Pfizer Inc. Saccharides and uses thereof
US9452205B2 (en) * 2012-09-24 2016-09-27 Montana State University Recombinant Lactococcus lactis expressing Escherichia coli colonization factor antigen I (CFA/I) fimbriae and their methods of use
US20140086950A1 (en) * 2012-09-24 2014-03-27 Montana State University Recombinant lactococcus lactis expressing escherichia coli colonization factor antigen i (cfa/i) fimbriae and their methods of use
US9931390B2 (en) 2012-09-24 2018-04-03 Montana State University Recombinant Lactococcus lactis expressing Escherichia coli colonization factor antigen I (CFA/I) fimbriae and their methods of use
US10391161B2 (en) * 2016-12-19 2019-08-27 I-Shou University FSBM recombinant protein
EP3872096A4 (en) * 2018-10-25 2022-11-30 Ajinomoto Co., Inc. Method for secretory production of protein

Also Published As

Publication number Publication date
EP1784211A2 (en) 2007-05-16
WO2006078318A3 (en) 2008-10-30
JP2011206061A (en) 2011-10-20
JP2008508320A (en) 2008-03-21
EP2612679A1 (en) 2013-07-10
US8778358B2 (en) 2014-07-15
CA2575548A1 (en) 2006-07-27
US20090317420A1 (en) 2009-12-24
EP1784211A4 (en) 2010-06-30
US20110110982A1 (en) 2011-05-12
WO2006078318A2 (en) 2006-07-27

Similar Documents

Publication Publication Date Title
US8778358B2 (en) Immunogenic compositions for gram positive bacteria such as Streptococcus agalactiae
US20100150943A1 (en) Immunogenic compositions for gram positive bacteria
JP2008508320A5 (en)
US20100074923A1 (en) Purification of bacterial antigens
EP2167531B1 (en) Streptococcus pneumoniae pilus antigens
US9102740B2 (en) Cna-B domain antigens in vaccines against gram positive bacteria
EP2537857B1 (en) Mutant forms of streptolysin O
US20110038879A1 (en) Immunogenic and therapeutic compositions for streptococcus pyogenes
US8945589B2 (en) Immunogenic compositions for Streptococcus agalactiae
RU2727476C2 (en) Antigens and antigen compositions
US8147849B2 (en) Protective antigens for group B Streptococcus hypervirulent strains
AU2013203028A1 (en) Cna_B domain antigens in vaccines against gram positive bacteria

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHIRON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TELFORD, JOHN L.;GRANDI, GUIDO;MORA, MARIROSA;AND OTHERS;REEL/FRAME:017376/0716;SIGNING DATES FROM 20051020 TO 20051215

AS Assignment

Owner name: CHIRON CORPORATION, CALIFORNIA

Free format text: RE-RECORD TO CORRECT SEVERAL ASSIGNORS NAMES ON A DOCUMENT PREVIOUSLY RECORDED AT REEL 017376, FRAME 0716. (ASSIGNMENT OF ASSIGNOR'S INTEREST);ASSIGNORS:TELFORD, JOHN L.;GRANDI, GUIDO;MORA, MARIROSA;AND OTHERS;REEL/FRAME:017817/0542;SIGNING DATES FROM 20051020 TO 20051215

AS Assignment

Owner name: NOVARTIS VACCINES AND DIAGNOSTICS, INC., CALIFORNI

Free format text: MERGER;ASSIGNOR:CHIRON CORPORATION;REEL/FRAME:022664/0792

Effective date: 20060419

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE