WO2005084306A2 - Immunogenic compositions for chlamydia pneunomiae - Google Patents

Immunogenic compositions for chlamydia pneunomiae Download PDF

Info

Publication number
WO2005084306A2
WO2005084306A2 PCT/US2005/006588 US2005006588W WO2005084306A2 WO 2005084306 A2 WO2005084306 A2 WO 2005084306A2 US 2005006588 W US2005006588 W US 2005006588W WO 2005084306 A2 WO2005084306 A2 WO 2005084306A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
acid sequence
protein
polypeptide
Prior art date
Application number
PCT/US2005/006588
Other languages
French (fr)
Other versions
WO2005084306A3 (en
Inventor
Guido Grandi
Ratti Giulio
Original Assignee
Chiron Corporation
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 Chiron Corporation filed Critical Chiron Corporation
Priority to EP05724183A priority Critical patent/EP1729800A4/en
Priority to BRPI0508365-6A priority patent/BRPI0508365A/en
Priority to RU2006134631/13A priority patent/RU2006134631A/en
Priority to JP2007501899A priority patent/JP2007526318A/en
Priority to CA002557353A priority patent/CA2557353A1/en
Publication of WO2005084306A2 publication Critical patent/WO2005084306A2/en
Publication of WO2005084306A3 publication Critical patent/WO2005084306A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/118Chlamydiaceae, e.g. Chlamydia trachomatis or Chlamydia psittaci
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention is in the field of immunology and vaccinology.
  • it relates to immunogenic compositions comprising combinations of immunogenic molecules from Chlamydia pneumoniae.
  • the bacteria of the genus Chlamydia (and Chlamydophila, according to the recently proposed but still controversial re-classification of Chlamydiaceae (Bush et ⁇ l (2001) Int J Syst Evol Microbiol 51: 203-20; Everett et ⁇ l (1999) Int J Syst Bacteriol 49: Pt2 415-40; Schachter et ⁇ l (2001) Int J Syst Evol Microbiol 51: 249, 251-3) are obligate intracellular parasites of eukaryotic cells, which have a unique biphasic life cycle involving two pleiomorphic developmental forms: an extracellular, metabolically inert, spore-like, infectious form (the elementary bodies, EBs) and an intracellular, non- infectious, replicative form (the reticulate bodies, Bs) which remains contained in a specialized cytoplasmic compartment (the Chl ⁇ mydi ⁇ l inclusion).
  • the EBs are responsible for the initial attachment to host cell surface and the establishment of the cytoplasmic inclusion where EBs can differentiate to RBs and thus initiate the replicative stage. Eventually RBs revert to infectious EB forms able to start new replicative cycles in neighbouring host cells.
  • Chlamydia infection is an intracellular infection
  • the currently accepted paradigm is that effective anti-Chlamydial immunisation would require both an adequate T-cell response and high serum levels of neutralising antibodies and that "an ideal vaccine should induce long lasting (neutralising) antibodies and a cell mediated immunity that can quickly respond upon exposure to Chlamydia".
  • WO 02/02404 provides information on the immunogenicity and immunoaccessibility of certain Chlamydia proteins and highlights that (i) current genomic annotations and/or (ii) predictions based on cellular location and/or cellular function based on in-silico analyses may not always be accurate.
  • Applicants have recently engaged in a whole-genome search (Montigiani et al (2002) Infection and Immunity 70:368-379) for possible vaccine candidates among proteins potentially associated with the outer membrane of C.pneumoniae.
  • mice antisera was prepared against over 100 recombinant His-tagged or Glutathione-S-transferase (GST) fusion proteins encoded by genes predicted by in silico analyses to be peripherally located in the Chlamydial cell.
  • GST Glutathione-S-transferase
  • 53 recombinant proteins derived from the genome of Chlamydia Chlamydophila pneumoniae (CPn) were described which induced mouse antibodies, capable of binding, in a FACS assay, to the surface of purified CPn cells.
  • the scope of the Montigiani study (ibid) was restricted to checking if polyclonal antisera produced in mice against the recombinantly expressed antibodies to CPn antigens were capable of binding to the surface of the CPn cells.
  • compositions capable of eliciting an immune response upon exposure to Chlamydia pneumoniae proteins. It is also desirable to provide improved compositions comprising one or more combinations of two or more selected CPn proteins with complementary immunological and/or biological profiles capable of providing immunity against Chlamydial induced disease and/or infection (such as in prophylactic vaccination) or (b) for the eradication of an established chronic Chlamydial infection (such as in therapeutic vaccination).
  • FIG. 1A Assay of in vitro neutralization of C.pneumoniae infectivity for LLC-MK2 cells by polyclonal mouse antisera to recombinant Chlamydial proteins.
  • Figure IB shows serum titres giving 50% neutralization of infectivity for 10 C.pneumoniae recombinant antigens. Each titer was assessed in 3 separate experiments (SEM values shown).
  • Figure 2 shows immunoblot analysis of two dimensional electrophoretic maps of C.pneumoniae EBs using the imune sera described in the text.
  • Figure 3 shows mean numbers of C.pneumoniae IFU recovered from equivalent spleen samples from immunized and mock-immunized hamsters following a systemic challenge.
  • Figure 4 shows flow cytometric analysis of splenocytes from DNA-immunized HLA- A2 transgenic and non transgenic mice.
  • Figure 5 shows a flow cytometric analysis of splenocytes from transgenic and non transgenic mice infected with C. pneumoniae EBs.
  • Figure 6 shows an alignment of the proteins in the 7105-7110 protein family.
  • Figure 7 shows an N-terminal alignment of Cpn0794 - Cpn0799.
  • Figure 8 shows a protein encoded by Cpn0796 and demonstrates a C-terminal domain comprising approximately residues from 1 to 648.
  • Figure 9 shows an alignment of the C-terminal (beta barrel) domains of the proteins encoded by the C.pneumoniae genes Cpn0795 and Cpn0796.
  • Table I shows a summary of data and properties of the C.pneumoniae antigens described in the text.
  • Table 2 shows results from hamster mouse model studies for hypothetical proteins.
  • Table 3 shows expressed genes of CPn EB selected by microarray.
  • Table 4 shows C. pneumoniae selected peptides: protein sources and HLA-A2 stabilization assay.
  • Table 5 shows ELISPOT assay with CD8+ T cells from DNA immunised HLA-A2 transgenic mice.
  • Table 6 shows IFN- ⁇ production from splenocytes of DNA immunized HLA-A2 transgenic and non transgenic mice.
  • the present invention relates to a polypeptide for use as an autotransporter antigen, the polypeptide comprising: (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79, (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a); or (c) an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids from an amino acid sequence of (a) or combinations thereof.
  • the present invention also relates to the use of a polypeptide in the preparation of a medicament for the prevention or treatment of a Chlamydia pneumoniae infection in an individual.
  • the use of the polypeptide may be as an autotransporter protein which immunoreacts with seropositive serum of an individual infected with Chlamydia pneumoniae.
  • the present invention further relates to a method of eliciting an immune response in an individual comprising administering to the individual a polypeptide comprising (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79 (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a), or (c) an amino acid sequence comprising one or more fragment of at least 1, 2, 3, 4, 5, 6, or 7 amino acids from an amino acid sequence of (a) or mixtures thereof.
  • a method for diagnosing an immune response in an individual comprising (a) contacting a biological sample obtained from the individual with a binding agent that binds to a polypeptide with an autotransporter function, (b) detecting in the biological sample the amount of the polypeptide that binds to the binding agent; and (c) comparing the amount of the polypeptide to a predetermined cut-off value and thereby determining the presence of an immune response in the individual.
  • a composition for eliciting an immune response in a subject comprising two or more Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof is also provided.
  • the composition may further comprise one or more immunostimulants.
  • polypeptide for use as an autotransporter antigen comprising an amino acid sequence corresponding to SEQ ID NO: 86, an amino acid sequence having at least 50% sequence identity to SEQ ID NO: 86, or an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids of SEQ ID NO: 86.
  • the present invention relates to a composition comprising a first biological molecule from a Chlamydia pneumoniae bacterium and a second biological molecule from a Chlamydia pneumoniae bacterium.
  • the first biological molecule is selected from the group consisting of SEQ ID No 1 to SEQ ID No 86, or the group consisting of SEQ ID No. l to 41.
  • composition may also contain the second biological molecule being selected from the group consisting of SEQ ID No 1 to SEQ ID No. 86 or SEQ ID No 1 to SEQ ID
  • composition may also comprise two or more biological molecules selected from the group consisting of SEQ ID Nos 1-41.
  • composition may also comprise one or more biological molecules selected from the group consisting of SEQ ID Nos 1-41 combined with one or more biological molecules selected from the group consisting of SEQ ID Nos 42-86.
  • composition according to any one of the previous claims further comprising an adjuvant such as an ADP-ribosylating exotoxin or a derivative thereof or an adjuvant is selected from the group consisting of cholera toxin (CT), Escherichia heat-labile exterotoxin (LT) and mutants thereof having adjuvant activity.
  • an adjuvant such as an ADP-ribosylating exotoxin or a derivative thereof or an adjuvant is selected from the group consisting of cholera toxin (CT), Escherichia heat-labile exterotoxin (LT) and mutants thereof having adjuvant activity.
  • CT cholera toxin
  • LT heat-labile exterotoxin
  • a vaccine and use of the vaccine is also provided comprising the composition of the present invention.
  • the vaccine may be used in the preparation of a medicament for the prevention or treatment of a Chlamydia infection and may be administered mucosally, intra-nasally or intra-vaginally, for example.
  • a method for treating a Chlamydia infection in a host subject wherein the method comprises the administration of a safe and effective amount of a vaccine.
  • an immunogenic composition comprising a combination of Chlamydia pneumoniae antigens, the combination comprising at least one Chlamydia pneumoniae antigen associated with elementary bodies of Chlamydia pneumoniae and at least one Chlamydia pneumoniae antigen associated with reticulate bodies of Chlamydia pneumoniae.
  • an immunogenic composition comprising a combination of Chlamydia pneumoniae antigens, the combination comprising at least one Chlamydia pneumoniae antigen of a first antigen group and at least one Chlamydia pneumoniae antigen of a second antigen group, said first antigen group comprising a Type III secretion system (TTSS) protein and said second antigen group comprising a Type III secretion system (TTSS) effector protein.
  • TTSS Type III secretion system
  • TTSS Type III secretion system
  • an immunogenic composition comprising a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
  • an immunogenic composition comprising a combination of Chlamydia pneumoniae antigens, the combination eliciting a Chlamydia pneumoniae specific TH1 immune response and a Chlamydia pneumoniae specific TH2 immune response.
  • the present invention further provides a method of monitoring the efficacy of treatment of a patient infected with Chlamydia pneumoniae comprising determining the level of Chlamydia pneumoniae specific antibody in the patient after administration of an immunogenic composition of the present invention to the patient.
  • compositions comprising a first biological molecule from a Chlamydia pneumoniae bacterium and a second biological molecule from a Chlamydia pneumoniae bacterium.
  • biological molecule includes proteins, antigens and nucleic acids.
  • the compositions may also comprise further biological molecules preferably also from Chlamydia pneumoniae. That is to say, the compositions may comprise two or more biological molecules (eg. 3, 4, 5, 6, 7, 8 etc.) at least two of which are from a Chlamydia pneumoniae bacterium (eg. 3, 4, 5, 6, 7, 8 etc.).
  • compositions include those comprising (i) two or more different Chlamydia pneumoniae proteins; (ii) two or more different Chlamydia pneumoniae nucleic acids, or (iii) mixtures of one or more Chlamydia pneumoniae protein and one or more Chlamydia pneumoniae nucleic acid.
  • an immunogenic composition comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response (such as a cell mediated or cellular immune response) and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 response (such as a humoral or antibody response).
  • the immunogenic composition may further comprise a THl adjuvant and a TH2 adjuvant.
  • an immunogenic composition comprising a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
  • an immunogenic composition comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 immune response, the combination comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
  • the immunogenic composition comprising at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 immmune response preferably comprises a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen associated with the EB of Chlamydia pneumoniae and at least one Chlamydia pneumoniae antigen associated with the RB of Chlamydia pneumoniae. Still further such combinations can comprise EB and/or RB antigens from one serovar combined with RB and/or EB antigens from at least one other serovar.
  • kits comprising a combination of Chlamydia pneumoniae antigens wherein at least one of the Chlamydia pneumoniae antigens is associated with the EB of Chlamydia pneumoniae and at least one of the Chlamydia pneumoniae antigens is associated with the RB of Chlamydia pneumoniae.
  • the kit may further include a THl adjuvant, a TH2 adjuvant and instructions.
  • the present invention further provides methods of eliciting a Chlamydia specific immune response by administering an immunogenic composition of this invention.
  • the present invention further provides a method of monitoring the efficacy of treatment of a subject infected with Chlamydia pneumoniae comprising determining the level of Chlamydia specific antibody or Chlamydia specific effector molecule in the subject after administration of an immunogenic composition of this invention.
  • the first and second biological molecules are from different Chlamydia pneumoniae species (for example, from different Chlamydia pneumoniae serovars) but they may be from the same species.
  • the biological molecules in the compositions may be from different serogroups or strains of the same species.
  • the first biological molecule is preferably selected from the group consisting of SEQ ID Nos 1-86. More preferably, it is selected from the group consisting of SEQ IDs l-41and/or SEQ ID Nos 42-86. It is preferably a purified or isolated biological molecule.
  • the second biological molecule is preferably selected from the group consisting of SEQ ID Nos 1-86.
  • compositions according to the invention therefore include those comprising: two or more biological molecules selected from the group consisting of SEQ ID Nos 1-41; one or more biological molecules selected from the group consisting of SEQ IDs 1-41 combined with one or more biological molecules selected from the group consisting of SEQ IDs 42-86.
  • One or both of the first and second biological molecules may be a Chlamydia pneumoniae biological molecule which is not specifically disclosed herein, and which may not have been identified, discovered or made available to the public or purified before this patent application was filed.
  • a combination of Chlamydia pneumoniae antigens comprising at least one Type III Secretion System (TTSS) protein and at least one Type III Secretion System (TTSS) secreted or effector protein or fragment thereof.
  • TTSS Type III Secretion System
  • TTSS proteins associated with the Chlamydial TTSS machinery.
  • TTSS is a complex protein secretion and delivery machine or apparatus, which may be located, either wholly or partially, on the Elementary Body (EB) and which allows an organism, such as Chlamydia, to maintain its intracellular niche by injecting proteins, such as bacterial effector proteins (which may act as anti-host virulence determinants) into the cytosol of a eukaryotic cell in order to establish the bacterial infection and to modulate the host cellular functions.
  • TTSS proteins exposed on the EB surface may play a role in adhesion and/or uptake into host cells.
  • the TTSS is a complex protein secretion and delivery machine or apparatus, which may be located on the Elementary Body (EB) and which allows an organism, such as Chlamydia, to maintain its intracellular niche by injecting proteins, such as bacterial effector proteins (which may act as anti-host virulence determinants) into the cytosol of a eukaryotic cell in order to establish the bacterial infection and to modulate the host cellular functions.
  • proteins such as bacterial effector proteins (which may act as anti-host virulence determinants) into the cytosol of a eukaryotic cell in order to establish the bacterial infection and to modulate the host cellular functions.
  • These injected proteins can have various effects on the host cell which include but are not limited to manipulating actin and other structural proteins and modification of host cell signal transduction systems.
  • the injected (or translocated) proteins or substrates of the TTTS system may also be processed and presented by MHC-class I molecules. Not all the proteins secreted by a Type III secretion system are delivered into the host cell or have effector function.
  • the Elementary Body (EB) is regarded as "metabolically inert", it has been postulated that the Chlamydial TTSS system located on the (EB) is triggered by membrane contact and is capable of releasing pre-formed "payload” proteins.
  • Type Three Secretion System becomes active during the intracellular phase of the chlamydial replicative cycle for the secretion of proteins into the host cell cytoplasm and for the insertion of chlamydial proteins (like the Inc set) into the inclusion membrane that separates the growing chlamydial microcolony from the host cell cytoplasm (see Montigiani et al (2002) Infection and Immunity 70(1); 386-379).
  • Proteins may be expressed and secreted by 2 hours (early cycle) after infection while the expression of other early and mid cycle Type III specific genes are not detectable until 6-12 hours (mid cycle). After 16-20 hours, the RBs begin to differentiate into EBs, and by 48-72 hours, the EBs predominate within the inclusion. Host cell lysis results in the release of the EBs to the extracellular space where they can infect more cells.
  • an early gene is one that is expressed (in terms of mRNA expression) early in infection
  • an intermediate gene is one that is expressed in the mid-cycle after infection
  • a late gene is one which is expressed during the terminal transition of RBs to EBs.
  • the present invention may comprise TTSS effector proteins.
  • the TTSS effector proteins as described are associated with the RB form of Chlamydia pneumoniae and may be identified, for example, using immunofluorescence microscopy (see Bannantine et al, Infection and Immunity 66(12); 6017-6021). Effector antibodies to putative Chlamydial TTSS effector proteins secreted by the TTSS machinery may be micro-injected into host cells at specified time points during Chlamydia pneumoniae infection (e.g., early, mid or late cycle).
  • TTSS effector proteins RB-associated Chlamydia pneumoniae proteins
  • a specific composition of the present invention may comprise a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, five or all six Chlamydia pneumoniae antigens of a first antigen group, said first antigen group consisting of: (1) pmp2; (2) pmplO; (3) Enolase; (4) OmpH-like protein; and (5) the products of CPn specific genes CPn0759 and CPn0042.
  • These antigens are referred to herein as the 'first antigen group'.
  • the composition of the invention comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of: (1) pmp2 and pmplO; (2) pmp2 and Enolase; (3) pmp2 and OmpH-like protein; (4) pmp2 and CPn0759; (5) pmp2 and CPn0042; (6) pmplO and Enolase; (7) pmplO and OmpH-like protein; (8) pmplO and CPn0759; (9) pmplO and CPn0042; (10) Enolase and OmpH-like protein (11) Enolase and CPn0759; (12) Enolase and CPn0042; (13) OmpH-like protein and CPn0759 (14) OmpH-like protein and CPn0042; (15) CPn0759 and CPn0042; (16) pmp2 and pmplO and Enolase; (17) pmp2 and pmplO and OmpH-like protein; (18) pmp
  • the composition of Chlamydia pneumoniae antigens consists of pmp2, pmplO, Enolase, OmpH-like protein and CPn0042.
  • the composition of Chlamydia pneumoniae antigens consists of pmp2, pmplO, Enolase, OmpH-like protein and CPn0759 and CPn0042.
  • the invention also provides for a slightly larger group of 12 Chlamydia pneumoniae antigens that are particularly suitable for immunisation purposes, particularly when used in combinations.
  • This second antigen group includes the six Chlamydia pneumoniae antigens of the first antigen group).
  • These 12 Chlamydia pneumoniae antigens form a second antigen group of (1) pmp2; (2) pmplO; (3) Enolase; (4) OmpH-like protein; (5) CPn0759; (6) CPn0042; (7) ArtJ; (8) HtrA; (9) AtoS; (10) OmcA; (11) CPn0498; and (12) CPn0525.
  • These antigens are referred to herein as the 'second antigen group'.
  • the invention therefore provides a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve Chlamydia pneumoniae antigens of the second antigen group.
  • the combination is selected from the group consisting of two, three, four or five Chlamydia pneumoniae antigens of the second antigen group.
  • the combination consists of six Chlamydia pneumoniae antigens of the second antigen group.
  • Each of the Chlamydia pneumoniae antigens of the first and second antigen group are described in more detail below.
  • pmplO protein is set forth as SEQ ID NO: 1 below (GenBank Accession No.GI: 14195016).
  • Preferred pmplO proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 1; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 1, wherein n is 7 or more (e.g.
  • pmp2 proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 1.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 1.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 1.
  • 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).
  • Pmp2 Polymorphic Outer Membrane Protein G Family (CPn 0013)
  • pmp2 protein is disclosed as SEQ ID NO : 139 and 140 in WO 02/02606.
  • SEQ ID NO: 139 and 140 are disclosed as SEQ ID NO: 139 and 140 in WO 02/02606.
  • Preferred pmp2 proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 2 SEQ ID NO: 2; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 1, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These pm ⁇ 2 proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 2.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 1.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 2.
  • 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).
  • Eno' protein is disclosed as SEQ ID NO s : 93 and 94 in WO 02/02606.
  • SEQ ID NO s 93 and 94 in WO 02/02606.
  • Preferred Eno proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 2 SEQ ID NO: 2; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 2, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • Eno proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 3.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 3.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 3.
  • 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).
  • OmpH-like' protein is disclosed as SEQ ID NO s : 77 & 78 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred OmpH-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 4 which is a fragment of at least n consecutive amino acids of SEQ ID NO: 3, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • OmpH-like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 4.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 4.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 4.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • SEQ ID No 4 1 MKKLIiFSTFI.
  • hypothetical protein is set forth as SEQ ID NO: 5 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 5; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 5, wherein n is 7 or more (e.g.
  • Hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 5.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 5.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g.
  • fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • proteins secreted by the autotransporter secretion mechanism possess an overall unifying structure, including an amino-terminal leader peptide (for secretion across the inner membrane), the secreted mature protein (or passenger domain), and a dedicated C-terminal domain, which forms a pore in the outer membrane through which the passenger domain passes to the cell surface. It is likely that requirements for secretion across the outer membrane are contained within a single molecule and secretion is an energy- independent process. Structural properties of the proteins may be confined by the size of the pore considering the biophysical constraints that may be imposed on secretion.
  • the autotransporter, or type V, secretion system is a dedicated protein translocation mechanism which allows the organism to secrete proteins to and beyond the bacterial surface. The secretion mechanism and the ability to develop a new autotransporter protein simply by a single recominbation event have presented bacteria with abundant opportunities to increase the efficiency of secretion of proteins that were developed as periplasmic or exported virulence factors.
  • proteins are exported by the autotransporter secretion mechanism and are translated as a polyproptein possessing domains.
  • the autotransporters possess an overall unifying structure comprising three functional domains: the amino-termianl leader sequence, the secreted mature proterin (passenger domain) and a carboxy-terminal (beta-) domain that forms a beta-barrel pore to allow secretion of the passenger protein.
  • the leader sequence directs secretion via the sec apparatus and is cleaved at the inner membrane by a signal peptidase releasing the remaining portion of the molecule into the periplasm.
  • the ⁇ -domain assumes a biophysically favored state characterized by a ⁇ -barrel shaped structure which inserts itself into the outer membrane to form a pore.
  • the passenger domain is translocated to the bacterial cell surface where it may remain intact or undergo processing.
  • a processed protein may be released into the extracellular milieu or remain associated with the bacterial cell surface.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 6; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 6, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • Hypothetical proteins include variants (e.g.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 6.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 6.
  • Other fragments omit one or more domains of the protein (e.g.
  • Cpn0795 SEQ ID NO: 6
  • a Cpn specific hypotlietical protein is a FACS positive protein which demonstrates significant immunoprotective activity in a hamster spleen model of Chlamydia pneumoniae infection.
  • Chlamydia trachomatis include: gi/4377105 (Cpn0794), gi 4377106 (Cpn0795), gi/4377107 (Cpn0796), gi 4377108 (Cpn0797), gi/4377109 (CPn0798), gi 4377110 (Cpn0799).
  • 'ArtJ' protein is disclosed as SEQ ID NO s : 73 & 74 in WO 02/02606.
  • SEQ ID NO s GenBank accession number: gi
  • Preferred ArtJ proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 7 SEQ ID NO: 7; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 7, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ArtJ proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 7.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 7.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 7.
  • 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).
  • the ArtJ protein may be bound to a small molecule like arginine or another amino acid.
  • HtrA DO Serine Protease (CPn0979)
  • 'HrtA' protein is disclosed as SEQ ID NO s : 111 & 112 in WO 02/02606.
  • SEQ ID NO: 8 GenBank accession number: gi
  • Preferred HrtA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • HrtA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 8.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 8.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably at least 16 to remove the signal peptide) from the N-terminus of SEQ ID NO: 8.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • SEQID N0 8 1 MITKQ RSWIi AVLVGSS IA LP SGQAVGK KESRVSELPQ DVLLKEISGG 51 FSKVATKATP AVVYIESFPK SQAVTHPSPG RRGPYENPFD YFNDEFFNRF 101 FG PSQREKP QSKEAVRGTG FLVSPDGYIV TNNHWEDTG KIHVTLHDGQ 151 KYPATVIGLD PKTD AVIKI KSQNLPY SF GNSDHLKVGD WAIAIGNPFG 201 LQATVTVGVI SAKGRNQLHI ADFEDFIQTD AAINPGNSGG P LNIDGQVI 251 GVNTAIVSGS GGYIGIGFAI PSLMANRIID QLIRDGQVTR GFLGVTLQPI 301 DAEL
  • AtoS two-component regulatory system sensor histidine kinase protein (CPn0584)
  • AtoS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 9; and/or (b) which is a fragment of at least ..
  • AtoS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 9.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 9.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and or one or more amino acids (e.g.
  • 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).
  • OmcA' protein is disclosed as SEQ ID NO s : 9 & 10 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred OmcA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 10 SEQ ID NO: 10; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 10, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 10.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 10.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 10.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • the protein may be lipidated (e.g.
  • SEQ ID NO: 11 One example of a hypothetical protein is set forth as SEQ ID NO: 11 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 11 ; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 11, wherein ra is 7 or more (e.g.
  • Hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 11.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 11.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 11.
  • fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • the protein may be lipidated (e.g. by a N-acyl diglyceride), and may thus have a ⁇ -terminal cysteine.
  • 'Cpn0525' protein is disclosed as SEQ ID ⁇ O s : 117 & 118 in WO 02/02606.
  • SEQ ID NO: 12 GenBank accession number: gi
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 12 SEQ ID NO: 12; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 12, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 12.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 12.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 12.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group.
  • Chlamydia pneumoniae antigens include a third antigen group consisting of (1) LcrE, (2) DnaK, (3) Omp85 homolog, (4) Mip-like; (5) OmcB (6) MurG (7) Cpn0186 and (8) fliY. These antigens are referred to herein as the "third antigen group".
  • LcrE' protein is disclosed as SEQ ID NO s : 29 & 30 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred LcrE proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • LcrE proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 13.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 13.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 13.
  • 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).
  • DnaK' protein is disclosed as SEQ ID NO s : 103 & 104 in WO 02/02606.
  • SEQ ID NO: 14 GenBank accessionnumber: gi
  • Preferred DnaK proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 14 which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 14, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • DnaK proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 14.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 14.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 14.
  • 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).
  • Omp85 Homolog protein is disclosed as SEQ ID NO s : 147 & 148 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred Omp85 proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 15 amino acids of SEQ ID NO: 15; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 15, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • DnaK proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 15.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 15.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 15.
  • 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).
  • Mip-like protein is disclosed as SEQ ID NO s : 55 & 56 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred Mip-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 16 amino acids of SEQ ID NO: 16; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 16, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • mip- like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 16.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 16.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 16.
  • 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).
  • OmcB protein is disclosed as SEQ ID NO s : 47 & 48 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred OmcB proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • OmcB proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 17.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 17.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 17.
  • 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) .
  • MurG peptidoglycan transferase protein (CPn0904)
  • MurG proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • MurG proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 18.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 18.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 18.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • the MurG may be lipidated e.g. with undecaprenyl.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 19; and/or (b) which is a fragment of at least « consecutive amino acids of SEQ ID NO: 19, wherein n is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 19.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 19.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 19.
  • 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).
  • FUY Glutamine Binding Protein (CPn0604)
  • SEQ ID NO s 11 & 12 in WO 02/02606.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 20 amino acids of SEQ ID NO: 20; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 20, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 20.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 20.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 20.
  • 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).
  • Chlamydia pneumoniae antigens include a fourth antigen group consisting one or more members of the PMP family. These antigens are referred to herein as the "fourth antigen group”. Each of the Chlamydia pneumoniae antigens of the fourth antigen group is described in more detail below. Fourth Antigen Group
  • Pmpl protein is set forth as SEQ ID NO s : 41 & 42 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 4 (CPn0017)
  • SEQ ID NO: 22 The sequence for pmp4 protein can be found at AE001587.1 GI.4376271.
  • Pmp 6 protein is set forth as SEQ ID NO s 31 & 32 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 7 protein is set forth as SEQ ID NO s 153 & 154 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 8 protein is set forth as SEQ ID NO s 45 & 46 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 9 protein is set forth as SEQ ID NO s 33 & 34 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 11 protein is set forth as SEQ ID NO s 115 & 116 in WO 02/02606. ⁇ GenBank accession number: gi
  • SEQ ID No 27 1 MKTSIPWVT ⁇ .
  • Pmp 12 protein is set forth as SEQ ID NO s 51 & 52 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 13 (CPn0453)
  • CPn0453 One example of a Pmp 13 protein is set forth as SEQ ID NO s 3 & 4 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 14 (CPn0454)
  • CPn0454 One example of a Pmp 14 protein is set forth as SEQ ID NO s 35 & 36 in WO 02/02606. ⁇ GenBank accession number: gi]4376737
  • Pmp 15 protein is set forth as SEQ ID NO s 5 & 6 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 16 protein is set forth as SEQ ID NO s 7 & 8 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 18 protein is set forth as SEQ ID No 33 below ⁇ GenBank accession number: gi
  • SEQ ID No 33 1 MQNNRSLSKS SFFVGALILG KTTILLNATP LSDYFDNQAN QLTTLFPLID TLTNMTPYSH 61 RATLFGVRDD TNQDIVLDHQ NSIES FENF SQDGGALSCK SLAITNTKNQ ILFLNSFAIK 121 RAGAMYVNGN FDLSENHGSI IFSGNLSFPN ASNFADTCTG GAVLCSKNVT ISKNQGTAYF 181 INNKAKSSGG AIQAAIINIK DNTGPCLFFN NAAGGTAGGA LFANACRIEN NSQPIYFLNN 241 QSGLGGAIRV HQECILTKNT GSVIFNNNFA MEADISANHS SGGAIYCISC SIKDNPGIAA 301 FDNNTAARDG GA
  • Pmp 19 (CPn0539)
  • SEQ ID No 34 GenBank accession number: gi
  • Pmp 20 protein is set forth as SEQ ID NO s 119 & 120 in WO 02/02606. ⁇ GenBank accession number: gi
  • Pmp 21 protein is set forth as SEQ ID NO s 83 & 84 in WO 02/02606. ⁇ GenBank accession number: gi
  • PMP proteins include variants (e.g., amino acids, amino acids, amino acids set forth for the pmp proteins above and/or (b) which is a fragment of at least ra consecutive amino acids of one of the polypeptide sequences set forth above wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • PMP proteins include variants (e.g.
  • Preferred fragments of (b) comprise an epitope from one of the polypeptide sequences set forth above.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of one of the polypeptide sequences set forth above.
  • Other fragments omit one or more domains of the protein (e.g.
  • Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group.
  • Such other Chlamydia pneumoniae antigens mclude a fifth antigen group consisting one or more cell surface exposed proteins. These antigens are referred to herein as the "fifth antigen group”.
  • Each of the Chlamydia pneumoniae antigens of the fifth antigen group is described in more detail below.
  • PorB protein is set forth as SEQ ID NO s : 67 & 68 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred PorB proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 37 SEQ ID NO: 37; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 37, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • PorB proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 37.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 37.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 37.
  • 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).
  • 76kDa Protein Homolog (CPn0728)
  • a 76kDa Protein Homolog protein is set forth as SEQ ID NO s : 13 & 14 in WO 02/02606.
  • SEQ ID NO s 13 & 14 in WO 02/02606.
  • Preferred 76kDa proteins homologs for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 38 amino acid sequence corresponding to SEQ ID NO: 38; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 21, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • variants e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 38.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 38.
  • 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).
  • OmpA conserved outer membrane protein protein is set forth as SEQ ID NO s : 59 & 60 in WO 02/02606.
  • SEQ ID NO s 59 & 60 in WO 02/02606.
  • Preferred ompA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 39 SEQ ID NO: 39; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 39, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 39.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 39.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 39.
  • 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).
  • PepA protein protein is set forth as SEQ ID NO s : 99 & 100 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred PepA proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g.
  • PepA proteins include variants (e.g.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 40.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 40.
  • 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).
  • conserved outer membrane protein protein is set forth as SEQ ID NO: 41 below.
  • GenBank Accession No. GL4376552; AAD18427.1 Preferred conserved outer membrane proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%,, 75%,, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 41; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 41, wherein ra is 7 or more (e.g.
  • conserved outer membrane proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 41.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 41.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 41.
  • 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).
  • Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group.
  • Chlamydia pneumoniae antigens include a sixth antigen group consisting one or more FACS positive CPn antigens. These antigens are referred to herein as the " sixth antigen group".
  • Each of the Chlamydia pneumoniae antigens of the sixth antigen group is described in more detail below.
  • Omp protein is set forth as SEQ ID NO s : 91 & 92 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred Omp proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 42 amino acids of SEQ ID NO: 42; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 42, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • Omp proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 42.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 42.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 42.
  • 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).
  • Omp protein is set forth as SEQ ID NO s : 49 & 50 in WO 02/02606.
  • SEQ ID NO s 49 & 50 in WO 02/02606.
  • Preferred Omp proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g.
  • SEQ ID NO: 43 SEQ ID NO: 43; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 43, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 43.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 43.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 43.
  • 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).
  • oligopeptide binding protein is set forth as SEQ ID NO s : 23 and 24 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred oligopeptide binding proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g.
  • SEQ ID NO: 44 amino acids of SEQ ID NO: 44, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 44.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 44.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 44.
  • 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).
  • CHLPS protein is set forth as SEQ ID NO: 45 below. GenBank Accession No. GI.4376854; AAD 18702.1.
  • Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 45 98%, 99%, 99.5% or more
  • SEQ ID NO: 45 amino acids which are 98%, 99%, 99.5% or more
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 45.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 45.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 45.
  • Other fragments omit one or more domains of the protein (e.g.
  • YscJ protein is set forth as SEQ ID NO s : 109 and 110 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred YscJ proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 46 SEQ ID NO: 46; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 46, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • YscJ proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 46.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 46.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 46.
  • 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).
  • a hypothetical protein is set forth as SEQ ID NO s : 101 and 102 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%) or more identity (e.g.
  • SEQ ID NO: 47 SEQ ID NO: 47; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 47, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 47.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 47.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 47.
  • 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).
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 48; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 48, wherein n is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 48.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 48.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 48.
  • 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).
  • SEQ ID No 48 1 MSNQLQPCIS LGCVSYINSF PLSLQLIKRN DIRCVLAPPA DLLNLLIEGK 51 LDVALTSSLG AISHNLGYVP GFGIAANQRI LSVNLYAAPT
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%o or more) to SEQ ID NO: 49; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 49, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 49.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 49.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 49.
  • 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).
  • SEQ ID No 49 1 MKFLLYVPLL VLVSTGCDA KPVSFEPFSG KLSTQRFEPQ HSAEEYFSQG 51 QEFLKKGNFR KALLCFGIIT HHFPRDILRN QAQYLIGVCY FTQDHPDLAD 101 KAFASYLQLP DAEYSEELFQ MKYAIAQRFA QGKRKRICRL EGFPKLMNAD 151 EDALRIYDEI LTAFPSKDLG AQALYSKAAL LIVKNDLTEA TKTLKKLTLQ 201 FPLHILSSEA FVRLSEIYLQ QAKKEPHNLQ YLHFAKLNEE AMKKQHPNHP 251 LNEWSANVG AMREHYARGL YATGRFYEKK KKAEAANI
  • a hypothetical protein is set forth as SEQ ID NO s : 123 and 124 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 50 amino acids of SEQ ID NO: 50, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 50.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 50.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 50.
  • 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).
  • hypothetical (CPn0792) One example of a hypothetical protein is set forth as SEQ ID NO s : 61 and 62 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 51 SEQ ID NO: 51; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 51, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 51.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 51.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 51.
  • 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).
  • a hypothetical protein is set forth as SEQ ID NO s : 113 and 114 in WO 02/02606. ⁇ GenBank accession number gi
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g.
  • SEQ ID NO: 52 SEQ ID NO: 52; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 52, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 52.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 52.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 52.
  • 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).
  • SEQ ID NO: 53 Hypothetical (CPn0126)
  • SEQ ID NO: 53 GenBank Accession No. GL4376390; AAD 18279.1
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 53; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 53, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 53.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 53.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 53.
  • 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).
  • PROPreferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 54; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 54, wherein n is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 54.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 54.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 54.
  • 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).
  • PROMAGE Identities are set forth as SEQ ID NO: 55 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 55; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 55, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 55.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 55.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 55.
  • Cpn0796 may be secreted from C. pneumoniae and is localized in the membrane of Chlamydia in young inclusions whereas an N-terminal part of Cpn0796 is secreted into the host cell cytoplasm at later times.
  • Cpn0796 was proposed to be an autotransporter and it is the first example of secretion into the host cell cytoplasm of a proposed Chlamydia autotrasporter.
  • One preferred protein for use with the invention comprises an ⁇ -terminal peptide of
  • the ⁇ -terminal peptide of Cpn0796 may form a beta-propeller structural conformation.
  • SEQ ID NO: 86 One example of the N-terminal peptide of Cpn0796 is set forth as SEQ ID NO: 86 below.
  • the N-terminal peptide of Cpn0796 for use with the invention may comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 86 amino acids of SEQ ID NO: 86; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 86, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 86.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 86.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 86.
  • PROMEQ ID NO: 56 One example of a hypothetical protein is set forth as SEQ ID NO: 56 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 56; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 56, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 56.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 56.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 56.
  • 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).
  • SEQ ID No S ⁇ 1 MSKKIKVLGH LTLCTLFRGV LCAAALSNIG YASTSQESPY QKSIEDWKGY TFTDLELLSK 61 EG SEAHAVS GNGSRIVGAS GAGQGSVTAV I ESHLIKHL GTLGGEASSA EGISKDGEW 121 VGWSDTREGY THAFVFDGRD MKDLGTLGAT YSVARGVSGD GSIIVGVSAT ARGEDYGWQV 181 GVKWEKGKIK QLKLLPQGLW SEANAISEDG TVIVGRGEIS RNHIVAVK N KNAVYSLGTL 241 GGSVASAEAI SANGKVIVGW STTNNGETHA FMHKDETMHD LGTLGGGFSV ATGVSADGRA 301 IVGFSAV
  • oligopeptide binding protein is set forth as SEQ ID NO s : 127 and 128 in WO 02/02606. ⁇ GenBank accession number G 4376467; AAD18349.1 'CPn0196'; SEQ ID NO: 76 below ⁇ .
  • Preferred oligopeptide binding proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 76 amino acids of SEQ ID NO: 76; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 76, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO 76.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 76.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 76.
  • 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).
  • Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group.
  • Such other Chlamydia pneumoniae antigens include a seventh antigen group consisting one or more hypothetical proteins (ie proteins which, for example, have no known cellular location and/or function. These antigens are referred to herein as the "seventh antigen group".
  • Each of the Chlamydia pneumoniae antigens of the seventh antigen group is described in more detail below.
  • polypeptide sequence (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 91%, 98%, 99%, 99.5% or more) to SEQ ID NO: 57; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 57, wherein ra is 7 or more (e.g.
  • SEQ ID No 57 a hypothetical protein that has been modified by SEQ ID No 57.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 57.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 57.
  • 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).
  • SEQ ID No 57 a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 58; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 21, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 58.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 58.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 58.
  • Other fragments omit one or more domains of the protein (e.g.
  • SEQ ID No 58 1 MLQSCKKALL SIWSILAFH PIPGMGVEAK SGFLGKVKG FSKKEIQEEA RILPVKDSLS 61 KRYDYTSSS GFSVEFPGEP DHSGQIVEVP QSEITIRYDT YVTETHPDNT VYWSV EYP 121 EKVDISRPEL NLQEGFSGMM QALPESQVLF MQARQIQGHK ALEF IVCED VYFRGMLISV 181 NHTLYQVFMV YKNKNPQALD KEYEAFSQSF KITKIREPRT IPSSVKKKVS L (59) Hypothetical (CPn0572) One example of a hypothetical protein is set forth as SEQ ID NO: 59 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 59; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 59, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 59.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 59.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 59.
  • Other fragments omit one or more domains of the protein (e.g.
  • Chlamydia pneumoniae antigens may be unproved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group of the third antigen group or the fourth antigen group or the fifth or the sixth antigen group or the seventh antigen group.
  • Chlamydia pneumoniae antigens include an eigth antigen group consisting one or more FACS positive CPn antigens. These antigens are referred to herein as the "eight antigen group".
  • Each of the Chlamydia pneumoniae antigens of the eight antigen group is described in more detail below.
  • Low Calcium Response Protein H is set forth as SEQ ID NO: 60 below. Genbank Accession No. GL4377123; AAD18949.1.
  • Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 60; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 60, wherein n is 7 or more (e.g.
  • These low calcium response proteins mclude variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 60.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 60.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 60.
  • 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).
  • SEQ ID No 60 1 mskpsprnan qpqkpsasfn kktrsrlael aaqkkakadd leqvhpvpte eeikkalgni 61 feglsngldl qqilglsdyl leeiytvayt fysqgkynea vglfqllaaa qpqnykymlg 121 lsscyhqlhl yneaafgffl afdaqpdnpi ppyyiadsll klqqpeesrm fldvtmdicg 181 nnpefkilke rcq
  • Yop Proteins Translocation Protein T is set forth as SEQ ID NO: 61 below. Genbank Accession No. G 4377135; AAD18960.1.
  • Preferred Yop proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 61; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 61, wherein « is 7 or more (e.g.
  • Yop proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 61.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 61.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 61.
  • 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).
  • SEQ ID No 61 1 mgislpelfs nlgsayldyi fq ppayvws vfllllarll pifavapflg aklfpspiki 61 gisls laii fpkvladtqi tnymdnnlfy vllvkemiig ivigfvlafp fyaaqsagsf 121 itnqqgiqgl egatslisie qtsphgilyh yfvtiifwlv gghrivisll lqtlevipih 181 sffpaemmsl sapiwitm ⁇ k mcqlclvmti qlsapaal
  • Yop Proteins Translocation Protein J is set forth as SEQ ID NO: 62 below Genbank Accession No. GL4377140; AAD18965.1.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 62; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 62, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 62.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 62.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 62.
  • 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).
  • SEQ ID No 62 1 mvrrsisfcl fflratllcct scnsrslivh glpgreanei vvllvskgva aqklpqaaaa 61 tagaateqmw diavpsaqit ealailnqag lprmkgtsll dlfakqglvp selqekiryq 121 eglseqmast irkmdgvvda svqisftten ednlpltasv yik rgvldn pnsimvskik 181 rliasavpgl vpenvsvvsd raa
  • OmpA (CPn0695)
  • SEQ ID NO: 63 One example of an OmPA encoded (MOMP) protein is set forth as SEQ ID NO: 63 below Genbank Accession No. GL4376998; AAD 18834.1.
  • Preferred OmpA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 63; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 63, wherein ra is 7 or more (e.g.
  • OmpA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 63.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 63.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 63.
  • 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).
  • Hypothetical Protein is set forth as SEQ ID NO: 64 below Genbank Accession No. G 4376482; AAD 18363.1.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 64; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 64, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 64.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 64.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 64.
  • 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).
  • SEQID No 64 1 mlvelealkr efahlkdqkp tsdqeitsly qcldhlefvl lglgqdkflk atededvlfe 61 sqkaidawna lltkardvlg lgdigaiyqt ieflgaylsk vnrrafcias ei flktair 121 dlnayylldf rwplckieef vdwgndcvei akrklctfek etkelnesll reehamekcs 181 iqdlqrklsd iiielhdvsl f
  • Low Calcium Response Locus Protein H (CPnl 021)
  • SEQ ID NO: 65 One example of a Low Calcium Response Protein H is set forth as SEQ ID NO: 65 below Genbank Accession No. GL4377352; AAD19158.1.
  • Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 65; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 65, wherein ra is 7 or more (e.g.
  • These low calcium response proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 65.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 65
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 65.
  • 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).
  • the immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group.
  • Such other Chlamydia pneumoniae antigens include a ninth antigen group. These antigens are referred to herein as the "ninth antigen group".
  • Each of the Chlamydia pneumoniae antigens of the ninth antigen group is described in more detail below.
  • Low Calcium Response Protein D (CPn0323)
  • SEQ ID NO: 66 One example of a Low Calcium Response Protein D is set forth as SEQ ID NO: 66 below Genbank Accession No. G 4376601; AAD18472.1.
  • Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 66; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 66, wherein ra is 7 or more (e.g.
  • These low calcium response proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 66.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 66.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 66.
  • 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).
  • CHLPS 43kDa Protein Homolog-1 (CPn0062)
  • One example of a CHLPS 43kDa Protein Homolog-1 is set forth as SEQ ID NO: 67 below Genbank Accession No. GL4376318; AAD18215.1.
  • Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 67 SEQ ID NO: 67; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 67, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 67.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 67.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 67.
  • 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).
  • CHLPS 43kDa Protein Homolog-1 is set forth as SEQ ID NO: 68 below Genbank Accession No. G 4376437; AAD18322.1.
  • Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 68; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 68, wherein n is 7 or more (e.g.
  • CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 68.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 68.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 68.
  • 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).
  • SEQ ID No 68 1 mknvgsecsq plvmelntqp lrnlcesrlv kitsfviall alvggitlta lagagilsfl 61 pwlvlgivlv vlcalfllfs ykfcpikelg vvyntdsqih qwfqkqrnkd lekatenpel 121 fgenraednn rsarsqvket lrdcdgnvlk kiyernldvl lfmnwvpktr ⁇ ddvdpvs ⁇ ds 181 irtviscykl ikackpefrs lisellrar ⁇ q sgl
  • PmpD family (CPn0963)
  • SEQ ID NO: 69 Genbank Accession No. GL4377287; AAD19099.1.
  • Preferred PmpD proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 69; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 69, wherein ra is 7 or more (e.g.
  • PmpD proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 69.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 69.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 69.
  • 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).
  • Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group.
  • Such other Chlamydia pneumoniae antigens include a tenth antigen group. Each of the Chlamydia pneumoniae antigens of the tenth antigen group is described in more detail below.
  • OmpH-like' protein is disclosed as SEQ ID NO s : 77 & 78 in WO 02/02606.
  • SEQ ID NO s 77 & 78 in WO 02/02606.
  • Preferred OmpH-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 4 which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 3, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • OmpH-like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 4.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 4.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 4.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • L7/L12 Ribosomal Protein (CPn0080)
  • CPn0080 One example of an L7/L12 Ribosomal protein is set forth as SEQ ID No 71 below ⁇ GenBank accession number: GL4376338; AAD18233.1 ⁇ . 'CPn0080'; SEQ ID NO: 71 below.
  • Preferred L7/L12 proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g.
  • L7/L12 ribosomal proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 71.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 71.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 71.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • AtoS two-component regulatory system sensor histidine kinase protein (CPn0584)
  • AtoS proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g.
  • AtoS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 72.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 72.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 72.
  • 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).
  • OmcA' protein is disclosed as SEQ ID NO s : 9 & 10 in WO 02/02606. ⁇ GenBank accession number: gi
  • Preferred OmcA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
  • SEQ ID NO: 73 SEQ ID NO: 73; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 73, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
  • OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 73.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 73.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 73.
  • Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • the protein may be lipidated (e.g. by a N-acyl diglyceride), and may thus have a ⁇ -terminal cysteine.
  • hypothetical (CPn0331) One example of a hypothetical protein is set forth as SEQ ID NO: 74 below and SEQ ID No 57 above. Genbank Accession No. GI.4376609; AAD 18480.1.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 74; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 74, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 74.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 74.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 74.
  • 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).
  • SEQ ID NO 74 1 mavsggggvq pssdpgk np alqgeqaegp splkesifse tkqassaakq eslvrsgstg 61 myatesqink akyrkaqdrs stspksklkg tfskmrasvq gfmsgfgsra srvsakrasd 121 sgegtsllpt emdvalkkgn rispemqgff ldasg ggss sdisqlslea lkssafsgar 181 slslsssess svasfgsfqk aiepmseekv na tvarlgg emvsslldpn vetsslvrra 241 matgnegmid lsdlgqeevs tamtsprave gkvk
  • PmpD protein is set forth as SEQ ID NO: 75 below Genbank Accession No. GI.4377287; AAD19099.1.
  • Preferred PmpD proteins for use with the invention comprise an amino acid sequence: (a) having 50%) or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%>, 99.5% or more) to SEQ ID NO: 75; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 75, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 75.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 75.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 75.
  • 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).
  • SEQ ID No 75 1 mvakktvrsy rssfshsviv ailsagiafe ahslhsseld Igvfnkqfee hsahveeaqt 61 svlkgsdpvn psqkesekvl ytqvpltqgs sgesldlada nflehfqhlf eettvfgidq 121 klvwsdldtr nfsqptqepd tsnavsekis sdtkenrkdl etedpskksg lkevssdlpk 181 spetavaais edleisenis ardp
  • SEQ ID NO: 78 One example of a hypothetical protein is set forth as SEQ ID NO: 78 below.
  • Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%,, 99.5% or more) to SEQ ID NO: 78; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 78, wherein ra is 7 or more (e.g.
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 78.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 78.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 78.
  • Other fragments omit one or rnore domains of the protein (e.g. omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
  • These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 79.
  • Preferred fragments of (b) comprise an epitope from SEQ ID NO: 79.
  • Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 79.
  • 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).
  • the composition of the invention comprises a combination of CPn antigens selected from the group consisting of: (1) CPn0301 and CPn0080; (2) CPn 0584 and CPn 0558; and (3) CPn 0331 and CPN 0963.
  • the composition comprises a combination of any one or more of groups (1), (2) and (3).
  • the composition of the present invention comprises a combination of CPn antigens selected from the group consisting of (1) CPn0385, CPn0324, CPn 0503, CPn0525 and CPn 0482.
  • CPn0385, CPn0324, CPn 0503, CPn0525 and CPn 0482 Preferably the composition is administered in the presence of alum and/or cPG.
  • the invention thus includes a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or six Chlamydia pneumoniae antigens of the first antigen group and two, three, four, five, or six Chlamydia pneumoniae antigens of the second antigen group.
  • the combination is selected from the group consisting of three, four, five or six Chlamydia pneumoniae antigens from the first antigen group and three, four, five or six Chlamydia pneumoniae antigens from the second antigen group.
  • the combination consists of six Chlamydia pneumoniae antigens from the first antigen group and three, four, five or six, Chlamydia pneumoniae antigens from the second antigen group.
  • the invention further includes a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or six, Chlamydia pneumoniae antigens of the second antigen group and two, three, four, five, six, seven or eight Chlamydia pneumoniae antigens of the third antigen group.
  • the combination is selected from the group consisting of three, four, five or six Chlamydia pneumoniae antigens from the second antigen group and three, four, five, six, seven or eight Chlamydia pneumoniae from the third antigen group.
  • the combination consists of six Chlamydia pneumoniae antigens from the second antigen group and three, four, five, six, seven or eight Chlamydia pneumoniae antigens of the third antigen group.
  • the number of Chlamydia pneumoniae antigens which will be in the compositions of the invention.
  • the number of Chlamydia pneumoniae antigens 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.
  • the number of Chlamydia pneumoniae antigens in a composition of the invention is less than 6, less than 5, or less than 4.
  • Chlamydia pneumoniae antigens 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.
  • the composition comprises one or more Chlamydia pneumoniae antigens from the fourth antigen group which includes porB.
  • the composition comprises one or more Chlamydia pneumoniae antigens from the fourth antigen group which includes porB.
  • Chlamydia pneumoniae antigens from the fourth antigen group includes one or more members of the pmp3 family.
  • 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 mechanism by which the immune system controls disease includes the induction of neutralising antibodies via humoral immunity and the generation of T-cell responses via cellular immunity.
  • immune response against an antigen refers to the development in a host mammalian subject of a humoral and/or a cellular immune response against that antigen.
  • humoral immune response refers to an immune response mediated by antibody molecules.
  • the antibodies generated by humoral immunity are primarily effective against extracellular infectious agents.
  • SEQ ID Nos 1-86 in the compositions of the invention may be supplemented or substituted with an antibody that binds to the protein.
  • This antibody may be monoclonal or polyclonal.
  • CMI cell mediated immune response
  • T-lymphocytes and/or other white blood cells The CMI immune mechanisms are generally more effective against intracellular infections and disease because the CMI mechanisms prime T cells in a way that, when an antigen appears at a later date, memory T cells are activated to result in a CMI response that destroys target cells that have the corresponding antigen or a portion thereof on their cell surfaces, and thereby the infecting pathogen.
  • the CMI response is focused on the destruction of the source of infection mediated by either effector cells that destroy infected cells of the host by direct cell-to-cell contact and/or by the release of molecules, such as cytokines, that possess anti-viral activity.
  • an immunogenic composition comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific Thl immune response (such as a cell mediated or cellular immune response) and at least one antigen that elicits a Chlamydia pneumoniae specific Th2 response (such as a humoral or antibody response).
  • the immunogenic composition may further comprise a Thl adjuvant and a Th2 adjuvant.
  • the invention provides a composition comprising a combination of Chlamydia pneumoniae antigens that elicit at least a Chlamydia pneumoniae specific Thl immune response.
  • the combination of Chlamydia pneumoniae antigens may include at least one antigen associated with reticulate bodies (RBs) of Chlamydia pneumoniae, including but not limited to antigens expressed, exposed on or translocated into, through or across on the inclusion membrane, antigens expressed, secreted, released or translocated into the cytosol of host cells, or antigens processed or degraded in the cytosol of host cells and/or expressed, exposed or presented on the surface of the host cell.
  • RBs reticulate bodies
  • compositions of the invention will preferably elicit both a cell mediated immune response as well as a humoral immune response in order to effectively address a Chlamydia intracellular infection.
  • This immune response will preferably induce long lasting (eg neutralising) antibodies and a cell mediated immunity that can quickly respond upon exposure to Chlamydia.
  • the invention also comprises an immunogenic composition comprising one or more immunoregulatory agents.
  • one or more of the immunoregulatory agents include an adjuvant.
  • the adjuvant may be selected from one or more of the group consisting of a Thl adjuvant and Th2 adjuvant, further discussed below.
  • the adjuvant may be selected from the group consisting of a mineral salt, such as an aluminium salt and an oligonucleotide containing a CpG motif.
  • the immunogenic composition includes both an aluminium salt and an oligonucleotide containing a CpG motif. Use of the combination of a mineral salt, such as an aluminium salt, and an oligonucleotide containing a CpG motif provide for an enhanced immune response.
  • the invention therefore includes an oligonucleotide containing a CpG motif, a mineral salt such as an aluminium salt, and an antigen, such as a Chlamydia pneumoniae antigen.
  • T cells At least two special types of T cells are required to initiate and/or to enhance CMI and and humoral responses.
  • the antigenic receptors on a particular subset of T cells which express a CD4 co-receptor can be T helper (Th) cells or CD4 T cells (herein after called T helper cells) and they recognise antigenic peptides bound to MHC class II molecules.
  • T helper cells T helper cells
  • CD8+ T cells CD8+ T cells
  • Helper T cells or CD4+ cells can be further divided into two functionally distinct subsets: Thl and Th2 which differ in their cytokine and effector function.
  • Thl and Th2 responses have been shown to be regulated not only in a positive but also in a negative way such that Thl cellular responses are augmented by Thl cytokines such as IL-2, IL-12 and IFN-gamma and decreased by Th2 cytokines such as E -4 and IL- 10.
  • Th2 cytokines such as IL-4 and IL-10
  • Thl cytokines such as IFN-gamma and another cytokine IL-12 that enhances IFN-gamma and is produced by monocytes.
  • Thl cytokines such as IFN-gamma, IL-2 and IL-12 can be regarded as immune co-factors that induce an effective inflammatory response.
  • the classic Th2 cytokines such as IL-4 and IL-10 can be regarded as cytokines that will suppress a severe inflammatory response.
  • CD8+ T cells may function in more than one way.
  • the best known function of CD8+ T cells is the killing or lysis of target cells bearing peptide antigen in the context of an MHC class I molecule. Hence the reason why these cells are often termed cytotoxic T lymphocytes (CTL).
  • CTL cytotoxic T lymphocytes
  • CD8+ T cells may secrete interferon gamma (IFN-gamma).
  • IFN-gamma interferon gamma
  • assays of lytic activity and of IFN-gamma release are both of value in measuring CD8+ T cell immune response (eg in an ELISPOT assay as set forth below).
  • ELISPOT assay as set forth below.
  • the present invention concerns methods, processes and compositions capable of enhancing and or modulating the CMI response in a host subject against a target antigen.
  • enhancing encompasses improvements in all aspects of the CMI response which include but are not limited to a stimulation and/or augmentation and/or potentiation and/or up-regulation of the magnitude and/or duration, and/or quality of the CMI response to an antigen or a nucleotide sequence encoding an antigen of interest.
  • the CMI response may be enhanced by either (i) enhancing the activation and/or production and/or proliferation of CD 8+ T cells that recognise a target antigen and/or (ii) shifting the CMI response from a Th2 to a Thl type response.
  • This enhancement of the Thl associated responses is of particular value in responding to intracellular infections because, as explained above, the CMI response is enhanced by activated Thl (such as, for example, IFN- gamma inducing) cells.
  • Such an enhanced immune response may be generally characterized by increased titers of interferon-producing CD4 + and/or CD8 + T lymphocytes, increased antigen- specific CD8+ T cell activity, and a T helper 1-like immune response (Thl) against the antigen of interest (characterized by increased antigen-specific antibody titers of the subclasses typically associated with cellular immunity (such as, for example IgG2a), usually with a concomitant reduction of antibody titers of the subclasses typically associated with humoral immunity (such as, for example IgGl)) instead of a T helper 2-like immune response (Th2).
  • Thl T helper 1-like immune response against the antigen of interest
  • the enhancement of a CMI response may be determined by a number of well-known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject (see, for example, Erickson et al. (1993) J. Immunol. 151: 4189-4199; and Doe et al. (1994) Eur. J. Immunol. 24: 2369-2376) or CD8+ T cell ELISPOT assays for measuring Interferon gamma production (Miyahara et al PNAS(USA) (1998) 95: 3954-3959).
  • the term "enhancing a T -cell response” encompasses improvements in all aspects of the T-cell response which include but are not limited to a stimulation and or augmentation and or potentiation and/or up-regulation of the magnitude and/or duration, and/or quality of the T-cell response to an antigen (which may be repeatedly administered) or a nucleotide sequence encoding an antigen.
  • the antigen may be a Chlamydia antigen, preferably a Chlamydia pneumoniae antigen.
  • the T-cell response may be enhanced by either enhancing the activation and/or production and/or distribution and/or proliferation of the induced T-cells and/or longevity of the T-cell response to T-cell inducing/modulating antigen or nucleotide sequence encoding an antigen.
  • the enhancement of the T-cell response in a host subject may be associated with the enhancement and/or modulation of the Thl immune response in the host subject.
  • the enhancement of the T-cell response may be determined by a number of well- known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T-cell cytotoxic cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject (see, for example, Erickson et al. (1993) J. Immunol. 151: 4189-4199; and Doe et al. (1994) Eur. J. Immunol. 24: 2369-2376) or CD8+ T-cell ELISPOT assays for measuring Interferon gamma production (Miyahara et al PNAS(USA) (1998) 95: 3954-3959).
  • lymphoproliferation lymphoproliferation
  • CD8+ T-cell cytotoxic cell assays or by assaying for T-lymphocytes specific for the epitope in a sensitized subject (see, for example, Erickson et al
  • Activated Thl cells enhance cellular immunity (including an increase in antigen- specific CTL production) and are therefore of particular value in responding to intracellular infections.
  • Activated Thl cells may secrete one or more of IL-2, IFN- gamma, and TNF-beta.
  • a Thl immune response may result in local inflammatory reactions by activating macrophages, NK (natural killer) cells, and CD8 cytotoxic T cells (CTLs).
  • a Thl immune response may also act to expand the immune response by stimulating growth of B and T cells with IL-12.
  • Thl stimulated B cells may secrete IgG2a.
  • 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 IgGl, IgE, IgA and memory B cells for future protection.
  • each disease causing agent or disease state has associated with it an antigen or immunodominant epitope on the antigen which is crucial in immune recognition and ultimate elimination or control of a disease causing agent or disease state in a host.
  • the host immune system In order to mount a humoral and/or cellular immune response against a particular disease, the host immune system must come in contact with an antigen or an immunodominant epitope on an antigen associated with that disease state.
  • the term "antigen” refers to any agent, generally a macromolecule, which can elicit an immunological response in an individual.
  • the term "antigen" is used interchangeably with the term "immunogen”.
  • the immunological response may be of B- and/or T-lymphocytic cells.
  • antigenic macromolecule may be used to refer to an individual macromolecule or to a homogeneous or heterogeneous population of antigenic macromolecules.
  • antigenic macromolecule is used to refer to a protein molecule or portion thereof which contains one or more antigenic determinants or epitopes.
  • antigen means an immunogenic peptide or protein of interest comprising one or more epitopes capable of inducing a CMI response to an infectious Chlamydia pathogen.
  • the antigen can mclude but is not limited to an auto-antigen, a self-antigen, a cross-reacting antigen, an alloantigen, a tolerogen, an allergen, a hapten, an immunogen or parts thereof as well as any combinations thereof.
  • epitope generally refers to the site on an antigen which is recognised by a T-cell receptor and/or an antibody. Preferably it is a short peptide derived from or as part of a protein antigen. However the term is also intended to include peptides with glycopeptides and carbohydrate epitopes. Several different epitopes may be carried by a single antigenic molecule. The term “epitope” also includes modified sequences of amino acids or carbohydrates which stimulate responses which recognise the whole organism. It is advantageous if the selected epitope is an epitope of an infectious agent, such as a Chlamydia bacterium, which causes the infectious disease.
  • an infectious agent such as a Chlamydia bacterium
  • SEQ ID Nos 1-86 in the compositions of the invention may be supplemented or substituted with molecules comprising fragments of SEQ ID Nos 1-86.
  • Such fragments may comprise at least n consecutive monomers from the molecules and. depending on the particular sequence, n is either (i) 7 or more for protein molecules (eg. 8 18, 20 or more), preferably such that the fragment comprises an epitope from the sequence, or (ii) 10 or more for nucleic acid molecules (eg 15, 18, 20, 25, 30, 35, 40 or more).
  • the epitope can be generated from knowledge the amino acid and corresponding
  • DNA sequences of the peptide or polypeptide as well as from the nature of particular amino acids (e.g., size, charge, etc.) and the codon dictionary, without undue experimentation. See, e.g., Ivan Roitt, Essential Immunoloqy, 1988; Kendrew, supra; Janis Kuby, Immunology, 1992 e.g., pp. 79-81.
  • Some guidelines in determining whether a protein will stimulate a response include: Peptide length — preferably the peptide is about 8 or 9 amino acids long to fit into the MHC class I complex and about 13-25 amino acids long to fit into a class II MHC complex. This length is a minimum for the peptide to bind to the MHC complex.
  • the peptides may be longer than these lengths because cells may cut peptides.
  • the peptide may contain an appropriate anchor motif which will enable it to bind to the various class I or class II molecules with high enough specificity to generate an immune response (See Bocchia, M. et al, Specific Binding of Leukemia Oncogene Fusion Protein Pentides to HLA Class I Molecules, Blood 85:2680-2684; Englehard, NH, Structure of peptides associated with class I and class II MHC molecules Ann. Rev. Immunol. 12:181 (1994)). This can be done, without undue experimentation, by comparing the sequence of the protein of interest with published structures of peptides associated with the MHC molecules. Thus, the skilled artisan can ascertain an epitope of interest by comparing the protein sequence with sequences listed in the protein data base.
  • one or more antigens of the present invention contain one or more T cell epitopes.
  • T cell epitope refers generally to those features of a peptide structure which are capable of inducing a T cell response.
  • T cell epitopes comprise linear peptide determinants that assume extended conformations within the peptide-binding cleft of MHC molecules (Unanue et al. (1987) Science 236: 551-557).
  • a T cell epitope is generally a peptide having at least about 3-5 amino acid residues, and preferably at least 5-10 or more amino acid residues.
  • T cell epitope encompasses any MHC Class I-or MHC Class II restricted peptide.
  • the ability of a particular T cell epitope to stimulate/enhance a CMI response may be dete ⁇ nined by a number of well-known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T-cell cytotoxic cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject.
  • lymphoproliferation lymphoproliferation
  • CD8+ T-cell cytotoxic cell assays or by assaying for T-lymphocytes specific for the epitope in a sensitized subject.
  • the antigens of the present invention comprisse CD8+ T-cell inducing epitopes.
  • a CD8+ T-cell -inducing epitope is an epitope capable of stimulating the formation, or increasing the activity, of specific CD 8+ T-cells following its administration to a host subject.
  • the CD8+ T-cell epitopes may be provided in a variety of different forms such as a recombinant string of one or two or more epitopes. CD8+ T-cell epitopes have been identified and can be found in the literature, for many different diseases. It is possible to design epitope strings to generate CD8+ T- cell response against any chosen antigen that contains such CD8+ T-cell epitopes.
  • CD8+ T-cell inducing epitopes may be provided in a string of multiple epitopes which are linked together without intervening sequences so that unnecessary nucleic acid material is avoided.
  • the antigens of the present invention comprise helper T lymphocyte epitopes.
  • helper T lymphocyte epitopes Various methods are available to identify T helper cell epitopes suitable for use in accordance herewith. For example, the amphipathicity of a peptide sequence is known to effect its ability to function as a T helper cell inducer. A full discussion of T helper cell-inducing epitopes is given in U.S. Patent 5,128,319, inco ⁇ orated herein by reference.
  • the antigens of the present invention comprise a mixture of CD8+ T-cell epitopes and B cell epitopes.
  • B cell epitope generally refers to the site on an antigen to which a specific antibody molecule binds. The identification of epitopes which are able to elicit an antibody response is readily accomplished using techniques well known in the art. See, e. g., Geysen et al. (1984) Proc. Natl. Acad. Sci. USA 81: 3998-4002 (general method of rapidly synthesizing peptides to determine the location of immunogenic epitopes in a given antigen); U. S. Patent No.
  • the antigen or antigen combination comprises a mixture of a CD8+ T-cell -inducing epitopes and a T helper cell-inducing epitopes.
  • T and B cell inducing epitopes are frequently distinct from each other and can comprise different peptide sequences. Therefore certain regions of a protein's peptide chain can possess either T cell or B cell epitopes. Therefore, in addition to the CD8+ T-cell epitopes, it may be preferable to include one or more epitopes recognised by T helper cells, to augment the immune response generated by the CD8+ T-cell epitopes.
  • T helper cell inducing agents The mechanism of enhancing a CD8+ T-cell induced response in vivo by T helper cell inducing agents is not completely clear. However, without being bound by theory, it is likely that the enhancing agent, by virtue of its ability to induce T helper cells, will result in increased levels of necessary cytokines that assist in the clonal expansion and dissemination of specific CD8+ T-cells. Regardless of the underlying mechanism, it is envisioned that the use of mixtures of helper T cell and CD8+ T-cell -inducing antigen combinations of the present invention will assist in the enhancement of the CMI response.
  • T helper cell epitopes are ones which are active in individuals of different HLA types, for example T helper epitopes from tetanus (against which most individuals will already be primed). It may also be useful to include B cell epitopes for stimulating B cell responses and antibody production. Synthetic nucleotide sequences may also be constructed to produce two types of immune responses: T cell only and T cell combined with a B cell response.
  • an individual is immunized with an antigen or combination of antigens or nucleotide sequence or combinations of nucleotide sequences encoding multiple epitopes of a target antigen
  • the majority of responding T lymphocytes will be specific for one or more linear epitopes from that target antigen and/or a majority of the responding B lymphocytes will be specific for one or more linear or conformational epitopes for the antigen or combination of antigens.
  • epitopes are referred to as "immunodominant epitopes”.
  • a single epitope may be the most dominant in terms of commanding a specific T or B cell response.
  • compositions of the present invention may be administered in conjunction with other immunoregulatory agents.
  • the compositions of the present i .nnvvpe/nnttiinonn m m-avy h bie. a aHdmmi.nni.ssttfe»rrfevd. w wiithh a ann . ai_d.j_u_ _vv.ainntt.
  • an adjuvant may be useful in further enhancing or modulating the CMI response.
  • An adjuvant may enhance the CMI response by enhancing the immunogenicity of a co-administered antigen in an immunized subject, as well inducing a Thl-like immune response against the co- administered antigen which is beneficial in a vaccine product.
  • An immune response and particularly a CMI response may be refined, by the addition of adjuvants to combinations of antigens or nucleotide sequences encoding combinations of antigens which lead to particularly effective compositions for eliciting a long lived and sustained enhanced CMI response.
  • adjuvant refers to any material or composition capable of specifically or non-specifically altering, enhancing, directing, redirecting, potentiating or initiating an antigen-specific immune response.
  • adjuvant includes but is not limited to a bacterial ADP-ribosylating exotoxin, a biologically active factor, immunomodulatory molecule, biological response modifier or immunostimulatory molecule such as a cytokine, an interleukin, a chemokine or a ligand or an epitope (such as a helper T cell epitope) and optimally combinations thereof which, when administered with an antigen, antigen composition or nucleotide sequence encoding such antigens enhances or potentiates or modulates the CMI response relative to the CMI response generated upon administration of the antigen or combination of antigens alone.
  • the adjuvant may be any adjuvant known in the art which is appropriate for human or animal use.
  • Immunomodulatory molecules such as cytokines (TNF-alpha, IL-6, GM-CSF, and IL- 2), and co-stimulatory and accessory molecules (B7-1, B7-2) may be used as adjuvants in a variety of combinations.
  • GM-CSF is not administered to subject before, in or after the administration regimen.
  • Simultaneous production of an immunomodulatory molecule and an antigen of interest at the site of expression of the antigen of interest may enhance the generation of specific effectors which may help to enhance the CMI response.
  • the degree of enhancement of the CMI response may be dependent upon the specific immunostimulatory molecules and/or adjuvants used because different immunostimulatory molecules may elicit different mechanisms for enhancing and or modulating the CMI response.
  • the different effector mechanisms/immunomodulatory molecules include but are not limited to augmentation of help signal (IL-2), recruitment of professional APC (GM-CSF), increase in T cell frequency (IL-2), effect on antigen processing pathway and MHC expression (IFN-gamma and TNF-alpha) and diversion of immune response away from the Thl response and towards a Th2 response (LTB) (see WO 97/02045).
  • IL-2 help signal
  • GM-CSF professional APC
  • IL-2 increase in T cell frequency
  • IFN-gamma and TNF-alpha effect on antigen processing pathway and MHC expression
  • LTB Th2 response
  • Unmethylated CpG containing oligonucleotides are also preferential inducers of a Thl response and are suitable for use in the present invention.
  • an adjuvant is advantageous because the adjuvant may help to enhance the CMI response to the expressed antigen by diverting the Th2 response to a Thl response and/or specific effector associated mechanisms to an expressed epitope with the consequent generation and maintenance of an enhanced CMI response (see, for example, the teachings in WO 97/02045).
  • an adjuvant with an antigen or nucleotide sequence encoding the antigen is also advantageous because it may result in a lower dose or fewer doses of the antigen/antigenic combination being necessary to achieve the desired CMI response in the subject to which the antigen or nucleotide sequence encoding the antigen is administered, or it may result in a qualitatively and/or quantitatively different immune response in the subject.
  • the effectiveness of an adjuvant can be determined by administering the adjuvant with the antigen in parallel with the antigen alone to animals and comparing antibody and/or cellular-mediated immunity in the two groups using standard assays such as radioimmunoassay, ELISAs, CD8+ T-cell assays, and the like, all well known in the art.
  • the adjuvant is a separate moiety from the antigen, although a single molecule (such for example, CTB) can have both adjuvant and antigen properties.
  • the term “genetic adjuvant” refers to an adjuvant encoded by a nucleotide sequence and which, when administered with the antigen enhances the CMI response relative to the CMI response generated upon administration of the antigen alone.
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention.
  • the protein is derived from E. coli (i.e., E. coli heat labile enterotoxin "LT), cholera ("CT”), or pertussis ("PT").
  • the genetic adjuvant is a bacterial ADP-ribosylating exotoxin.
  • ADP-ribosylating bacterial toxins are a family of related bacterial exotoxins and include diphtheria toxin (DT), pertussis toxin (PT), cholera toxin (CT), the E. coli heat-labile toxins (LT1 and LT2), Pseudomonas endotoxin A, Pseudomonas exotoxin S, B. cereus exoenzyme, B. sphaericus toxin, C. botulinum C2 and C3 toxins, C. limosum exoenzyme, as well as toxins from C. perfringens, C. spiriforma and C.
  • DT diphtheria toxin
  • PT pertussis toxin
  • CT cholera toxin
  • LT1 and LT2 the E. coli heat-labile toxins
  • Pseudomonas endotoxin A Pseudomonas exo
  • ADP-ribosylating bacterial toxin mutants such as CRM] 9 , a non-toxic diphtheria toxin mutant (see, e.g., Bixler et al. (1989) Adv. Exp. Med. Biol. 251:175; and Constantino et al. (1992) Vaccine).
  • Most ADP- ribosylating bacterial toxins are organized as an A:B multimer, wherein the A subunit contains the ADP-ribosyltransferase activity, and the B subunit acts as the binding moiety.
  • Preferred ADP-ribosylating bacterial toxins for use in the compositions of the present invention include cholera toxin and the E. coli heat-labile toxins.
  • Cholera toxin (CT) and the related E. coli heat labile enterotoxins (LT) are secretion products of their respective enterotoxic bacterial strains that are potent immunogens and exhibit strong toxicity when administered systemically, orally, or mucosally. Both CT and LT are known to provide adjuvant effects for antigen when administered via the intramuscular or oral routes. These adjuvant effects have been observed at doses below that required for toxicity.
  • the two toxins are extremely similar molecules, and are at least about 70-80%, homologous at the amino acid level.
  • the genetic adjuvant is cholera toxin (CT), enterotoxigenic E. Coli heat- labile toxin (LT), or a derivative, subunit, or fragment of CT or LT which retains adjuvanticity.
  • CT cholera toxin
  • LT enterotoxigenic E. Coli heat- labile toxin
  • the genetic adjuvant is LT.
  • the genetic adjuvant may be CTB or LTB.
  • the entertoxin is a non-toxic enterotoxin.
  • the use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in WO 95/17211 and as parenteral adjuvants in WO 98/42375.
  • the toxin or toxoid is preferably in the form of a holotoxin, comprising both A and B subunits.
  • the A subunit contains a detoxifying mutation; preferably the B subunit is not mutated.
  • the adjuvant is a detoxified LT mutant such as LT-K63, LT- R72, and LTR192G.
  • 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 inco ⁇ orated by reference herein in their entirety (Beignon, et al. Infection and Immunity (2002) 70(6):3012 - 3019; Pizza, et al., Vaccine (2001) 19:2534 - 2541; Pizza, et al, Int. J. Med. Microbiol (2000) 290(4- 5):455-461; Scharton-Kersten et al. Infection and Immunity (2000) 68(9):5306 - 5313; Ryan et al.
  • At least one of the entertoxin subunit coding regions may be genetically modified to detoxify the subunit peptide encoded thereby, for example wherein the truncated A subunit coding region has been genetically modified to disrupt or inactivate ADP-ribosyl transferase activity in the subunit peptide expression product (see, for example, WO 03/004055).
  • this genetic adjuvant is particularly desirable where an even more enhanced CMI response is desired.
  • Other desirable genetic adjuvants include but are not limited to nucleotide sequences encoding IL-10, IL-12, IL-13, the interferons (IFNs) (for example, IFN-alpha, IFN-ss, and IFN-gamma), and preferred combinations thereof.
  • IFNs interferons
  • Still other such biologically active factors that enhance the CMI response may be readily selected by one of skill in the art, and a suitable plasmid vector containing same constructed by known techniques.
  • Preferred further adjuvants include, but are not limited to, one or more of the following set forth below: Mineral Containing Compositions
  • Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminium salts and calcium salts.
  • the invention includes mineral salts such as hydroxides (e.g. oxyhydroxides), phosphates (e.g. hydroxyphoshpates, orthophosphates), sulphates, etc. ⁇ e.g. see chapters 8 & 9 of ref. Bush and Everett (2001) Int J Syst Evol Microbiol 51: 203-220), or mixtures of different mineral compounds, with the compounds taking any suitable form (e.g. gel, crystalline, amo ⁇ hous, etc.), and with adso ⁇ tion being preferred.
  • the mineral containing compositions may also be formulated as a particle of metal salt. See WO 00/23105.
  • Aluminum salts may be included in immunogenic compositions and/or vaccines of the invention such that the dose of Al 3+ is between 0.2 and 1.0 mg per dose.
  • the adjuvant is alum, preferably an aluminium salt such as aluminium hydroxide (AIOH) or aluminium phospate or aluminium sulfate. Still more preferably the adjuvant is aluminium hydroxide (AIOH).
  • a mineral salt such as an aluminium salt, is combined with and another adjuvant, such as an oligonucleotide containing a CpG motif or an ADP ribosylating toxin. Still more preferably, the mineral salt is combined with an oligonucleotide containing a CpG motif. 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, 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) 21:4234-4237. MF59 is used as the adjuvant in the FLUADTM influenza virus bivalent subunit vaccine.
  • Particularly preferred adjuvants for use in the compositions are submicron oil-inwater 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 TM (polyoxyelthylenesorbitan monooleate), and/or 0.25-1.0%) Span 85TM (sorbitan trioleate), and, optionally, N-acetylmuramyl-L-alanyl-D-isogluatminyl-L- alanine-2-( -2 , -dipalmitoyl-5 , ra-glycero-3-huydroxyphosphophoryloxy)-ethylamine
  • MTP-PE submicron oil-in-water emulsion
  • MF59 submicron oil-in-water emulsion
  • International Publication No. WO90/14837 US Patent Nos. 6,299,884 and 6,451,325, inco ⁇ orated 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.
  • MTP-PE may be present in an amount of about 0-500 ⁇ g/dose, more preferably 0-250 ⁇ g/dose and most preferably, 0-100 ⁇ g/dose.
  • 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.
  • MF59-100 contains 100 ⁇ g 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 80TM, and 0.75% w/v Span 85TM and optionally MTP-PE.
  • MF75 also known as SAF, containing 10% squalene, 0.4% Tween 80TM, 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 ⁇ g 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. WO90/14837 and US Patent Nos. 6,299,884 and 6,45 1,325, inco ⁇ orated herein by reference in their entireties.
  • Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IF A) may also be used as adjuvants in the invention.
  • Saponin formulations may also be used as adjuvants in the invention.
  • Saponins are a heterologous group of sterol glycosides and trite ⁇ enoid 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. Patent No. 5,057,540. Saponin formulations may also comprise a sterol, such as cholesterol (see WO 96/33739).
  • a sterol such as cholesterol
  • ISCOMs Combinations of saponins and cholesterols can be used to form unique particles called ___nmunostim.ula.ing Complexs (ISCOMs).
  • ISCOMs typically also include a phospholipid such as phosphatidylethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs.
  • the ISCOM includes one or more of Quil A, QHA and QHC.
  • ISCOMS may be devoid of additional detergent. See WO 00/07621.
  • VLPs Virosomes and Virus Like Particles
  • Virosomes and Virus Like Particles 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.
  • 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, Q ⁇ -phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein pi).
  • 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 Norwalk virus
  • human Papilloma virus HIV
  • RNA- phages Q ⁇ -phage (such as coat proteins)
  • GA-phage such as fr-phage,
  • VLPs are discussed further in WO 03/024480, WO 03/024481; Niikura et al Virology (2002) 293:273 - 280; Lenz et al Journal of Immunology (2001) 5246 - 5355; Pinto, et al Journal of Infectious Diseases (2003) 188:327 - 338; and Gerber et al Journal of Virology (2001) 75(10):4752 - 47601; Virosomes are discussed further in, for example, Gluck et al Vaccine (2002) 20:B10 -B16.
  • Bacterial or Microbiol Derivatives are discussed further in, for example, Gluck et al Vaccine (2002) 20:B10 -B16.
  • Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as: 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 tlirough 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) BioorgMed Chem Lett 9:2273-2278.
  • 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. Vaccine (2003) 21:2485 - 2491; Pajak, et al Vaccine (2003) 21:836 - 842.
  • 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.
  • the guanosine may be replaced with an analog such as 2'-deoxy-7-deazaguanosine. See Kandimalla, et al Nucleic Acids Research (2003) 31(9): 2393 - 2400; WO 02/26757 and WO 99/62923 for examples of possible analog substitutions.
  • the CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT. See Kalman et al (1999) (Nature Genetics 21: 385-389).
  • the CpG sequence may be specific for inducing a Thl 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 J. Immunol. (2003) 170(8):4061 - 4068; Krieg BBRC (2003) 306:948 - 953; and WO 01/95935.
  • the CpG is a CpG-A ODN.
  • the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition.
  • two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers". See, for example, Kandimalla, et al (2003) 31(part 3):664 - 658; Bhagat et al BBRC (2003) 300:853 - 861 and WO 03/035836.
  • the adjuvant is CpG. Even more preferably, the adjuvant is Alum and an oligonucleotide containg a CpG motif or AIOH and an oligonucleotide containing a CpG motif.
  • Human immunomodulators suitable for use as adjuvants in the invention include cytokinfes, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g. interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • cytokinfes such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g. interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • ADP-ribosylating toxins and detoxified derivatives thereof.
  • Bacterial ADP-ribosylating toxins and detoxified derivatives thereof may be used as adjuvants in the invention.
  • the protein is derived from E. coli (i.e., E. coli heat labile enterotoxin "LT), cholera ("CT"), or pertussis ("PT").
  • LT E. coli heat labile enterotoxin
  • CT cholera
  • PT pertussis
  • the use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in W095/17211 and as parenteral adjuvants in W098/42375.
  • the adjuvant is a detoxified LT mutant such as LT-K63, LT-R72, and LTR192G.
  • 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 inco ⁇ orated 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.
  • Vaccine Differential Effects of the Nontoxic AB Complex and Enzyme Activity on Thl 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.
  • 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 inco ⁇ orated herein by reference in its entirety.
  • the adjuvant is an ADP-ribosylating toxin and an oligonucleotide containing a CpG motif (see for example, WO 01/34185)
  • the adjuvant is a detoxified ADP-ribosylating toxin and an oligonucleotide containing a CpG motif.
  • the detoxified ADP-ribosylating toxin is LTK63 or LTK72.
  • the adjuvant is LTK63.
  • the adjuvant is LTK72.
  • the adjuvant is LTK63 and an oligonucleotide containing a CpG motif.
  • the adjuvant is LTK72 and an oligonucleotide containing a CpG motif.
  • Bioadhesives and mucoadhesives may also be used as adjuvants in the invention.
  • Suitable bioadhesives include esterified hyaluronic acid microspheres (Singh et al.
  • 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. See for example, WO99/27960.
  • Microparticles may also be used as adjuvants in the invention.
  • Microparticles i.e. a particle of -lOOnm to ⁇ 150 ⁇ m in diameter, more preferably ⁇ 200nm to ⁇ 30 ⁇ m in diameter, and most preferably ⁇ 500nm to ⁇ 10 ⁇ m in diameter
  • materials that are biodegradable and non-toxic e.g. a poly( ⁇ -hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.
  • a negatively- charged surface e.g. with SDS
  • a positively-charged surface e.g. with a cationic detergent, such as CTAB
  • liposome formulations suitable for use as adjuvants are described in U.S.
  • Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters (W099/52549). Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol (WOO 1/21207) as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol (WO01/21152).
  • 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.
  • PCPP Polyphosphazene
  • PCPP formulations are described, for example, in Andrianov et al Biomaterials
  • muramyl peptides suitable for use as adjuvants in the invention include N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L- alanyl-D-isoglutamine (nor-MDP), and N-acetyhnuramyl-L-alanyl-D-isoglutaminyl-L- alanine-2-(r-2 , -dipalmitoyl-5ra-glycero-3-hydroxyphosphoryloxy)-ethylamine MTP- PE).
  • thr-MDP N-acetyl-muramyl-L-threonyl-D-isoglutamine
  • nor-MDP N-acetyl-normuramyl-L- alanyl-D-isoglutamine
  • 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.
  • the following adjuvant compositions may be used in the invention:
  • a saponin e.g. QS21
  • 3dMPL + IL-12 optionally + a sterol (W098/57659); combinations of 3dMPL with, for example, QS21 and/or oil-in-water emulsions (European patent applications 0835318, 0735898 and 0761231).
  • 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.
  • Ribi adjuvant system (RAS), (Ribi Immunochem) containing 2%> Squalene, 5 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 (DetoxTM); and (7) one or more mineral salts (such as an aluminum salt) + a non-toxic derivative of LPS (such as 3dPML).
  • MPL monophosphorylipid A
  • TDM trehalose dimycolate
  • CWS cell wall skeleton
  • LPS such as 3dPML
  • Bacterial toxins and bioadhesives are preferred adjuvants for use with mucosally-delivered vaccines, such 20 as nasal vaccines.
  • the composition may include an antibiotic.
  • the compositions of the present invention are administered with alum and/or CpG sequences.
  • 25 Nucleic Acid The antigens or epitopes of the present invention may be administered as nucleotide sequences encoding the antigens or epitopes.
  • nucleotide sequence refers to one of more nucleotide sequences which encode one or more 30 epitopes which are used in the compositions or combinations of the present invention.
  • NOI nucleotide sequence
  • polynucleotide or “nucleic acid”.
  • the NOI may be DNA or RNA of genomic or synthetic or of recombinant origin.
  • the NOI may be double-stranded or single-stranded whether representing the sense or antisense strand or combinations thereof.
  • the NOI is DNA.
  • the NOI is prepared by use of recombinant DNA techniques (e.g. recombinant DNA).
  • the NOI is cDNA.
  • the NOI may be the same as the naturally occurring form.
  • 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 pu ⁇ oses).
  • 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.
  • 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).
  • a process for producing nucleic acid of the invention comprising the step of synthesising at least part of the nucleic acid by chemical means.
  • an antigen or antigenic combination or NOI encoding same is administered directly to a host subject.
  • a vector comprising an NOI is administered to a host subject.
  • the NOI is prepared and/or administered using a genetic vector.
  • a vector is a tool that allows or faciliates the transfer of an entity from one environment to another.
  • some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a host and/or a target cell for the pu ⁇ ose of replicating the vectors comprising the NOI of the present invention and/or expressing the antigens or epitopes of the present invention encoded by the NOI.
  • examples of vectors used in recombinant DNA techniques include but are not limited to plasmids, chromosomes, artificial chromosomes or viruses.
  • the term “vector” includes expression vectors and/or transformation vectors.
  • expression vector means a construct capable of in vivo or in vitro/ex vivo expression.
  • transformation vector means a construct capable of being transferred from one species to another.
  • the vectors comprising the NOI of the present invention may be administered directly as "a naked nucleic acid construct", preferably further comprising flanking sequences homologous to the host cell genome.
  • naked DNA refers to a plasmid comprising the NOI of the present invention together with a short promoter region to control its production. It is called “naked” DNA because the plasmids are not carried in any delivery vehicle.
  • a host cell such as a eukaryotic cell, the proteins it encodes are transcribed and translated within the cell.
  • the vectors comprising the NOI of the present invention may be introduced into suitable host cells using a variety of viral techniques which are known in the art, such as for example infection with recombinant viral vectors such as retroviruses, he ⁇ es simplex viruses and adenoviruses.
  • the vector may be a recombinant viral vectors.
  • Suitable recombinant viral vectors include but are not limited to adeno virus vectors, adeno-associated viral (AAV) vectors, he ⁇ es-virus vectors, a retroviral vector, lentiviral vectors, baculoviral vectors, pox viral vectors or parvovirus vectors (see Kestler et al 1999 Human Gene Ther 10(10): 1619-32).
  • AAV adeno-associated viral
  • he ⁇ es-virus vectors he ⁇ es-virus vectors
  • retroviral vector lentiviral vectors
  • baculoviral vectors pox viral vectors or parvovirus
  • target vector refers to a vector whose ability to infect or transfect or transduce a cell or to be expressed in a host and/or target cell is restricted to certain cell types within the host subject, usually cells having a common or similar phenotype.
  • the NOI of the present invention which is inserted into a vector is operably linked to a control sequence that is capable of providing for the expression of the antigens or epitopes by the host cell, i.e. the vector is an expression vector.
  • the agent produced by a host cell may be secreted or may be contained intracellularly depending on the NOI and/or the vector used.
  • expression vectors containing the NOI can be designed with signal sequences which direct secretion of the EOI through a particular prokaryotic or eukaryotic cell membrane.
  • Chlamydia pneumoniae antigens 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, 18, 19 or 20) of the antigens are expressed as a single polypeptide chain (a 'hybrid' polypeptide).
  • Hybrid 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 hybrid 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 hybrid polypeptide may comprise two or more polypeptide sequences from the first antigen group.
  • the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a Chlamydia bactgerium, preferably a Chlamydia pneumoniae antigen or a fragment thereof of the first antigen group.
  • the first and second amino acid sequences in the hybrid polypeptide comprise different epitopes.
  • the hybrid polypeptide may comprise two or more polypeptide sequences from the second antigen group.
  • the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a Chlamydia pneumoniae antigen or a fragment thereof of the second antigen group.
  • the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes.
  • the hybrid polypeptide may comprise one or more polypeptide sequences from the first antigen group and one or more polypeptide sequences from the second antigen group.
  • the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group and said second amino acid sequence selected from a Chlamydia bactgerium, preferably a Chlamydia pneumoniae antigen or a fragment thereof from the second antigen group.
  • the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes.
  • the hybrid polypeptide may comprise one or more polypeptide sequences from the first antigen group and one or more polypeptide sequences from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.
  • the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group and said second amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.
  • the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes.
  • the hybrid polypeptide may comprise one or more polypeptide sequences from the second antigen group and one or more polypeptide sequences from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.
  • the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the second antigen group and said second amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.
  • the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes.
  • Hybrids consisting of amino acid sequences from two, tliree, four, five, six, seven, eight, nine, or ten Chlamydia pneumoniae antigens are preferred.
  • hybrids consisting of amino acid sequences from two, three, four, or five Chlamydia pneumoniae antigens are preferred.
  • Different hybrid polypeptides may be mixed together in a single formulation.
  • a Chlamydia pneumoniae 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.
  • Two-antigen hybrids for use in the invention may comprise any one of the combinations disclosed above.
  • Hybrid polypeptides can be represented by the formula NH 2 -A- ⁇ -X-L- ⁇ diligent-B-COOH, wherein: X is an amino acid sequence of a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group, the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.; 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 ra is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15.
  • a -X- moiety has a leader peptide sequence in its wild-type form, this may be included or omitted in the hybrid protein.
  • 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 Xi will be retained, but the leader peptides of X 2 ... X n will be omitted. This is equivalent to deleting all leader peptides and using the leader peptide of Xi as moiety -A-.
  • linker amino acid sequence -L- may be present or absent.
  • the hybrid may be NH 2 -X ⁇ -L ⁇ -X 2 -L 2 -COOH, NH 2 -Xj- X 2 -COOH, NH 2 -X!-L ⁇ -X 2 -COOH, NH 2 -X ! -X 2 -L 2 -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).
  • Other suitable linker amino acid sequences will be apparent to those skilled in the art.
  • a useful linker is GSGGGG (SEQ ID No 77), with the Gly-Ser dipeptide being formed from a BamYS. 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.
  • Other suitable N-terminal amino acid sequences will be apparent to those skilled in the art. If Xi lacks its own N-terminus methionine, -A- is preferably an oligopeptide (e.g.
  • the invention also provides nucleic acid encoding hybrid polypeptides of the invention. Furthermore, the invention provides nucleic acid which can hybridise to this nucleic acid, preferably under "high stringency" conditions (e.g. 65°C in a O.lxSSC, 0.5%> SDS solution).
  • "high stringency" conditions e.g. 65°C in a O.lxSSC, 0.5%> SDS solution.
  • the NOI of the present invention may be expressed as a fusion protein comprising an adjuvant and/or a biological response modifier and/or immunomodulator fused to the antigens or epitopes of the present invention to further enhance and/or augment the CMI response obtained.
  • the biological response modifier may act as an adjuvant in the sense of providing a generalised stimulation of the CMI response.
  • the antigens or epitopes may be attached to either the amino or carboxy terminus of the biological response modifier.
  • 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 Chlamydial or host cell proteins).
  • 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 further provides a process for producing a composition according to the invention comprising the step of bringing one or more of SEQ IDs 1-86 into combination with one or more other of SEQ IDs 1-86
  • Preferred polypeptides of the invention comprise an amino acid sequence found in
  • the individual antigens within the hybrid may be from one or more strains.
  • X 2 may be from the same strain as Xj or from a different strain.
  • heterologous host Whilst expression of the polypeptides of the invention may take place in Chlamydia, the invention preferably utilises 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), yeasts, etc.
  • compositions include a protein that exists in different nascent and mature forms
  • the mature form of the protein is preferably used.
  • the mature form of the Chlamydia pneumoniae protein lacking the signal peptide may be used
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal ⁇ e.g. see
  • the invention may be used to elicit systemic and/or mucosal immunity.
  • the compositions of the present invention may be administered, either alone or as part of a composition, via a variety of different routes. Certain routes may be favoured for certain compositions, as resulting in the generation of a more effective immune response, prefereably a CMI response, or as being less likely to induce side effects, or as being easier for administration.
  • compositions of the present invention may be administered via a systemic route or a mucosal route or a transdermal route or it may be administrered directly into a specific tissue.
  • systemic administration includes but is not limited to any parenteral routes of administration.
  • parenteral administration includes but is not limited to subcutaneous, intraperitoneal, intravenous, intraarterial, intramuscular, or intrasternal injection, intravenous, intraarterial, or kidney dialytic infusion techniques.
  • the systemic, parenteral administration is intramuscular injection.
  • the compositions of the present invention are administered via a transdermal route.
  • transdermal administration of a composition may be preferred because it more efficiently activates the cell mediated immune (CMI) arm of the immune system.
  • CMI cell mediated immune
  • transdermal delivery intends intradermal (e.g., into the dermis or epidermis), transdermal (e.g.,”percutaneous") and transmucosal administration, i.e., delivery by passage of an agent into or through skin or mucosal tissue.
  • Transdermal Drug Delivery Developmental Issues and Research Initiatives, Hadgraft and Guy (eds.), Marcel Dekker, Inc., (1989); Controlled Drug Delivery: Fundamentals and Applications, Robinson and Lee (eds.), Marcel Dekker Inc.,(1987); and Transdermal Delivery of Drugs, Vols. 1-3, Kydonieus and Berner (eds.), CRC Press, (1987).
  • the term encompasses delivery of an agent using a particle delivery device (e.g., a needleless syringe) such as those described in U.S. Patent No. 5,630,796, as well as delivery using particle-mediated delivery devices such as those described in U.S. Patent No. 5,865,796.
  • a particle delivery device e.g., a needleless syringe
  • particle-mediated delivery devices such as those described in U.S. Patent No. 5,865,796.
  • the term “mucosal administration” includes but is not limited to oral, intranasal, intravaginal, intrarectal, intratracheal, intestinal and ophthalmic administration.
  • Mucosal routes particularly intranasal, intratracheal, and ophthalmic are preferred for protection against natural exposure to environmental pathogens such as RSV, flu virus and cold viruses or to allergens such as grass and ragweed pollens and house dust mites.
  • the enhancement of the immune response preferably the CMI response will enhance the protective effect against a subsequently encountered target antigen such as an allergen or microbial agent.
  • the compositions of the present invention may be administered to cells which have been isolated from the host subject.
  • the composition is administered to professional antigen presenting cells (APCs), such as dendritic cells.
  • APCs may be derived from a host subject and modified ex vivo to express an antigen of interest and then transferred back into the host subject to induce an enhanced CMI response.
  • Dendritic cells are believed to be the most potent APCs for stimulating enhanced CMI responses because the expressed epitopes of the antigen of interest must be acquired, processed and presented by professional APCs to T cells (both Thl and Th2 helper cells as well as CD8+ T-cells) in order to induce an enhanced CMI response.
  • Particle-mediated methods for delivering the compositions of the present invention are known in the art.
  • the above-described antigens or NOI encoding same can be coated onto core carrier particles using a variety of techniques known in the art.
  • Carrier particles are selected from materials which have a suitable density in the range of particle sizes typically used for intracellular delivery from a gene gun device. The optimum carrier particle size will, of course, depend on the diameter of the target cells.
  • core carrier is meant a carrier on which a guest antigen or guest nucleic acid (e.g., DNA, RNA) is coated in order to impart a defined particle size as well as a sufficiently high density to achieve the momentum required for cell membrane penetration, such that the guest molecule can be delivered using particle-mediated techniques (see, e.g., U.S. Patent No. 5,100,792).
  • Core carriers typically include materials such as tungsten, gold, platinum, ferrite, polystyrene and latex. See e.g., Particle Bombardment Technology for Gene Transfer, (1994) Yang, N. ed., Oxford University Press, New York, NY pages 10-11. Tungsten and gold particles are preferred.
  • Tungsten particles are readily available in average sizes of 0.5 to 2.0 microns in diameter.
  • Gold particles or microcrystalline gold e. g., gold powder A1570, available from Engelhard Co ⁇ ., East Newark, NJ
  • Gold particles provide uniformity in size (available from Alpha Chemicals in particle sizes of 1-3 microns, or available from Degussa, South Plainfield, NJ in a range of particle sizes including 0.95 microns).
  • Microcrystalline gold provides a diverse particle size distribution, typically in the range of 0.5-5 microns. However, the irregular surface area of microcrystalline gold provides for highly efficient coating with nucleic acids. A number of methods are known and have been described for coating or precipitating NOIs onto gold or tungsten particles.
  • Most such methods generally combine a predetermined amount of gold or tungsten with plasmid DNA, CaC12 and spermidine.
  • the resulting solution is vortexed continually during the coating procedure to ensure uniformity of the reaction mixture.
  • the coated particles can be transferred to suitable membranes and allowed to dry prior to use, coated onto surfaces of a sample module or cassette, or loaded into a delivery cassette for use in particular gene gun instruments.
  • the particle compositions or coated particles are administered to the individual in a manner compatible with the dosage formulation, and in an amount that will be effective for the pu ⁇ oses of the invention.
  • the amoimt of the composition to be delivered e. g., about 0.1 mg to 1 mg, more preferably 1 to 50 ug of the antigen or allergen, depends on the individual to be tested. The exact amount necessary will vary depending on the age and general condition of the individual to be treated, and an appropriate effective amount can be readily determined by one of skill in the art upon reading the instant specification.
  • the term "host mammalian subject” means any member of the subphylum cordata, including, without limitation, humans and other primates, including non-human primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like.
  • the terms do not denote a particular age. Thus, both adult and newborn individuals are intended to be covered.
  • the methods described herein are intended for use in any of the above vertebrate species, since the immune systems of all of these vertebrates operate similarly. If a mammal, the subject will preferably be a human, but may also be a domestic livestock, laboratory subject or pet animal.
  • the mammal is preferably a human.
  • the human is preferably a child (e.g. a toddler or infant) or a teenager; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult.
  • a vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
  • 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 and to the preparation of a vaccine to prevent and/or treat an disorder associated with a Chlamydia bacterium. It is to be appreciated that all references herein to treatment include curative, palliative and prophylactic treatment.
  • the administration of antigenic combinations of the present invention or a composition comprising the NOI encoding the antigenic combinations may be for either "prophylactic” or "therapeutic” pu ⁇ ose.
  • therapeutic or “treatment” includes any of following: the prevention of infection or reinfection; the reduction or elimination of symptoms; and the reduction or complete elimination of a pathogen. Treatment may be effected prophylactically (prior to infection) or therapeutically (following infection).
  • Prophylaxis or therapy includes but is not limited to eliciting an effective immune response, preferably a CMI immune response and/or alleviating, reducing, curing or at least partially arresting symptoms and/or complications resulting from a T cell mediated immune disorder.
  • the composition of the present invention When provided prophylactically, the composition of the present invention is typically provided in advance of any symptom.
  • the prophylactic administration of the composition of the present invention is to prevent or ameliorate any subsequent infection or disease.
  • the composition of the present invention When provided therapeutically, the composition of the present invention is typically provided at (or shortly after) the onset of a symptom of infection or disease.
  • the composition of the present invention may be provided either prior to the anticipated exposure to a disease causing agent or disease state or after the initiation of an infection or disease.
  • prophylactic or therapeutic administration is the more appropriate will usually depend upon the nature of the disease.
  • immunotherapeutic composition of the present invention could be used in immunotherapy protocols to actively inducing immunity by vaccination. This latter form of treatment is advantageous because the immunity is prolonged.
  • a vaccine composition will preferably, though not necessarily be used prophylactically to induce an effective CMI response against subsequently encountered antigens or portions thereof (such as epitopes) related to the target antigen.
  • Chlamydia e.g. trachoma, pelvic inflammatory disease, epididymitis, infant pneumonia, artherosclerosis, cardiovascular disease etc.
  • the compositions may also be effective against C.pneumoniae.
  • composition dose administrated to a host subject should be sufficient to effect a beneficial prophylactic or therapeutic immune response, preferably a CMI response in the subject over time.
  • 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.
  • the method may raise a booster response.
  • prophylactically or therapeutically effective dose means a dose in an amount sufficient to elicit an enhanced immune response, preferably a CMI response to one or more antigens or epitopes and/or to alleviate, reduce, cure or at least partially arrest symptoms and/or complications from a T cell mediated immune disorder.
  • Immunogenic compositions used as vaccines comprise an immunologically effective amount of antigen(s), as well as any other components, as needed.
  • '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. 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.
  • the mammal is preferably a human.
  • the human is preferably a child (e.g. a toddler or infant) or a teenager or an adult; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult.
  • a vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
  • the human is a teenager. More preferably, the human is a pre-adolescent teenager. Even more preferably, the human is a pre- adolescent female or male Preferably the pre-adolescent male or female is around 9- 12 years of age.
  • One 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 or by protein or DNA microarray. 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.
  • One way of checking efficacy of therapeutic treatment involves monitoring Chlamydia infection after administration of the composition of the invention.
  • One way of checking efficacy of prophylactic treatment involves monitoring immune responses against the Chlamydia antigen, such as the Chlamydia pneumoniae antigen in the compositions of the invention after administration of the composition.
  • checking efficacy of prophylactic treatment may involve monitoring immune responses both sys temically (such as monitoring the level of IgGl and IgG2a production) and mucosally (such as monitoring the level of IgA production) against the Chlamydia pneumoniae antigens in the compositions of the invention after administration of the composition.
  • serum Chlamydia specific antibody responses are determined post-immunization but pre-challenge whereas mucosal Chlamydia specific antibody body responses are determined post-immunization and post-challenge.
  • Chlamydia pneumoniae e.g. pneumonia, bronchitis, pharyngitis, sinusitis, erythema nodosum, asthma, atherosclerosis, stroke, myocardial infarctions, coronary artery disease, etc.
  • a disease caused by Chlamydia pneumoniae e.g. pneumonia, bronchitis, pharyngitis, sinusitis, erythema nodosum, asthma, atherosclerosis, stroke, myocardial infarctions, coronary artery disease, etc.
  • 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.
  • in vitro neutralization by Peterson et al (1988) is suitable for testing vaccine compositions directed toward Chlamydia, preferably Chlamydia pneumoniae.
  • Hyper-immune antisera is diluted in PBS containing 5%> guinea pig serum, as a complement source.
  • Chlamydia pneumoniae (10 4 IFU; inclusion forming units) are added to the antisera dilutions.
  • the antigen-antibody mixtures are incubated at 37°C for 45 minutes and inoculated into duplicate confluent Hep-2 or HeLa cell monolayers contained in glass vials (e.g., 15 by 45 mm), which have been washed twice with PBS prior to inoculation.
  • the monolayer cells are infected by centrifugation at 1000X g for 1 hour followed by stationary incubation at 37°C for 1 hour.
  • Infected monolayers are incubated for 48 or 72 hours, fixed and stained with Chlamydia specific antibody, such as anti-MOMP. Inclusion-bearing cells are counted in ten fields at a magnification of 200X. Neutralization titer is assigned on the dilution that gives 50%o inhibition as compared to control monolayers/IFU.
  • the efficacy of immunogenic compositions can also be determined in vivo by challenging animal models of Chlamydia pneumoniae infection, e.g., guinea pigs or mice, with the immunogenic compositions.
  • the immunogenic compositions may or may not be derived from the same serovars as the challenge serovars.
  • the immunogenic compositions are derivable from the same serovars as the challenge serovars.
  • the serovars of the present invention are obtainable from clinical isolates or from culture collections such as the American Tissue Culture Collection (ATCC).
  • ATCC American Tissue Culture Collection
  • In vivo efficacy models include but are not limited to: (i) A murine infection model using human Chlamydia pneumoniae serotypes; (ii) a murine disease model which is a murine model using a mouse-adapted Chlamydia pneumoniae strain, such as the Chlamydia pneumoniae mouse pneumonitis (MoPn) strain also known as Chlamydia muridarum; and (iii) a primate model using human Chlamydia pneumoniae isolates.
  • the MoPn strain is a mouse pathogen while human Chlamydia pneumoniae serotypes are human pathogens (see for example, Brunham et al (2000) J Infect Dis 181 (Suppl 3) S538-S543; Murdin et al (2000) J Infect Dis 181 (Suppl 3) S544-S551 and Read et al (2000) NAR 28(6); 1397-1406).
  • human Chlamydia pneumoniae serotypes can be used in mouse models although they normally require high inocula or pretreatment with progesterone.
  • Progesterone is generally used because it seems to render the epithelium more susceptible to chlamydial infection (see Pal et al 2003 Vaccine 21: 1455-1465).
  • MoPn which was originally isolated from mouse tissues, is thought to be a natural murine pathogen and thus offers an evolutionarily adapted pathogen for analysis of host-pathogen interactions.
  • the MoPn serovar is thought to have a high degree of DNA homology to the human Chlamydia serovars, it may also have some unique properties (see for example, Pal et al (2002) Infection and Immunity 70(9); 4812-4817.
  • mice 7 to 12 weeks of age receive 2.5 mg of depoprovera subcutaneously at 10 and 3 days before vaginal infection.
  • Post-vaccination mice are infected in the genital tract with 1,500 inclusion- forming units of Chlamydia pneumoniae contained in 5ml of sucrose-phosphate- glutamate buffer, pH 7.4.
  • the course of infection is monitored by determining the percentage of inclusion-bearing cells by indirect immunofluorescence with Chlamydia pneumoniae specific antisera, or by a Giemsa-stained smear from a scraping from the genital tract of an infected mouse.
  • the presence of antibody titers in the serum of a mouse is determined by an enzyme-linked immunosorbent assay.
  • the immunogenic compositions of the present invention can be administered using a number of different immunization routes such as but not limited to intra-muscularly (i.m.), intra- peritoneal (i.p.), intra-nasal (i.n.), sub-cutaneous (s.c) or transcutaneous (t.c) routes.
  • the challenge serovars may be administered by a number of different routes.
  • the challenge serovars are administered mucosally, such as but not limited to an intranasal (i.n) challenge.
  • guinea pig models For example, in vivo vaccine composition challenge studies in the guinea pig model of Chlamydia pneumoniae infection can be performed. A description of one example of this type of approach follows. Female guinea pigs weighing 450 - 500 g are housed in an environmentally controlled room with a 12 hour light-dark cycle and immunized with vaccine compositions via a variety of immunization routes. Post-vaccination, guinea pigs are infected in the genital tract with the agent of guinea pig inclusion conjunctivitis (GPIC), which has been grown in HeLa or McCoy cells (Rank et al. (1988)).
  • GPIC guinea pig inclusion conjunctivitis
  • Each animal receives approximately 1.4xl0 7 inclusion forming units (IFU) contained in 0.05 ml of sucrose-phosphate-glutamate buffer, pH 7.4 (Schacter, 1980).
  • IFU inclusion forming units
  • the course of infection monitored by determining the percentage of inclusion-bearing cells by indirect immunofluorescence with GPIC specific antisera, or by Giemsa- stained smear from a scraping from the genital tract (Rank et al 1988).
  • Antibody titers in the serum is determined by an enzyme-linked immunosorbent assay.
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or mucosally, such as by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (See e.g. W099/27961) or transcutaneous (See e.g. WO02/074244 and WO02/064162), intranasal (See e.g. WO03/028760), ocular, aural, pulmonary or other mucosal administration.
  • parenteral injection e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue
  • mucosally such as by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (See e.g. W099
  • Prophylaxis or therapy can be accomplished by a single direct administration at a single time point or multiple time points. Administration can also be delivered to a single or to multiple sites. Some routes of administration, such as mucosal administration via ophthalmic drops may require a higher dose. Those skilled in the art can adjust the dosage and concentration to suit the particular route of delivery.
  • 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.
  • HOMOLOGUES a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc.
  • SEQ IDs 1-86 in the compositions of the invention may be supplemented or substituted with molecules comprising sequences homologous (ie. sharing sequence identitv) to SEQ ID Nos 1-86.
  • Proteins (including protein antigens) as used in the invention may have homology and/or sequence identity with naturally occurring forms. Similarly coding sequences capable of expressing such proteins will generally have homology and/or sequence identity with naturally occurring sequences.
  • Techniques for determining nucleic acid and amino acid "sequence identity" also are known in the art. Typically, such techniques include determining the nucleotide sequence of the mRNA for a gene and/or determining the amino acid sequence encoded thereby, and comparing these sequences to a second nucleotide or amino acid sequence.
  • identity refers to an exact nucleotide-to-nucleotide or amino acid-to- amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more sequences (polynucleotide or amino acid) can be compared by determining their "percent identity.” The percent identity of two sequences, whether nucleic acid or amino acid sequences, is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100.
  • a preferred method of establishing percent identity in the context of the present invention is to use the MPSRCH package of programs copyrighted by the University of Edinburgh, developed by John F. Collins and Shane S.
  • homology can be determined by hybridization of polynucleotides under conditions which form stable duplexes between homologous regions, followed by digestion with single-stranded-specific nuclease (s), and size determination of the digested fragments.
  • Two DNA, or two polypeptide sequences are "substantially homologous" to each other when the sequences exhibit at least about 80%>-85%>, preferably at least about 90%,, and most preferably at least about 95%-98%> sequence identity over a defined length of the molecules, as determined using the methods above.
  • substantially homologous or homologous also refers to sequences showing complete identity to the specified DNA or polypeptide sequence.
  • DNA sequences that are substantially homologous or homologous can be identified in a Southern hybridization experiment under, for example, stringent conditions, as defined for that particular system.
  • stringent hybridization conditions can include 50%> formamide, 5x Denhardt's Solution, 5x SSC, 0.1% SDS and 100 pg/ml denatured salmon sperm DNA and the washing conditions can include 2x SSC, 0.1 %> SDS at 37 C followed by lx SSC, 0.1% SDS at 68 C.
  • the degree of identity is preferably greater than 50% (eg. 65%. 80%. 90%. or more) and include mutants and allelic variants.
  • SEQ IDs 1-86 in the compositions of the invention may be supplemented or substituted with nucleic acid which can hybridise to the Chlamydia nucleic acid, preferably underv"high stringency" conditionsv(c. 65 C in an 0.1 x SSC, 0.5%, SDS solution).
  • hypothetical protein refers to a protein which lacks a known cellular location or a known cellular function. Typically, a hypothetical protein lacks significant homologies with known well characterised proteins.
  • the invention also provides the compositions of the invention for use as medicaments (eg. as immunogenic compositions or vaccines) or as diagnostic reagents for detecting a Chylamydia infectioin in a host subject. It also provides the use of the compositions in the manufacture of: (i) a medicament for treating or preventing infection due to Chlamydia pneumoniae bacteria: (ii) a diagnostic reagent for detecting the presence of Chlamydia Pneumonaie bacteria or of antibodies raised against Chlamydia Pneumonaie bacteria; and/or (iii) a reagent which can raise antibodies against Chlamydia pneumonaie bacteria.
  • the invention also provides a method of treating a patient, comprising administering to the patient a therapeutically effective amount of a composition according to the invention.
  • the present invention provides compositions that are useful for preventing and/or treating T cell mediated immune disorders.
  • the composition is a pharmaceutical composition.
  • the composition is an immunotherapeutic composition.
  • the composition is a vaccine composition.
  • the composition may also comprise a carrier such as a pharmaceutically or immunologically acceptable carrier.
  • Pharmaceutically acceptable carriers or immunologically acceptable carriers are determined in part by the particular composition being administered as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions or vaccine compositions or immunotherapeutic compositions of the present invention.
  • 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.
  • the invention includes a method for the therapeutic or prophylactic treatment of Chlamydia pneumoniae infection in an animal susceptible to Chlamydial infection comprising administering to said animal a therapeutic or prophylactic amount of the immunogenic compositions of the invention.
  • the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or all six Chlamydia pneumoniae antigens of the first antigen group. Still more preferably, the combination consists of all six Chlamydia pneumoniae antigens of the first antigen group.
  • the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve Chlamydia pneumoniae antigens selected from the first antigen group and the second antigen group.
  • the combination is selected from the group consisting of three, four, or five Chlamydia pneumoniae antigens selected from the second antigen group.
  • the combination consists of five Chlamydia pneumoniae antigens selected from the second antigen group.
  • the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, or five Chlamydia pneumoniae antigens of the first antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group.
  • the combination consists of three, four or five Chlamydia pneumoniae antigens of the first antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group.
  • the immunigenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, five, six, seven, eight, nine, ten, eleven or twelve Chlamydia pneumoniae antigens of the first antigen group and the second antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group.
  • the combination is selected from the group consisting of three, four, or five Chlamydia pneumoniae antigens from the second antigen group and three, four or five Chlamydia pneumoniae from the third antigen group.
  • the combination consists of five Chlamydia pneumoniae antigens from the second antigen group and three, four or five Chlamydia pneumoniae antigens of the third antigen group.
  • the composition comprises molecules from different Chlamydia species.
  • the composition may comprise molecules from different serogroups and/or strains of the same Chlamydia species. Further embodiments comprise mixtures of one or more Chlamydia molecules from different strains.
  • Many proteins are relatively conserved between different species serogroups and strains of Chlamydia trachomatis and Chlamydia pneumoniae. To ensure maximum cross-strain recognition and reactivity, regions of proteins that are conserved between different Chlamydia species, serogroups and strains can be used in the compositions of the present invention. The invention therefore provides proteins which comprise stretches of amino acid sequence that are shared across the majority of Chlamydia strains.
  • the composition comprises a protein comprising a fragment of a Chlamydia pneumoniae protein (preferably a protein from SEQ ID Nos 1-86 or more preferably SEQ ID Nos 1-41 wherein said fragment consists of n consecutive conserved amino acids.
  • compositions of the invention may further comprise antigen derived from one or more sexually transmitted diseases in addition to Chlamydia trachomatis.
  • the antigen is derived from one or more of the following sexually transmitted diseases: N.gonorrhoeae ⁇ e.g. i, ii, iii, iv ⁇ ; human papiloma virus; Treponema pallidum; he ⁇ es simplex virus (HSV-1 or HSV-2); HIV (HIV-1 or HIV-2); and Haemophilus ducreyi.
  • a preferred composition comprises: (1) at least t of the Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group, where t is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13, preferably t is five; (2) one or more antigens from another sexually transmitted disease.
  • the sexually transmitted disease is selected from the group consisting of he ⁇ es simplex virus, preferably HSV-1 and/or HSV-2; human papillomavirus; N.gonorrhoeae; Treponema pallidum; and Haemophilus ducreyi.
  • compositions can thus provide protection against the following sexually-transmitted diseases: Chlamydia, genital he ⁇ es, genital warts, gonorrhoea, syphilis and chancroid (see Stephens et al (1998) Science 282: 754-759).
  • a saccharide or carbohydrate antigen is used, it is preferably conjugated to a carrier /protein i order to enhance immunogenicity (For example, Ramsay et al.
  • Preferred carrier proteins are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids.
  • the CRM ⁇ 9 diphtheria toxoid is particularly preferred (Research Disclosure, 453077 (Jan 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 (W093/17712, WO94/03208) pertussis proteins (W098/58668, EP-A-0471177) protein D from H.influenzae (WO00/56360) cytokines (WO91/01146), lymphokines, hormones, growth factors, toxin A or B from C.difficile (WO00/61761) iron-uptake proteins WO01/72337) etc.
  • a mixture comprises capsular saccharides from both serogroups A and C
  • 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.
  • a diphtheria antigen is included in the composition it is preferred also to include tetanus antigen and pertussis antigens.
  • a tetanus antigen is included it is preferred also to include diphtheria and pertussis antigens.
  • 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 ⁇ g/ml each. In general, the concentration of any given antigen will be sufficient to elicit an immune response against that antigen.
  • nucleic acid encoding the antigen may be used Robinson & Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al.
  • 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.
  • nucleic acid preferably DNA e.g. in the form of a plasmid
  • compositions of the present invention- may be used to prevent and/or treat disorders such as but not limited to: pneumonia, cardiovascular diseases, atherosclerosis, bronchitis, pharyngitis, laryngitis, sinusitis, obstructive lung diseases, asthma, chronic obstructive pulmonary disease, reactive arthritis, otitis media, abdominal aortic aneurysm, erythema nodosum, Reiter syndrome, sarcoidosis, Alzheimer's disease, multiple sclerosis, lymphogranuloma venereum, ocular trachoma, pelvic inflammatory disease, inclusion conjunctivitis, genital trachoma, infant pneumonitis, incipient trachoma, keratitis, papillary hypertrophy, corneal infiltration, vulvovaginitis, mucopurulent rhinitis, salpingitis, cervicitis, cervical follicles, prostatitis, proctitis, urethritis
  • compositions of the invention may be prepared in various forms.
  • 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.
  • kits may comprise one or more antigens in liquid form and one or more lyophilised antigens.
  • 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).
  • lipid aggregates such as oil droplets or liposomes.
  • the vaccines may also contain diluents, such as water, saline, glycerol, etc.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like, may be present.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like.
  • the biological molecules of the present invention be formulated into a pharmaceutical composition or an immunotherapeutic composition or a vaccine composition.
  • Such formulations comprise biological molecules combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline.
  • a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline.
  • Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative.
  • Formulations include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • the active ingredient is provided in dry (for eg, a powder or granules) form for reconstitution with a suitable vehicle (e. g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • a suitable vehicle e. g., sterile pyrogen-free water
  • the pharmaceutical compositions may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution.
  • This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein.
  • Such sterile injectable formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example.
  • a non-toxic parenterally-acceptable diluent or solvent such as water or 1,3-butane diol, for example.
  • Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono-or di-glycerides.
  • Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, in a liposomal preparation, or as a component of a biodegradable polymer systems.
  • Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • kits for enhancing a CMI response to the biological molecules of the present invention may comprise an antigenic composition or nucleotide sequence encoding same.
  • the kit may also include an adjuvant, preferably a genetic adjuvant is administered with or as part of the biological molecule and instructions for administering the biological molecule.
  • Other preferred components of the kit include an applicator for administering the biological molecule.
  • the term "applicator” refers to any device including but not limited to a hypodermic syringe, gene gun, particle acceleration device, nebulizer, dropper, bronchoscope, suppository, impregnated or coated vaginally-insertable material such as a tampon, douche preparation, solution for vaginal irrigation, retention enema preparation, suppository, or solution for rectal or colonic irrigation for applying the NOI either systemically or mucosally or transdermally to the host subject.
  • the invention also provides for a kit comprising comprising a combination of Chlamydia pneumoniae antigens.
  • the combination of Chlamydia pneumoniae antigens may be one or more of the immunogenic compositions of the invention.
  • the kit may further include a second component comprising one or more of the following: instructions, syringe or other delivery device, adjuvant, or pharmaceutically acceptable formulating solution.
  • the invention also provides a delivery device pre- filled with the immunogenic compositions of the invention.
  • FIG. 1A Assay of in vitro neutralization of C.pneumoniae infectivity for LLC-MK2 cells by polyclonal mouse antisera to recombinant Chlamydial proteins. Results are shown as reduction in the number of inclusions obtained when monolayers were infected with antiserum-treated infectious EBs, as compared to inclusion numbers given by untreated EBs. Percent reduction values are plotted against the reciprocal of the corresponding serum dilution. For each dilution inclusion counts were corrected for background inhibition of infectivity observed with the corresponding dilution of the pre- immune serum.
  • FIG. 1 shows results obtained with serial dilutions of antibodies raised against a 'neutralizing' antigen (v), a 'non-neutralizing' FACS- positive antigen (v), and against the GST polypeptide, used in the fusion constructs, alone ( ⁇ ).
  • Figure IB shows serum litres giving 50% neutralization of infectivity for the 10 C.pneumoniae recombinant antigens described in the text. Each titer was assessed in 3 separate experiments (SEM values shown).
  • FIG. 2 shows immunoblot analysis of two dimensional electrophoretic maps of C.pneumoniae EBs using the imune sera described in the text. Immunoblots were obtained from either of two EB gels (panels A and B at the top) covering different pH intervals, according to which of the two allowed the best detection of a given antigen.
  • the arrows in the HtrA immunoblot show which of the signals had a corresponding stained spot in the gel (arrows in panel A) which was subjected to MALDI-TOF identification.
  • the two patterns in the HtrA blot are both suggestive of typical electrophoretic 'trains' composed of single charge variants of the same protein.
  • Figure 4 shows flow cytometric analysis of splenocytes from DNA-immunized HLA- A2 transgenic and non transgenic mice.
  • Groups of 4 mice were immunized 3 times i.m. with 50 ⁇ g of plasmid DNA expressing C. pneumoniae Low Calcium Response Protein H.
  • IFN- ⁇ production from splenocytes was monitored following either a 6h (ex-vivo) or a 6 day (restimulated) pulse with peptide CH-6 (lO ⁇ g/ml).
  • Equal numbers of gated live lymphocyte cells were acquired with a LSRII FACS System (Becton Dickinson) and percentages of IFN- ⁇ producing CD8 + T cells were calculated using DIVA Software (Becton Dickinson).
  • Figure 5 shows a flow cytometric analysis of splenocytes from transgenic and non transgenic mice infected with C. pneumoniae EBs.
  • HLA-A2 transgenic mice were intranasally infected twice with 5x10 5 C. pneumoniae FB/96 EBs and splenocytes were stimulated for 6 days in the presence of relevant peptides before determining IFN- ⁇ production by CD8 + T cells as described in the legend of Figure 4.
  • B HLA-A2 transgenic and non transgenic mice were infected together with the same EBs preparation and CD8 + T cells were subjected to FACS analysis as reported in (A). Table I shows a summary of data and properties of the C.pneumoniae antigens described in the text.
  • the neutralization titer is reported is as the reciprocal of the antiserum dilution causing a 50% reduction in the number of inclusions in the in vitro infectivity assay.
  • Table 2 shows results from hamster mouse model studies for hypothetical proteins.
  • Table 3 shows expressed genes of CPn EB selected by microarray.
  • Table 4 shows C. pneumoniae selected peptides: protein sources and HLA-A2 stabilization assay.
  • Table 5 shows ELISPOT assay with CD8+ T cells from DNA immunised HLA-A2 transgenic mice.
  • Table 6 shows IFN- ⁇ production from splenocytes of DNA immunized HLA-A2 transgenic and non transgenic mice.
  • METHODS AND MATERIALS (Examples 1-4) (see Reference Section 1)
  • C.pneumoniae FB/96 a clinical isolate obtained from a patient with pneumonia at the Sant'Orsola Polyclinic, Bologna, Italy, was grown in LLC-MK2 cells seeded in individual wells of a six- well plastic plate (7). Cells were harvested 72 hr after infection with a sterile rubber, sonically disrupted and the elementary bodies (EB) purified by gradient centrifugation as described (26). Purified Chlamydiae were resuspended in sucrose-phosphate-glutamic acid (SPG) transport buffer, and stored in 0.5 ml aliquots, at -80°C until used. When required, prior to storage, EB infectivity was heat-inactivated by 3 hour incubation at 56°C.
  • SPG sucrose-phosphate-glutamic acid
  • ORFs Open reading frames (ORFs), selected from the C. pneumoniae CWL029 genome sequence (16), were PCR-cloned into plasmid expression vectors and purified from E.coli cultures, essentially as previously described (25). Recombinant Chlamydial proteins were obtained as GST fusion proteins by using pGEX-KG derived vectors (12) in E. coli BL21 (Novagen). PCR primers were designed so as to amplify genes without the N-terminal signal peptide coding sequence. When a signal peptide or processing site was not clearly predictable, the ORF sequence was cloned as annotated by Kalman and coworkers (16).
  • Cells were collected by centrifugation ' 3 hr after induction and broken in a French Press (SLM Aminco, Rochester, NY). After centrifugation at 30.000 g, the supernatants were loaded onto Glutathione Sepharose 4B columns (Amersham Pharmacia Biotech) and column bound proteins were eluted with 50 mM Tris-HCl, 10 mM reduced glutathione, pH 8.0. Protein concentrations in the samples were determined using the Bradford method.
  • mice Groups of four 5/6-week old CD1 female mice (Charles River, Como, Italy) were immunized intraperitoneally at day 1 with 20ug of protein in Complete Freund's adjuvant (CFA) and boosted at day 15 and 28 with 20ug of recombinant protein in Incomplete Freund's adjuvant (IF A).
  • CFA Complete Freund's adjuvant
  • IF A Incomplete Freund's adjuvant
  • Pre-immune and immune sera were prepared from blood samples collected on days 0, 27 and 42. In order to reduce the amount of antibodies possibly elicited by contaminating E. coli antigens, the immune sera were incubated overnight at 4°C with nitrocellulose strips adsorbed with a total protein extract from E . coli BL21.
  • Protein samples (200 or 20 ⁇ g of protein for Coomassie Blue stained reference gels, or gels to be processed for immunoblotting, respectively) were adsorbed overnight on Immobiline DryStrips (7 cm, pH 3-10 NL, or pH 4-7). Electrofocusing was performed in an IPGphor Isoelectric Focusing Unit (Amersham Biosciences, Uppsala, Sweden). The focused strips were equilibrated as described (15) and loaded on linear 9-16.5 % acrylamide gradients (7x 4 cm, 1.5 mm thick), for SDS-PAGE separation in a Mini Protean III Cell (Bio-Rad, Hercules, CA). Gels were stained with colloidal Coomassie Blue (Novex, San Diego, CA) (4) and the protein maps so obtained were scanned with a Personal Densitometer SI (Molecular Dynamics) at 12 bits and 50 mm per pixel.
  • the proteins separated in the 2DE maps were transferred onto nitrocellulose membranes, overnight at 30 Volts, using a Protean III apparatus (BioRad, Hercules, CA). Membranes were stained with a 0.05% (w/v) CPTS (Copper(II) phthalocyanine-3,4',4",4'"-tetrasulfonic acid tetrasodium salt) in 12 mM HCl, and marked peripherally with 8 India-ink dots to provide anchors for subsequent image superimposition and matching.
  • CPTS Copper(II) phthalocyanine-3,4',4",4'"-tetrasulfonic acid tetrasodium salt
  • the membranes were destained with 0,5 M NaHC03, incubated with the mouse sera to be analyzed (either pre-immune or specific immune sera, diluted 1:1000), and then with a peroxidase-conjugated anti-mouse antibody (Amersham Biosciences, Uppsala, Sweden). After washing with PBS, 0.1 %> Tween-20, blots were developed using the Opti-4CN Substrate Kit (Biorad, Hercules, CA), and the images of the immunostained blots again acquired as above. Images were analysed with the computer program Image Master 2D Elite, version 4.01 (Amersham Biosciences, Uppsala, Sweden).
  • Tryptic peptides were desalted and concentrated using Zip-Tip (Millipore, Bedford, MA). Peptides were directly eluted and loaded onto a SCOUT 384 Anchor Chip multiprobe plate (400 ⁇ m, Bruker Daltonics, Bremen, Germany) with a solution of 2-5 dihydroxybenzoic acid (5g/l), in 50% acetonitrile, 0.1% trifluoroacetic acid. Spectra were acquired on a Bruker Biflex III matrix-assisted laser deso ⁇ tion ionization-time of flight (MALDI- TOF) apparatus. Resulting values for monoisotopic peaks were used for database searches using the Mascot software (32), as available at the website http://www.matrixscience.com/.
  • Antibody-EB interaction was allowed to proceed for 30 min at 37°C on a slowly rocking platform.
  • the lOOul of reaction mix of each sample was used to inoculate PBS-washed LLC-MK2 confluent monolayers (in triplicate for each serum dilution), in a 24-well tissue culture plate, and centrifuged at 805 x g for 1 hour at 37°C. After centrifugation Eagle's minimal essential medium containing Earle's salts, 20% fetal bovine serum and lug/ml cycloheximide was added. Infected cultures were incubated at 37°C in 5%>C0 2 for 72 hours.
  • the monolayers were fixed with methanol and the Chlamydial inclusions were detected by staining with mouse a ti-Chlamydia fluorescein-conjugated monoclonal antibody (Merifluor Chlamydia, Meridian Diagnostics, Inc.) and quantified by counting 10 fields per well at a magnification of 40X.
  • the inhibition of infectivity due to EBs interaction with the immune sera was calculated as percentage reduction in mean IFU number as compared to the SPG (buffer only)/EBs control. In this calculation the IFU counts obtained with immune sera were corrected for background inhibition of infection due to the corresponding pre-immune mouse serum.
  • the sera were considered as "neutralizing” if they could cause a 50% > or greater reduction in infectivity.
  • the corresponding neutralizing titer was defined as the serum dilution at which a 50% reduction of infectivity was observed.
  • Experimental variability was evaluated by calculating the standard error of measurement (SEM), from three titration experiments for each recombinant antigen, as shown in Fig. IB.
  • the clarified homogenates (0.2 ml) were inoculated in duplicate onto LLC-MK2 cells seeded in plastic individual well of a 24 well plate, incubated at 37°C for 72 h and fixed in acetone before detection and counting of numbers of Chlamydial inclusions per well by immunofluorescence microscopy.
  • the protocol was approved by the ethical committee of the University of Bologna.
  • pmp 10 and pmp2 encoding two members of the heterogeneous Chlamydial PMP family of polymo ⁇ hic membrane proteins; • artl, encoding a putative extracellular solute (possibly Arginine) binding protein of an aminoacid transport system;
  • htrA encoding a putative chaperone with heat-shock inducible protease activity
  • Cpn0301 "hypothetical" gene encoding a protein homologous to the ompH family of bacterial proteins, some members of which have been shown to be chaperones involved in outer membrane biosynthesis
  • a limit of the hamster model is that, because of the absence of immunological reagents, the relative contribution of humoral and cell-mediated immunity cannot be assessed.
  • Example 4 Two 'hypothetical'proteins 6784 and 6814 (encoded by the ORFs Cpn0498 and Cpn0525) yielded FACS-positive sera which, however, were not able to neutralize host cell infection in vitro. However, these antigens performed remarkably well in the hamster-spleen test. Table 2
  • cytoplasmic inclusions which can be stained with Chlamydia specific fluorescence-labeled monoclonal antibodies and counted with an UV light microscope.
  • cycloheximide which inhibits host cell protein synthesis and favours Chlamydial intracellular growth with the consequent formation of typical cytoplasmic inclusions which can be stained with Chlamydia specific fluorescence-labeled monoclonal antibodies and counted with an UV light microscope.
  • an anti-serum is labelled as 'neutralizing' when the reduction of infectivity is equal or greater than 50%>, and the serum dilution yielding a 50% reduction in infectivity is referred to as the 50% end- point neutralization titer.
  • hamster model Using a recently described in vivo model of systemic infection (hamster model), hamsters immunised with 6 of the in vitro neutralising antigens, when challenged with CPn EBs, showed a greater than 80%> reduction of spleen infection as compared with non-immunised controls.
  • proteins identified by the present work can be divided in 3 groups: • proteins which have an annotation compatible with (could be reasonably expected to have) an expected/predicted exposure on the Chlamydial cell surface and with the possibility that antibodies binding to them may actually interfere with host cell attachment and entry (ie proteins which could possibly induce neutralising antibodies)
  • the first group includes the 2 polymo ⁇ hic outer membrane proteins (Pmp's) Pmp2 and PmplO (10, 11, 14, 30), the outer membrane protein OmpH-like, and OmcA, which is annotated (Chlamydia Genome Project at htkp-J/Chlamydia- www.berkeley.edu:4231/) as "predicted 9-kD cysteine-rich, outer membrane protein, lipoprotein".
  • the Pmp family of Chlamydia-specific proteins is generally thought to comprise probable pathogenicity factors, with an autonomous secretion capacity (autotransporters), important for adhesion to host cells and are generally considered as promising vaccine candidates.
  • autotransporters important for adhesion to host cells and are generally considered as promising vaccine candidates.
  • this apart from very recent unpublished results on Pmp21, this is the first time that antisera to recombinant Pmp's are reported to have neutralizing properties.
  • OmcA 2 polymo ⁇ hic outer membrane proteins
  • OmcA is the product of a gene co-transcribed in the same operon with the 60 kDa OmcB cystein-rich protein which is a major structural component of the Chlamydial outer membrane and a major immunogen in human C. trachomatis infections.
  • OmcB and OmcA are likely to interact in some as yet unknown outer membrane structure, so it is possible that antibodies to OmcA can interfere with EB infectivity.
  • Chlamydial OmpH is probably a member of the OmpH (Skp) family of proteins which have been reported to have chaperonin activities in other bacteria very important for the correct biosynthesis of the outer membrane. These proteins appear to cooperate in this task with HtrA (see below). In fact, in E.coli single KO mutants of either OmpH (Skp) or HtrA (DegP) are still viable, but double mutants do not grow
  • Second Group of Selected Proteins (ArtJ, AtoS, HtrA and Enolase)
  • the second group which represents a somehow smprising finding, includes ArtJ, AtoS, HtrA and Enolase. If the current annotation (justified by analogy with homologous genes in other bacteria) is correct, all these proteins would be expected to have a periplasmic location in gram-negative bacteria, and to be surface-exposed only in a gram-positive bacterium. It is possible that owing to their atypical life cycle, requiring an efficient passage from a dormant spore-like status (the EB) to an active form needing to adapt quickly to host-cell responses to invasion, Chlamydiae in fact display some sensors directly on the outer surface of their infectious form.
  • EB dormant spore-like status
  • HtrA (DegP), which in other bacteria has a complex hexameric structure, has been described as having multiple functions (3, 5, 18, 19, 27, 38) : a chaperonin assisting a correct outer membrane biogenesis, inducible protease for the elimination of misfolded membrane proteins, and also a sensor of 'stress' conditions.
  • a chaperonin assisting a correct outer membrane biogenesis
  • inducible protease for the elimination of misfolded membrane proteins
  • a sensor of 'stress' conditions In Chlamydia none of these properties has been demonstrated yet, however we find that in purified EB HtrA is present in two forms one of which appears to be processed by being deprived of the N-terminal fragment. This fragment, if aligned with the homologous HtrA sequence from Thermologa maritima (18), would comprise a predicted loop acting as a structural lid controlling the access to the protease active.
  • HtrA could have a similar protease activity and the two forms identified on the 2-D map represent the active and inactive species.
  • C. trachomatis HtrA ortholog is recognized by human sera from patients who had a Chlamydial genital infection (35), and a similarly HtrA is one of the antigens in the immunoproteome of Helicobacter pylori (13).
  • the homologue protein in Haemophilus influenzae is a protective antigen in both a passive infant rat model of bacteremia and the active chinchilla model of otitis media (23) .
  • Cpn enolase Also in the second group of proteins expected to be located elsewhere than the cell surface, is Cpn enolase. This protein aligns with the well known family of conserved glycosylases, which are essentially cytoplasmic enzymes, but in Streptococci enolase has been shown to have also a cell surface location, and extracellular matrix binding properties (1, 28, 29)). Interestingly, Gaston and colleagues (8) also showed that in patients with reactive arthritis induced by C. trachomatis, enolase induces specific CD4 + T-cell responses.
  • the third of the 3 groups in which we propose to divide, just for the sake of discussion, the 10 neutralizing antigens above described, comprises two proteins which are still annotated in public Chlamydial databases as the hypothetical products of two CPn-specific genes: Cpn0759 and Cpn0042.
  • the Cpn0759 gene is the second gene in a cluster of 6 Cpn-specific hypothetical genes (from Cpn0794 to Cpn0799) immediately upstream of the enolase gene. With the exception of Cpn0759 the products of all the other genes in the cluster share similarities of 30 to 40% over long stretches of amino acids.
  • the Cpn0042 gene encodes a hypothetical protein, with 4 coiled-coil regions, which has been described as a member of a new family of hypervariable outer membrane proteins (33).
  • the hypervariability of these proteins could be due to a strand-slippage mechanism induced by the presence of a poly(C) stretch within the coding region of the corresponding genes, a mechanism already described in the Pmp's family for the pmpl 0 gene (30).
  • the function of these proteins is still unknown, and our observations provide the first experimental indication of a possible function related to the Chlamydial infection process.
  • Cpn0795 SEQ ID NO: 6
  • a Cpn specific hypothetical protein is a FACS positive protein which demonstrates significant immunoprotective activity in a hamster spleen model of Chlamydia pneumoniae infection.
  • Fig. 6 shows an alignment of the proteins in the 7105-7110 protein family. This
  • Alignment shows a new family of proteins expected to constitute a system of antigens probably delivered on the Cpn surface or secreted by a type V (autotransporter) secretion mechanism. This alignment was generated as follows:
  • 7107 can be predicted to fold in a beta-barrel structure which can form a translocation pore for secretion across the outer membrane.
  • Cpn0795 and Cpn0796 have C terminal ends that may form transmembrane pores (see alignment, FIG. 9).
  • CPn0794, Cpn0797, Cpn0798, and Cpn0799 have N-terminal ends indicating that all proteins have N-terminal and C-terminal ends.
  • Fig. 7 shows alignment of Cpn0794 - Cpn 0799. Proteins encoded by the genes Cpn0794, Cpn0795, Cpn0796, and Cpn0797 have been identified as likely to be exposed on the surface of the chlamydia cell and as possible vaccine candidates. These proteins are shown to be actually expressed by Cpn in vivo (WB data and FACS data). In the case of Cpn0797 we also showed that the level of expression in CPn EBs is high enough to be detected by mass spectrometry analysis on 2DE maps of protein extracts (see Montigiani et al.)
  • Cpn0794, Cpn0796 and Cpn0797 proteins can be aligned according to a set of imperfect repeats present within their aminoacid sequences (see FIG. 7) , whereas the putative product of CPn0795 can be mostly aligned to the C-terminal portion of the Cpn0796 protein.
  • proteins encoded by genes Cpn0798 or Cpn0799 can alse be aligned to the above proteins according to the above mentioned repeated sequence motifs (see FIG. 7). Overall alignment of the 6 genes demonstrates that the genes encode for a family of functionally-related proteins.
  • FIG. 8 illustrates Cpn0796.
  • Cpn0796 forms a beta-barrel structure and is capable of forming a pore across the bacterial outer membrane (OM).
  • OM bacterial outer membrane
  • the molecule may form a pore in the OM through which the N-terminal domain may pass (the 'passenger' domain) to the outside of the bacterial cell.
  • these molecules may either remain anchored to the bacterial surface or undergo a proteolytic cut which releases the 'passenger domain' or a portion of it into the medium surrounding the bacterial cell an example of which is represented in the following sequence:
  • amino acid residues 365-385 represent an alpha helix conformation that spans the beta barrel pore
  • the N-terminal passenger domain may be cleaved via a specific proteolytic action from the membrane-anchored pore structure.
  • a linker domain comprising the peptide sequence PSPAPV (SEQ ID NO: 84) as shown in bold in the following sequence illustrates a site at which cleavage of the N-terminal passenger domain may occur:
  • the N-terminal peptide may be secreted to be exposed on the bacterial cell surface and can also become detached via the proteolytic event described above.
  • the peptide may form a structural conformation known as beta-propellers indicated in the following sequence:
  • N-terminal passenger domain can also possess a specific protease activity, such as a serine protease-like activity.
  • the protease activity may act on the membrane anchored form of the molecule such that the N-terminal passenger domain is cleaved off form the surface of the chlamydial cell.
  • the serine protease like activity is supported by the presence of a consensus serine protease triad of adequately spaced amino acid residues (namely H, D and S) which can be located on the virtual structure of the 'passenger' domain modelled on a set of experimentally-determined templates, e.g. InrO (PDB identification code)
  • the gene Cpn0796 gene encodes for a protein which promotes its own secretion on the EB surface and may also mediate or promote its own release into the surrounding medium.
  • the secreted passenger peptide has several activities, including: 1. actin binding peptide, part of a chlamydial surface layer, and instrumental to the process of establishing the host cell infection 2.
  • specific protease activity within the host cell cytoplasm instrumental to the intracellular survival of infecting chlamydiae.
  • beta propeller domain Another function of the above N-terminal beta propeller domain is the regulation modulation of the activity of a cytosolic protease of the host cell in order to alter host cell properties in favour of chlamydial development, survival or persistence. See Fulop V, Bocskei Z, Polgar L. in "Prolyl oligopeptidase: an unusual beta-propeller domain regulates proteolysis.” Cell. 1998 Jul 24;94(2): 161-70.
  • the proteins encoded by Cpn0794, Cpn0797, Cpn0798, Cpn0799 - all comprising variants of the above described Cpn0796 structure - also provide beta propeller structures with activities similar and/or complementary to the ones described above.
  • a family of proteins cooperating to a common function either by generating - through events of site specific recombination - new molecules with structures and activities similar to the above described Cpn0796 product, OR by independently contributing to a multi-protein structure requiring a coordinated action of several related components.
  • the main stages in the Chlamydial life cycle are: (i) the binding to the host cell surface and entry into the cytoplasm through a specialised vacuole (the Chlamydial inclusion) by an extracellular sporelike infective form, called the elementary body (EB); and (ii) the conversion of the EB to a non-infective replicative form called a reticulate body (RB) that replicates by binary fission a number of times within the inclusion to form a microcolony.
  • EB extracellular sporelike infective form
  • Protein microarrays are used for high throughput protein analysis by detecting proteins and monitoring their expression levels. Through use of protein microarrays, complex screening of thousands of proteins and interactions with proteins may be performed in parallel.
  • a protein array typically includes a surface, such as glass, membrane, microtiter wells, mass spectrometer plates, beads or other particles, for binding ligands, proteins, or antibodies. For example, antibodies may be bound to the microarray to form a capture array. The capture array may be contacted with a biological sample to quantify the proteins in the biological sample.
  • proteins may be bound to the microarray and contacted with a biological sample to quantify protein-protein or protein-ligand interactions.
  • protein microarrays may also be used in diagnostics in which multiple immunoassays may be conducted in parallel such that levels of proteins in different samples may be quantified and compared for applications in the treatment or diagnosis of disease.
  • a capture array antibodies are bound to the microarray and exposed to a biological sample. Proteins and ligands that bind to the antibody array may be detected by direct labelling of the bound proteins. If a higher sensitivity or specificity is desired, a sandwich technique may be employed in which pairs of antibodies are directed to the same protein ligand. This technique is particularly useful if the amount of protein to be detected is low or if there are modifications to the protein. In addition, the use of sandwich assays minimizes the risk of cross-reactivity in highly multiplexed assays by providing dual level target recognition, i.e. two levels of specificity for each locus in the array. Alternatively, the bound proteins may be detected via label-free detection methods such as including mass spectrometry, surface plasmon resonance and atomic force microscopy. This technique is useful if modification or alteration of the protein is to be avoided.
  • label-free detection methods such as including mass spectrometry, surface plasmon resonance and atomic force microscopy. This technique is useful if modification or alteration of
  • Large-scale functional chips containing large numbers of immobilized purified proteins may be used to assay a wide range of biochemical functions, such as protein interactions with other proteins, drug-target interactions, enzyme-substrates, etc.
  • proteins may be purified from an expression library, for example, and the protein array can be used to screen libraries to select specific binding partners, including antibodies, synthetic scaffolds, peptides and aptamers. In this way, 'library against library' screening can be carried out, such as screening of drug candidates in combinatorial chemical libraries against an array of protein targets identified from genome projects.
  • Protein microarray technology permits analysis of the proteins themselves rather than inferring protein function, interactions and characteristics through mRNA expression. In many cases, mRNA expression does not correlate accurately with protein abundance. Furthermore, mRNA expression analysis does not provide sufficient information on protein-protein interaction or post-translational modifications. Thus, direct analysis of proteins via protein microarrays provides an advantage by providing more accurate information of proteins and protein-protein interactions that may not be readily available through measurment of mRNA expression.
  • DNA microarray techniques permit profiling of gene expression at the mRNA level as a function of the cellular state. This can lead to the identification of genes or clusters of genes whose up- or down-regulation is associated to a particular state of the cell and to the identification of therapeutically relevant targets.
  • DNA fragments representing specific portions of all genes belonging to a given organism are chemically bound to the surfaces of solid supports (chips) at high densities and in an ordered manner.
  • chips solid supports
  • RNA is prepared, labelled with fluorescent dyes and finally hybridised to the DNA fragments fixed to the surface of the chip.
  • fluorescence signals emitted by each spot upon excitation with a laser beam is carefully quantified to define the transcription activity of all the arrayed genes.
  • CPn DNA microarrays have been developed to look at the transcriptional events which occur when a given CPn pathogen gets into contact with the host cells, both in in vivo and in vitro settings.
  • DNA chips carrying the entire genome of a particular bacterium, such as the CPn bacterium can be prepared in a very short period of time so that whole genome expression analysis can be determined.
  • a genomic DNA (open reading frame probes) microarray approach for gene expression in CPn bacteria was adopted.
  • an array was prepared for the analysis of the CPn life cycle on the basis of the published annotation of the complete genome.
  • the Chlamydia DNA chips carry about 1000 PCR-derived DNA fragments, which have an average size of 400-700bp and correspond to internal portions of all CPn annotated genes.
  • Table 3(i)-(xi) shows transcriptional activity for expressed genes for CPn EB selected by microarray.
  • the data in Tables 3(i)-(iv) shows different rnRNAs in order of abundance from cells in their infectious "spore-like" (EB) form.
  • Data in Tables 3(v)- (xi) correlates and summarizes mRNA expression levels of genes for CPn. The cells were used at the end of their cycle where gene expression is likely to be at its highest. As values less than approximately 10000 is likely to be background, the top set of proteins (approx top 30) with more intense signals are likely to be the most interesting proteins.
  • Table 3(v)-(xi) shows the transcriptional activity for expressed genes for CPn EB selected by microarray.
  • the Table shows different mRNA in order of abundance from cells in their infectious "spore-like" (EB) form. The cells were used at the end of the cycle where gene expression is likely to be at its highest.
  • Chlamdydia late gene products have been described more frequently than early gene products. This is primarily because of the presence of late gene products in EBs but not RBs and that it is easier to study EBs rather than RBs.
  • Late gene functions appear to be predominantly those associated with the terminal differentiation of RBs back to EBs (Shaw et al., Mol Microbiology 37(4), 2000, 913-925). Late gene products appear to function in the termination of bacterial cell division and constitute structural components and remodelling activities involved in the formation of the cross-linked outer membrane complex that functions in the attachment and invasion of new host cells.
  • an important aspect of the secondary differentiation process (RB to infectious EB) is the expression of genes that encode proteins that form the highly disulfide cross-linked bacterial outer membrane (OM) complex.
  • Cpn 0384 whose CT equivalent is CT046 (hctB) has been shown to be associated with differentiation from RB to EB (see Belland et al, PNAS (USA) 100(14), 2003, 8478-83). We also found Cpn0384 to have relatively high levels of transcriptional activity (again see top of Table 3(v)-(xi)). Other Cpn antigens thought to be involved in the Type III secretion system were found to have moderate expression levels in terms of transcriptional activity.
  • Chlamydial Pmps The function of Chlamydial Pmps remains unknown, although based on sequence prediction and experimental testing, these Pmps are regarded as surface proteins and thus, likely to be critical for Chlamydial virulence. Like the Inclusion (Inc) Membrane proteins, the Pmp proteins are regarded, at present, as unique to the Chlamydiae family (see Rockey et al (2000) Infection and Immunity 69(10) 5473-5479). The findings disclosed here and by others, such as Grimwood et al, demonstrates that the Chlamydia organism appears to expend a considerable metabolic cost in Pmp transcription, such as Pmp 19 transcription, despite the potential lack of production of a functional Pmp proteins, such as the Pmp 19 protein.
  • T cell epitope prediction was carried out on the genomic sequence of C. pneumoniae CWL029 strain (Accession numbers NC 000922 or AE001363. using the BIMAS algorithm [24]. Synthetic peptides (purity > 80%o) were synthesized by Primm Sri (Milan, Italy), suspended in 100% DMSO and kept at -20° C before use.
  • the T cell lymphoma murine cell line RMA-S stably transfected with HLA-A2 was kindly provided by Dr. Barnaba, Universita degli Studi “La Sapienza", Rome, Italy, and cultured at 37° C in RPMI-1640 (GIBCO) supplemented with heat inactivated 10%) FCS, 100 RJ/ml penicillin/streptomycin, 2 mM Lglutamine (GIBCO) and 5x10-5 M 2-ME (Sigma).
  • H2-b HLA-A2 transgenic mice [35] were housed in a pathogen-free environment and screened for HLA-A2 expression by FCM carried out on total blood samples using the BB7.2 anti-A2 mAb [48]. Only mice with percentages of A2 expressing cells higher than 70-80 %> were used for DNA immunization and C. pneumoniae infection experiments. Animals which showed no HLA-A2 expression were mated in order to obtain an HLA-A2 non transgenic population, to be used as a control in the experiments. Epitope stabilization assay
  • RMA-S/A2 cells (3-5 x 10 5 /well) were seeded in serum-free RPMI medium, supplemented with human ⁇ 2 microglobulin (3 ⁇ g/ml, Sigma), without or with the test peptide (lO ⁇ M). Following overnight incubation at 26°C in humidified 5% C0 2 atmosphere, cells were shifted to 37° C for 2 h before determining the HLA-A2 expression level at the cell surface using the BB7.2 anti-A2 mAb and a PE-conjugated anti-mouse IgG (Jackson ImmunoResearch). Fluorescence intensity on living cells, which did not inco ⁇ orate propidium iodide, was analyzed by FCM. As controls, corresponding samples without peptide and samples with peptide but treated only with the anti-mouse secondary antibody, were used.
  • mice were intranasally infected twice with a month interval, using 5xl0 5
  • C. pneumoniae FB/96 EBs [4] diluted in 50 ⁇ l of PBS.
  • C. pneumoniae antigen coding genes were amplified by PCR using FB/96 genomic DNA, cloned into plasmid pcmvKaSF2120 [49] and verified by DNA sequence analysis.
  • Splenocytes from DNA immunized mice were prepared one week after the third immunization using Cell Strainer (Falcon) filters. Following red blood cells lysis, CD8 + T cells from spleen cells suspensions were enriched by positive selection using magnetic activated cell sorting (MACS-Miltenyi Biotec) with CD8a (Ly-2) microbeads. CD8 + T cells purity was higher than 90%>, as determined by FMC. Multiscreen 96-well nitrocellulose plates (Millipore) were coated with 5 ⁇ g/ml of the anti-mouse IFN- ⁇ antibody (R4-6A2, PharMingen) in 100 ⁇ l of carbonate buffer, pH 9.2.
  • CD8 + 5xl0 4
  • spleen cells from non immunized HLA-A2 transgenic mice as a source of antigen-presenting cells (2xl0 5 /well), 10 ⁇ g/ml of peptide and lOU/ml of human r-IL-2 (Chiron Co ⁇ oration).
  • Splenocytes from infected mice were isolated one week after the second infection with C. pneumoniae Ebs.
  • 2xl0 6 splenocytes were seeded in the presence of the test peptide (lO ⁇ g/ml) and anti-mouse CD28 antibody (1 ⁇ g/ml, PharMingen) as co-stimulus.
  • the test peptide lO ⁇ g/ml
  • anti-mouse CD28 antibody (1 ⁇ g/ml, PharMingen
  • Brefeldin A 10 ⁇ g/ml, Sigma
  • mice For analysis of IFN- ⁇ production by short term T cell lines, 5-10xl0 6 splenocytes from infected mice were cultured for 6 days in the presence of the test peptide (20 ⁇ g/ml), with rIL-2 (10 ⁇ g/ml) being added after the first two days. At the end of the incubation period, cells were washed twice in RPMI, pulsed again for 6 h in the presence of the test peptide (lO ⁇ g/ml), lxlO 5 freshly prepared CD8 depleted antigen presenting cells from HLA-A2 transgenic mice (irradiated at 3000 rad) and anti-mouse CD28 antibody (1 ⁇ g/ml, PharMingen) as co-stimulus. After a two h incubation at 37° C, 5 % C0 2 , Brefeldin A (10 ⁇ g/ml, Sigma) was added, the incubation was extended for 4 additional hours and IFN- ⁇ production was analyzed by FCM.
  • Chlamydial proteins have been reported to induce autoimmune responses [25-28]
  • the predicted binding score of 157.22, obtained for the well characterized HIV-1 pl7 gag epitope 77 SLYNTVATL 85 [29] was taken as an arbitrary cut-off for peptide selection.
  • the capacity of the selected peptides to bind to HLA-A2 was assessed using an in vitro MHC class I stabilization assay, carried out with the murine transporter associated with antigen processing (TAP)-deficient cell line RMA-S/A2, stably transfected with the human class I A2 gene.
  • MHC class I molecules cultured at 37° C, are unstably expressed on the cell surface of TAP-deficient cells [30-32]. Culturing the cells at 37° C in the presence of binding peptides, results in formation of a more stable MHC/peptide complex which can be monitored by flow cytometric analysis.
  • RMA-S/A2 cells were therefore cultured overnight at 26° C in the presence of the test peptides, shifted to 37° C for 2 hours and the surface level of stabilized A2 molecules was quantified by direct staining with an anti-HLA-A2 specific mAb.
  • Two known HLA-A2 restricted CTL epitopes were used as positive controls for binding to A2, the HIN-1 pl7 gag peptide [29] and the influenza matrix Ml protein peptide FluMP58 [33], while the Hepatitis B virus envelope antigen peptide HbenvAgl25 (HepB) was used as a negative control [34].
  • the binding results obtained are shown in Table 4 and allowed the identification of 15 peptides with a net mean fluorescence intensity (Net MFI) higher than 92.3, corresponding to the value obtained with the HIV-1 pl7 gag positive control peptide, 8 peptides with a Net MFI intermediate between the values 92.3 and 63.1, obtained with the two positive control peptides, and 12 peptides with an Net MFI ranging between 29.6 and 63.
  • Net MFI net mean fluorescence intensity
  • HLA-A2 binders are recognized by CD8 + T cells from DNA-immunized transgenic mice
  • the in vitro assay with RMA-S/A2 cells allowed the definition of a set of peptides which were able to bind and stabilize the HLA-A2 molecules on the cell surface.
  • peptide recognition by CD8 + T cells was studied under conditions in which the related complete antigen was intracellularly expressed and presented in vivo.
  • the ORF sequences were selected among those containing either one or more epitopes positive in the in vitro assay or a combination of positive and negative epitopes.
  • the ORF sequence corresponding to the outer membrane protein A (OMPA, CPn 0695) was included in this analysis, since human MHC-I-restricted epitopes have already been reported for this protein in C. trachomatis [18;36].
  • One coding sequence, related to gene CPn 0131 was chosen, which included four epitopes, all negative in the in vitro stabilization assay.
  • transgenic mice were sacrificed, spleen CD8 + T cells were isolated, stimulated for 20 hour with the corresponding peptide and ex vivo IFN- ⁇ production was assessed using an enzyme- linked immunospot (ELISpot) assay,
  • Example 8 To test the capacity of peptides to amplify specific CD8 + T cell populations in vitro, some of these plasmids were used to repeat the DNA immunization experiment and to determine by flow cytometry the intracellular IFN- ⁇ production by CD8 + T cells, both ex vivo and after a 6 day stimulation in the presence of the relevant peptides. In the attempt to establish a direct correlation between IFN- ⁇ production by CD8 + T cells and HLA-A2 specific restriction, the experiment was carried out with both transgenic and non transgenic syngenic mice.
  • the plasmids used contained genes CPn 0695, CPn 0811 and CPn 0823, including peptides CH-13, CH-6 and CH-7 respectively, which were highly positive in the in vitro binding and in the ELISpot assays and gene CPn 0323, including six different peptides, all of them with ELISpot values slightly higher than background
  • CD8 + T cells of transgenic mice infected with C. pneumoniae recognize HLA-A2 binders in vivo It has been recently shown that infection of mice with C. pneumoniae elicits a pathogen-specific murine class I-restricted immune response [22]. Therefore, we asked whether any of the A2 in vitro binders could be recognized by specific CD8 T cells that are clonally selected during the immune response raised against the corresponding native antigen in C. pneumoniae infected cells.
  • HLA-A2 transgenic mice were intranasally infected with a non lethal dose of C. pneumoniae EBs and challenged with an equal dose of bacteria one month later, before being sacrificed to obtain splenocytes that were used to measure IFN- ⁇ production by CD8 + T cells. Since no appreciable IFN- ⁇ -production could be observed if splenocytes from infected mice were tested directly ex vivo (data not shown), spleen cells were cultured with each individual peptide or with the HepB irrelevant peptide for 6 days. The resulting short-term TCLs were then pulsed again for 6 hours with the same peptides and intracellular IFN- ⁇ production by CD8 + T cells was assessed.
  • Fig. 5A The results obtained with 40 tested peptides are shown in Fig. 5A.
  • Sixteen peptides (CH-2, CH-7, CH-8, CH-10, CH-13, CH-15, CH-20, CH-21, CH-28, CH-35, CH-37, CH-45, CH-46, CH-47, CH-50 and CH-55) elicited the strongest CD8 + responses (1 to 7.1 % of IFN- ⁇ -producing CD8 + T cells), while 19 peptides elicited low but consistent responses (percentages of CD8 + /IFN- ⁇ + T cells between 0.3 and 0.9). Five peptides did not induce percentages of IFN- ⁇ -producing CD8 + T cells significantly higher than those observed in response to the HepB control peptide.
  • pneumoniae antigens can indeed reach the cytosol of infected cells and enter the MHC-I presentation pathway, i.e. during remodeling that occurs during Chlamydia replication or following autolysis of developing bacterial particles [22].
  • Kuon et al. [42] recently reported the identification of 11 C. trachomatis-de ⁇ ved HLA-B27-restricted peptides, capable of stimulating CD8 + T cells obtained from patients with Chlamydia-m ⁇ uced reactive arthritis. Importantly, 8 of them overlapped those selected by analyzing splenocytes of HLA-B27 transgenic mice infected with C. trachomatis, indicating that antigen processing can be closely reproduced using the murine animal model, although differences between murine and human antigen processing and T cell repertoires have been hypothesized [43].
  • CH-8 which is the most reactive peptide in the assay with the infected mice, does not seem to be recognized by a specific T cell population when the corresponding antigen is expressed by DNA immunization (Tables 5 and 6). This may be due to different factors, i.e. low in vivo expression level of the injected DNA or altered protein conformation.
  • pneumoniae infected mammalian cells and, possibly, to correlate the identified T cell epitopes with CD8 + T cell populations naturally induced in C. pneumoniae infected patients.
  • results obtained with DNA-mediated expression of distinct antigens can represent an initial step towards the definition of a significant set of C.
  • Example 9 Immunizations with Combinations of the First Antigen Group
  • the five antigens of the first antigen group (OmpH-like protein, pmp 10, pmp2, Enolase, OmpH-like, CPn0042 and CPn00795 were prepared as described in the Materials and Methods Section above for Examples 1-4.
  • the antigens are expressed and purified.
  • Compositions of antigen combinations are then prepared comprising five antigens per composition (and containing 15 ⁇ g of each antigen per composition).
  • CD1 mice are divided into seven groups (5-6 mice per group for groups 1 through 4; 3 to 4 mice for groups 5, 6 and 7), and immunized as follows:
  • mice are immunized at two week intervals. Two weeks after the last immunization, all mice are challenged by intravaginal infection with Chlamydia pneumoniae serovars. Experiment 9 was repeated with another group of CPn antigens. These were: CPn0385 (PepA), CPn0324 (LcrE), CPn0503 (DnaK), CPn0525 (Hypothetical) and CPn0482 (ArtJ). These antigens are combined and administered with and without alum and CpG as described in Experiment 9. Summary
  • CPn proteins with desirable immunological and or biological properties. Specifically, at least twelve CPn proteins have been identified which are capable of inducing the production of antibodies, which can neutralise, in a dose-dependent manner, the infectivity of C. pneumoniae in in vitro cell cultures. The induction of neutralising antibodies is important because it prevents infectious EBs from invading human tissues. Furthermore, at least six of these CPn proteins were also capable of attenuating Chlamydial (C. pneumoniae) infection in a in vivo hamster model. In addition, some of these CPn proteins were also capable of inducing not only adequate T-cell responses but also high serum levels of neutralising antibodies.
  • C. pneumoniae Chlamydial
  • this paper describes a group of recombinant antigens which can induce antibodies inhibiting the infectivity of C. pneumoniae in vitro and have protective effects in vivo.
  • alpha-Enolase of Streptococcus pneumoniae is a plasmin(ogen)-binding protein displayed on the bacterial cell surface.
  • Chlamydia pneumoniae major outer membrane protein is a surface-exposed antigen that elicits antibodies primarily directed against conformation-dependent determinants.
  • Rappuoli,R. Reverse vaccinology. Current Opinion in Microbiology 2000. 3: 445- 450. 3. Rappuoli,R., Reverse vaccinology, a genome-based approach to vaccine development. Vaccine 2001. 19: 2688-2691.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Communicable Diseases (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention relates to polypeptides for use as an autotransporter antigen. The invention further relates to methods and uses of a polypeptide for an autotransporter function in preparation of a medicament for the prevention or treatment of a Chlamydia pneumoniae infection or for the preparation of an assay for the diagnosis of a Chlamydia pneumoniae infection in an individual. Also, a method is provided for raising an immune response in an individual by administering to the individual a polypeptide for use as an autotransporter antigen.

Description

IMMUNOGENIC COMPOSITIONS FOR CHLAMYDIA PNEUNOMIAE
This application claims the benefit of U.S. Provisional Application 60/542,832, filed March 2, 2004; U.S. Provisional Application 60/643,110, filed January 12, 2005; and U.S. Provisional Application 60/644,552, filed January 19, 2005, all of which are incorporated herein in their entireties.
All documents cited herein are incorporated by reference in their entirety. Field
The invention is in the field of immunology and vaccinology. In particular, it relates to immunogenic compositions comprising combinations of immunogenic molecules from Chlamydia pneumoniae. Background Art
The bacteria of the genus Chlamydia (and Chlamydophila, according to the recently proposed but still controversial re-classification of Chlamydiaceae (Bush et αl (2001) Int J Syst Evol Microbiol 51: 203-20; Everett et αl (1999) Int J Syst Bacteriol 49: Pt2 415-40; Schachter et αl (2001) Int J Syst Evol Microbiol 51: 249, 251-3) are obligate intracellular parasites of eukaryotic cells, which have a unique biphasic life cycle involving two pleiomorphic developmental forms: an extracellular, metabolically inert, spore-like, infectious form (the elementary bodies, EBs) and an intracellular, non- infectious, replicative form (the reticulate bodies, Bs) which remains contained in a specialized cytoplasmic compartment (the Chlαmydiαl inclusion). The EBs are responsible for the initial attachment to host cell surface and the establishment of the cytoplasmic inclusion where EBs can differentiate to RBs and thus initiate the replicative stage. Eventually RBs revert to infectious EB forms able to start new replicative cycles in neighbouring host cells. As Chlamydia infection is an intracellular infection, the currently accepted paradigm is that effective anti-Chlamydial immunisation would require both an adequate T-cell response and high serum levels of neutralising antibodies and that "an ideal vaccine should induce long lasting (neutralising) antibodies and a cell mediated immunity that can quickly respond upon exposure to Chlamydia". Several sometimes contradictory studies have indicated that both CD4+ and CD 8 positive T cells have a role in Chlamydia! clearance (Loomis and Starnback (2002) Curr Opin Microbiol 5: 87-91). Indeed, there now appears to be a prevailing consensus that specific CD4+ T cells and B cells are critical to the complete clearance of intracellular Chlamydia and for mediating recall immunity to Chlamydia infection (see Igietseme, Black and Caldwell (2002) Biodrugs 16: 19-35 and Igietseme et al (1999) Immunology 98: 510-519).
Whilst it is now possible to carry out searches of the whole Chlamydia pneumoniae genome, there is still insufficient information available on parallel proteo e characterisation. By way of example, while sequence data is available for many of the Chlamydia pneumoniae antigens, there is insufficient characterisation of the Chlamydia antigens in terms of their immunological and/or biological function. By way of example, whilst applications such as WO 99/28475 and WO 99/27105 disclose sequence information, there is no characterisation of these sequences in terms of their immunological and or biological function. In contrast, WO 02/02404 provides information on the immunogenicity and immunoaccessibility of certain Chlamydia proteins and highlights that (i) current genomic annotations and/or (ii) predictions based on cellular location and/or cellular function based on in-silico analyses may not always be accurate. Applicants have recently engaged in a whole-genome search (Montigiani et al (2002) Infection and Immunity 70:368-379) for possible vaccine candidates among proteins potentially associated with the outer membrane of C.pneumoniae. For this study, mouse antisera was prepared against over 100 recombinant His-tagged or Glutathione-S-transferase (GST) fusion proteins encoded by genes predicted by in silico analyses to be peripherally located in the Chlamydial cell. From this screening study, 53 recombinant proteins derived from the genome of Chlamydia (Chlamydophila pneumoniae (CPn) were described which induced mouse antibodies, capable of binding, in a FACS assay, to the surface of purified CPn cells. The scope of the Montigiani study (ibid) was restricted to checking if polyclonal antisera produced in mice against the recombinantly expressed antibodies to CPn antigens were capable of binding to the surface of the CPn cells. No studies were carried out to test whether antisera against the recombinant FACS positive antigens were capable of interfering with EB in vitro infectivity of host cells - that is, whether the murine antibodies raised against the recombinantly expressed antigens could inhibit CPn infectivity in vitro to an extent greater than 50%, a property that common practice qualifies such antigens as "neutralising".
Indeed, so far, only few C. pneumoniae antigens with 'neutralizing' properties have been described in the literature: notably, a protein identified as 76-kDa-homolog protein (Perez-Melgosa et al (1994) Infect Immunity 62: 880-6), the surface-exposed outer membrane proteins MOMP (Wolf et al (2001) Infect Immun 69: 3082-91), PorB (Kawa et al (2002) J Immunol 168 : 5184-91 and Kubo et al (2000) Mol Microbiol 38 : 772-80), and very recently also the Pmp21 member of the Chlamydia-specific polymorphic family of outer membrane proteins (A.Szczepek, personal comunication). All these proteins were in fact selected in the earlier FACS-based screening study (Montigiani et al (2002) ibid). It can be however noted that outer membrane antigens, as it is the case for MOMP and PorB, could possibly present some kind of practical problems for a recombinant vaccine development project. For instance both MOMP and PorB are integral membrane proteins which appear to require a native conformation to maintain neutralizing epitopes which are discontinuous and conformation-dependent. The production of such proteins may require special processing steps (refolding) which could be undesirable in the preparation of an hypothetical vaccine. Other general problems may arise from the extent of allelic variation, and from regulated proteins which are not always expressed in all Chlamydial cell or all Chlamydial isolates.
Thus, it is desirable to provide improved compositions capable of eliciting an immune response upon exposure to Chlamydia pneumoniae proteins. It is also desirable to provide improved compositions comprising one or more combinations of two or more selected CPn proteins with complementary immunological and/or biological profiles capable of providing immunity against Chlamydial induced disease and/or infection (such as in prophylactic vaccination) or (b) for the eradication of an established chronic Chlamydial infection (such as in therapeutic vaccination). Brief description of the drawings and tables
Figure 1A. Assay of in vitro neutralization of C.pneumoniae infectivity for LLC-MK2 cells by polyclonal mouse antisera to recombinant Chlamydial proteins.
Figure IB shows serum titres giving 50% neutralization of infectivity for 10 C.pneumoniae recombinant antigens. Each titer was assessed in 3 separate experiments (SEM values shown). Figure 2 shows immunoblot analysis of two dimensional electrophoretic maps of C.pneumoniae EBs using the imune sera described in the text.
Figure 3 shows mean numbers of C.pneumoniae IFU recovered from equivalent spleen samples from immunized and mock-immunized hamsters following a systemic challenge.
Figure 4 shows flow cytometric analysis of splenocytes from DNA-immunized HLA- A2 transgenic and non transgenic mice. Figure 5 shows a flow cytometric analysis of splenocytes from transgenic and non transgenic mice infected with C. pneumoniae EBs.
Figure 6 shows an alignment of the proteins in the 7105-7110 protein family. Figure 7 shows an N-terminal alignment of Cpn0794 - Cpn0799.
Figure 8 shows a protein encoded by Cpn0796 and demonstrates a C-terminal domain comprising approximately residues from 1 to 648. Figure 9 shows an alignment of the C-terminal (beta barrel) domains of the proteins encoded by the C.pneumoniae genes Cpn0795 and Cpn0796.
Table I shows a summary of data and properties of the C.pneumoniae antigens described in the text.
Table 2 shows results from hamster mouse model studies for hypothetical proteins.
Table 3 shows expressed genes of CPn EB selected by microarray. Table 4 shows C. pneumoniae selected peptides: protein sources and HLA-A2 stabilization assay.
Table 5 shows ELISPOT assay with CD8+ T cells from DNA immunised HLA-A2 transgenic mice.
Table 6 shows IFN-γ production from splenocytes of DNA immunized HLA-A2 transgenic and non transgenic mice.
Summary of the Invention The present invention relates to a polypeptide for use as an autotransporter antigen, the polypeptide comprising: (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79, (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a); or (c) an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids from an amino acid sequence of (a) or combinations thereof.
The present invention also relates to the use of a polypeptide in the preparation of a medicament for the prevention or treatment of a Chlamydia pneumoniae infection in an individual. For example, the use of the polypeptide may be as an autotransporter protein which immunoreacts with seropositive serum of an individual infected with Chlamydia pneumoniae. The present invention further relates to a method of eliciting an immune response in an individual comprising administering to the individual a polypeptide comprising (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79 (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a), or (c) an amino acid sequence comprising one or more fragment of at least 1, 2, 3, 4, 5, 6, or 7 amino acids from an amino acid sequence of (a) or mixtures thereof.
Also, a method is provided for diagnosing an immune response in an individual comprising (a) contacting a biological sample obtained from the individual with a binding agent that binds to a polypeptide with an autotransporter function, (b) detecting in the biological sample the amount of the polypeptide that binds to the binding agent; and (c) comparing the amount of the polypeptide to a predetermined cut-off value and thereby determining the presence of an immune response in the individual. A composition for eliciting an immune response in a subject comprising two or more Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof is also provided. The composition may further comprise one or more immunostimulants.
Also provided is a polypeptide for use as an autotransporter antigen comprising an amino acid sequence corresponding to SEQ ID NO: 86, an amino acid sequence having at least 50% sequence identity to SEQ ID NO: 86, or an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids of SEQ ID NO: 86. The present invention relates to a composition comprising a first biological molecule from a Chlamydia pneumoniae bacterium and a second biological molecule from a Chlamydia pneumoniae bacterium. The first biological molecule is selected from the group consisting of SEQ ID No 1 to SEQ ID No 86, or the group consisting of SEQ ID No. l to 41.
The composition may also contain the second biological molecule being selected from the group consisting of SEQ ID No 1 to SEQ ID No. 86 or SEQ ID No 1 to SEQ ID The composition may also comprise two or more biological molecules selected from the group consisting of SEQ ID Nos 1-41.
The composition may also comprise one or more biological molecules selected from the group consisting of SEQ ID Nos 1-41 combined with one or more biological molecules selected from the group consisting of SEQ ID Nos 42-86.
The composition according to any one of the previous claims further comprising an adjuvant such as an ADP-ribosylating exotoxin or a derivative thereof or an adjuvant is selected from the group consisting of cholera toxin (CT), Escherichia heat-labile exterotoxin (LT) and mutants thereof having adjuvant activity.
A vaccine and use of the vaccine is also provided comprising the composition of the present invention. The vaccine may be used in the preparation of a medicament for the prevention or treatment of a Chlamydia infection and may be administered mucosally, intra-nasally or intra-vaginally, for example.
Further, a method is provided for treating a Chlamydia infection in a host subject wherein the method comprises the administration of a safe and effective amount of a vaccine.
In another aspect of the invention, an immunogenic composition is provided comprising a combination of Chlamydia pneumoniae antigens, the combination comprising at least one Chlamydia pneumoniae antigen associated with elementary bodies of Chlamydia pneumoniae and at least one Chlamydia pneumoniae antigen associated with reticulate bodies of Chlamydia pneumoniae.
In another aspect of the invention, an immunogenic composition is provided comprising a combination of Chlamydia pneumoniae antigens, the combination comprising at least one Chlamydia pneumoniae antigen of a first antigen group and at least one Chlamydia pneumoniae antigen of a second antigen group, said first antigen group comprising a Type III secretion system (TTSS) protein and said second antigen group comprising a Type III secretion system (TTSS) effector protein. In yet another aspect of the invention, an immunogenic composition is provided comprising a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
In still another aspect of the invention, an immunogenic composition is provided comprising a combination of Chlamydia pneumoniae antigens, the combination eliciting a Chlamydia pneumoniae specific TH1 immune response and a Chlamydia pneumoniae specific TH2 immune response.
The present invention further provides a method of monitoring the efficacy of treatment of a patient infected with Chlamydia pneumoniae comprising determining the level of Chlamydia pneumoniae specific antibody in the patient after administration of an immunogenic composition of the present invention to the patient.
Description of the Invention The present invention provides compositions comprising a first biological molecule from a Chlamydia pneumoniae bacterium and a second biological molecule from a Chlamydia pneumoniae bacterium. The term "biological molecule" includes proteins, antigens and nucleic acids. The compositions may also comprise further biological molecules preferably also from Chlamydia pneumoniae. That is to say, the compositions may comprise two or more biological molecules (eg. 3, 4, 5, 6, 7, 8 etc.) at least two of which are from a Chlamydia pneumoniae bacterium (eg. 3, 4, 5, 6, 7, 8 etc.). Such compositions include those comprising (i) two or more different Chlamydia pneumoniae proteins; (ii) two or more different Chlamydia pneumoniae nucleic acids, or (iii) mixtures of one or more Chlamydia pneumoniae protein and one or more Chlamydia pneumoniae nucleic acid.
In one aspect of the present invention, an immunogenic composition is provided comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response (such as a cell mediated or cellular immune response) and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 response (such as a humoral or antibody response). The immunogenic composition may further comprise a THl adjuvant and a TH2 adjuvant. In another aspect of the present invention, an immunogenic composition is provided comprising a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
In yet another aspect of the present invention, an immunogenic composition is provided comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 immune response, the combination comprising at least one Chlamydia pneumoniae antigen that is conserved over at least two serovars.
In another aspect of the present invention, the immunogenic composition comprising at least one antigen that elicits a Chlamydia pneumoniae specific THl immune response and at least one antigen that elicits a Chlamydia pneumoniae specific TH2 immmune response preferably comprises a combination of Chlamydia pneumoniae antigens comprising at least one Chlamydia pneumoniae antigen associated with the EB of Chlamydia pneumoniae and at least one Chlamydia pneumoniae antigen associated with the RB of Chlamydia pneumoniae. Still further such combinations can comprise EB and/or RB antigens from one serovar combined with RB and/or EB antigens from at least one other serovar.
In an additional aspect of the present invention, a kit is provided comprising a combination of Chlamydia pneumoniae antigens wherein at least one of the Chlamydia pneumoniae antigens is associated with the EB of Chlamydia pneumoniae and at least one of the Chlamydia pneumoniae antigens is associated with the RB of Chlamydia pneumoniae. The kit may further include a THl adjuvant, a TH2 adjuvant and instructions.
The present invention further provides methods of eliciting a Chlamydia specific immune response by administering an immunogenic composition of this invention. The present invention further provides a method of monitoring the efficacy of treatment of a subject infected with Chlamydia pneumoniae comprising determining the level of Chlamydia specific antibody or Chlamydia specific effector molecule in the subject after administration of an immunogenic composition of this invention.
In one preferred embodiment the first and second biological molecules are from different Chlamydia pneumoniae species (for example, from different Chlamydia pneumoniae serovars) but they may be from the same species. The biological molecules in the compositions may be from different serogroups or strains of the same species. The first biological molecule is preferably selected from the group consisting of SEQ ID Nos 1-86. More preferably, it is selected from the group consisting of SEQ IDs l-41and/or SEQ ID Nos 42-86. It is preferably a purified or isolated biological molecule. The second biological molecule is preferably selected from the group consisting of SEQ ID Nos 1-86. More preferably, it is selected from the group consisting of SEQ IDs 1-41 and/or SEQ ID Nos 42-86. It is preferably a purified or isolated biological molecule. Specific compositions according to the invention therefore include those comprising: two or more biological molecules selected from the group consisting of SEQ ID Nos 1-41; one or more biological molecules selected from the group consisting of SEQ IDs 1-41 combined with one or more biological molecules selected from the group consisting of SEQ IDs 42-86. One or both of the first and second biological molecules may be a Chlamydia pneumoniae biological molecule which is not specifically disclosed herein, and which may not have been identified, discovered or made available to the public or purified before this patent application was filed.
In another embodiment, a combination of Chlamydia pneumoniae antigens is provided, the combination comprising at least one Type III Secretion System (TTSS) protein and at least one Type III Secretion System (TTSS) secreted or effector protein or fragment thereof. There are many methods for identifying TTSS proteins (i.e., TTSS proteins associated with the Chlamydial TTSS machinery). TTSS is a complex protein secretion and delivery machine or apparatus, which may be located, either wholly or partially, on the Elementary Body (EB) and which allows an organism, such as Chlamydia, to maintain its intracellular niche by injecting proteins, such as bacterial effector proteins (which may act as anti-host virulence determinants) into the cytosol of a eukaryotic cell in order to establish the bacterial infection and to modulate the host cellular functions. TTSS proteins exposed on the EB surface may play a role in adhesion and/or uptake into host cells. By way of background information, the TTSS is a complex protein secretion and delivery machine or apparatus, which may be located on the Elementary Body (EB) and which allows an organism, such as Chlamydia, to maintain its intracellular niche by injecting proteins, such as bacterial effector proteins (which may act as anti-host virulence determinants) into the cytosol of a eukaryotic cell in order to establish the bacterial infection and to modulate the host cellular functions. These injected proteins (the TTSS effector proteins) can have various effects on the host cell which include but are not limited to manipulating actin and other structural proteins and modification of host cell signal transduction systems. The injected (or translocated) proteins or substrates of the TTTS system may also be processed and presented by MHC-class I molecules. Not all the proteins secreted by a Type III secretion system are delivered into the host cell or have effector function. Although the Elementary Body (EB) is regarded as "metabolically inert", it has been postulated that the Chlamydial TTSS system located on the (EB) is triggered by membrane contact and is capable of releasing pre-formed "payload" proteins. The current hypothesis is that Type Three Secretion System (TTSS) becomes active during the intracellular phase of the chlamydial replicative cycle for the secretion of proteins into the host cell cytoplasm and for the insertion of chlamydial proteins (like the Inc set) into the inclusion membrane that separates the growing chlamydial microcolony from the host cell cytoplasm (see Montigiani et al (2002) Infection and Immunity 70(1); 386-379).
Proteins may be expressed and secreted by 2 hours (early cycle) after infection while the expression of other early and mid cycle Type III specific genes are not detectable until 6-12 hours (mid cycle). After 16-20 hours, the RBs begin to differentiate into EBs, and by 48-72 hours, the EBs predominate within the inclusion. Host cell lysis results in the release of the EBs to the extracellular space where they can infect more cells. For purposes of this description, an early gene is one that is expressed (in terms of mRNA expression) early in infection, an intermediate gene is one that is expressed in the mid-cycle after infection and a late gene is one which is expressed during the terminal transition of RBs to EBs. There may be a time lag between surface expression of early, mid and late stage proteins and their transcriptional and translational profiles because mRNA abundance may not always correlate with protein abundance.
In one example, the present invention may comprise TTSS effector proteins. The TTSS effector proteins as described are associated with the RB form of Chlamydia pneumoniae and may be identified, for example, using immunofluorescence microscopy (see Bannantine et al, Infection and Immunity 66(12); 6017-6021). Effector antibodies to putative Chlamydial TTSS effector proteins secreted by the TTSS machinery may be micro-injected into host cells at specified time points during Chlamydia pneumoniae infection (e.g., early, mid or late cycle). Host cell reaction to Chlamydia pneumoniae (e.g., actin remodeling, inhibition of endosomal maturation, host lipid acquisition, and MHC Class I and Class II molecule downregulation) associated with Chlamydia pneumoniae entry into host cells is then observed. Based on these temporal observations, TTSS effector proteins (RB-associated Chlamydia pneumoniae proteins) may be detected.
A specific composition of the present invention may comprise a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, five or all six Chlamydia pneumoniae antigens of a first antigen group, said first antigen group consisting of: (1) pmp2; (2) pmplO; (3) Enolase; (4) OmpH-like protein; and (5) the products of CPn specific genes CPn0759 and CPn0042. These antigens are referred to herein as the 'first antigen group'.
Preferably, the composition of the invention comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of: (1) pmp2 and pmplO; (2) pmp2 and Enolase; (3) pmp2 and OmpH-like protein; (4) pmp2 and CPn0759; (5) pmp2 and CPn0042; (6) pmplO and Enolase; (7) pmplO and OmpH-like protein; (8) pmplO and CPn0759; (9) pmplO and CPn0042; (10) Enolase and OmpH-like protein (11) Enolase and CPn0759; (12) Enolase and CPn0042; (13) OmpH-like protein and CPn0759 (14) OmpH-like protein and CPn0042; (15) CPn0759 and CPn0042; (16) pmp2 and pmplO and Enolase; (17) pmp2 and pmplO and OmpH-like protein; (18) pmp2 and pmplO and CPn0759; (19) pmp2 and pmplO and CPn0042; (20) pmp2 and Enolase and OmpH-like protein; (21) pmp2 and Enolase and Cpn0759; (22) pmp2 and Enolase and CPn0042; (23) pmp2 and OmpH- like protein and CPn0759; (24) pmp2 and OmpH-like protein and CPn0042; (25) pmp2 and Cpn0759 and CPn0042; and (26) pmplO and Enolase and OmpH-like protein; (27) pmplO and Enolase and CPn0759; (28) pmplO and Enolase and CPn0042; (29) Enolase and OmpH-like protein and CPn0759; (30) Enolase and OmpH-like protein and CPn0042; (31) OmpH-like protein and CPn0759 and CPn0042. Preferably, the composition of Chlamydia pneumoniae antigens consists of pmp2, pmplO, Enolase, OmpH-like protein and CPn0759.
Preferably, the composition of Chlamydia pneumoniae antigens consists of pmp2, pmplO, Enolase, OmpH-like protein and CPn0042. Preferably, the composition of Chlamydia pneumoniae antigens consists of pmp2, pmplO, Enolase, OmpH-like protein and CPn0759 and CPn0042.
The invention also provides for a slightly larger group of 12 Chlamydia pneumoniae antigens that are particularly suitable for immunisation purposes, particularly when used in combinations. (This second antigen group includes the six Chlamydia pneumoniae antigens of the first antigen group). These 12 Chlamydia pneumoniae antigens form a second antigen group of (1) pmp2; (2) pmplO; (3) Enolase; (4) OmpH-like protein; (5) CPn0759; (6) CPn0042; (7) ArtJ; (8) HtrA; (9) AtoS; (10) OmcA; (11) CPn0498; and (12) CPn0525. These antigens are referred to herein as the 'second antigen group'.
The invention therefore provides a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve Chlamydia pneumoniae antigens of the second antigen group. Preferably, the combination is selected from the group consisting of two, three, four or five Chlamydia pneumoniae antigens of the second antigen group. Still more preferably, the combination consists of six Chlamydia pneumoniae antigens of the second antigen group. Each of the Chlamydia pneumoniae antigens of the first and second antigen group are described in more detail below.
(1) PmplO (CPn0449)
One example of a pmplO protein is set forth as SEQ ID NO: 1 below (GenBank Accession No.GI: 14195016). Preferred pmplO proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 1; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 1, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These pmp2 proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 1. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 1. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 1. 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).
SEQ ID No 1 1 MKSQFSWLVL SSTLACFTSC STVFAATAEN IGPSDSFDGS TNTGTYTPKN TTTGIDYTLT 61 GDITLQN GD SAALTKGCFS DTTESLSFAG KGYSLSFLNI KSSAEGAALS VTTDKNLS T 121 GFSS TFLAA PSSVITTPSG KGAVKCGGDL TFDNNGTI F KQDYCEENGG AISTKN SLK 181 NSTGSISFEG NKSSATGKKG GAICATGTVD ITNNTAPTLF SNNIAEAAGG AINSTGNCTI 241 TGNTS VFSE NSVTATAGNG GA SGDADVT ISGNQSVTFS GNQAVANGGA IYAKKLTLAS 301 GGGGGISFSN NIVQGTTAGN GGAISI AAG ECSLSAEAGD ITFNGNAIVA TTPQTTKRNS 361 IDIGSTAKIT NLRAISGHSI FFYDPITANT AADSTDTLNL NKADAGNSTD YSGSIVFSGE 421 K SEDEAKVA DNLTST1KQP VTLTAGNLVL RGVTLDTKG FTQTAGSSVI DAGTT KAS 481 TEEVTLTGLS IPVDS GEGK KWIAASAAS KNVALSGPIL LLDNQGNAYE NHDLGKTQDF 541 SFVQLSALGT ATTTDVPAVP TVATPTHYGY QGTWGMTWVD DTASTPKTKT ATLAWTNTGY 601 LPNPERQGPL VPNSL GSFS DIQAIQGVIE RSALTLCSDR GF AAGVANF LDKDKKGEKR 661 KYRHKSGGYA IGGAAQTCSE N ISFAFCQL FGSDKDFLVA KNHTDTYAGA FYIQHITECS 721 GFIGC LDKL PGS SHKPLV LEGQ AYSHV SND KTKYTA YPEVKGSWGN NAFNMMLGAS 781 SHSYPEY HC FDTYAPYIKL NLTYIRQDSF SEKGTEGRSF DDSN FN S PIGVKFEKFS 841 DCNDFSYDLT LSYVPDLIRN DPKCTTALVI SGAS ETYAN NLARQALQVR AGSHYAFSPM 901 FEVLGQFVFE VRGSSRIYNV DLGGKFQF
(2) Pmp2 = Polymorphic Outer Membrane Protein G Family (CPn 0013)
One example of a pmp2 protein is disclosed as SEQ ID NO : 139 and 140 in WO 02/02606. {GenBank accession number: gi|4376270|gb|AAD18172.1 'CPn0013'; SEQ ID NO: 2 below}. Preferred pmp2 proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 2; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 1, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These pmρ2 proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 2. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 1. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 2. 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). SEQ ID No 2 1 M IPLRFL I SLVPTLSMSN LLGAATTEE SASNSFDGTT STTSFSSKTS 51 SATDGTNYVF KDSWIENVP KTGETQSTSC FKNDAAAGDL NFLGGGFSFT 101 FSNIDATTAS GAAIGSEAAN KTVTLSGFSA LSFLKSPAST VTNG GAINV 151 KGN SLLDND KV IQDNFST GDGGAINCAG SLKIA NKSL SFIGNSSSTR 201 GGAIHTKN T LSSGGET FQ GNTAPTAAGK GGAIAIADSG TLSISGDSGD 251 I1FEGNTIGA TGTVSHSAID LGTSAKITAL RAAQGHTIYF YDPITVTGST 301 SVADALNINS PDTGDNKEYT GTIVFSGEKL TEAEAKDEKN RTSKLLQNVA 351 FKNGTVVLKG DWLSANGFS QDANSKLIMD GTSLVANTE SIELTN E1N 401 IDS RNGKKI KLSAATAQKD IRIDRPWLA ISDESFYQNG FLNEDHSYDG 451 ILELDAGKDI VISADSRSID AVQSPYGYQG KWTIN STDD KKATVSWAKQ 501 SFNPTAEQEA P VPNLL GS FIDVRSFQNF IELGTEGAPY EKRF VAGIS 551 NVLHRSGREN QRKFRHVSGG AWGASTRMP GGDTLS GFA QLFARDKDYF 601 MNTNFAKTYA GSLRLQHDAS YSWSIL G EGGLREILLP YVSKTLPCSF 651 YGQLSYGHTD HRMKTESLPP PPPT STDHT SWGGYV AGE GTRVAVENT 701 SGRGFFQEYT PFVKVQAVYA RQDSFVELGA ISRDFSDSH YNLAIP GIK 751 LEKRFAEQYY HWAMYSPDV CRSNPKCTTT LLSNQGSWKT KGSNLARQAG 801 IVQASGFRSL GAAAE FGNF GFEWRGSSRS YNVDAGSKIK F*
(3) Enolase (Cpn0800)
One example of an 'Eno' protein is disclosed as SEQ ID NOs: 93 and 94 in WO 02/02606. {GenBank accession number: gi|4377111|gb|AAD18938.1| ' Cpn0800'; SEQ ID NO: 3 below} .Preferred Eno proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 2; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 2, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Eno proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 3. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 3. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 3. 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). SEQ ID No 3 1 MFEAVIAD1Q AREI DSRGY PTLHVKVTTS TGSVGEARVP SGASTGKKEA 51 LEFRDTDSPR YQGKGVLQAV KNVKEILFPL VKGCSVYEQS IDSLMMDSD 101 GSPNKETLGA NAILGVSLAT AHAAAATLRR PLYRY GGCF ACSLPCPMMN 151 INGGMHADN G EFQEFMIR PIGASSIKEA VNMGADVFHT LKKLLHERGL 201 STGVGDEGGF APN ASNEEA LE LLLAIEK AGFTPGKDIS ALDCAASSF 251 YNVKTGTYDG RHYEEQIAIL SNLCDRYPID SIEDG AEED YDGWA LTEV 301 LGEKVQIVGD DLFVTNPELI EGISNG AN SVLIKPNQIG TLTETVYAIK 351 LAQMAGYTTI ISHRSGETTD TTIADLAVAF NAGQIKTGSL SRSERVAKYN 401 R MEIEEELG SEAIFTDΞNV FSYEDSEE*
(4) OmpH-like outer membrane protein (CPn0301)
One example of 'OmpH-like' protein is disclosed as SEQ ID NOs: 77 & 78 in WO 02/02606. {GenBank accession number: gi|4376577|gb|AAD18450.1| 'CPn0301'; SEQ ID NO: 4 below}. Preferred OmpH-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 4; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 3, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmpH-like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 4. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 4. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 4. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). SEQ ID No 4 1 MKKLIiFSTFI. LVLGSTSAAH ANLGYVNLKR C EESDLGKK ETEELEAMKQ 51 QFVKNAEKIE EELTSIYNKL QDEDYMESLS DSASEE RKK FEDLSGEYNA 101 YQSQYYQS1N QSNVKRIQKL IQEVKIAAES VRSKEK EAI NEEAVLAIA 151 PGTDKTTEII AILNESFKKQ N*
(5) CPn0042 (Hypothetical)
One example of hypothetical protein is set forth as SEQ ID NO: 5 below. GenBank accession number: gi|4376296|gb|AAD 18195.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 5; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 5, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 5. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 5. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 5 Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
SEQ ID No 5 1 MEEVSEY QQ VENQ ESCSK RLTK ETFAL GVRLEAKEEI ESIILSDVVN RFEVLCRDIE 61 DMLSRVEEIE RM RMAELPL LPIKEALTKA FVQHNSCKEK TKVEPYFKE SPAY TSEER 121 QSLNQTLQR AYKESQKVSG ESEVRACRE QLKDQVRQFE TQGVS IKEE I FVTSTFRT 181 KFSYHSFRLH VPCMRLYEEY YDDIDLERTR ARWMAMSERY RDAFQAFQEM LKEGLVEEAQ 241 ALRETEY LY REERKSKKKH
(6) CPn0795 (Hypothetical)
One example of hypothetical protein is disclosed as SEQ ID NOs: 63 & 64 in WO 02/02606. {GenBank accession number: gi|4377106|gb|AAD18933.1| 'CPn0795'; SEQ ID NO: 6 below). As the examples demonstrate, we have shown for the first time that CPn0795 and related proteins in the group Cpn0794 - Cpn0799 have a secreted autotransporter function. It has been shown that proteins secreted by the autotransporter secretion mechanism possess an overall unifying structure, including an amino-terminal leader peptide (for secretion across the inner membrane), the secreted mature protein (or passenger domain), and a dedicated C-terminal domain, which forms a pore in the outer membrane through which the passenger domain passes to the cell surface. It is likely that requirements for secretion across the outer membrane are contained within a single molecule and secretion is an energy- independent process. Structural properties of the proteins may be confined by the size of the pore considering the biophysical constraints that may be imposed on secretion. The autotransporter, or type V, secretion system is a dedicated protein translocation mechanism which allows the organism to secrete proteins to and beyond the bacterial surface. The secretion mechanism and the ability to develop a new autotransporter protein simply by a single recominbation event have presented bacteria with abundant opportunities to increase the efficiency of secretion of proteins that were developed as periplasmic or exported virulence factors.
In one model of autotransporter (type V) secretion mechanism, proteins are exported by the autotransporter secretion mechanism and are translated as a polyproptein possessing domains. The autotransporters possess an overall unifying structure comprising three functional domains: the amino-termianl leader sequence, the secreted mature proterin (passenger domain) and a carboxy-terminal (beta-) domain that forms a beta-barrel pore to allow secretion of the passenger protein. The leader sequence directs secretion via the sec apparatus and is cleaved at the inner membrane by a signal peptidase releasing the remaining portion of the molecule into the periplasm. Once in the periplasm the β-domain assumes a biophysically favored state characterized by a β-barrel shaped structure which inserts itself into the outer membrane to form a pore. After insertion into the outer membrane the passenger domain is translocated to the bacterial cell surface where it may remain intact or undergo processing. A processed protein may be released into the extracellular milieu or remain associated with the bacterial cell surface. (Henderson and Nataro, "Virulence Functions of Autotransporter Proteins", Infection and Immunity, Vol. 69, No. 3, March 2001, pages 1231-1243).
Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 6; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 6, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 6. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 6. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 6. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). As the Examples demonstrate, we have shown for the first time that CPn0795 appears to be present and accessible to antibodies on the surface of the infectious EB form which makes this protein a good component of an immunogenic composition or vaccine.
Table 1 of this application demonstrates that Cpn0795 (SEQ ID NO: 6) a Cpn specific hypotlietical protein is a FACS positive protein which demonstrates significant immunoprotective activity in a hamster spleen model of Chlamydia pneumoniae infection. We have found evidence to demonstrate that other Cpn proteins in this group of Cpn specific hypothetical proteins have now been found to have a secreted autotransporter function. These proteins, which are absent from Chlamydia trachomatis include: gi/4377105 (Cpn0794), gi 4377106 (Cpn0795), gi/4377107 (Cpn0796), gi 4377108 (Cpn0797), gi/4377109 (CPn0798), gi 4377110 (Cpn0799). SEQ ID No 6 1 MKDLGTLGGT SSTAKTVSPD GKVIMGRSQI ADGSWHAFMC HTDFSSNNVL 51 FD DNTYKTL RENGRQ NSI FNLQNMMLQR ASDHEFTEFG RSNIALGAGL 101 YVNALQN PS NLAAQYFGIA YKIRPKYR G VFLDHNFSSH VPNNFNVSHN 151 R WMGAFIG QDSDALGSSV KVSFGYGKQK ATITREQLEN TEAGSGESHF 201 EGVAAQIEGR YGKS GGHVR VQPFLGLQFV HITRKEYTEN AVQFPVHYDP 251 IDYSTGWYL GIGSHIA VD SLHVGTRMGM EQNFAAHTDR FSGSIASIGN 301 FVFEKLDVTH TRAFAEMRVN YELPYLQS N LILRVNQQPL QGVMGFSSD 351 RYALGF*
(7) ArtJ arginine periplasmic-binding protein (CPn 0482)
One example of 'ArtJ' protein is disclosed as SEQ ID NOs: 73 & 74 in WO 02/02606. {GenBank accession number: gi|4376767|gb|AAD18622.1| 'CPn0482'; SEQ ID NO: 7 below} . Preferred ArtJ proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 7; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 7, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These ArtJ proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 7. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 7. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 7. 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). The ArtJ protein may be bound to a small molecule like arginine or another amino acid. SEQ ID No 7 1 MIKQIGRFFR AFIFIMPLSL TSCESKIDRN RI IVGTNAT YPPFEYVDAQ 51 GEWGFDIDL AKAISEK GK Q EVREFAFD A ILNLKKHR IDAILAGMSI 101 TPSRQKEIAL LPYYGDEVQE LMWSKRS E TPV PLTQYS SVAVQTGTFQ 151 EHY LSQPGI CVRSFDSTLE VIMEVRYGKS PVAVLEPSVG RW KDFPNL 201 VATR ELPPE CWVLGCGLGV AKDRPEEIQT IQQAITDLKS EGVIQSLTKK WQLSEVAYE*
(8) HtrA DO Serine Protease (CPn0979) One example of an 'HrtA' protein is disclosed as SEQ ID NOs: 111 & 112 in WO 02/02606. {GenBank accession number: gi|4377306|gb|AAD19116.1| 'CPn0979'; SEQ ID NO: 8 below}. Preferred HrtA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 8; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 8, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These HrtA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 8. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 8. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably at least 16 to remove the signal peptide) from the N-terminus of SEQ ID NO: 8. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). In relation to SEQ ID NO: 8, distinct domains are residues: 1-16; 17-497; 128-289; 290-381; 394- 485; and 394-497. SEQID N0 8 1 MITKQ RSWIi AVLVGSS IA LP SGQAVGK KESRVSELPQ DVLLKEISGG 51 FSKVATKATP AVVYIESFPK SQAVTHPSPG RRGPYENPFD YFNDEFFNRF 101 FG PSQREKP QSKEAVRGTG FLVSPDGYIV TNNHWEDTG KIHVTLHDGQ 151 KYPATVIGLD PKTD AVIKI KSQNLPY SF GNSDHLKVGD WAIAIGNPFG 201 LQATVTVGVI SAKGRNQLHI ADFEDFIQTD AAINPGNSGG P LNIDGQVI 251 GVNTAIVSGS GGYIGIGFAI PSLMANRIID QLIRDGQVTR GFLGVTLQPI 301 DAELAACYK EKVYGA VTD VVKGSPADKA GLKQEDVIIA YNGKEVDS S 351 MFRNAVSLMN PDTRIVLKW REGKVIEIPV TVSQAPKEDG MSA QRVGIR 401 VQNLTPETAK KLGIAPETKG I IISVEPGS VAASSGIAPG QLI AVNRQK 451 VSSIEDLNRT LKDSNNENIL LMVSQGDVIR FIALKPEE*
(9) AtoS two-component regulatory system sensor histidine kinase protein (CPn0584)
One example of 'AtoS' protein is disclosed as SEQ ID NOs: 105 & 106 in WO 02/02606. {GenBank accession number: gi|4376878|gb|AAD18723.1| 'CPn0584'; SEQ ID NO: 9 below}. Preferred AtoS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 9; and/or (b) which is a fragment of at least .. consecutive amino acids of SEQ ID NO: 9, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These AtoS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 9. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 9. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 9. 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).
SEQ ID No 9 1 MNVPDSKNLH PPAYE EIK ARITQSYKEA SAILTAIPDG I LLSETGHF 51 LICNSQAREI LGIDENLEI NRSFTDVLPD TCLGFSIQEA LESLKVPKTL 101 RLSLCKESKE KEVELFIRKN EISGYLFIQI RDRSDYKQLE NAIERYKNIA 151 ELGKMTATLA HEIRNP SGI VGFASI KKE ISSPRHQRM SSIISGTRSL 201 NN VSSMLEY TKSQPLNLKI INLQDFFSSL IP LSVSFPN CKFVREGAQP 251 LFRSIDPDRM NSWWNLVKN AVETGNSPIT LTLHTSGDIS VTNPGTIPSE 301 IMDKLFTPFF TTKREGNGLG LAEAQKIIRL HGGDIQLKTS DSAVSFFIII 351 PE LAA PKE RAAS* (10) OmcA 9kDa-cysteine-rich Upoprotein(CPn0558)
One example of OmcA' protein is disclosed as SEQ ID NOs: 9 & 10 in WO 02/02606. {GenBank accession number: gi|4376850|gb|AAD18698.1| 'CPn0558', 'OmcA', 'Omp3'; SEQ ID NO: 10 below}. Preferred OmcA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 10; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 10, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 10. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 10. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 10. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). The protein may be lipidated (e.g. by a N- acyl diglyceride), and may thus have a Ν-terminal cysteine. SEQID No lO 1 MKKAVLIAA FCGWSLSSC CRIVDCCFED PCAPSSCΝPC EVIRKKERSC 51 GGΝACGSYVP SCSΝPCGSTE CΝSQSPQVKG CTSPDGRCKQ * (11) CPn0498 (Hypothetical)
One example of a hypothetical protein is set forth as SEQ ID NO: 11 below. (GenBank Accession No. GL4376784; AAD18638.1). Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 11 ; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 11, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 11. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 11. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 11. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). The protein may be lipidated (e.g. by a N-acyl diglyceride), and may thus have a Ν-terminal cysteine.
SEQID No ll 1 MΝRRKAR VV ALFAMTA IS VGCCPWSQAK SRCSIDKYIP VV RLLEVCG LPEAEΝVEDL 61 IESSSAWVLT PEERFSGELV SICQVKDEHA FYΝDLSLLH TQAVPSYSAT YDCAWFGGP 121 LPALRQR DF LVRE QRGVR FKKIVFLCGE RGRYQSIEEQ EHFFDSRYΝP FPTEEΝ ESG 181 ΝRVTPSSEEE IAKFVWMQML LPRA RDSTS GVRVTF LAK PEEΝRWAΝR KDT LLFRSY 241 QEAFPGRVLF VSSQPFIGLD ACRVGQFFKG ESYDLAGPGF AQGV YHWA PRIC HT AE 301 KETΝGCLΝ ISEGCFG
(12) CPn 0525 (hypothetical)
One example of 'Cpn0525' protein is disclosed as SEQ ID ΝOs: 117 & 118 in WO 02/02606. {GenBank accession number: gi|4376814|gb|AAD18665.1| 'CPn0525', SEQ ID NO: 12 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 12; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 12, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 12. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 12. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 12. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). SEQ ID No 12 1 MHDALLSILA IQELDIKMIR LMRVKKEHQK ELAKVQS KS DIRRKVQEKE 51 LEMENLKTQI RDGENRIQEI SEQINKLENQ QAAVKKMDEF NALTQEMTTA 101 NKERRSLEHQ LSD MDKQAG GEDLIVSLKE SLASTENSSS VIEKEIFESI 151 K INEEGKA LEQRTELKHA TNPELLSIYE RLLNNKKDRV WPIENRVCS 201 GCHIVLTPQH EN VRKKDRL IFCEHCSRIL Y QESQVNAQ ENSTAKRRRR 251 RAAV*
Third Antigen Group
The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group. Such other Chlamydia pneumoniae antigens include a third antigen group consisting of (1) LcrE, (2) DnaK, (3) Omp85 homolog, (4) Mip-like; (5) OmcB (6) MurG (7) Cpn0186 and (8) fliY. These antigens are referred to herein as the "third antigen group".
(13) LcrE low calcium response E protein (CPn0324)
One example of a 'LcrE' protein is disclosed as SEQ ID NOs: 29 & 30 in WO 02/02606. {GenBank accession number: gi|4376602|gb|AAD18473.1| 'CPn0324'; SEQ ID NO: 13 below}. Preferred LcrE proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 13; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 13, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These LcrE proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 13. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 13. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 13. 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).
SEQ ID No 13 1 MAASGGTGGL GGTQGVNLAA VEAAAAKADA AEWASQEGS E NMIQQSQD 51 TNPAAATRT KKKEEKFQTL ESRKKGEAGK AEKKSESTEE KPDTD ADKY 101 ASGNSEISGQ ELRGLRDAIG DDASPEDILA LVQEKIKDPA QSTALDYLV 151 QTTPPSQGKL KEALIQARNT HTEQFGRTAI GAKNILFASQ EYADQLNVSP 201 SG RSLYLEV TGDTHTCDQL LSM QDRYTY QDMAIVSSFL MKGMATELKR 251 QGPYVPSAQL QV MTETRNL QAVLTSYDYF ESRVPILLDS KAEGIQTPS 301 DLNFVKVAES YHKIINDKFP TASKVEREVR NLIGDDVDSV TGVLNLFFSA 351 RQTSSR FS SADKRQQLGA MIANALDAVN INNEDYPKAS DFPKPYPWS* (14) DnaK heat-shock protein 70 (chaperone) (CPn0503)
One example of 'DnaK' protein is disclosed as SEQ ID NOs: 103 & 104 in WO 02/02606. {GenBank accessionnumber: gi|4376790|gb|AAD18643.1| 'CPn0503'; SEQ ID NO: 14 below. Preferred DnaK proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 14; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 14, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These DnaK proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 14. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 14. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 14. 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).
SEQ ID No 14 1 MSEHKKSSKI IGID GTTNS CVSVMEGGQA KVITSSEGTR TTPSIVAFKG 51 NEKLVGIPAK RQAVTNPEKT LGSTKRFIGR KYSEVASEIQ TVPYTVTSGS 101 KGDAVFEVDG KQYTPEEIGA QILMKMKETA EAY GETVTE AVITVPAYFN 151 DSQRASTKDA GRIAGLDVKR IIPEPTAAAL AYGIDKVGDK K1AVFDLGGG 201 TFDISILEIG DGVFEVLSTN GDTLLGGDDF DEVIIKWMIE EFKKQEGIDL 251 SKDNMALQRL KDAAEKAKIE LSGVSSTEIN QPFITMDAQG PKHLA TLTR 301 AQFEKLAAS IERTKSPCIK ALSDAK SAK DIDDVLLVGG MSRMPAVQET 351 VKE FGKEPN KGVNPDEWA IGAAIQGGVL GGEVKDVLLL DVIP SLGIE 401 TLGGVMTTLV ERNTTIPTQK KQIFSTAADN QPAVTIWLQ GERPMAKDNK 451 EIGRFDLTDI PPAPRGHPQI EVSFDIDANG IFHVSAKDVA SGKEQKIRIE 501 ASSGLQEDEI QRMVRDAEJN KEEDKKRREA SDAKNEADSM IFRAEKAIKD 551 YKEQIPET V KEIEERIENV RNALKDDAPI EKIKEVTED SKHMQKIGES 601 MQSQSASAAA SSAANAKGGP NINTED KKH SFSTKPPSNN GSSEDHIEEA 651 DVEIIDNDDK*
(15) Omp85 homolog (Cpn0300)
One example of an Omp85 Homolog protein is disclosed as SEQ ID NOs: 147 & 148 in WO 02/02606. {GenBank accession number: gi|4376576|gb|AAD18449.1| 'CPn0300'; SEQ ID NO: 15 below}. Preferred Omp85 proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 15; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 15, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These DnaK proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 15. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 15. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 15. 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). SEQ ID No 15 1 MLIMRNKVIL QISI A IQT PLTLFSTEKV KEGHWVDSI TIITEGENAS 51 NKHPLPKLKT RSGA FSQLD FDEDLRI1AK EYDSVEPKVE FSEGKTNIA 101 H IAKPSIRN IHISGNQWP EHKILKT QI YRNDLFEREK FLKGLDDLRT 151 YYLKRGYFAS SVDYSLEHNQ EKGHIDVLIK INEGPCGKIK Q TFSGISRS 201 E SDIQEFIQ TKQHSTTTSW FTGAG YHPD IVEQDSLAIT NYLHNNGYAD 251 AIVNSHYDLD DKGNILLYMD IDRGSRYTLG HVHIQGFEV PKRLIEKQSQ 301 VGPNDLYCPD KI DGAHKIK QTYAKYGYIN TNVDVLFIPH ATRPIYDVTY 351 EVSEGSPYKV G IKITGNTH TKSDVILHET SLFPGDTFNR LKLEDTEQRL 401 RNTGYFQSVS VYTVRSQLDP MGNADQYRDI FVEVKETTTG NLGLFLGFSS 451 LDN FGGIEL SESNFDLFGA RNIFSKGFRC RGGGEHLF KANFGDKVTD 501 YT K TKPHF LNTP ILGIE LDKSINRA S KDYAVQTYGG NVSTTYILNE 551 HLKYGLFYRG SQTSLHEKRK FLLGPNIDSN KGFVSAAGV LNYDSVDSPR 601 TPTTGIRGGV TFEVSG GGT YHFTK SLNS SIYRKLTRKG ILKIKGEAQF 651 IKPYSNTTAE GVPVSERFFL GGETTVRGYK SFIIGPKYSA TEPQGGLSSL 701 LISEEFQYPL IRQPNISAFV F DSGFVG Q EYKIS KDLR SSAGFG RFD 751 VMNNVPVMLG FGWPFRPTET LNGEKIDVSQ RFFFALGGMF *
(16) Mip-like FKBP-type peptidyl-prolyl cis-trans (CP11O66I)
One example of a Mip-like protein is disclosed as SEQ ID NOs: 55 & 56 in WO 02/02606. {GenBank accession number: gi|4376960|gb|AAD18800.1| 'CPn0661'; SEQ ID NO: 16 below}. Preferred Mip-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%o or more) to SEQ ID NO: 16; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 16, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These mip- like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 16. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 16. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 16. 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).
SEQ ID No 16 1 I_-S[RRWt. VIA TVALA SVAS CDVRSKDKDK DQGSLVEYKD N DTNDIE S 51 DNQKLSRTFG HLLARQ RKS EDMFFDIAEV AKGLQAE VC KSAPLTETEY 101 EEKMAEVQKL VFEKKSKENL SLAEKFLKEN SKNAGWEVQ PSKLQYKIIK 151 EGAGKAISGK PSALLHYKGS FINGQVFSSS EGNNEPI LP LGQTIPGFA 201 G QGMKEGET RVLYIHPDLA YGTAGQLPPN SLLIFEINLI QASADEVAAV 251 PQEGNQGE*
(17) OmcB 60 kDa Cysteine rich OMP (CPn0557)
One example of an OmcB protein is disclosed as SEQ ID NOs: 47 & 48 in WO 02/02606. {GenBank accession number: gi|4376849|gb|AAD18697.1| ' CPn0557'; SEQ ID NO: 17 below}. Preferred OmcB proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%) or more) to SEQ ID NO: 17; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 17, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmcB proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 17. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 17. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 17. 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) .. SEQ ID No 17 1 MSK IRRWT VLALTS ASC FASGGIEAAV AESLITKIVA SAETKPAPVP 51 MTAKKVRLVR RNKQPVEQKS RGAFCDKEFY PCEEGRCQPV EAQQESCYGR 101 LYSVKVNDDC NVEICQSVPE YATVGSPYPI EI AIGKKDC VDWITQQLP 151 CEAEFVSSDP ETTPTSDGKL VWKIDRLGAG DKCKITVWVK PLKEGCCFTA 201 ATVCACPELR SYTKCGQPAI CIKQEGPDCA CLRCPVCYKI EVVNTGSAIA 251 RNVTVDNPVP DGYSHASGQR V SFN GDMR PGDKKVFTVE FCPQRRGQIT 301 NVATVTYCGG HKCSANVTTV VNEPCVQVNI SGAD SYVCK PVEYSISVSN 351 PGD VLHDW IQDTLPSGVT V EAPGGEIC CNKW RIKE MCPGET QFK 401 LWKAQVPGR FTNQVAVTSE SNCGTCTSCA ETTTHWKGLA ATHMCVLDTN 451 DPICVGENTV YRICVTNRGS AEDTNVSLIL KFSKELQPIA SSGPTKGTIS 501 GNTWFDA P KLGSKESVEF SVTLKGIAPG DARGEAI SS DTLTSPVSDT 551 ENTHVY*
(18) MurG peptidoglycan transferase protein (CPn0904)
One example of a 'MurG' protein is disclosed as SEQ ID NOs: 107 & 108 in WO 02/02606. {GenBank accession number: gi|4377224|gb|AAD19042.1| 'CPn0904'; SEQ ID NO: 18 below}. Preferred MurG proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 18; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 18, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These MurG proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 18. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 18. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 18. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). The MurG may be lipidated e.g. with undecaprenyl. ID No 18 1 MMKKIRKVAL AVGGSGGHIV PALSVKEAFS REGIDVL LG KG KNHPS Q 51 QGISYREIPS GLPTVLNPIK I SRTLSLCS GYLKARKELK IFDPDLVIGF 110011 GSYHSLPV L AGLSHKIP F HEQNLVPGK VNQLFSRYAR GIGVNFSPVT 151 HFRCPAEEV F PKRSFS G SPMMKRCTNH TPTICWGGS QGAQI NTCV 201 PQALVKLVNK YPNMYVHHIV GPKSDVMKVQ HVYNRGEV C CVKPFEEQ L 251 DVLLAADLVI SRAGATI EE I WAKVPGIL IPYPGAYGHQ EVNAKFFVDV 301 LEGGTMILEK ELTEK LVEK VTFALDSHNR EKQRNSLAAY SQQRSTKTFH 335511 AFICEC * (19) CPn0186 (Hypothetical)
One example of a hypothetical protein is set forth as SEQ ID NO: 19 below}. (GenBank Accession No. GL4376456; AAD 18339.1). Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 19; and/or (b) which is a fragment of at least « consecutive amino acids of SEQ ID NO: 19, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 19. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 19. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 19. 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).
SEQ ID No l9 1 MSSPVNNTPS APNIPIPAPT TPGIPTTKPR SSFIEKVIIV AKYI FAIAA TSGALGTILG 61 LSGALTPGIG IALLVIFFVS MVLLGLILKD SISGGEERRL REEVSRFTSE NQRLTVITTT 121 LETEVKDLKA AKDQLTLEIE AFRNENGNLK TTAEDLEEQV ΞKLSEQLEAL ERINQLIQAN 181 AGDAQEISSE LKKLISG DS KWEQINTSI QALKVLLGQE VQEAQTHVK AMQEQIQALQ 241 AEILGMHNQS TALQKSVENL LVQDQALTRV VGELLESENK LSQACSALRQ EIEKLAQHET 301 SLQQRIDAML AQEQNLAEQV TALEKMKQEA QKAESEFIAC VRDRTFGRRE TPPPTTPWE 361 GDESQEEDEG GTPPVSQPSS PVDRATGDGQ
(20) FUY Glutamine Binding Protein (CPn0604) One example of a hypothetical protein is set forth as SEQ ID NOs: 11 & 12 in WO 02/02606. {GenBank accession number: gi|4376900|gb|AAD18743.1| 'CPn0604'; SEQ ID NO: 20 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%) or more) to SEQ ID NO: 20; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 20, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 20. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 20. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 20. 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).
SEQID No 20 1 MKIKFS KVN FLICLLAVGL IFFGCSRVKR EVLVGRDAT FPKQFGIYTS 51 DTNAFLNDLV SEINYKENLN INIVNQD VH LFENLDDKKT QGAFTSVLPT 101 LEMLEHYQFS DPILLTGPVL VVAQDSPYQS IEDLKGRLIG VYKFDSSVLV 151 AQNIPDAVIS LYQHVPIALE ALTSNCYDAL LAPVIEVTAL IETAYKGRLK 201 IISKPLNADG LRLAILKGTN GDLLEGFNAG LVKTRRSGKY DAIKQRYRLP The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group. Such other Chlamydia pneumoniae antigens include a fourth antigen group consisting one or more members of the PMP family. These antigens are referred to herein as the "fourth antigen group". Each of the Chlamydia pneumoniae antigens of the fourth antigen group is described in more detail below. Fourth Antigen Group
(21) Polymorphic Membrane Proteins (PMP)
A family of twenty one Chlamydia pneumoniae genes encoding predicted polymorphic membrane proteins (PMP) have been identified (pmpl to pmp2I).
Pmpl (CPn0005)
One example of a Pmpl protein is set forth as SEQ ID NOs: 41 & 42 in WO 02/02606. {GenBank accession number: gi|4376260|gb|AAD18163.1 'CPn0005'; SEQ ID NO: 21 below}.
SEQ ID No 21 1 MRFSLCGFBL VFSFTLLSVF DTSLSATTIS LTPEDSFHGD SQNAERSYNV 51 QAGDVYSLTG DVSISNVDNS ALNKACFNVT SGSVTFAGNH HGLYFNNISS 110011 GTTKEGAVLC CQDPQATARF SGFSTLSFIQ SPGDIKEQGC LYS NALMLL 151 NNYWRFEQN QSKTKGGAIS GANVTIVGNY DSVSFYQNAA TFGGAIHSSG 201 PLQIAVNQAE IRFAQNTAKN GSGGALYSDG DIDIDQNAYV LFRENEALTT 251 AIGKGGAVCC LPTSGSSTPV PIVTFSDNKQ LVFERNHSIM GGGAIYARKL 301 SISSGGPTLF INNISYANSQ NLGGAIAΓDT GGEISLSAEK GTITFQGNRT 335511 SLPFLNGIHL LQNAKFLKLQ ARNGYSIEFY DPITSEADGS TQLNINGDPK 401 NKEYTGTILF SGEKSLANDP RDFKSTIPQN VNLSAGYLVI KEGAEVTVSK 451 FTQSPGSHLV LDLGTKLIAS KEDIAITGLA IDIDSLSSSS TAAVIKANTA 501 NKQISVTDSI ELISPTGNAY EDLRMRNSQT FPLLSLEPGA GGSVTVTAGD 551 FLPVSPHYGF QGN KLA TG TGNKVGEFF DKINYKPRPE KEGNLVPNIL 660011 GNAVDVRSL MQVQETHASS LQTDRGL ID GIGNFFHVSA SEDNIRYRHN 651 SGGYVLSVNN EITPKHYTSM AFSQLFSRDK DYAVSNNEYR MYLGSYLYQY 701 TTSLGNIFRY ASRNPNVNVG ILSRRFLQNP LMIFHFLCAY GHATNDMKTD 751 YANFPMVKNS WRNNC AIEC GGSMPLLVFE NGRLFQGAIP FMKLQLVYAY 801 QGDFKETTAD GRRFSNGSLT SISVPLGIRF EKLALSQDVL YDFSFSYIPD 885511 IFRKDPSCEA ALVISGDSWL VPAAHVSRHA FVGSGTGRYH FNDYTELLCR 901 GSIECRPHAR NYNINCGSKF RF*
Pmp 4 (CPn0017) One example of a Pmp 4 protein is designated SEQ ID NO: 22. The sequence for pmp4 protein can be found at AE001587.1 GI.4376271.
Pmp 6 (CPn0444)
One example of a Pmp 6 protein is set forth as SEQ ID NOs 31 & 32 in WO 02/02606. {GenBank accession number: gi|4376727|gb|AAD18588.1| 'CPn0444'; SEQ ID NO: 23 below}.
SEQ ID No 23 1 MKYSLPWLLT SSALVFSLHP LMAANTDLSS SDNYENGSSG SAAFTAKETS 51 DASGTTYTLT SDVSITNVSA ITPADKSCFT NTGGALSFVG ADHSLVLQTI 101 ALTHDGAAIN NTNTALSFSG FSSLLIDSAP ATGTSGGKGA ICVTNTEGGT 151 ATFTDNASVT LQKNTSEKDG AAVSAYSIDL AKTTTAALLD QNTSTKNGGA 201 LCSTANTTVQ GNSGTVTFSS NTATDKGGGI YSKEKDSTLD ANTGVVTFKS 251 NTAKTGGA S SDDNLALTGN TQVLFQENKT TGSAAQANNP EGCGGAICCY 301 LATATDKTGL AISQNQEMSF TSNTTTANGG AIYATKCTLD GNTTLTFDQN 351 TATAGCGGAI YTETEDFSLK GSTGTVTFST NTAKTGGALY SKGNSSLTGN 401 TNLLFSGNKA TGPSNSSA Q EGCGGAILAF IDSGSVSDKT GLSIANNQEV 451 S TSNAATVS GGAIYATKCT LTGNGSLTFD GNTAGTSGGA IYTETEDFTL 501 TGSTGTVTFS TNTAKTGGAL YSKGNNSLSG NTNLLFSGNK ATGPSNSSAN 551 QEGCGGAILS FLESASVSTK KGL IEDNEN VSLSGMTATV SGGAIYATKC 601 ALHGNTTLTF DGNTAETAGG AIYTETEDFT LTGSTGTVTF STNTAKTAGA 651 LHTKGNTSFT KNKALVFSGN SATATATTTT DQEGCGGAIL CNISESDIAT 701 KSLTLTENES LSFINNTAKR SGGGIYAPKC VISGSESINF DGNTAETSGG 751 AIYSKNLSIT ANGPVSFTNN SGGKGGAIYI ADSGELSLEA IDGDITFSGN 801 RATEGTSTPN SIHLGAGAKI TKLAAAPGHT IYFYDPITME APASGGTIEE 851 LVINPVVKAI VPPPQPKNGP IASVPWPVA PANPNTGTIV FSSGKLPSQD 901 ASIPANTTTI LNQKINLAGG NWLKEGATL QVYSFTQQPD STVFMDAGTT 951 LETTTTNNTD GSIDLKNLSV NLDALDGKRM IT AVNSTSG GLKISGDLKF 1001 HNNEGSFYDN PGLKANLNLP FLDLSSTSGT V LDDFNPIP SSMAAPDYGY 1051 QGSWTLVPKV GAGGKVTLVA EWQALGYTPK PELRATLVPN SL NAYVNIH 1101 S1QQEIATAM SDAPSHPGI IGGIGNAFHQ DKQKENAGFR LISRGYIVGG 1151 SMTTPQEYTF AVAFSQLFGK SKDYWSDIK SQVYAGSLCA QSSYVIPLHS 1201 SLRRHVLSKV LPELPGETPL VLHGQVSYGR NHHN TTKLA NNTQGKSD D 1251 SHSFAVEVGG SLPVDLNYRY LTSYSPYVKL QWSVNQKGF QEVAADPRIF 1301 DASHLVNVSI PMGLTFKHES AKPPSALLLT LGYAVDAYRD HPHCLTSLTN 1351 GTS STFATN LSRQAFFAEA SGHLKLLHGL DCFASGSCEL RSSSRSYNAN 1401 CGTRYSF*
Pmp 7 (CPn0445)
One example of a Pmp 7 protein is set forth as SEQ ID NOs 153 & 154 in WO 02/02606. {GenBank accession number: gi|4376728|gb|AAD 18589.11 'CPn0445'; SEQ ID NO: 24 below}. SEQ ID No 24 1 MKSSVSWLFF SSIPLFSSLS IVAAEVTLDS SNNSYDGSNG TTFTVFSTTD I 51 AAAGTTYSLL SDVSFQNAGA LGIPLASGCF LEAGGDLTFQ GNQHALKFAF 101 INAGSSAGTV ASTSAADKNL LFNDFSRLSI ISCPSLLLSP TGQCALKSVG 151 NLSLTGNSQI IFTQNFSSDN GGVINTKNFL LSGTSQFASF SRNQAFTGKQ 201 GGWYATGTI TIENSPGIVS FSQNLAKGSG GALYSTDNCS ITDNFQVIFD 251 GNSA EAAQA QGGAICCTTT DKTVTLTGNK NLSFTNNTAL TYGGAISGLK 301 VSISAGGPTL FQSNISGSSA GQGGGGAINI ASAGELALSA TSGDITFNNN 351 QVTNGSTSTR NAINIIDTAK VTSIRAATGQ SIYFYDPITN PGTAASTDTL 401 NLNLADANSE IEYGGAIVFS GEKLSPTEKA IAANVTSTIR QPAVLARGDL 451 VLRDGVTVTF KDLTQSPGSR ILMDGGTTLS AKEANLSLNG LAVNLSSLDG 501 TNKAALKTEA ADKNISLSGT IALIDTEGSF YENHNLKSAS TYPLLELTTA 551 GANGTITLGA LSTLTLQEPE THYGYQGN Q LS ANATSSK IGSINWTRTG 601 YIPSPERKSN LPLNSLWGNF IDIRSINQLI ETKSSGEPFE REL LSGIAN 651 FFYRDSMPTR HGFRHISGGY ALGITATTPA EDQLTFAFCQ LFARDRNH1T 701 GKNHGDTYGA SLYFHHTEGL FDIANFL GK ATRAP VLSE 1SQIIPLSFD 751 AKFSYLHTDN HMKTYYTDNS IIKGS RNDA FCADLGASLP FVISVPYLLK 801 EVEPFVKVQY IYAHQQDFYE RHAEGRAFNK SELINVEIPI GVTFERDSKS 851 EKGTYDLTLM YILDAYRRNP KCQTSLIASD ANWMAYGTNL ARQGFSVRAA 901 NHFQVNPHME IFGQFAFEVR SSSRNYNTNL GSKFCF*
Pmp 8 (CPn0446)
One example of a Pmp 8 protein is set forth as SEQ ID NOs 45 & 46 in WO 02/02606. {GenBank accession number: gi|4376729|gb|AAD 18590.1] 'CPn0446'; SEQ ID NO: 25 below}. SEQ ID No 25 1 MKIPLHKLLI SSTLVTPILL SIATYGADAS LSPTDSFDGA GGSTFTPKST 51 ADANGTNYVL SGNVYINDAG KGTALTGCCF TETTGDLTFT GKGYSFSFNT 101 VDAGSNAGAA ASTTADKALT FTGFSNLSFI AAPGTTVASG KSTLSSAGAL 151 NLTDNGTILF SQNVSNEANN NGGAITT TL SISGNTSSIT FTSNSAKKLG 201 GAIYSSAAAS ISGNTGQLVF MNNKGETGGG ALGFEASSSI TQNSSLFFSG 251 NTATDAAGKG GAIYCEKTGE TPTLTISGNK SLTFAENSSV TQGGAICAHG 301 LDLSAAGPTL FSNNRCGNTA AGKGGAIAIA DSGSLSLSAN QGDITFLGNT 351 LTSTSAPTST RNAIYLGSSA KITNLRAAQG QSIYFYDPIA SNTTGASDVL 401 TINQPDSNSP LDYSGTIVFS GEKLSADEAK AADNFTSILK QPLALASGTL 451 ALKGNVELDV NGFTQTEGST LLMQPGTKLK ADTEAISLTK LVVDLSALEG 501 NKSVSIETAG ANKTITLTSP LVFQDSSGNF YESHTINQAF TQPLVVFTAA 551 TAASDIYIDA LLTSPVQTPE PHYGYQGH E AT ADTSTAK SGTMTWVTTG 601 YNPNPERRAS WPDSLWASF TDIRTLQQIM TSQANSIYQQ RGLWASGTAN 651 FFHKDKSGTN QAFRHKSYGY IVGGSAEDFS ENIFSVAFCQ LFGKDKDLFI 701 VENTSHNYLA SLYLQHRAFL GGLPMPSFGS ITDMLKDIPL ILNAQLSYSY 751 TKNDMDTRYT SYPEAQGS T NNSGALELGG SLALYLPKEA PFFQGYFPFL 801 KFQAVYSRQQ NFKESGAEAR AFDDGDLVNC SIPVGIRLEK ISEDEKNNFE 851 ISLAYIGDVY RKNPRSRTSL MVSGAS TSL CKNLARQAFL ASAGSHLTLS 901 PHVELSGEAA YELRGSAHIY NVDCGLRYSF *
Pmp 9 (CPn0447)
One example of a Pmp 9 protein is set forth as SEQ ID NOs 33 & 34 in WO 02/02606. {GenBank accession number: gi|4376731|gb|AAD18591.1| 'CPn0447'; SEQ ID NO: 26 below}.
SEQID No 26 1 M SSLHWFLI SSSLALPLSL NFSAFAAWE INLGPTNSFS GPGTYTPPAQ 51 TTNADGTIYN LTGDVSITNA GSPTALTASC FKETTGNLSF QGHGYQFLLQ 101 NIDAGANCTF TNTAANKLLS FSGFSYLSLI QTTNATTGTG AIKSTGACSI 151 QSNYSCYFGQ NFSNDNGGAL QGSSISLSLN PNLTFAKNKA TQKGGALYST 201 GGITINNTLN SASFSENTAA NNGGAIYTEA SSFISSNKAI SFINNSVTAT 251 SATGGAIYGS STSAPKPVLT LSDNGELNFI GNTAITSGGA IYTDNLVLSS 301 GGPTLFKNNS AIDTAAPLGG AIAIADSGSL SLSALGGDIT FEGNTWKGA 351 SSSQTTTRNS INIGNTNAKI VQLRASQGNT IYFYDPITTS ITAALSDALN 401 LNGPDLAGNP AYQGTIVFSG EKLSEAEAAE ADNLKSTIQQ PLTLAGGQLS 451 LKSGVTLVAK SFSQSPGSTL LMDAGTTLET ADGITINNLV LNVDSLKETK 501 KATLKATQAS QTVTLSGSLS LVDPSGNVYE DVSWNNPQVF SCLTLTADDP 551 ANIHITDLAA DPLEKNPIHW GYQGN ALS QEDTATKSKA ATLT TKTGY 601 NPNPERRGTL VANTL GSFV DVRSIQQLVA TKVRQSQETR GIWCEGISNF 651 FHKDSTKINK GFRHISAGYV VGATTTLASD NLITAAFCQL FGKDRDHFIN 701 KNRASAYAAS LHLQHLATLS SPSLLRYLPG SESEQPVLFD AQISYIYSKN 751 T KTYYTQAP KGESSWYNDG CALELASSLP HTALSHEGLF HAYFPFIKVE 801 ASYIHQDSFK ERNTTLVRSF DSGDLINVSV PIGITFERFS RNERASYEAT 851 VIYVADVYRK NPDCTTALLI NNTSWKTTGT NLSRQAGIGR AGIFYAFSPN 901 LEVTSNLSME IRGSSRSYNA DLGGKFQF* Pmp 11 (CPn0451)
One example of a Pmp 11 protein is set forth as SEQ ID NOs 115 & 116 in WO 02/02606. {GenBank accession number: gi|4376733|gb|AAD18593.1| 'CPn0451';
SEQ ID NO: 27 below). SEQ ID No 27 1 MKTSIPWVTΛ. SSVIAFSCHL QSLANEELLS PDDSFNGNID SGTFTPKTSA 51 TTYSLTGDVF FYEPGKGTPL SDSCFKQTTD NLTFLGNGHS LTFGFIDAGT 101 HAGAAASTTA NKNLTFSGFS LLSFDSSPST TVTTGQGTLS SAGGVNLENI 151 RKLWAGNFS TADGGAIKGA SFLLTGTSGD ALFSNNSSST KGGAIATTAG 201 ARIANNTGYV RFLSNIASTS GGAIDDEGTS ILSNNKFLYF EGNAAKTTGG 251 A1CNTKASGS PELIISNNKT LIFASNVAET SGGAIHAKKL ALSSGGFTEF 301 LRNNVSSATP KGGAISIDAS GELSLSAETG NITFVRNTLT TTGSTDTPKR 351 NAINIGSNGK FTELRAAKNH TIFFYDPITS EGTSSDVLKI NNGSAGALNP 401 YQGTILFSGE TLTADELKVA DNLKSSFTQP VSLSGGKLLL QKGVTLESTS 451 FSQEAGSLLG DSGTTLSTT AGSITITNLG INVDSLGLKQ PVSLTAKGAS 501 NKVIVSGKLN LIDIEGNIYE SHMFSHDQLF SLLKITVDAD VDTNVDISSL 551 IPVPAEDPNS EYGFQGQ NV NWTTDTATNT KEATAT TKT GFVPSPERKS 601 ALVCNTLWGV FTDIRSLQQL VEIGATGMEH KQGFWVSSMT NFLHKTGDEN 651 RKGFRHTSGG YVIGGSAHTP KDDLFTFAFC HLFARDKDCF IAHNNSRTYG 701 GTLFFKHSHT LQPQNYLRLG RAKFSESA1E KFPREIPLAL DVQVSFSHSD 751 NRMETHYTSL PESEGS SNE ClAGGIGLDL PFVLSNPHPL FKTFIPQMKV 801 EMVYVSQNSF FESSSDGRGF S1GRLLNLSI PVGAKFVQGD IGDSYTYDLS 851 GFFVSDVYRN NPQSTATLVM SPDS KIRGG NLSRQAFLLR GSNNYVYNSN 901 CELFGHYAME LRGSSRNYNV DVGTKLRF* Pmp 12 (CPn0452)
One example of a Pmp 12 protein is set forth as SEQ ID NOs 51 & 52 in WO 02/02606. {GenBank accession number: gi|4376735|gb|AAD18594.1 'CPn0452'; SEQ ID NO: 28 below). SEQ ID No 28 1 MTILRNFLTC SALFLALPAA AQWYLHESD GYNGAINNKS LEPKITCYPE 51 GTSYIFLDDV RISNVKHDQE DAGVFINRSG NLFFMGNRCN FTFHNLMTEG 101 FGAAISNRVG DTTLTLSNFS YLAFTSAPLL PQGQGAIYSL GSVMIENSEE 151 VTFCGNYSS SGAAIYTPYL LGSKASRPSV NLSGNRYLVF RDNVSQGYGG 201 AISTHNLTLT TRGPSCFENN HAYHDVNSNG GAIAIAPGGS ISISVKSGDL 251 IFKGNTASQD GNTIHNSIHL QSGAQFKNLR AVSESGVYFY DPISHSESHK 301 ITDLVINAPE GKETYEGTIS FSGLCLDDHE VCAENLTSTI LQDVTLAGGT 351 LSLSDGVTLQ LHSFKQEASS TLTMSPGTTL LCSGDARVQN LHILIEDTDN 401 FVPVRIRAED DALVSLEKL KVAFEAYWSV YDFPQFKEAF TIPLLELLGP 451 SFDSLLLGET TLERTQVTTE NDAVRGF SL SWEEYPPSLD KDRRITPTKK 501 TVFLTWNPEI TSTP*
Pmp 13 (CPn0453) One example of a Pmp 13 protein is set forth as SEQ ID NOs 3 & 4 in WO 02/02606. {GenBank accession number: gi|4376736|gb|AAD18595.1 'CPn0453'; SEQ ID NO: 29 below}.
SEQ ID No 29 1 MKTSIRKFLI STTLAPCFAS TAFTVEVIMP SENFDGSSGK IFPYTTLSDP 51 RGTLCIFSGD LYIANLDNAI SRTSSSCFSN RAGALQILGK GGVFSFLNIR 101 SSADGAAISS VITQNPELCP LSFSGFSQMI FDNCESLTSD TSASNVIPHA 151 SAIYATTPML FTNNDSILFQ YNRSAGFGAA IRGTSITIEN TKKSLLFNGN 201 GSISNGGALT GSAAINLINN SAPVIFSTNA TGIYGGAIYL TGGSMLTSGN 251 LSGVLFVNNS SRSGGAIYAN GNVTFSNNSD LTFQNNTASP QNSLPAPTPP 301 PTPPAVTPLL GYGGAIFCTP PATPPPTGVS LTISGENSVT FLENIASEQG 351 GALYGKKISI DSNKSTIFLG NTAGKGGAIA IPESGELSLS ANQGDILFNK 401 NLSITSGTPT RNSIHFGKDA KFATLGATQG YTLYFYDPIT SDDLSAASAA 451 ATWVNPKAS ADGAYSGTIV FSGETLTATE AATPANATST LNQKLELEGG 501 TLALRNGATL NVHNFTQDEK SWIMDAGTT LATTNGANNT DGAITLNKLV 551 INLDSLDGTK AAWNVQSTN GALTISGTLG LVKNSQDCCD NHGMFNKDLQ 601 QVPILELKAT SNTVTTTDFS LGTNGYQQSP YGYQGTWEFT IDTTTHTVTG 651 NWKKTGYLPH PERLAPLIPN SL ANVIDLR AVSQASAADG EDVPGKQLSI 701 TGITNFFHAN HTGDARSYRH MGGGYLINTY TRITPDAALS LGFGQLFTKS 751 KDYLVGHGHS NVYFATVYSN ITKSLFGSSR FFSGGTSRVT YSRSNEKVKT 801 SYTKLPKGRC SWSNNCWLGE LEGNLPITLS SRILNLKQII PFVKAEVAYA 851 THGGIQENTP EGRIFGHGHL LNVAVPVGVR FGKNSHNRPD FYTIIVAYAP 901 DVYRHNPDCD TTLPINGAT TSIGNNLTRS TLLVQASSHT SVNDVLEIFG 951 HCGCDIRRTS RQYTLDIGSK LRF*
Pmp 14 (CPn0454) One example of a Pmp 14 protein is set forth as SEQ ID NOs 35 & 36 in WO 02/02606. {GenBank accession number: gi]4376737|gb|AAD 18596.1 'CPn0454'; SEQ ID NO: 30 below}. SEQID No 30 1 MELSFKSSSF CLLACLCSAS CAFAETRLGG NFVPPITNQG EEILLTSDFV 51 CSNFLGASFS SSFINSSSNL SLLGKGLSLT FTSCQAPTNS NYALLSAAET 101 LTFKNFSSIN FTGNQSTGLG GLIYGKDIVF QSIKDLIFTT NRVAYSPASV 151 TTSATPAITT VTTGASALQP TDSLTVENIS QSIKFFGNLA NFGSAISSSP 201 TAVVKFINNT ATMSFSHNFT SSGGGVIYGG SSLLFENNSG CIIFTANSCV 251 NSLKGVTPSS GTYALGSGGA ICIPTGTFEL KNNQGKCTFS YNGTPNDAGA 301 IYAETCNIVG NQGALLLDSN TAARNGGAIC AKVLNIQGRG PIEFSRNRAE 351 KGGAIFIGPS VGDPAKQTST LTILASEGDI AFQGN LNTK PGIRNAITVE 401 AGGEIVSLSA QGGSRLVFYD PITHSLPTTS PSNKDITINA NGASGSWFT 451 SKGLSSTELL LPANTTTILL GTVKIASGEL KITDNAW V LGFATQGSGQ 501 LTLGSGGTLG LATPTGAPAA VDFTIGKLAF DPFSFLKRDF VSASVNAGTK 551 NVTLTGALVL DEHDVTDLYD MVSLQTPVAI PIAVFKGATV TKTGFPDGEI 601 ATPSHYGYQG KWSYTWSRPL LIPAPDGGFP GGPΞPSANTL YAV NSDTLV 651 RSTYILDPER YGEIVSNSLW ISFLGNQAFS DILQDVLLID HPGLSITAKA 701 LGAYVEHTPR QGHEGFSGRY GGYQAALS N YTDHTTLGLS FGQLYGKTNA 751 NPYDSRCSEQ MYLLSFFGQF PIVTQKSEAL IS KAAYGYS KNHLNTTYLR 801 PDKAPKSQGQ HNNSYYVLI SAEHPFLNWC LLTRPLAQA DLSGFISAEF 851 LGGWQSKFTE TGDLQRSFSR GKGYNVSLPI GCSSQWFTPF KKAPSTLTIK 901 LAYKPDIYRV NPHNIVTWS NQESTSISGA NLRRHGLFVQ IHDWDLTED 951 TQAFLNYTFD GKNGFTNHRV STGLKSTF*
Pmp 15 (CPn0466)
One example of a Pmp 15 protein is set forth as SEQ ID NOs 5 & 6 in WO 02/02606. {GenBank accession number: gi|4376751|gb|AAD18608.1 'CPn0466'; SEQ ID NO:
31 below}.
SEQ ID No 31 1 MRFFCFGM L PFTFVLANEG LQLPLETYIT LSPEYQAAPQ VGFTHNQNQD 51 LAIVGNHNDF ILDYKYYRSN GGALTCKNLL ISENIGNVFF EKNVCPNSGG 101 AIYAAQNCTI SKNQNYAFTT NLVSDNPTAT AGSLLGGALF AINCSITNNL 151 GQGTFVDNLA LNKGGALYTE TNLSIKDNKG PIIIKQNRAL NSDSLGGGIY 201 SGNSLNIEGN SGAIQITSNS SGSGGGIFST QTLTISSNKK LIEISENSAF 251 ANNYGSNFNP GGGGLTTTFC TILNNREGVL FNNNQSQSNG GAIHAKSIII 301 KENGPVYFLN NTATRGGALL NLSAGSGNGS FILSADNGDI IFNNNTASKH 351 ALNPPYRNAI HSTPNMNLQI GARPGYRVLF YDPIEHELPS SFPILFNFET 401 GHTGTVLFSG EHVHQNFTDE MNFFSYLRNT SELRQGVLAV EDGAGLACYK 451 FFQRGGTLLL GQGAVITTAG TIPTPSSTPT TVGSTITLNH IAIDLPSILS 501 FQAQAPKI I YPTKTGSTYT EDSNPTITIS GTLTLRNSNN EDPYDSLDLS 551 HSLEKVPLLY IVDVAAQKIN SSQLDLSTLN SGEHYGYQGI WSTYWVETTT 601 ITNPTSLLGA NTKHKLLYAN WSPLGYRPHP ERRGEFITNA LWQSAYTALA 651 GLHSLSSWDE EKGHAASLQG IGLLVHQKDK NGFKGFRSHM TGYSATTEAT 701 SSQSPNFSLG FAQFFSKAKE HESQNSTSSH HYFSGMCIEN TLFKEWIRLS 751 VSLAYMFTSE HTHTMYQGLL EGNSQGSFHN HTLAGALSCV FLPQPHGESL 801 QIYPFITALA IRGNLAAFQE SGDHAREFSL HRPLTDVSLP VGIRASWKNH 851 HRVPLV LTE ISYRSTLYRQ DPELHSKLLI SQGTWTTQAT PVTYNALGIK 901 VKNTMQVFPK VTLSLDYSAD ISSSTLSHYL NVASRMRF* Pmp 16 (CPn0467)
One example of a Pmp 16 protein is set forth as SEQ ID NOs 7 & 8 in WO 02/02606. {GenBank accession number: gi|4376752|gb|AAD18609.1| 'CPn0467'; SEQ ID NO:
32 below}. SEQ ID No 32 1 MFGMTPAVYS LQTDSLEKFA LERDEEFRTS FPLLDSLSTL TGFSPITTFV 51 GNRHNSSQDI VLSNYKSIDN ILLLWTSAGG AVSCNNFLLS NVEDHAFFSK 101 NLAIGTGGAI ACQGACTITK NRGPLIFFSN RGLNNASTGG ETRGGAIACN 151 GDFTISQNQG TFYFVNNSVN N GGALSTNG HCRIQSNRAP LLFFNNTAPS 201 GGGALRSENT TISDNTRPIY FKNNCGNNGG AIQTSVTVAI KNNSGSVIFN 251 NNTALSGSIN SGNGSGGAIY TTNLSIDDNP GTILFNNNYC IRDGGAICTQ 301 FLTIKNSGHV YFTNNQGN G GALMLLQDST CLLFAEQGNI AFQNNEVFLT 351 TFGRYNAIHC TPNSNLQLGA NKGYTTAFFD PIEHQHPTTN PLIFNPNANH 401 QGTILFSSAY IPEASDYENN FISSSKNTSE LRNGVLSIED RAGWQFYKFT 451 QKGGILKLGH AASIATTANS ETPSTSVGSQ VIINNLAINL PSILAKGKAP 501 TLWIRPLQSS APFTEDNNPT ITLSGPLT1L NEENRDPYDS IDLSEPLQNI 551 HLLSLSDVTA RHINTDNFHP ESLNATEHYG YQGIWSPY V ETITTTNNAS 601 IETANTLYRA LYANWTPLGY KVNPEYQGDL ATTPLWQSFH TMFSLLRSYN 651 RTGDSDIERP FLEIQGIADG LFVHQNSIPG APGFRIQSTG YSLQASSETS 701 LHQKISLGFA QFFTRTKEIG SSNNVSAHNT VSSLYVELP FQEAFATSTV 751 LAYGYGDHHL HSLHPSHQEQ AEGTCYSHTL AAAIGCSFP QQKSYLHLSP 801 FVQAIAIRSH QTAFEEIGDN PRKFVSQKPF YNLTLPLGIQ GK QSKFHVP 851 TE TLELSYQ PVLYQQNPQI GVTLLASGGS DILGHNYVR NALGYKVHNQ 901 TALFRSLDLF LDYQGSVSSS TSTHHLQAGS TLKF*
Pmp 18 (CPn0471)
One example of a Pmp 18 protein is set forth as SEQ ID No 33 below {GenBank accession number: gi|4376753|gb|AAD18610.1| 'CPn0471'. SEQ ID No 33 1 MQNNRSLSKS SFFVGALILG KTTILLNATP LSDYFDNQAN QLTTLFPLID TLTNMTPYSH 61 RATLFGVRDD TNQDIVLDHQ NSIES FENF SQDGGALSCK SLAITNTKNQ ILFLNSFAIK 121 RAGAMYVNGN FDLSENHGSI IFSGNLSFPN ASNFADTCTG GAVLCSKNVT ISKNQGTAYF 181 INNKAKSSGG AIQAAIINIK DNTGPCLFFN NAAGGTAGGA LFANACRIEN NSQPIYFLNN 241 QSGLGGAIRV HQECILTKNT GSVIFNNNFA MEADISANHS SGGAIYCISC SIKDNPGIAA 301 FDNNTAARDG GAICTQSLTI QDSGPVYFTN NQGTWGGAIM LRQDGACTLF ADQGDIIFYN 361 NRHFKDTFSN HVSVNCTRNV SLTVGASQGH SATFYDPILQ RYTIQNSIQK FNPNPEHLGT 421 ILFSSTYIPD TSTSRDDFIS HFRNHIGLYN GTLALEDRAE WKVYKFDQFG GTLRLGSRAV 481 FSTTDEEQSS SSVGSVININ NLAINLPSIL GNRVAPKLWI RPTGSSAPYS EDNNPIINLS 541 GPLSLLDDEN LDPYDTADLA QPIAEVPLLY LLDVTAKHIN TDNFYPEGLN TTQHYGYQGV 601 WSPYWIETIT TSDTSSEDTV NTLHRQLYGD TPTGYKVNP ENKGDIALSA F QSFHNLFA 661 TLRYQTQQGQ IAPTASGEAT RLFVHQNSNN DAKGFHMEAT GYSLGTTSNT ASNHSFGVNF 721 SQLFSNLYES HSDNSVASHT TTVALQINNP WLQERFSTSA SLAYSYSNHH IKASGYSGKI 781 QTEGKCYSTT LGAALSCSLS LQ RSRPLHF TPFIQAIAVR SNQTAFQESG DKARKFSVHK 841 PLYNLTVPLG IQSA ESKFR LPTYWNIELA YQPVLYQQNP EINVSLESSG SSWLLSGTTL 901 ARNAIAFKGR NQIFIFPKLS VFLDYQGSVS SSTTTHYLHA GTTFKF
Pmp 19 (CPn0539) One example of a Pmp 19 protein is set forth as SEQ ID No 34 below {GenBank accession number: gi|4376829|gb|AADl 8679.1 'CPn0539'; SEQ ID NO: 34 below}.
SEQ ID No 34 1 MKQMRLWGFL FLSSFCQVSY LRANDVLLPL SGIHSGEDLE -LFTLRSSSPT KTTYSLRKDF 61 IVCDFAGNSI HKPGAAFLNL KGDLFFINST PLAALTFKNI HLGARGAGLF SESNVTFKGL 121 HSLVLENNES WGGVLTTSGD LSFINNTSVL CQNNISYGPG GALLLQGRKS KALFFRDNRG 181 TILFLKNKAV NQDESHPGYG GAVSSISPGS PITFADNQEI LFQENEGELG GAIYNDQGAI 241 TFENNFQTTS FFSNKASFGG AVYSRYCNLY SQWGDTLFTK NAAAKVGGAI HADYVHIRDC 301 KGSIVFEENS ATAGGAIAVN AVCDINAQGP VRFINNSALG LNGGAIYMQA TGSILRLHA 361 QGDIEFCGNK VRSQFHSHIN STSNFTNNAI TIQGAPREFS LSANEGHRIC FYDPIISATE 421 NYNSLYINHQ RLLEAGGAVI FSGARLSPEH KKENKNKTSI INQPVRLCSG VLSIEGGAIL 481 AVRSFYQEGG LLALGPGSKL TTQGKNSEKD KIVITNLGFN LENLDSSDPA EIRATEKASI 541 EISGVPRVYG HTESFYENHE YASKPYTTSI ILSAK LVTA PSRPEKDIQN LIIAESEYMG 601 YGYQGS EFS SPNDTKEKK TIIASWTPTG EFSLDPKRRG SFIPTTL ST FSGLNIASNI 661 VNNNYLNNSE VIPLQHLCVF GGPVYQIMEQ NPKQSSNNLL VQHAGHNVGA RIPFSFNTIL 721 SAALTQLFSS SSQQNVADKS HAQILIGTVS LNKSWQALSL RSSFSYTEDS QVMKHVFPYK 781 GTSRGSWRNY G SGSVGMSY AYPKGIRYLK MTPFVDLQYT KLVQNPFVET GYDPRYFSSS 841 EMTNLSLPIG IALEMRFIGS RSSLFLQVST SYIKDLRRVN PQSSASLVLN HYT DIQGVP 901 LGKEALNITL NST1KYKIVT AY GISSTQR EGSNLSANAH AGLSLSF
As the Examples demonstrate, we and others have demonstrated (Grimwood et al (2001), Infection and Immunity 69(4), 2383-2389) using Flow cytometry (FACS) analyses and Western Blot analyses that PMP 19 does not appear to be surface exposed. However, high levels of mRNA expression is nevertheless observed in gene microarray analysis of pmp 19 (CPn0539). Pmp 20 (CPn0540)
One example of a Pmp 20 protein is set forth as SEQ ID NOs 119 & 120 in WO 02/02606. {GenBank accession number: gi|4376830|gb|AAD18680.1 'CPn0540'; SEQ ID NO: 35 below}. SEQID No 35 1 MKWLPATAVF AAVLPALTAF GDPASVEIST SHTGSGDPTS DAALTGFTQS 51 STETDGTTYT IVGDITFSTF TNIPVPWTP DANDSSSNSS KGGSSSSGAT 101 SLIRSSNLHS DFDFTKDSVL DLYHLFFPSA SNTLNPALLS SSSSGGSSSS 151 SSSSSSGSAS AWAADPKGG AAFYSNEANG TLTFTTDSGN PGSLTLQNLK 201 MTGDGAAIYS KGPLVFTGBK NLTFTGNESQ KSGGAAYTEG ALTTQAIVEA 251 VTFTGNTSAG QGGAIYVKEA TLFNALDSLK FEKNTSGQAG GGIYTESTLT 301 ISNITKSIEF ISNKASVPAP APEPTSPAPS SLINSTTIDT STLQTRAASA 351 TPAVAPVAAV TPTPISTQET AGNGGAIYAK QGISISTFKD LTFKSNSASV 401 DATLTVDSST IGESGGAIFA ADSIQIQQCT GTTLFSGNTA NKSGGGIYAV 451 GQVTLEDIAN LKMTNNTCKG EGGAIYTKKA LTINNGAILT TFSGNTSTDN 501 GGAIFAVGGI TLSDLVEVRF SKNKTGNYSA PITKAASNTA PVVSSSTTAA 551 SPAVPAAAAA PVTNAAKGGA LYSTEGLTVS GITS1LSFEN NECQNQGGGA 601 YVTKTFQCSD SHRLQFTSNK AADEGGGLYC GDDVTLTNLT GKTLFQENSS 651 EKHGGGLSLA SGKSLTMTSL ESFCLNANTA KENGGGANVP ENIVLTFTYT 701 PTPNEPAPVQ QPVYGEALVT GNTATKSGGG IYTKNAAFSN LSSVTFDQNT 751 SSENGGALLT QKAADKTDCS FTYITNVNIT NNTATGNGGG IAGGKAHFDR 801 IDNLTVQSNQ AKKGGGVYLE DALILEKVIT GSVSQNTATE SGGGIYAKDI 851 QLQALPGSFT ITDNKVETSL TTSTNLYGGG IYSSGAVTLT NISGTFGITG 901 NSVINTATSQ DADIQGGGIY ATTSLSINQC NTPILFSNNS AATKKTSTTK 951 QIAGGAIFSA AVTIENNSQP IIFLNNSAKS EATTAATAGN KDSCGGAIAA 1001 NSVTLTNNPE ITFKGNYAET GGAIGCIDLT NGSPPRKVSI ADNGSVLFQD 1051 NSALNRGGAI YGETIDISRT GATFIGNSSK HDGSAICCST ALTLAPNSQL 1101 IFENNKVTET TATTKASINN LGAAIYGNNE TSDVTISLSA ENGSIFFKNN 1151 LCTATNKYCS IAGNVKFTAI EASAGKAISF YDAVNVSTKE TNAQELKLNE 1201 KATSTGTILF SGELHENKSY IPQKVTFAHG NLILGKNAEL SWSFTQSPG 1251 TTITMGPGSV LSNHSKEAGG IAINNVIIDF SEIVPTKDNA TVAPPTLKLV 1301 SRTNADSKD IDITGTVTLL DPNGNLYQNS YLGEDRDITL FNIDNSASGA 1351 VTATNVTLQG NLGAKKGYLG T NLDPNSSG SKIILKWTFD KYLR PYIPR 1401 DNHFYINSI GAQNSLVTVK QGILGNMLNN ARFEDPAFNN F ASAIGSFL 1451 RKEVSRNSDS FTYHGRGYTA AVDAKPRQEF ILGAAFSQVF GHAESEYHLD 1501 NYKHKGSGHS TQASLYAGNI FYFPAIRSRP ILFQGVATYG Y QHDTTTYY 1551 PSIEEKNMAN WDSIALFDL RFSVDLKEPQ PHSTARLTFY TEAEYTRIRQ 1601 EKFTELDYDP RSFSACSYGN LAIPTGFSVD GALA REIIL YNKVSAAYLP 1651 VILRNNPKAT YEVLSTKEKG NWNVLPTRN AARAEVSSQI YLGSYWTLYG 1701 TYTIDASMNT LVQMANGGIR FVF*
Pmp21 (CPn0963)
One example of a Pmp 21 protein is set forth as SEQ ID NOs 83 & 84 in WO 02/02606. {GenBank accession number: gi|4377287|gb|AAD19099.1| 'CPn0963'; SEQ ID NO: 36 below}.
SEQ ID No 36 1 MVAKKTVRSY RSSFSHSVIV AILSAGIAFE AHSLHSSELD LGVFNKQFEE 51 HSAHVEEAQT SVL GSDPVN PSQKESEKVL YTQVPLTQGS SGESLDLADA 101 NFLEHFQHLF EETTVFGIDQ KLVWSDLDTR NFSQPTQEPD TSNAVSEKIS 151 SDTKENRKDL ETEDPSKKSG LKEVSSDLPK SPETAVAAIS EDLEISENIS 201 ARDPLQGLAF FYKNTSSQSI SEKDSSFQGI IFSGSGANSG LGFENLKAPK 251 SGAAVYSDRD IVFENLVKGL SFISCESLED GSAAGVNIW THCGDVTLTD 301 CATGLDLEAL RLVKDFSRGG AVFTARNHEV QNNLAGGILS WGNKGAIW 351 EKNSAEKSNG GAFACGSFVY SNNENTALWK ENQALSGGAI SSASDIDIQG 401 NCSAIEFSGN QSLIALGEHI GLTDFVGGGA LAAQGTLTLR NNAWQCVKN 451 TSKTHGGAIL AGTVDLNETI SEVAFKQNTA ALTGGALSAN DKVIIANNFG 501 EILFEQNEVR NHGGA1YCGC RSNPKLEQKD SGENINIIGN SGAITFLKNK 551 ASVLEVMTQA EDYAGGGALW GHNVLLDSNS GNIQFIGNIG GSTFWIGEYV 601 GGGAILSTDR VTISNNSGDV VFKGNKGQCL AQKYVAPQET APVESDASST 651 NKDEKSLNAC SHGDHYPPKT VEEEVPPSLL EEHPWSSTD IRGGGAILAQ 701 HIFITDNTGN LRFSGNLGGG EESSTVGDLA IVGGGALLST NEVNVCSNQN 751 WFSDNVTSN GCDSGGAILA KKVDISANHS VEFVSNGSGK FGGAVCALNE 801 SVNITDNGSA VSFSKNRTRL GGAGVAAPQG SVTICGNQGN IAFKENFVFG 851 SENQRSGGGA IIANSSV IQ DNAGDILFVS NSTGSYGGAI FVGSLVASEG 901 SNPRTLTITG NSGDILFAKN STQTAASLSE KDSFGGGAIY TQNLKIVKNA 951 GNVSFYGNRA PSGAGVQIAD GGTVCLEAFG GDILFEGNIN FDGSFNAIHL 1001 CGNDSKIVEL SAVQDKNIIF QDAITYEENT IRGLPDKDVS PLSAPSLIFN 1051 SKPQDDSAQH HEGTIRFSRG VSKIPQIAAI QEGTLALSQN AELWLAGLKQ 1101 ETGSSIVLSA GSILRIFDSQ VDSSAPLPTE NKEETLVSAG VQINMSSPTP 1151 NKDKAVDTPV LADIISITVD LSSFVPEQDG TLPLPPEIII PKGTKLHSNA 1201 IDLKIIDPTN VGYENHALLS SHKDIPLISL KTAEGMTGTP TADASLSN1K 1251 IDVSLPSITP ATYGHTGV S ESKMEDGRLV VGWQPTGYKL NPEKQGALVL 1301 NNL SHYTDL RALKQEIFAH HTIAQRMELD FSTNVWGSGL GWEDCQNIG 1351 EFDGFKHHLT GYALGLDTQL VEDFLIGGCF SQFFGKTESQ SYKAKNDVKS 1401 YMGAAYAGIL AGPWLIKGAF VYGNINNDLT TDYGTLGIST GS IGKGFIA 1451 GTSIDYRYIV NPRRFISAIV STWPFVEAE YVRIDLPEIS EQGKEVRTFQ 1501 KTRFENVAIP FGFALEHAYS RGSRAEVNSV QLAYVFDVYR KGPVSLITLK 1551 DAAYSWKSYG VDIPCKAWKA RLSNNTE NS YLSTYLAFNY EWREDLIAYD 1601 FNGGIRIIF* Preferred PMP proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to one of the polypeptide sequences set forth for the pmp proteins above and/or (b) which is a fragment of at least ra consecutive amino acids of one of the polypeptide sequences set forth above wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These PMP proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of the polypeptide sequences set forth above. Preferred fragments of (b) comprise an epitope from one of the polypeptide sequences set forth above. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of one of the polypeptide sequences set forth above. 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). Fifth Antigen Group The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group. Such other Chlamydia pneumoniae antigens mclude a fifth antigen group consisting one or more cell surface exposed proteins. These antigens are referred to herein as the "fifth antigen group". Each of the Chlamydia pneumoniae antigens of the fifth antigen group is described in more detail below. (37) PorB Outer Membrane Protein B (CPn0854)
One example of a PorB protein is set forth as SEQ ID NOs: 67 & 68 in WO 02/02606. {GenBank accession number: gi|4377170|gb|AAD18992.1| 'CPn0854'; SEQ ID NO: 37 below}. Preferred PorB proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 37; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 37, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These PorB proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 37. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 37. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 37. 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).
SEQID No 37 1 MNSK KHLR IATLSFSMFF GIVSSPAVYA LGAGNPAAPV LPGVNPEQTG 51 WCAFQLCNSY DLFAALAGSL KFGFYGDYVF SESAHITNVP VITSVTTSGT 101 GTTPTITSTT KNVDFDLNNS SISSSCVFAT IALQETSPAA IPLLDIAFTA 151 RVGGLKQYYR LPLNAYRDFT SNPLNAESEV TDGLIEVQSD YGIV GLSLQ 201 KVLWKDGVSF VGVSADYRHG SSPINYIIVY NKANPEIYFD ATDGNLSYKE 251 WSASIGISTY LNDYVLPYAS VSIGNTSRKA PSDSFTELEK QFTNFKFKIR 301 KITNFDRVNF CFGTTCCISN NFYYSVEGR GYQRAINITS GLQF*
(38) 76kDa Protein Homolog (CPn0728) One example of a 76kDa Protein Homolog protein is set forth as SEQ ID NOs: 13 & 14 in WO 02/02606. {GenBank accession number: gi|4377033|gb]AAD18867.1| 'CPn0728'; SEQ ID NO: 38 below}. Preferred 76kDa proteins homologs for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%), 99%, 99.5% or more) to SEQ ID NO: 38; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 21, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These 76kDa protein homologs include Variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 38. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 38. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 38. 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).
SEQID No 38 1 MV PIGPGPI DETERTPPAD LSAQGLEASA ANKSAEAQRI AGAEAKPKES 51 KTDSVER SI LRSAVNALMS LADKLGIASS NSSSSTSRSA DVDSTTATAP 101 TPPPPTFDDY KTQAQTAYDT IFTSTSLADI QAALVSLQDA VTNIKDTAAT 151 DEETAIAAE ETKNADAVKV GAQITELAKY ASDNQAILDS LGKLTSFDLL 201 QAALLQSVAN NNKAAELLKE MQDNPWPGK TPAIAQSLVD QTDATATQIE 251 KDGNAIRDAY FAGQNASGAV ENAKSNNSIS NIDSAKAAIA TAKTQIAEAQ 301 KKPPDSPILQ EAEQMVIQAE KDL NIKPAD GSDVPNPGTT VGGSKQQGSS 351 IGSIRVSMLL DDAENETASI LMSGFRQMIH MFNTENPDSQ AAQQELAAQA 401 RAAKAAGDDS AAAALADAQK ALEAALGKAG QQQGILNALG QIASAAWSA 451 GVPPAAASSl GSSV QLYKT SKSTGSDYKT QISAGYDAYK SINDAYGRAR 501 NDATRDVINN VSTPALTRSV PRARTEARGP EKTDQALARV ISGNSRTLGD 551 VYSQVSALQS VMQI1QSNPQ ANNEEIRQKL TSAVTKPPQF GYPYVQLSND 601 STQKFIAKLE SLFAEGSRTA AEIKALSFET NSLFIQQVLV NIGSLYSGYL 651 Q*
(39) OmpA conserved outer membrane protein (CPn0695)
One example of an OmpA conserved outer membrane protein protein is set forth as SEQ ID NOs: 59 & 60 in WO 02/02606. {GenBank accession number: gi|4376998|gb|AAD18834.1| 'CPn0695'; SEQ ID NO: 39 below}. Preferred ompA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 39; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 39, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 39. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 39. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 39. 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).
SEQID No 39 1 MKK KSALL SAAFAGSVGS LQALPVGNPS DPSLL1DGTI WEGAAGDPCD 51 PCATWCDAIS LRAGFYGDYV FDRILKVDAP KTFSMGAKPT GSAAANYTTA 101 VDRPNPAYNK HLHDAEWFTN AGFIALNIWD RFDVFCTLGA SNGYIRGNST 151 AFNLVGLFGV KGTTVNANEL PNVΞLSNGW ELYTDTSFS SVGARGAL E 220011 CGCATLGAEF QYAQSKPKVE ELNVICNVSQ FSVNKPKGYK GVAFPLPTDA 251 GVATATGTKS ATINYHEWQV GASLSYRLNS LVPYIGVQWS RATFDADNIR 301 IAQPKLPTAV LNLTA NPSL LGNATALSTT DSFSDFMQIV SCQINKFKSR 351 KACGVTVGAT LVDADK SLT AEARLINERA AHVSGQFRF* (40) PepA (CPn0385)
One example of a PepA protein protein is set forth as SEQ ID NOs: 99 & 100 in WO 02/02606. {GenBank accession number: gi|4376664|gb|AAD 18529.1 'CPn0385'; SEQ ID NO: 40 below}. Preferred PepA proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%>, 65%>, 70%>, 75%>, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 40; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 40, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These PepA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 40 Preferred fragments of (b) comprise an epitope from SEQ ID NO: 40. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 40. 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). SEQID No 40 1 MVLFHAQASG RNRVKADAIV LPF HFKDAK NAASFEAEFE PSYLPALENF 51 QGKTGEIELL YSSPKAKEKR IVLLGLGKNE ELTSDWFQT YATLTRVLRK 101 AKCSTVNIIL PTISELRLSA EEFLVGLSSG ILSLNYDYPR YNKVDRNLET 151 PLS VTVIGI VPKMADAIFR KEAAIFEGVY LTRDLVNRNA DEITPKKLAE 201 VALNLGKEFP SIDTKVLGKD AIAKEKMGLL LAVSKGSCVD PHFIWRYQG 251 RPKSKDHTVL IGKGVTFDSG GLDLKPGKSM LT KEDMAGG ATVLGILSAL 330011 AVLELPINVT GIIPATENAI DGASYKMGDV YVGMSGLSVE ICSTDAEGRL 351 ILADAITYAL KYCKPTRIID FATLTGAMW SLGEEVAGFF SNNDVLAEDL 401 LEASAETSEP LWRLPLVKKY DKTLHSDIAD MKNLGSNRAG AITAALFLQR 451 FLEESSVA A HLDIAGTAYH EKEEDRYPKY ASGFGVRSIL YYLENSLSK*
(41) Conserved Outer Membrane Protein (Cpn0278)
One example of a conserved outer membrane protein protein is set forth as SEQ ID NO: 41 below. GenBank Accession No. GL4376552; AAD18427.1. Preferred conserved outer membrane proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%,, 75%,, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 41; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 41, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These conserved outer membrane proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 41. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 41. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 41. 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).
SEQ ID No 41 1 MKKKLSLLVG LIFVLSSCHK EDAQNKIRIV ASPTPHAELL ESLQEEAKDL GIKLKILPVD 61 DYR1PNRLLL DKQVDANYFQ HQAFLDDECE RYDCKGELW IAKVHLEPQA IYSKKHSSLE 121 RLKSQKKLTI AIPVDRTNAQ RALHLLEECG LIVCKGPANL NMTAKDVCGK ENRSINILEV 181 SAPLLVGSLP DVDAAVIPGN FAIAANLSPK KDSLCLEDLS VSKYTNLWI RSEDVGSPKM 241 IKLQKLFQSP SVQHFFDTKY HGNILTMTQD NG
Sixth Antigen Group
The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group. Such other Chlamydia pneumoniae antigens include a sixth antigen group consisting one or more FACS positive CPn antigens. These antigens are referred to herein as the " sixth antigen group". Each of the Chlamydia pneumoniae antigens of the sixth antigen group is described in more detail below.
(42) Predicted Omp (CPn0020)
One example of a predicted Omp protein is set forth as SEQ ID NOs: 91 & 92 in WO 02/02606. {GenBank accession number gi|4376272|gb|AAD18173.1: 'CPn0020'; SEQ ID NO: 42 below). Preferred Omp proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%,, 70%,, 75%,, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 42; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 42, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Omp proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 42. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 42. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 42. 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). SEQ ID No 42 1 MKRCFLFLAS FVLMGSSADA LTHQEAVKKK NSYLSHFKSV SGIVTIEDGV 51 LNIHNNLRIQ ANKVYVENTV GQSLKLVAHG NVMVNYRAKT LVCDYLEYYE 101 DTDSCLLTNG RFA YPWFLG GSMITLTPET IVIRKGYIST SEGPKKDLCL 151 SGDYLEYSSD SLLSIGKTTL RVCRIPILFL PPFSIMPMEI PKPPINFRGG 201 TGGFLGSYLG MSYSPISRKH FSSTFFLDSF FKHGVGMGFN LHCSQKQVPE 251 NVFNMKSYYA HRLAIDMAEA HDRYRLHGDF CFTHKHVNFS GEYHLSDSWE 301 TVADIFPNNF MLKNTGPTRV DCT NDNYFE GYLTSSVKVN SFQNANQELP 351 YLTLRQYPIS IYNTGVYLEN IVECGYLNFA FSDHIVGENF SSLRLAARPK 401 LHKTVPLPIG TLSSTLGSSL IYYSDVPEIS SRHSQLSAKL QLDYRFLLHK 451 SYIQRRHIIE PFVTFITETR PLAKNEDHYI FSIQDAFHSL NLLKAGIDTS 501 VLSKTNPRFP RIHAKLWTTH ILSNTESKPT FPKTACELSL PFGKKNTVSL 551 DAE IWKKHC WDHMNIRWE IGNDNVA TL ESLHRSKYSL IKCDRENFIL 601 DVSRPIDQLL DSPLSDHRNL ILGKLFVRPH PCWNYRLSLR YGWHRQDTPN 651 YLEYQMILGT KIFEHWQLYG VYERREADSR FFFFLKLDKP KKPPF*
(43) Predicted Omp (CPn0021)
One example of a predicted Omp protein is set forth as SEQ ID NOs: 49 & 50 in WO 02/02606. {GenBank accession numbe gi|4376273|gb|AAD18174.1: 'CPn0021'; SEQ ID NO: 43 below}. Preferred Omp proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g. 60%, 65%>, 70%>, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 43; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 43, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 43. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 43. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 43. 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). SEQID No 43 1 GLFHLTLFG CSLPISL VAKFPESVGH KILYISTQST QQAIATYLEA 51 LDAYGDHDFF VLRKIGEDYL KQSIHSSDPQ TRKSTIIGAG LAGSSEALDV 101 LSQAMETADP LQQLLVLSAV SGHLGKTSDD LLFKALASPY PVIRLEAAYR 151 LANLKNTKVI DHLHSFIHKL PEEIQCLSAA IFLRLETEES DAYIRDLLAA 201 KKSAIRSATA LQIGEYQQKR FLPTLRNLLT SASPQDQEAI LYALGKLKDG 251 QSYYNIKKQL QKPDVDVTLA AAQALIALGK EEDALPVIKK QALEERPRAL 301 YALRHLPSEI GIP1ALPIFL KTKNSEAKLN VALALLELGC DTPKLLEYIT 351 ERLVQPHYNE TLALSFSKGR TLQNWKRVNI IVPQDPQERE RLLSTTRGLE 401 EQILTFLFRL PKEAYLPCIY KLLASQKTQL ATTAISFLSH TSHQEALDLL 451 FQAAKLPGEP IIRAYADLAI YNLTKDPEKK RSLHDYAKKL IQETLLFVDT 501 ENQRPHPSMP YLRYQVTPES RTKLMLDILE TLATSKSSED IRLLIQLMTE 551 GDAKNFPVLA GLLIKIVE* (44) Oligopeptide Binding Protein Oppa-1 Lipoprotein (CPn0195)
One example of an oligopeptide binding protein is set forth as SEQ ID NOs: 23 and 24 in WO 02/02606. {GenBank accession number gi|4376466|gb|AAD18348.1: 'CPn0195'; SEQ ID NO: 44 below}. Preferred oligopeptide binding proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 44; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 44, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 44. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 44. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 44. 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). ID No 44 11 RKISVGICI TIL SLSW QGCKESSHSS TSRGELAINI RDEPRSLDPR 51 QVRLLSEISL VKHIYEGLVQ ENNLSGNIEP ALAEDYSLSS DGLTYTFKLK 101 SAFWSNGDPL TAEDFIESWK QVATQEVSGI YAFALNPIKN VRKIQEGHLS 151 IDHFGVHSPN ESTLWTLES PTSHFLKLLA LPVFFPVHKS QRTLQSKSLP 201 IASGAFYPKN IKQKQWIKLS KNPHYYNQSQ VETKTITIHF IPDA TAAKL 225511 FNQGKLNWQG PPWGERIPQE TLSNLQSKGH LHSFDVAGTS LTFNINKFP 301 LNNMKLREAL ASALDKEALV STIFLGRAKT ADHLLPTNIH SYPEHQKQEM 351 AQRQAYAKKL FKEALEELQI TAKDLEHLNL IFPVSSSASS LLVQLIREQ 401 KESLGFAIPI VGKEFALLQA DLSSGNFSLA TGGWFADFAD PMAFLTIFAY 451 PSGVPPYAIN HKDFLEILQN IEQEQDHQKR SELVSQASLY LETFHIIEPI 550011 YHDAFQFAMN KKLSNLGVSP TGWDFRYAK EN*
(45) CHLPS 43 kDa Protein Homologue-1 (CPn0562)
One example of a CHLPS protein is set forth as SEQ ID NO: 45 below. GenBank Accession No. GI.4376854; AAD 18702.1. Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g.
60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,
98%, 99%, 99.5% or more) to SEQ ID NO: 45; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 45, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more).
These CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 45. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 45. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 45. 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). SEQID No 45 1 MSIAIAREQY AAILDMHPKP SIA FSSEQA RTSWEKRQAH PYLYRLLEII WGWKFLLGL 61 IFFIPLGLFW VLQKICQNFI LLGAGG IFR PICRDSNLLR QAYAARLFSA SFQDHVSSVR 121 RVCLQYDEVF IDGLELRLPN AKPDRWMLIS NGNSDCLEYR TVLQGEKDWI FRIAEESQSN 181 1LIFNYPGVM KSQGNITRNN WKSYQACVR YLRDEPAGPQ ARQIVAYGYS LGASVQAEAL 241 SKEIADGSDS VRWFWKDRG ARSTGAVAKQ FIGSLGV LA NLTHWNINSE KRSKDLHCPE 301 LFIYGKDSQG NLIGDGLFKK ETCFAAPFLD PKNLEECSGK KIPVAQTGLR HDHILSDDVI 361 KEVAGHIQRH FDN
(46) YscJ (Yop translocation J protein) (CPn0828)
One example of a YscJ protein is set forth as SEQ ID NOs: 109 and 110 in WO 02/02606. {GenBank accession number gi|4377140|gb|AAD18965.1| 'CPn0828'; SEQ ID NO: 46 below. Preferred YscJ proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 46; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 46, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These YscJ proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 46. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 46. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 46. 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).
SEQID No 46 11 MVRRSISFC FFIMTLLCCT SCNSRSLIVH GLPGREANEI WLLVSKGVA 51 AQKLPQAAAA TAGAATEQM DIAVPSAQIT EALAILNQAG LPRMKGTSLL 101 DLFAKQGLVP SELQEKIRYQ EGLSEQMAST IRKMDGWDA SVQISFTTEN 151 EDNLPLTASV YIKHRGVLDN PNSIMVSKIK RLIASAVPGL VPENVSWSD 201 RAAYSDITIN GP GLTEEID YVSV GIILA KSSLTKFRLI FYVLILILFV 225511 ISCGLL VIW KTHTLIMTMG GTKGFFNPTP YTKNALEAKK AEGAAADKEK 301 KEDADSQGES KNAETSDKDS SDKDAPEGSN EIEGA*
(47) Hypothetical (CPn 0415)
One example of a hypothetical protein is set forth as SEQ ID NOs: 101 and 102 in WO 02/02606. {GenBank accession number gi|4376696|gb|AAD18559.1| 'CPn0415'; SEQ ID NO: 47 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%) or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 47; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 47, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 47. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 47. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 47. 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). SEQ ID No 47 1 MTLIFVIIIV WCNAFLIKLC VIMGLQSRLQ HCIEVSQNSN FDSQVKQFIY 51 ACQDKTLRQS VLKIFRYHPL LKIHDIARAV YBLMALEEGE DLGLSFLNVQ 101 QYPSGAVELF SCGGFP KGL PYPAEHAEFG LLLLQIAEFY EESQAYVSKM 151 SHFQQALFDH QGSVFPSL S QENSRLLKEK TTLSQSFLFQ LGMQIHPEYS 201 LEDPALGF M QRTRSSSAFV AASGCQSSLG AYSSGDVGVI AYGPCSGDIS 251 DCYYFGCCGI AKEFVCQKSH QTTEISFLTS TGKPHPRNTG FSYLRDSYVH 301 LPIRCKITIS DKQYRVHAAL AEATSAMTFS IFCKGKNCQV VDGPRLRSCS 351 LDSYKGPGND IMILGENDAI NIVSASPYME IFALQGKEKF NADFLINIP 401 YKEEGVMLIF EKKVTSEKGR FFTKMN* (48) Hypothetical (CPn0514) One example of a hypothetical protein is set forth as SEQ ID NOs: 87 and 88 in WO 02/02606. {GenBank accession number gi|4376802|gb|AAD18654.1| 'CPn0514'; SEQ ID NO: 48 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 48; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 48, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 48. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 48. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 48. 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). SEQ ID No 48 1 MSNQLQPCIS LGCVSYINSF PLSLQLIKRN DIRCVLAPPA DLLNLLIEGK 51 LDVALTSSLG AISHNLGYVP GFGIAANQRI LSVNLYAAPT FFNSPQPRIA 101 ATLESRSSIG LLKVLCRHL RIPTPHILRF ITTKVLRQTP ENYDGLLLIG 151 DAALQHPVLP GFVTYDLASG YDLTKLPFV FALLLHSTS KEHPLPNLAM 201 EEALQQFESS PEEVLKEAHQ HTGLPPSLLQ EYYALCQYRL GEEHYESFEK 251 FREYYGTLYQ QARL* (49) Hypothetical (CPn0668) One example of a hypothetical protein is set forth as SEQ ID NOs: 57 and 58 in WO 02/02606. {GenBank accession number gi|4376968|gb|AAD 18807.1 'CPn0668'; SEQ ID NO: 49 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%o or more) to SEQ ID NO: 49; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 49, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 49. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 49. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 49. 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). SEQ ID No 49 1 MKFLLYVPLL VLVSTGCDA KPVSFEPFSG KLSTQRFEPQ HSAEEYFSQG 51 QEFLKKGNFR KALLCFGIIT HHFPRDILRN QAQYLIGVCY FTQDHPDLAD 101 KAFASYLQLP DAEYSEELFQ MKYAIAQRFA QGKRKRICRL EGFPKLMNAD 151 EDALRIYDEI LTAFPSKDLG AQALYSKAAL LIVKNDLTEA TKTLKKLTLQ 201 FPLHILSSEA FVRLSEIYLQ QAKKEPHNLQ YLHFAKLNEE AMKKQHPNHP 251 LNEWSANVG AMREHYARGL YATGRFYEKK KKAEAANIYY RTAITNYPDT 301 LLVAKCQKRL DRISKHTS* (50) Hypothetical (CPn0791)
One example of a hypothetical protein is set forth as SEQ ID NOs: 123 and 124 in WO 02/02606. {GenBank accession number gi|4377101|gb|AAD18929.1| 'CPn0791'; SEQ ID NO: 50 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, or more) to SEQ ID NO: 50; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 50, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 50. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 50. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 50. 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).
SEQ ID No 50 1 MYSCYSKGIS HNYLLHPMSR LDIFVFDSLI ANQDQNLLEE IFCSEDTVLF 51 KAYRTTALQS PLAAKNLNIA RKVANYILAD NGEIDTVKLV EAIHHLSQCT 101 YPLGPHRHNE AQDREHLLKM LKALKENPKL KESIKTLFVP SYSTIQNLIR 151 HTLALNPQTI LSTIHVRQAA LTALFTYLRQ DVGSCFATAP AILIHQEYPE 201 RFLKDLNDLI SSGKLSRIVN QREIAVPINL SGCIGELFKP LR1LDLYPDP 251 LVKLSSSPGL KKAFSAANLI ETLGDSEAQI QQLLSHQYLM QKLQNVHETL 301 TANDIIKSTL LHYYQLQEST VRAIFFKEGL FSKEQVAFST QHPRELSEIQ 351 RVYHYLHAYE EAKSAFIHDT QNPLLKAWEY TLATLADASQ PTISNHIRLA 401 LGWKSEDPHS LVSLVTHFVE EEVENIRILV QQCEQTYHEA RSQLEYIEGR 451 MRNPLNNQDS QILTMDHMRF RQELNKALYE WDSAQEKAKK FLHLPEFLLS 501 FYTKQIPLYF RSSYDAFIQE FAHLYANAPA GFRILFTHGR THPNTWSPIY 551 SINEFIRFLS EFFTSTESEL LGKHAVINLE KETSRLVHNI TA LHTDVFQ 601 EALLTRILEA YQLPVPPSIL NHLDQLSQTP VYVSGGTVD TLLLDYFESS 651 EPLTLTEKHP ENPHELAAFY ADALKDLPTG IKSYLEEGSH SLLSSSPTHV 701 FSIIAGSPLF REAWDND YS YT LRDV VK QHQDFLQDTI LPQLSIYAFI 751 ENFCNKYALQ HWHDFHDFC SDHSLTLPEL YDKGSRFLSS LFTKDKTVAL 801 IYIRRLLYLM VREVPYVSEQ QLPEVLDNVS SYLGISSRIT YEKFRSLIEE 851 TIPKMTLLSS ADLRHIYKGL LMQSYQKIYT EEDTYLRLTT AMRHHNLAYP 901 APLLFADSNW PSIYFGFILN PGTTEIDL K FNYAGLQGQP LDNIQELFAT 951 SRPWTLYANP IDYGMPPPPG YRSRLPKEFF *
(51) Hypothetical (CPn0792) One example of a hypothetical protein is set forth as SEQ ID NOs: 61 and 62 in WO 02/02606. {GenBank accession number gi|4377102|gb|AAD18930.1| 'CPn0792'; SEQ ID NO: 51 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 91%, 98%, 99%, 99.5%, or more) to SEQ ID NO: 51; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 51, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 51. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 51. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 51. 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).
SEQID No 51 1 KHTFTKRVL FFFFLVIPIP LLLN MVVGF FSFSAAKAML VQVLHTRATN 5511 LSIEFEKKLT IHKLFLDRLA NTLALKSYAS PSAEPYAQAY NEMMALSNTD 101 FSLCLIDPFD GSVRTKNPGD PFIRYLKQHP EMKKKLSAAV GKAFLLTIPG 151 KPLLHYLILV EDVASWDSTT TSGLLVSFYP MSFLQKDLFQ SLHITKGNIC 201 LVNKYGEVLF CAQDSESSFV FSLDLPNLPQ FQARSPSAIE IEKASGILGG 251 ENLITVSINK KRYLGLVLNK IPIQGTYTLS LVPVSDLIQS ALKVPLNICF 330011 FYVLAFLLMW IFSKINTKL NKPLQELTFC MEAAWRGNHN VRFEPQPYGY 351 EFNELGNIFN CTLLLLLNSI EKADIDYHSG EKLQKELGIL SSLQSALLSP 401 DFPTFPKVTF SSQHLRRRQL SGHFNG TVQ DGGDTLLGII GLAGDIGLPS 451 YLYALSARSL FLAYASSDVS LQKISKDTAD SFSKTTEGNE AWAMTFIKY 501 VEKDRSLELL SLSEGAPTMF LQRGESFVRL PLETHQALQP GDRLICLTGG 555511 EDILKYFSQL PIEELLKDPL NPLNTENLID SLTMMLNNET EHSADGTLTI 601 LSFS*
[52) Hypothetical (CPn0820)
One example of a hypothetical protein is set forth as SEQ ID NOs: 113 and 114 in WO 02/02606. {GenBank accession number gi|4377132|gb|AAD18958.1| 'CPn0820'; SEQ ID NO: 52 below}. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 52; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 52, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 52. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 52. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 52. 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). SEQ ID No 52 1 MCNSIAMKKQ KRGFVLMELL MSFTLIALLL GTLGF YRK1 YTVQKQKERI 51 YNFYIEESRA YKQLRTLFSM SLSSSYEEPG SLFSLIFDRG VYRDPKLAGA 101 VRASLHHDTK DQRLELRICN IKDQSYFETQ RLLSHVTH LSFQRNPDPE 151 KLPETIALTI TREPKAYPPR TLTYQFAVGK*
(53) Hypothetical (CPn0126) One example of a hypothetical protein is set forth as SEQ ID NO: 53 below. GenBank Accession No. GL4376390; AAD 18279.1 Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 53; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 53, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 53. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 53. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 53. 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).
SEQID No 53 1 MVFSYYCMGL FFFSGAISSC GLLVSLGVGL GLSVLGVLLL LLAGLLLFKI QSMLREVPKA 61 PDLLDLEDAS ERLRVKASRS LASLPKEISQ LESYIRSAAN DLNTIKTWPH KDQRLVETVS 121 RKLERLAAAQ NYMISELCEI SEILEEEEHH LILAQESLEW IGKSLFSTFL DMESFLNLSH 181 LSEVRPYLAV NDPRLLEITE ES EWSHFI NVTSAFKKAQ ILFKNNEHSR MKKKLESVQE 241 LLETFIYKSL KRSYRELGCL SEKMRIIHDN PLFPWVQDQQ KYAHAKNEFG EIARCLEEFE 301 KTFFWLDEEC AISYMDCWDF LNESIQNKKS RVDRDYISTK KIALKDRART YAKVLLEENP 361 TTEGKIDLQD AQRAFERQSQ EFYTLEHTET KVRLEALQQC FSDLREATNV RQVRFTNSEN 421 ANDLKESFEK IDKERVRYQK EQRLYWETID RNEQELREEI GESLRLQNRR KGYRAGYDAG 481 RLKGLLRQWK KNLRDVEAHL EDATMDFEHE VSKSELCSVR ARLEVLEEEL MDMSPKVADI 541 EELLSYEERC ILPIRENLER AYLQYNKCSE ILSKAKFFFP EDEQLLVSEA NLREVGAQLK 601 QVQGKCQERA QKFAIFEKHI QEQKSLIKEQ VRSFDLAGVG FLKSELLSIA CNLYIKAWK 661 ESIPVDVPCM QLYYSYYEDN EAWRNRLLN MTERYQNFKR SLNSIQFNGD VLLRDPVYQP 721 EGHETRLKER ELQETTLSCK KLKVAQDRLS ELESRLSRR
(54) Hypothetical (CPn0794)
One example of a hypothetical protein is set forth as SEQ ID NO: 54 below. GenBank Accession No. G 4377105; AAD18932.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 54; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 54, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 54. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 54. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 54. 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).
SEQ ID No 54 1 MSLYQK NS QLKKSLCYST VAALIFMIPS QESFADSLID LNLGLDPSVE CLSGDGAFSV 61 GYFTKAGSTP VEYQPFKYDV SKKTFTILSV ETANQSGYAY GISYDGTITV GTCSLGAGKY 121 NGAKWSADGT LTPLTGITGG TSHTEARAIS KDTQVIEGFS YDASGQPKAV QWASGATTVT 181 QLADISGGSR SSYAYAISDD GTIIVGSMES TITRKTTAVK VNNVPTYLG TLGGDASTGL 241 YISGDGTVIV GAANTATVTN GNQESHAYMY KDNQMKD
(55) Hypothetical (CPn0796)
One example of a hypothetical protein is set forth as SEQ ID NO: 55 below. GenBank Accession No. GI.4377107; AAD18934.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 55; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 55, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 55. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 55. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 55. 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). Cpn0796 may be secreted from C. pneumoniae and is localized in the membrane of Chlamydia in young inclusions whereas an N-terminal part of Cpn0796 is secreted into the host cell cytoplasm at later times. Cpn0796 was proposed to be an autotransporter and it is the first example of secretion into the host cell cytoplasm of a proposed Chlamydia autotrasporter. Te finding in the host cell cytoplasm of Cpn0796 suggests that an unknown transport mechanism exists for translocation over the inclusion membrane (Nandahl, "Proteome analysis of Chlamydia pneumoniae - proteins at the Chlamydia- host cell Interface," Abstract of PhD Dissertation, Dan Med Bull 2004: 51:306).
SEQID No 55 1 MQPCLΝMSIV RΝSALPLPCL SRSETFKKVR SH KFMKVLT PWIYRKDL V TAFLLTAIPG 61 SFAHTLVDIA GEPRHAAQAT GVSGDGKIVI GMKVPDDPFA ITVGFQYIDG HLQPLEAVRP 121 QCSVYPΝGIT PDGTVIVGTΝ YAIGMGSVAV KWVΝGKVSEL PMLPDTLDSV ASAVSADGRV 181 IGGΝRΝIΝLG ASVAVKWEDD VITQLPSLPD AMΝACVΝGIS SDGSIIVGTM VDVSWRΝTAV 241 Q IGDQLSVI GTLGGTTSVA SAISTDGTVI VGGSEΝADSQ THAYAYKΝGV MSDIGTLGGF 301 YSLAHAVSSD GSVIVGVSTΝ SEHRYHAFQY ADGQMVDLGT LGGPESYAQG VSGDGKVIVG 361 RAQVPSGD H AFLCPFQAPS PAPVHGGSTV VTSQΝPRGMV DIΝATYSSLK ΝSQQQLQRLL 421 IQHSAKVESV SSGAPSFTSV KGAISKQSPA VQΝDVQKGTF LSYRSQVHGΝ VQΝQQLLTGA 481 FMD KLASAP KCGFKVALHY GSQDALVERA ALPYTEQGLG SSVLSGFGGQ VQGRYDFΝLG 541 ETWLQPFMG IQVLHLSREG YSEKΝVRFPV SYDSVAYSAA TSFMGAHVFA SLSPKMSTAA 601 TLGVERDLΝS HIDEFKGSVS AMGΝFVLEΝS TVSVLRPFAS LAMYYDVRQQ QLVTLSVVMΝ 661 QQPLTGTLSL VSQSSYΝLSF
One preferred protein for use with the invention comprises an Ν-terminal peptide of
Cpn0796 that may be secreted to be exposed on the bacterial cell surface and can also become detached via a proteolytic event. In one embodiment, the Ν-terminal peptide of Cpn0796 may form a beta-propeller structural conformation. One example of the N-terminal peptide of Cpn0796 is set forth as SEQ ID NO: 86 below. The N-terminal peptide of Cpn0796 for use with the invention may comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 86; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 86, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 86. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 86. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 86.
SEQ ID NO: 86 HTLVDIAGEPRHAAQΆTGVSGDGKIVIGMKVPDDPFAITVGFQYIDGHLQP EAVRPQCSVYPNG ITPDGTVIVGTNYAIGMGSVAVK V GKVSE PMLPDTLDSVASAVSADGRVIGGNRNINLGASV AVKWEDDVITQLPS PDA NACVNGISSDGSIIVGTMVDVS R TAVQWIGDQLSVIGTLGGTTS VASAISTDGTVIVGGSENADSQTHAYAYKNGVMSDIGTLGGFYSLAHAVSSDGSVIVGVSTNSEH RYHAFQYADGQMVDLGT GGPESYAQGVSGDGKVIVGRAQVPSGDWHAF C
(56) Hypothetical (CPn0797)
One example of a hypothetical protein is set forth as SEQ ID NO: 56 below. GenBank Accession No. GL4377108; AAD18935.1 Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 56; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 56, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 56. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 56. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 56. 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). SEQ ID No Sό 1 MSKKIKVLGH LTLCTLFRGV LCAAALSNIG YASTSQESPY QKSIEDWKGY TFTDLELLSK 61 EG SEAHAVS GNGSRIVGAS GAGQGSVTAV I ESHLIKHL GTLGGEASSA EGISKDGEW 121 VGWSDTREGY THAFVFDGRD MKDLGTLGAT YSVARGVSGD GSIIVGVSAT ARGEDYGWQV 181 GVKWEKGKIK QLKLLPQGLW SEANAISEDG TVIVGRGEIS RNHIVAVK N KNAVYSLGTL 241 GGSVASAEAI SANGKVIVGW STTNNGETHA FMHKDETMHD LGTLGGGFSV ATGVSADGRA 301 IVGFSAVKTG EIHAFYYAEG EMEDLTTLGG EEARVFDISS EGNDIIGSIK TDAGAERAYL 361 FHIHK
(76) Oligopeptide Binding Protein Oppa-2 Lipoprotein (CPn0196)
One example of an oligopeptide binding protein is set forth as SEQ ID NOs: 127 and 128 in WO 02/02606. {GenBank accession number G 4376467; AAD18349.1 'CPn0196'; SEQ ID NO: 76 below}. Preferred oligopeptide binding proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%,, 99.5% or more) to SEQ ID NO: 76; and or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 76, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO 76. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 76. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 76. 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).
SEQ ID No 76 1 mlrffavfis tlwlitsgcs psqsskgifv vnmkemprsl dpgktrliad qtlmxhlyeg 61 lveehsqnge ikpalaesyt isedgtrytf kiknilwsng dpltaqdfvs swkeilkeda 121 ssvylyaflp iknaraifdd tespenlgvr aldkrhleiq letpcahflh fltlpiffpv 181 hetlrnysts feempitcga frpvslekgl rlhleknpmy hnksrvklhk iivqfisnan 241 taailfk kk ld qgpp ge pippeisasl qddqlfsip gastt llfn iqkkpwnnak 301 lrkalslaid kdmltkvvyq glaeptdhil hprlypgtyp erkrqneril eaqqlfeeal 361 delqmtredl eketltfstf sfsygricqm lreqwkkvlk ftipivgqef ftiqknfleg 421 nysltvnq t aafidpmsyl mifanpggis pyhlqdshfq tllikitqeh kk lrnqlii 481 ealdylehch ileplchpnl rialnknikn fnlfvrrtsd frfiekl
Seventh Antigen Group The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group. Such other Chlamydia pneumoniae antigens include a seventh antigen group consisting one or more hypothetical proteins (ie proteins which, for example, have no known cellular location and/or function. These antigens are referred to herein as the "seventh antigen group". Each of the Chlamydia pneumoniae antigens of the seventh antigen group is described in more detail below. (57) Hypothetical (CPn0331)
One example of a hypothetical protein is set forth as SEQ ID NO: 57 below. GenBank Accession No. GL4376609; AAD 18480.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 91%, 98%, 99%, 99.5% or more) to SEQ ID NO: 57; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 57, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 57. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 57. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 57. 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). SEQ ID No 57
> 1 MAVSGGGGVQ PSSDPGKWNP ALQGEQAEGP SPLKESIFSE TKQASSAAKQ ESLVRSGSTG 61 MYATESQINK AKYRKAQDRS STSPKSKLKG TFSKMRASVQ GFMSGFGSRA SRVSAKRASD 121 SGEGTSLLPT EMDVALKKGN RISPEMQGFF LDASGMGGSS SDISQLSLEA LKSSAFSGAR 181 SLSLSSSESS SVASFGSFQK AIEPMSEEKV NA TVARLGG EMVSSLLDPN VETSSLVRRA 24 .1.. MATGNEGMID LSDLGQEEVS TAMTSPRAVE GKVKVSSSDS PEANPTGIPN SNTLERAEKE 330011 AAEEKKQQEESSRREEQQLL SEDQMMLARA MAGLLTGAAP QEVLSNSV S GPSTVFPPPK FSGTLPTQRS 361 GDKSKHKSPG IEKSTNHTNF SPLREGTVKS AEVKSLPHPE SMYRFPKDSI VSREEPEAW 421 KESTAFKNPE NSSQNFLPIA VESVFPKESG TGGALGSDAV SSSYHFLAQR GVSLLAPLPR 481 ATDDYKEKLE AHKGPGGPPD PLIYQYRNVA VEPPIVLRSP QPFSGSSRLS VQGKPEAASV 541 HDDGGGGNSG GFSGDQRRGS SGQKASRQEK KGKKLSTDI (58) Hypothetical (CPn0234) One example of a hypothetical protein is set forth as SEQ ID NO: 58 below. GenBank Accession No. gi|4376508|gb| AAD18387.1 Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 58; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 21, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 58. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 58. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 58. 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). SEQ ID No 58 1 MLQSCKKALL SIWSILAFH PIPGMGVEAK SGFLGKVKG FSKKEIQEEA RILPVKDSLS 61 KRYDYTSSS GFSVEFPGEP DHSGQIVEVP QSEITIRYDT YVTETHPDNT VYWSV EYP 121 EKVDISRPEL NLQEGFSGMM QALPESQVLF MQARQIQGHK ALEF IVCED VYFRGMLISV 181 NHTLYQVFMV YKNKNPQALD KEYEAFSQSF KITKIREPRT IPSSVKKKVS L (59) Hypothetical (CPn0572) One example of a hypothetical protein is set forth as SEQ ID NO: 59 below. Genbank Accession No. gi|4376866|gb|; AAD18712.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 59; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 59, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 59. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 59. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 59. 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). SEQID No 59 1 MAAPINQPST TTQITQTGQT TTTTTVGSLG EHΞVTTTGSG AAAQTSQTVT LIADHEMQEI 61 ASQDGSAVSF SAEHSFSTLP PETGSVGATA QSAQSAGLFS LSGRTQRRDS EISSSSDGSS 121 ISRTSSNASS GETSRAESSP DLGDLDSLSG SERAEGAEGP EGPGGLPEST IPHYDPTDKA 181 SILNFLKNPA VQQKMQTKGG HFVYVDEARS SFIFVRNGD STAESIKVSN AKTKENITKP 241 ADLEMCIAKF CVGYETIHSD TGRVKPTME ERSGATGNYN HL LSMKFKT AWYGP NAK 301 ESSSGYTPSA RRGAKVETG PIWDDVGGLK GINWKTTPAP DFSFINETPG GGAHSTSHTG 361 PGTPVGATW PNVNVNLGGI KVDLGGINLG GITTNVTTEE GGGTNITSTK STSTDDKVSI 421 TSTGSQSTIE EDTIQFDDPG QGEDDNAIPG TNTPPPPGPP PNLSSSRLLT ISNASLNQVL 481 QNVRQHLNTA YDSNGNSVSD LNQDLGQWK NSENGVNFPT VILPKTTGDT DPSGQATGGV 541 TEGGGHIRNI IQRNTQSTGQ SEGATPTPQP TIAKIVTSLR KANVSSSSVL PQPQVATTIT 601 PQARTASTST TSIGTGTEST STTSTGTGTG SVSTQSTGVG TPTTTTRSTG TSATTTTSSA 661 STQTPQAPLP SGTRHVATIS LVRNAAGRSI VLQQGGRSQS FPIPPSGTGT QNMGAQLWAA 721 ASQVASTLGQ VVNQAATAGS QPSSRRSSPT SPRRK
Eighth Antigen Group
The immunogenicity of other Chlamydia pneumoniae antigens may be unproved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group of the third antigen group or the fourth antigen group or the fifth or the sixth antigen group or the seventh antigen group. Such other Chlamydia pneumoniae antigens include an eigth antigen group consisting one or more FACS positive CPn antigens. These antigens are referred to herein as the "eight antigen group". Each of the Chlamydia pneumoniae antigens of the eight antigen group is described in more detail below.
(60) Low Calcium Response Protein H (CPnOSll)
One example of a Low Calcium Response Protein H is set forth as SEQ ID NO: 60 below. Genbank Accession No. GL4377123; AAD18949.1. Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 60; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 60, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These low calcium response proteins mclude variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 60. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 60. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 60. 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). SEQ ID No 60 1 mskpsprnan qpqkpsasfn kktrsrlael aaqkkakadd leqvhpvpte eeikkalgni 61 feglsngldl qqilglsdyl leeiytvayt fysqgkynea vglfqllaaa qpqnykymlg 121 lsscyhqlhl yneaafgffl afdaqpdnpi ppyyiadsll klqqpeesrm fldvtmdicg 181 nnpefkilke rcqimkqsie kqmagetkka ptkkpagksk tttnkksgkk r
(61) Yop Proteins Translocation Protein T (CPn0823)
One example of a Yop Proteins Translocation Protein T is set forth as SEQ ID NO: 61 below. Genbank Accession No. G 4377135; AAD18960.1. Preferred Yop proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 61; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 61, wherein « is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These Yop proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 61. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 61. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 61. 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). SEQ ID No 61 1 mgislpelfs nlgsayldyi fq ppayvws vfllllarll pifavapflg aklfpspiki 61 gisls laii fpkvladtqi tnymdnnlfy vllvkemiig ivigfvlafp fyaaqsagsf 121 itnqqgiqgl egatslisie qtsphgilyh yfvtiifwlv gghrivisll lqtlevipih 181 sffpaemmsl sapiwitm±k mcqlclvmti qlsapaalam lmsdlflgii nrmapqvqvi 241 yllsalkafm gllfltlaw fiikqidyft lawfkevpim llgsnpqvl
(62) Yop Proteins Translocation Protein J
One example of a Yop Proteins Translocation Protein J is set forth as SEQ ID NO: 62 below Genbank Accession No. GL4377140; AAD18965.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 62; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 62, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 62. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 62. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 62. 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). SEQ ID No 62 1 mvrrsisfcl fflratllcct scnsrslivh glpgreanei vvllvskgva aqklpqaaaa 61 tagaateqmw diavpsaqit ealailnqag lprmkgtsll dlfakqglvp selqekiryq 121 eglseqmast irkmdgvvda svqisftten ednlpltasv yik rgvldn pnsimvskik 181 rliasavpgl vpenvsvvsd raaysditin gp glteeid yvsvwgiila kssltkfrli 241 fyvlililfv iscgll vi kthtlimtmg gtkgffnptp ytknaleakk aegaaadkek 301 kedadsqges knaetsdkds sdkdapegsn eiega
(63) OmpA (CPn0695) One example of an OmPA encoded (MOMP) protein is set forth as SEQ ID NO: 63 below Genbank Accession No. GL4376998; AAD 18834.1. Preferred OmpA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 63; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 63, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmpA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 63. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 63. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 63. 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).
SEQ ID No 63 1 mkkllksall saafagsvgs lqalpvgnps dpsllidgti wegaagdpcd pcat cdais 61 lragfygdyv fdrilkvdap ktfs gakpt gsaaanytta vdrpnpaynk hlhdae ftn 121 agfialniwd rfdvfotlga sngyirgnst afnlvglfgv kgttvnanel pnvslsngvv 181 elytdtsfsw svgargal e cgcatlgaef qyaqskpkve elnvicnvsq fsvnkpkgyk 241 gvafplptda gvatatgtks atinyhewqv gaslsyrlns Ivpyigvqws ratfdadnir 301 iaqpklptav lnltawnpsl lgnatalstt dsfsdfmqiv scqinkfksr kacgvtvgat 361 lvdadkwslt aearlinera ahvsgqfrf
(64) Hypothetical (CPn0210)
One example of a Hypothetical Protein is set forth as SEQ ID NO: 64 below Genbank Accession No. G 4376482; AAD 18363.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 64; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 64, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 64. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 64. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 64. 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). SEQID No 64 1 mlvelealkr efahlkdqkp tsdqeitsly qcldhlefvl lglgqdkflk atededvlfe 61 sqkaidawna lltkardvlg lgdigaiyqt ieflgaylsk vnrrafcias ei flktair 121 dlnayylldf rwplckieef vdwgndcvei akrklctfek etkelnesll reehamekcs 181 iqdlqrklsd iiielhdvsl fcfsktpsqe eyqkdclyqs rlryllllye ytllcktstd 241 fqeqarakee firekfslle lekgikqtke lefaiakskl ergclvmrky eaaakhslds 301 mfeeetvksp rkdte
(65) Low Calcium Response Locus Protein H (CPnl 021) One example of a Low Calcium Response Protein H is set forth as SEQ ID NO: 65 below Genbank Accession No. GL4377352; AAD19158.1. Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 65; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 65, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These low calcium response proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 65. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 65 Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 65. 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).
SEQ ID No 65 1 mshlnyllek iaasskedfp fpddlesyle gyvpdknial dtyqkifkis sedlekvyke 61 gyhayldkdy aksitvfr l vffnpfvskf fslgaslhm seqysqalha ygvtavlrdk 121 dpyphyyayi cytltnehee aekale'mawv raqhkplyne lkeeildirk hk
Ninth Antigen Group The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group. Such other Chlamydia pneumoniae antigens include a ninth antigen group. These antigens are referred to herein as the "ninth antigen group". Each of the Chlamydia pneumoniae antigens of the ninth antigen group is described in more detail below.
(66) Low Calcium Response Protein D (CPn0323) One example of a Low Calcium Response Protein D is set forth as SEQ ID NO: 66 below Genbank Accession No. G 4376601; AAD18472.1. Preferred low calcium response proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 66; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 66, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These low calcium response proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 66. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 66. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 66. 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).
SEQ ID No 66 1 mnkllnfvsr tlggdtalnm inkssdlila l mmgvvl i iiplpppivd lmitinlsis 61 vfllmvalyi psalqlsvfp slllittmfr lginisssrq illkayaghv iqafgdfvvg 121 gny vgfiif liitiiqfiv vtkgaervae vaarfrldam pgkqmaidad lragmidatq 181 ardkraqiqk eselygamdg amkfikgdvi agivislini vggltigvam hgmdlaqaah 241 vytllsigdg lvsqipslli altagivttr vssdkntnlg keistqlvke pralllagaa 301 tlgvgffkgf pl sfsilal ifvalgilll tkksaagkkg ggsgasttvg aagdgaatvg 361 dnpddysltl pvilelgkdl skliqhktks gqsfvdd ip kmrqalyqdi girypgihvr 421 tdspslegyd ymillnevpy vrgkipphhv ltnevednls rynlpfityk naaglpsa v 481 sedakailek aaiky tple viilhlsyff hkssqeflgi qevrs iefm ersfpdlvke 541 vtrliplqkl teifkrlvqe qisikdlrti leslsewaqt ekdtvlltey vrsslklyis 601 fkfsqgqsai svylldpeie emirgaikqt sagsylaldp dsvnlilksm rntitptpag 661 gqppvlltai dvrryvrkli etefpdiavi syqeilpeir iqplgriqif
(67) CHLPS 43kDa Protein Homolog-1 (CPn0062) One example of a CHLPS 43kDa Protein Homolog-1 is set forth as SEQ ID NO: 67 below Genbank Accession No. GL4376318; AAD18215.1. Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 67; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 67, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 67. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 67. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 67. 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).
SEQ ID No 67 1 mmsskrtski avlsilltft hsigfanans svglgtvyit sevvkkpqkg serkqakkep 61 rarkgylvps srtlsaraqk mknssrkess ggcneisans tprsvklrrn kraeqkaakq 121 gfsafsnltl ksllpklpsk qktsiherek atsrfvnesq lssarkryct pssaapslfl 181 eteivrapve rtkelqdnei hip vqvqtn pkeqntkttk qlasqasiqq segteqslre 241 laqgaslpvl vrsnpevsvq rqkeellkel vaerrqckrk svrqalears ltkkvarggs 301 vtstlrydpe kaaeiksrrn ckvspeareq kyssckrdar angkqdkttp sedasqeeqq 361 tgaglvrktp ksqvasnaqn fyrnskntni dsyltanqys csseetdwpc sscvskrrth 421 nsisvctmvv tviamivgal iianatesqt tsdptpptpt p
(68) Hypothetical (CPn0169)
One example of a CHLPS 43kDa Protein Homolog-1 is set forth as SEQ ID NO: 68 below Genbank Accession No. G 4376437; AAD18322.1. Preferred CHLPS proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 68; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 68, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These CHLPS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 68. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 68. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 68. 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). SEQ ID No 68 1 mknvgsecsq plvmelntqp lrnlcesrlv kitsfviall alvggitlta lagagilsfl 61 pwlvlgivlv vlcalfllfs ykfcpikelg vvyntdsqih qwfqkqrnkd lekatenpel 121 fgenraednn rsarsqvket lrdcdgnvlk kiyernldvl lfmnwvpktrα ddvdpvsβds 181 irtviscykl ikackpefrs lisellrarαq sglgllsrcs ryqeraktvs hkdaplfcpt 241 hsyyrdgylt plragpryii nrai
(69) PmpD family (CPn0963) One example of a PmpD protein is set forth as SEQ ID NO: 69 below Genbank Accession No. GL4377287; AAD19099.1. Preferred PmpD proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 69; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 69, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These PmpD proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 69. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 69. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 69. 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).
SEQID No 69 1 mvakktvrsy rssfshsviv ailsagiafe ahslhsseld lgvfnkqfee hsahveeaqt 61 svlkgsdpvn psqkesekvl ytqvpltqgs sgesldlada nflehfqhlf eettvfgidq 121 klvwsdldtr nfsqptqepd tsnavsekis sdtkenrkdl etedpskksg lkevssdlpk 181 spetavaais edleisenis ardplqglaf fykntssqsi sekdssfqgi ifsgsgansg 241 lgfenlkapk sgaavysdrd ivfenlvkgl sfiscesled gsaagvni v thcgdvtltd 301 catgldleal rlvkdfsrgg avftarnhev qrmlaggils vgnkgaiw eknsaeksng 361 gafacgsfvy snnentalwk enqalsggai ssasdidiqg ncsaiefsgn qslialgehi 421 gltdfvggga laaqgtltlr nnavvqcvkn tskthggail agtvdlneti sevafkqnta 481 altggalsan dkviiannfg eilfeqnevr nhggaiycgc rsnpkleqkd sgeniniign 541 sgaitflknk asvlevmtqa edyagggalw ghnvlldsns gniqfignig gstfwigeyv 601 gggailstdr vtisnnsgdv vfkgnkgqcl aqkyvapqet apvesdasst nkdekslnac 661 shgdhyppkt veeevppsll eehpvvsstd irgggailaq hifitdntgn lrfsgnlggg 721 eesstvgdla ivgggallst nevnvcsnqn vvfsdnvtsn gcdsggaila kkvdisanhs 781 vefvsngsgk fggavcalne svnitdngsa vsfsknrtrl ggagvaapqg svticgnqgn 841 iafkenfvfg senqrsggga iianssvnlq dnagdilfvs nstgsyggai fvgslvaseg 901 snprtltitg nsgdilfakn stqtaaslse kdsfgggaiy tqnlkivkna gnvsfygnra 961 psgagvqiad ggtvcleafg gdilfegnin fdgsfnaihl cgndskivel savqdkniif 1021 qdaityeent irglpdkdvs plsapslifn skpqddsaqh hegtirfsrg vskipqiaai 1081 qegtlalsqn ael laglkq etgssivlsa gsilrifdsq vdssaplpte nkeetlvsag 1141 vqinmssptp nkdkavdtpv ladiisitvd lssfvpeqdg tlplppeiii pkgtklhsna 1201 idlkiidptn vgyenhalls shkdiplisl ktaegmtgtp tadaslsnik idvslpsitp 1261 atyghtgv s eskmedgrlv vg qptgykl npekqgalvl nnl shytdl ralkqeifah 1321 htiaqrraeld fstnvwgsgl g vedcqnig efdgfkhhlt gyalgldtql vedfliggcf 1381 sqffgktesq sykakndvks ymgaayagil agpwlikgaf vygnirmdlt tdygtlgist 1441 gswigkgfia gtsidyryiv nprrfisaiv stvvpfveae yvridlpeis eqgkevrtfq 1501 ktrfenvaip fgfalehays rgsraevnsv qlayvfdvyr kgpvslitlk daayswksyg 1561 vdipckawka rlsnnte ns ylstylafny e redliayd fnggiriif
Tenth Antigen Group
The immunogenicity of other Chlamydia pneumoniae antigens may be improved by combination with two or more Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group. Such other Chlamydia pneumoniae antigens include a tenth antigen group. Each of the Chlamydia pneumoniae antigens of the tenth antigen group is described in more detail below.
(70) OmpH-like outer membrane protein (CPn0301)
One example of 'OmpH-like' protein is disclosed as SEQ ID NOs: 77 & 78 in WO 02/02606. {GenBank accession number: gi|4376577|gb|AAD18450.1| 'CPn0301'; SEQ ID NO: 70 below and SEQ ID No 4 above}. Preferred OmpH-like proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 4; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 3, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmpH-like proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 4. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 4. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 4. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
SEQ ID No 70 1 MK LLFSTFL LVLGSTSAAH ANLGYVNLKR CLEESDLGKK ETEELEAMKQ 51 QFVKNAEKIE EELTSIYNKL QDEDYMESLS DSASEELRKK FEDLSGEYNA 101 YQSQYYQSIN QSNVKR1QKL IQEVKIAAES VRSKEKLEAI LNEEAVLAIA 151 PGTDKTTEII AILNESFKKQ N*
(71) L7/L12 Ribosomal Protein (CPn0080) One example of an L7/L12 Ribosomal protein is set forth as SEQ ID No 71 below{GenBank accession number: GL4376338; AAD18233.1}. 'CPn0080'; SEQ ID NO: 71 below. Preferred L7/L12 proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g. 60%>, 65%>, 70%>, 75%o, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 71; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 71, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These L7/L12 ribosomal proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 71. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 71. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 19 or more, to remove the signal peptide) from the N-terminus of SEQ ID NO: 71. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). SEQ ID No 71 1 mttesletlv eklsnltvle lsqlkkllee k dvtasapv vavaagggge apvaaeptef 61 avtledvpad kkigvlk vr evtglalkea kemteglpkt vkektsksda edtvkklqda 121gakasfkgl
(72) AtoS two-component regulatory system sensor histidine kinase protein (CPn0584)
One example of 'AtoS' protein is disclosed as SEQ ID NOs: 105 & 106 in WO 02/02606. {GenBank accession number: gi|4376878|gb|AAD18723.1| 'CPn0584'; SEQ ID NO: 72 below and SEQ ID No 9 above}. Preferred AtoS proteins for use with the invention comprise an amino acid sequence: (a) having 50%> or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 72; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 72, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These AtoS proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 72. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 72. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 72. 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).
SEQ ID No 72 1 MNVPDSKNLH PPAYELLEIK ARITQSYKEA SAILTAIPDG ILLLSETGHF 51 LICNSQAREI LGIDENLEIL NRSFTDVLPD TCLGFSIQEA LESLKVPKTL 101 RLSLCKESKE KEVELFIRKN EISGYLFIQI RDRSDYKQLE NAIERYKNIA 151 ELGKMTATLA HEIRNPLSGI VGFASILKKE ISSPRHQRML SSIISGTRSL 201 NNLVSSMLEY TKSQPLNLKI INLQDFFSSL IPLLSVSFPN CKFVREGAQP 251 LFRSIDPDRM NSWWNLVKN AVETGNSPIT LTLHTSGDIS VTNPGTIPSE 301 IMDKLFTPFF TTKREGNGLG LAEAQKIIRL HGGDIQLKTS DSAVSFFIII 351 PELLAALPKE RAAS*
(73) OmcA 9kDa-cysteine-rich Upoprotein(CPn0558)
One example of OmcA' protein is disclosed as SEQ ID NOs: 9 & 10 in WO 02/02606. {GenBank accession number: gi|4376850|gb|AAD18698.1| 'CPn0558', 'OmcA', 'Omp3'; SEQ ID NO: 73 below and SEQ ID No 10 above}. Preferred OmcA proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 73; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 73, wherein n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These OmcA proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 73. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 73. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more; preferably 18 or more to remove the signal peptide) from the N-terminus of SEQ ID NO: 73. Other fragments omit one or more domains of the protein (e.g. omission of a signal peptide as described above, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain). The protein may be lipidated (e.g. by a N-acyl diglyceride), and may thus have a Ν-terminal cysteine.
SEQID No 73 1 MKKAVLIAAM FCGWSLSSC CRIVDCCFED PCAPSSCΝPC EVIRKKERSC 51 GGΝACGSYVP SCSΝPCGSTE CΝSQSPQVKG CTSPDGRCKQ *
(74) Hypothetical (CPn0331) One example of a hypothetical protein is set forth as SEQ ID NO: 74 below and SEQ ID No 57 above. Genbank Accession No. GI.4376609; AAD 18480.1. Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 74; and/or (b) which is a fragment of at least n consecutive amino acids of SEQ ID NO: 74, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 74. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 74. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C-terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 74. 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).
SEQ ID NO 74 1 mavsggggvq pssdpgk np alqgeqaegp splkesifse tkqassaakq eslvrsgstg 61 myatesqink akyrkaqdrs stspksklkg tfskmrasvq gfmsgfgsra srvsakrasd 121 sgegtsllpt emdvalkkgn rispemqgff ldasg ggss sdisqlslea lkssafsgar 181 slslsssess svasfgsfqk aiepmseekv na tvarlgg emvsslldpn vetsslvrra 241 matgnegmid lsdlgqeevs tamtsprave gkvkvsssds peanptgipn sntleraeke 301 aekqesreql sedqmrαlara aglltgaap qevlsnsvws gpstvfpppk fsgtlptqrs 361 gdkskhkspg iekstnhtnf splregtvks aevkslphpe smyrfpkdsi vsreepea v 421 kestafknpe nssqnflpia vesvfpkesg tggalgsdav sssyhflaqr gvsllaplpr 481 atddykekle ahkgpggppd pliyqyrnva veppivlrsp qpfsgssrls vqgkpeaasv 541 hddggggnsg gfsgdqrrgs sgqkasrqek kgkklstdi
(75) PmpD family (CPn0963)
One example of a PmpD protein is set forth as SEQ ID NO: 75 below Genbank Accession No. GI.4377287; AAD19099.1. Preferred PmpD proteins for use with the invention comprise an amino acid sequence: (a) having 50%) or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%>, 99.5% or more) to SEQ ID NO: 75; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 75, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 75. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 75. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 75. 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). SEQ ID No 75 1 mvakktvrsy rssfshsviv ailsagiafe ahslhsseld Igvfnkqfee hsahveeaqt 61 svlkgsdpvn psqkesekvl ytqvpltqgs sgesldlada nflehfqhlf eettvfgidq 121 klvwsdldtr nfsqptqepd tsnavsekis sdtkenrkdl etedpskksg lkevssdlpk 181 spetavaais edleisenis ardplqglaf fykntssqsi sekdssfqgi ifsgsgansg 241 lgfenlkapk sgaavysdrd ivfenlvkgl sfiscesled gsaagvni v thcgdvtltd 301 catgldleal rlvkdfsrgg avftarnhev qnnlaggils vvgnkgaiw eknsaeksng 361 gafacgsfvy snnental k enqalsggai ssasdidiqg ncsaiefsgn qslialgehi 421 gltdfvggga laaqgtltlr nnavvqcvkn tskthggail agtvdlneti sevafkqnta 481 altggalsan dkviiannfg eilfeqnevr nhggaiycgc rsnpkleqkd sgeniniign 541 sgaitflknk asvlevmtqa edyagggalw ghnvlldsns gniqfignig gstfwigeyv 601 gggailstdr vtisnnsgdv vfkgnkgqcl aqkyvapqet apvesdasst nkdekslnac 661 shgdhyppkt veeevppsll eehp vsstd irgggailaq hifitdntgn lrfsgnlggg 721 eesstvgdla ivgggallst nevnvcsnqn vfsdnvtsn gcdsggaila kkvdisanhs 781 vefvsngsgk fggavcalne svnitdngsa vsfsknrtrl ggagvaapqg svticgnqgn 841 iafkenfvfg senqrsggga iianssvniq dnagdilfvs nstgsyggai fvgslvaseg 901 snprtltitg nsgdilfakn stqtaaslse kdsfgggaiy tqnlkivkna gnvsfygnra 961 psgagvqiad ggtvcleafg gdilfegnin fdgsfnaihl cgndskivel savqdkniif 1021 qdaityeent irglpdkdvs plsapslifn skpqddsaqh hegtirfsrg vskipqiaai 1081 qegtlalsqn ael laglkq etgssivlsa gsilrifdsq vdssaplpte nkeetlvsag 1141 vqinmssptp nkdkavdtpv ladiisitvd Issfvpeqdg tlplppeiii pkgtklhsna 1201 idlkiidptn vgyenhalls shkdiplisl ktaegmtgtp tadaslsnik idvslpsitp 1261 atyghtgv s eskmedgrlv vgwqptgykl npekqgalvl nnl shytdl ralkqeifah 1321 htiaqr eld fstnv gsgl g vedcqnig efdgfkhhlt gyalgldtql vedfliggcf 1381 sqffgktesq sykakndvks ymgaayagil agp likgaf vygninndlt tdygtlgist 1441 gs igkgfia gtsidyryiv nprrfisaiv stvvpfveae yvridlpeis eqgkevrtfq 1501 ktrfenvaip fgfalehays rgsraevnsv qlayvfdvyr kgpvslitlk daayswksyg 1561 vdipckawka rlsnntewns ylstylafny e redliayd fnggiriif
(76) Hypothetical (CPn0798)
One example of a hypothetical protein is set forth as SEQ ID NO: 78 below. GenBank Accession No. GL4377109; AAD 18936 Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%,, 99.5% or more) to SEQ ID NO: 78; and/or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 78, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 78. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 78. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and/or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 78. Other fragments omit one or rnore domains of the protein (e.g. omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, or of an extracellular domain).
SEQ ID No 78 1 mkktccqnyr sigvvfs vl fvlttqtlfa ghfidigtsg lyswargvsg dgrv vgyeg 61 gnafkyvdge kflleglvpr sealvfkasy dgsviigisd qdpscravkw vngalvdlgi 121 fsegmqsfae gvssdgktiv gclysddtet nfavk dβtg mvvlpnlped rhscawdase 181 dgsvivgdam gseeiakavy wkdgeqhlls nipgakrssa havskdgsfi vgefiseene 241 vhafvyhngv ikdigtlggd ysvatgvsrd gkvivghstr tdgeyrafky vdgrmidlgt 301 lggsasfafg vsddgktivg kfetelgec afiyldd
(77) Hypothetical (CPn0799) One example of a hypothetical protein is set forth as SEQ ID NO: 79 below. GenBank Accession No. GI: 15618708; AAD18937 Preferred hypothetical proteins for use with the invention comprise an amino acid sequence: (a) having 50%, or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%,, 99%, 99.5% or more) to SEQ ID NO: 79; and or (b) which is a fragment of at least ra consecutive amino acids of SEQ ID NO: 79, wherein ra is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more). These hypothetical proteins include variants (e.g. allelic variants, homologs, orthologs, paralogs, mutants, etc.) of SEQ ID NO: 79. Preferred fragments of (b) comprise an epitope from SEQ ID NO: 79. Other preferred fragments lack one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the C- terminus and or one or more amino acids (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 or more) from the N-terminus of SEQ ID NO: 79. 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).
SEQ ID No 79 1 maaikqilrs mlsqsslwmv lfslyslsgy cyvitdkped dfhsssavkw dh gkttlsr 61 lsnkkasaka vsgtgattvg fikdt srty avr ny gtk elptss vkk skatgissdg 121 siiagivene lsqsfavtwk nnemyllpst avqskaygi ssdgsvivgs akda srtfa 181 vkwtgheaqv lpvgwavksv ansvsangsi ivgsvqdasg ilyavk egn tithlgtlgg 241 ysaiakavsn ngkvivgrse tyygevhafc hkngvmsdlg tlggsysaak gvsatgkviv 301 gmsttangkl hafkyvggrm idlgeyswke acanavsidg eiivgvqse
Preferably the composition of the invention comprises a combination of CPn antigens selected from the group consisting of: (1) CPn0301 and CPn0080; (2) CPn 0584 and CPn 0558; and (3) CPn 0331 and CPN 0963. Preferably the composition comprises a combination of any one or more of groups (1), (2) and (3).
Even more preferably, the composition of the present invention comprises a combination of CPn antigens selected from the group consisting of (1) CPn0385, CPn0324, CPn 0503, CPn0525 and CPn 0482. Preferably the composition is administered in the presence of alum and/or cPG.
The invention thus includes a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or six Chlamydia pneumoniae antigens of the first antigen group and two, three, four, five, or six Chlamydia pneumoniae antigens of the second antigen group. Preferably, the combination is selected from the group consisting of three, four, five or six Chlamydia pneumoniae antigens from the first antigen group and three, four, five or six Chlamydia pneumoniae antigens from the second antigen group. Still more preferably, the combination consists of six Chlamydia pneumoniae antigens from the first antigen group and three, four, five or six, Chlamydia pneumoniae antigens from the second antigen group.
The invention further includes a composition comprising a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or six, Chlamydia pneumoniae antigens of the second antigen group and two, three, four, five, six, seven or eight Chlamydia pneumoniae antigens of the third antigen group. Preferably, the combination is selected from the group consisting of three, four, five or six Chlamydia pneumoniae antigens from the second antigen group and three, four, five, six, seven or eight Chlamydia pneumoniae from the third antigen group. Still more preferably, the combination consists of six Chlamydia pneumoniae antigens from the second antigen group and three, four, five, six, seven or eight Chlamydia pneumoniae antigens of the third antigen group.
There is an upper limit to the number of Chlamydia pneumoniae antigens which will be in the compositions of the invention. Preferably, the number of Chlamydia pneumoniae antigens 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 Chlamydia pneumoniae antigens in a composition of the invention is less than 6, less than 5, or less than 4. The Chlamydia pneumoniae antigens 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.
In either of the above combinations, preferably the composition comprises one or more Chlamydia pneumoniae antigens from the fourth antigen group which includes porB. Or, alternatively, in either of the above combinations, preferably the
Chlamydia pneumoniae antigens from the fourth antigen group includes one or more members of the pmp3 family.
Other aspects of the present invention are presented in the accompanying claims and in the following description and drawings. These aspects are presented under separate section headings. However, it is to be understood that the teachings under each section are not necessarily limited to that particular section heading.
Before describing the present invention in detail, it is to be understood that this invention is not limited to particularly exemplified molecules or process parameters as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting. In addition, the practice of the present invention will employ, unless otherwise indicated, conventional methods of virology, microbiology, molecular biology, recombinant DNA techniques and immunology all of which are within the ordinary skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, et al., Molecular Cloning: A Laboratory Manual
(2nd Edition, 1989); DNA Cloning: A Practical Approach, vol. I & II (D. Glover, ed.); Oligonucleotide Synthesis (N. Gait, ed., 1984); A Practical Guide to Molecular Cloning (1984); and Fundamental Virology, 2nd Edition, vol. I & II (B.N. Fields and
D.M. Knipe, eds.).
All publications, patents and patent applications cited herein, whether supra or infra, are hereby incoφorated by reference in their entirety. It must be noted that, as used in this specification and the appended claims, the singular forms "a", "an" and "the" include plural referents unless the content clearly dictates otherwise. All scientific and technical terms used in this application have meanings commonly used in the art unless otherwise specified. As used in this application, the following words or phrases have the meanings specified. 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.
IMMUNE RESPONSE
The mechanism by which the immune system controls disease includes the induction of neutralising antibodies via humoral immunity and the generation of T-cell responses via cellular immunity. As used herein, the term "immune response" against an antigen refers to the development in a host mammalian subject of a humoral and/or a cellular immune response against that antigen.
As used herein, the term "humoral immune response" refers to an immune response mediated by antibody molecules. The antibodies generated by humoral immunity are primarily effective against extracellular infectious agents.
SEQ ID Nos 1-86 in the compositions of the invention may be supplemented or substituted with an antibody that binds to the protein. This antibody may be monoclonal or polyclonal.
As used herein, the term "cell mediated immune (CMI) response" is one mediated by T-lymphocytes and/or other white blood cells. The CMI immune mechanisms are generally more effective against intracellular infections and disease because the CMI mechanisms prime T cells in a way that, when an antigen appears at a later date, memory T cells are activated to result in a CMI response that destroys target cells that have the corresponding antigen or a portion thereof on their cell surfaces, and thereby the infecting pathogen. The CMI response is focused on the destruction of the source of infection mediated by either effector cells that destroy infected cells of the host by direct cell-to-cell contact and/or by the release of molecules, such as cytokines, that possess anti-viral activity. Thus the CMI response, which is characterised by a specific T lymphocyte cellular response, is crucial to produce resistance to diseases caused by cancer, viruses, pathogenic and other intracellular microorganisms. In one aspect of the present invention, an immunogenic composition is provided comprising a combination of at least one antigen that elicits a Chlamydia pneumoniae specific Thl immune response (such as a cell mediated or cellular immune response) and at least one antigen that elicits a Chlamydia pneumoniae specific Th2 response (such as a humoral or antibody response). The immunogenic composition may further comprise a Thl adjuvant and a Th2 adjuvant. In one embodiment, the invention provides a composition comprising a combination of Chlamydia pneumoniae antigens that elicit at least a Chlamydia pneumoniae specific Thl immune response. As an example, the combination of Chlamydia pneumoniae antigens may include at least one antigen associated with reticulate bodies (RBs) of Chlamydia pneumoniae, including but not limited to antigens expressed, exposed on or translocated into, through or across on the inclusion membrane, antigens expressed, secreted, released or translocated into the cytosol of host cells, or antigens processed or degraded in the cytosol of host cells and/or expressed, exposed or presented on the surface of the host cell. The compositions of the invention will preferably elicit both a cell mediated immune response as well as a humoral immune response in order to effectively address a Chlamydia intracellular infection. This immune response will preferably induce long lasting (eg neutralising) antibodies and a cell mediated immunity that can quickly respond upon exposure to Chlamydia.
The invention also comprises an immunogenic composition comprising one or more immunoregulatory agents. Preferably, one or more of the immunoregulatory agents include an adjuvant. The adjuvant may be selected from one or more of the group consisting of a Thl adjuvant and Th2 adjuvant, further discussed below. The adjuvant may be selected from the group consisting of a mineral salt, such as an aluminium salt and an oligonucleotide containing a CpG motif. Most preferably, the immunogenic composition includes both an aluminium salt and an oligonucleotide containing a CpG motif. Use of the combination of a mineral salt, such as an aluminium salt, and an oligonucleotide containing a CpG motif provide for an enhanced immune response. This improved immune response is wholly unexpected and could not be predicted from the use of either agent alone. The invention therefore includes an oligonucleotide containing a CpG motif, a mineral salt such as an aluminium salt, and an antigen, such as a Chlamydia pneumoniae antigen.
T CELLS IMPLICATED IN THE CMI RESPONSE
At least two special types of T cells are required to initiate and/or to enhance CMI and and humoral responses. The antigenic receptors on a particular subset of T cells which express a CD4 co-receptor can be T helper (Th) cells or CD4 T cells (herein after called T helper cells) and they recognise antigenic peptides bound to MHC class II molecules. In contrast, the antigenic receptors on a particular subset of T cells which express a CD8 co-receptor are called Cytotoxic T lymphocytes (CTLs) or CD8+ T cells (hereinafter called CD8+ T cells) and they react with antigens displayed on MHC Class I molecules.
HELPER T CELLS
Helper T cells or CD4+ cells can be further divided into two functionally distinct subsets: Thl and Th2 which differ in their cytokine and effector function. Thl and Th2 responses have been shown to be regulated not only in a positive but also in a negative way such that Thl cellular responses are augmented by Thl cytokines such as IL-2, IL-12 and IFN-gamma and decreased by Th2 cytokines such as E -4 and IL- 10. In contrast, antibody responses are enhanced by Th2 cytokines such as IL-4 and IL-10 but are downregulated by Thl cytokines such as IFN-gamma and another cytokine IL-12 that enhances IFN-gamma and is produced by monocytes. Thus, classic Thl cytokines such as IFN-gamma, IL-2 and IL-12 can be regarded as immune co-factors that induce an effective inflammatory response. In contrast, the classic Th2 cytokines such as IL-4 and IL-10 can be regarded as cytokines that will suppress a severe inflammatory response. CD8+ T CELLS
CD8+ T cells may function in more than one way. The best known function of CD8+ T cells is the killing or lysis of target cells bearing peptide antigen in the context of an MHC class I molecule. Hence the reason why these cells are often termed cytotoxic T lymphocytes (CTL). However, another function, perhaps of greater protective relevance in certain infections is the ability of CD8+ T cells to secrete interferon gamma (IFN-gamma). Thus assays of lytic activity and of IFN-gamma release are both of value in measuring CD8+ T cell immune response (eg in an ELISPOT assay as set forth below). In infectious diseases there is evidence to suggest that CD8+ T cells can protect by killing an infectious agent comprising an infectious antigen at the early stages of a disease before any symptoms of disease are produced.
ENHANCED CMI RESPONSE
The present invention concerns methods, processes and compositions capable of enhancing and or modulating the CMI response in a host subject against a target antigen. As used herein, the term "enhancing" encompasses improvements in all aspects of the CMI response which include but are not limited to a stimulation and/or augmentation and/or potentiation and/or up-regulation of the magnitude and/or duration, and/or quality of the CMI response to an antigen or a nucleotide sequence encoding an antigen of interest. By way of example, the CMI response may be enhanced by either (i) enhancing the activation and/or production and/or proliferation of CD 8+ T cells that recognise a target antigen and/or (ii) shifting the CMI response from a Th2 to a Thl type response. This enhancement of the Thl associated responses is of particular value in responding to intracellular infections because, as explained above, the CMI response is enhanced by activated Thl (such as, for example, IFN- gamma inducing) cells.
Such an enhanced immune response may be generally characterized by increased titers of interferon-producing CD4+ and/or CD8+ T lymphocytes, increased antigen- specific CD8+ T cell activity, and a T helper 1-like immune response (Thl) against the antigen of interest (characterized by increased antigen-specific antibody titers of the subclasses typically associated with cellular immunity (such as, for example IgG2a), usually with a concomitant reduction of antibody titers of the subclasses typically associated with humoral immunity (such as, for example IgGl)) instead of a T helper 2-like immune response (Th2).
The enhancement of a CMI response may be determined by a number of well-known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject (see, for example, Erickson et al. (1993) J. Immunol. 151: 4189-4199; and Doe et al. (1994) Eur. J. Immunol. 24: 2369-2376) or CD8+ T cell ELISPOT assays for measuring Interferon gamma production (Miyahara et al PNAS(USA) (1998) 95: 3954-3959). ENHANCED T-CELL RESPONSE
As used herein, the term "enhancing a T -cell response" encompasses improvements in all aspects of the T-cell response which include but are not limited to a stimulation and or augmentation and or potentiation and/or up-regulation of the magnitude and/or duration, and/or quality of the T-cell response to an antigen (which may be repeatedly administered) or a nucleotide sequence encoding an antigen. The antigen may be a Chlamydia antigen, preferably a Chlamydia pneumoniae antigen. By way of example, the T-cell response may be enhanced by either enhancing the activation and/or production and/or distribution and/or proliferation of the induced T-cells and/or longevity of the T-cell response to T-cell inducing/modulating antigen or nucleotide sequence encoding an antigen. The enhancement of the T-cell response in a host subject may be associated with the enhancement and/or modulation of the Thl immune response in the host subject. The enhancement of the T-cell response may be determined by a number of well- known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T-cell cytotoxic cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject (see, for example, Erickson et al. (1993) J. Immunol. 151: 4189-4199; and Doe et al. (1994) Eur. J. Immunol. 24: 2369-2376) or CD8+ T-cell ELISPOT assays for measuring Interferon gamma production (Miyahara et al PNAS(USA) (1998) 95: 3954-3959).
Activated Thl cells enhance cellular immunity (including an increase in antigen- specific CTL production) and are therefore of particular value in responding to intracellular infections. Activated Thl cells may secrete one or more of IL-2, IFN- gamma, and TNF-beta. A Thl immune response may result in local inflammatory reactions by activating macrophages, NK (natural killer) cells, and CD8 cytotoxic T cells (CTLs). A Thl immune response may also act to expand the immune response by stimulating growth of B and T cells with IL-12. Thl stimulated B cells may secrete IgG2a.
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 IgGl, IgE, IgA and memory B cells for future protection.
ANTIGEN
Each disease causing agent or disease state has associated with it an antigen or immunodominant epitope on the antigen which is crucial in immune recognition and ultimate elimination or control of a disease causing agent or disease state in a host. In order to mount a humoral and/or cellular immune response against a particular disease, the host immune system must come in contact with an antigen or an immunodominant epitope on an antigen associated with that disease state. As used herein, the term "antigen" refers to any agent, generally a macromolecule, which can elicit an immunological response in an individual. The term "antigen" is used interchangeably with the term "immunogen". The immunological response may be of B- and/or T-lymphocytic cells. The term may be used to refer to an individual macromolecule or to a homogeneous or heterogeneous population of antigenic macromolecules. As used herein, "antigen" is used to refer to a protein molecule or portion thereof which contains one or more antigenic determinants or epitopes. As used herein, the term "antigen" means an immunogenic peptide or protein of interest comprising one or more epitopes capable of inducing a CMI response to an infectious Chlamydia pathogen. The antigen can mclude but is not limited to an auto-antigen, a self-antigen, a cross-reacting antigen, an alloantigen, a tolerogen, an allergen, a hapten, an immunogen or parts thereof as well as any combinations thereof.
EPITOPE
As used herein, the term "epitope" generally refers to the site on an antigen which is recognised by a T-cell receptor and/or an antibody. Preferably it is a short peptide derived from or as part of a protein antigen. However the term is also intended to include peptides with glycopeptides and carbohydrate epitopes. Several different epitopes may be carried by a single antigenic molecule. The term "epitope" also includes modified sequences of amino acids or carbohydrates which stimulate responses which recognise the whole organism. It is advantageous if the selected epitope is an epitope of an infectious agent, such as a Chlamydia bacterium, which causes the infectious disease.
SEQ ID Nos 1-86 in the compositions of the invention may be supplemented or substituted with molecules comprising fragments of SEQ ID Nos 1-86. Such fragments may comprise at least n consecutive monomers from the molecules and. depending on the particular sequence, n is either (i) 7 or more for protein molecules (eg. 8 18, 20 or more), preferably such that the fragment comprises an epitope from the sequence, or (ii) 10 or more for nucleic acid molecules (eg 15, 18, 20, 25, 30, 35, 40 or more).
SOURCE OF EPITOPES
The epitope can be generated from knowledge the amino acid and corresponding
DNA sequences of the peptide or polypeptide, as well as from the nature of particular amino acids (e.g., size, charge, etc.) and the codon dictionary, without undue experimentation. See, e.g., Ivan Roitt, Essential Immunoloqy, 1988; Kendrew, supra; Janis Kuby, Immunology, 1992 e.g., pp. 79-81. Some guidelines in determining whether a protein will stimulate a response, include: Peptide length — preferably the peptide is about 8 or 9 amino acids long to fit into the MHC class I complex and about 13-25 amino acids long to fit into a class II MHC complex. This length is a minimum for the peptide to bind to the MHC complex. It is preferred for the peptides to be longer than these lengths because cells may cut peptides. The peptide may contain an appropriate anchor motif which will enable it to bind to the various class I or class II molecules with high enough specificity to generate an immune response (See Bocchia, M. et al, Specific Binding of Leukemia Oncogene Fusion Protein Pentides to HLA Class I Molecules, Blood 85:2680-2684; Englehard, NH, Structure of peptides associated with class I and class II MHC molecules Ann. Rev. Immunol. 12:181 (1994)). This can be done, without undue experimentation, by comparing the sequence of the protein of interest with published structures of peptides associated with the MHC molecules. Thus, the skilled artisan can ascertain an epitope of interest by comparing the protein sequence with sequences listed in the protein data base.
T CELL EPITOPES
Preferably one or more antigens of the present invention contain one or more T cell epitopes. As used herein, the term "T cell epitope" refers generally to those features of a peptide structure which are capable of inducing a T cell response. In this regard, it is accepted in the art that T cell epitopes comprise linear peptide determinants that assume extended conformations within the peptide-binding cleft of MHC molecules (Unanue et al. (1987) Science 236: 551-557). As used herein, a T cell epitope is generally a peptide having at least about 3-5 amino acid residues, and preferably at least 5-10 or more amino acid residues. However, as used herein, the term "T cell epitope" encompasses any MHC Class I-or MHC Class II restricted peptide. The ability of a particular T cell epitope to stimulate/enhance a CMI response may be deteπnined by a number of well-known assays, such as by lymphoproliferation (lymphocyte activation) assays, CD8+ T-cell cytotoxic cell assays, or by assaying for T-lymphocytes specific for the epitope in a sensitized subject. See, e. g., Erickson et al. (1993) J. Immunol. 151: 4189-4199; and Doe et al. (1994) Eur. J. Immunol. 24: 2369-2376 or CD8+ T-cell ELISPOT assays for measuring Interferon gamma production (Miyahara et al PNAS(USA) (1998) 95: 3954-3959). CD8+ T-CELL EPITOPES
Preferably the antigens of the present invention comprisse CD8+ T-cell inducing epitopes. A CD8+ T-cell -inducing epitope is an epitope capable of stimulating the formation, or increasing the activity, of specific CD 8+ T-cells following its administration to a host subject. The CD8+ T-cell epitopes may be provided in a variety of different forms such as a recombinant string of one or two or more epitopes. CD8+ T-cell epitopes have been identified and can be found in the literature, for many different diseases. It is possible to design epitope strings to generate CD8+ T- cell response against any chosen antigen that contains such CD8+ T-cell epitopes. Advantageously, CD8+ T-cell inducing epitopes may be provided in a string of multiple epitopes which are linked together without intervening sequences so that unnecessary nucleic acid material is avoided.
T HELPER EPITOPES
Preferably the antigens of the present invention comprise helper T lymphocyte epitopes. Various methods are available to identify T helper cell epitopes suitable for use in accordance herewith. For example, the amphipathicity of a peptide sequence is known to effect its ability to function as a T helper cell inducer. A full discussion of T helper cell-inducing epitopes is given in U.S. Patent 5,128,319, incoφorated herein by reference.
B CELL EPITOPES
Preferably the antigens of the present invention comprise a mixture of CD8+ T-cell epitopes and B cell epitopes. As used herein, the term "B cell epitope" generally refers to the site on an antigen to which a specific antibody molecule binds. The identification of epitopes which are able to elicit an antibody response is readily accomplished using techniques well known in the art. See, e. g., Geysen et al. (1984) Proc. Natl. Acad. Sci. USA 81: 3998-4002 (general method of rapidly synthesizing peptides to determine the location of immunogenic epitopes in a given antigen); U. S. Patent No. 4,708,871 (procedures for identifying and chemically synthesizing epitopes of antigens); and Geysen et α/.(1986) Molecular Immunology 23: 709-715 (technique for identifying peptides with high affinity for a given antibody).
COMBINATION OF EPITOPES In a preferred embodiment of the present invention, the antigen or antigen combination comprises a mixture of a CD8+ T-cell -inducing epitopes and a T helper cell-inducing epitopes. As is well known in the art, T and B cell inducing epitopes are frequently distinct from each other and can comprise different peptide sequences. Therefore certain regions of a protein's peptide chain can possess either T cell or B cell epitopes. Therefore, in addition to the CD8+ T-cell epitopes, it may be preferable to include one or more epitopes recognised by T helper cells, to augment the immune response generated by the CD8+ T-cell epitopes.
The mechanism of enhancing a CD8+ T-cell induced response in vivo by T helper cell inducing agents is not completely clear. However, without being bound by theory, it is likely that the enhancing agent, by virtue of its ability to induce T helper cells, will result in increased levels of necessary cytokines that assist in the clonal expansion and dissemination of specific CD8+ T-cells. Regardless of the underlying mechanism, it is envisioned that the use of mixtures of helper T cell and CD8+ T-cell -inducing antigen combinations of the present invention will assist in the enhancement of the CMI response. Particularly suitable T helper cell epitopes are ones which are active in individuals of different HLA types, for example T helper epitopes from tetanus (against which most individuals will already be primed). It may also be useful to include B cell epitopes for stimulating B cell responses and antibody production. Synthetic nucleotide sequences may also be constructed to produce two types of immune responses: T cell only and T cell combined with a B cell response.
IMMUNODOMINANT EPITOPE
When an individual is immunized with an antigen or combination of antigens or nucleotide sequence or combinations of nucleotide sequences encoding multiple epitopes of a target antigen, in many instances the majority of responding T lymphocytes will be specific for one or more linear epitopes from that target antigen and/or a majority of the responding B lymphocytes will be specific for one or more linear or conformational epitopes for the antigen or combination of antigens.. For the puφoses of the present invention, then, such epitopes are referred to as "immunodominant epitopes". In an antigen having several immunodominant epitopes, a single epitope may be the most dominant in terms of commanding a specific T or B cell response.
As the Examples show, at least sixteen peptides of the present invention were recognised by IFN-gamma positive CD8+ T cell populations which were actually expanded as a result of bacterial infection.
ADJUVANTS
The compositions of the present invention may be administered in conjunction with other immunoregulatory agents. In particular, the compositions of the present i .nnvvpe/nnttiinonn m m-avy h bie. a aHdmmi.nni.ssttfe»rrfevd. w wiithh a ann . ai_d.j_u_ _vv.ainntt.
The inclusion of an adjuvant and in particular, a genetic adjuvant may be useful in further enhancing or modulating the CMI response. An adjuvant may enhance the CMI response by enhancing the immunogenicity of a co-administered antigen in an immunized subject, as well inducing a Thl-like immune response against the co- administered antigen which is beneficial in a vaccine product.
An immune response and particularly a CMI response may be refined, by the addition of adjuvants to combinations of antigens or nucleotide sequences encoding combinations of antigens which lead to particularly effective compositions for eliciting a long lived and sustained enhanced CMI response.
As used herein, the term "adjuvant" refers to any material or composition capable of specifically or non-specifically altering, enhancing, directing, redirecting, potentiating or initiating an antigen-specific immune response.
The term "adjuvant" includes but is not limited to a bacterial ADP-ribosylating exotoxin, a biologically active factor, immunomodulatory molecule, biological response modifier or immunostimulatory molecule such as a cytokine, an interleukin, a chemokine or a ligand or an epitope (such as a helper T cell epitope) and optimally combinations thereof which, when administered with an antigen, antigen composition or nucleotide sequence encoding such antigens enhances or potentiates or modulates the CMI response relative to the CMI response generated upon administration of the antigen or combination of antigens alone. The adjuvant may be any adjuvant known in the art which is appropriate for human or animal use.
Immunomodulatory molecules such as cytokines (TNF-alpha, IL-6, GM-CSF, and IL- 2), and co-stimulatory and accessory molecules (B7-1, B7-2) may be used as adjuvants in a variety of combinations. In one embodiment GM-CSF is not administered to subject before, in or after the administration regimen. Simultaneous production of an immunomodulatory molecule and an antigen of interest at the site of expression of the antigen of interest may enhance the generation of specific effectors which may help to enhance the CMI response. The degree of enhancement of the CMI response may be dependent upon the specific immunostimulatory molecules and/or adjuvants used because different immunostimulatory molecules may elicit different mechanisms for enhancing and or modulating the CMI response. By way of example, the different effector mechanisms/immunomodulatory molecules include but are not limited to augmentation of help signal (IL-2), recruitment of professional APC (GM-CSF), increase in T cell frequency (IL-2), effect on antigen processing pathway and MHC expression (IFN-gamma and TNF-alpha) and diversion of immune response away from the Thl response and towards a Th2 response (LTB) (see WO 97/02045). Unmethylated CpG containing oligonucleotides (see WO96/02555) are also preferential inducers of a Thl response and are suitable for use in the present invention.
Without being bound by theory, the inclusion of an adjuvant is advantageous because the adjuvant may help to enhance the CMI response to the expressed antigen by diverting the Th2 response to a Thl response and/or specific effector associated mechanisms to an expressed epitope with the consequent generation and maintenance of an enhanced CMI response (see, for example, the teachings in WO 97/02045).
The inclusion of an adjuvant with an antigen or nucleotide sequence encoding the antigen is also advantageous because it may result in a lower dose or fewer doses of the antigen/antigenic combination being necessary to achieve the desired CMI response in the subject to which the antigen or nucleotide sequence encoding the antigen is administered, or it may result in a qualitatively and/or quantitatively different immune response in the subject. The effectiveness of an adjuvant can be determined by administering the adjuvant with the antigen in parallel with the antigen alone to animals and comparing antibody and/or cellular-mediated immunity in the two groups using standard assays such as radioimmunoassay, ELISAs, CD8+ T-cell assays, and the like, all well known in the art. Typically, the adjuvant is a separate moiety from the antigen, although a single molecule (such for example, CTB) can have both adjuvant and antigen properties.
As used herein, the term "genetic adjuvant" refers to an adjuvant encoded by a nucleotide sequence and which, when administered with the antigen enhances the CMI response relative to the CMI response generated upon administration of the antigen alone.
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"). In one preferred embodiment, the genetic adjuvant is a bacterial ADP-ribosylating exotoxin.
ADP-ribosylating bacterial toxins are a family of related bacterial exotoxins and include diphtheria toxin (DT), pertussis toxin (PT), cholera toxin (CT), the E. coli heat-labile toxins (LT1 and LT2), Pseudomonas endotoxin A, Pseudomonas exotoxin S, B. cereus exoenzyme, B. sphaericus toxin, C. botulinum C2 and C3 toxins, C. limosum exoenzyme, as well as toxins from C. perfringens, C. spiriforma and C. difficile, Staphylococcus aureus EDIN, and ADP-ribosylating bacterial toxin mutants such as CRM]9 , a non-toxic diphtheria toxin mutant (see, e.g., Bixler et al. (1989) Adv. Exp. Med. Biol. 251:175; and Constantino et al. (1992) Vaccine). Most ADP- ribosylating bacterial toxins are organized as an A:B multimer, wherein the A subunit contains the ADP-ribosyltransferase activity, and the B subunit acts as the binding moiety. Preferred ADP-ribosylating bacterial toxins for use in the compositions of the present invention include cholera toxin and the E. coli heat-labile toxins. Cholera toxin (CT) and the related E. coli heat labile enterotoxins (LT) are secretion products of their respective enterotoxic bacterial strains that are potent immunogens and exhibit strong toxicity when administered systemically, orally, or mucosally. Both CT and LT are known to provide adjuvant effects for antigen when administered via the intramuscular or oral routes. These adjuvant effects have been observed at doses below that required for toxicity. The two toxins are extremely similar molecules, and are at least about 70-80%, homologous at the amino acid level.
Preferably the genetic adjuvant is cholera toxin (CT), enterotoxigenic E. Coli heat- labile toxin (LT), or a derivative, subunit, or fragment of CT or LT which retains adjuvanticity. In an even more preferred embodiment, the genetic adjuvant is LT. In another preferred embodiment, the genetic adjuvant may be CTB or LTB.
Preferably the entertoxin is a non-toxic enterotoxin.
The use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in WO 95/17211 and as parenteral adjuvants in WO 98/42375. The toxin or toxoid is preferably in the form of a holotoxin, comprising both A and B subunits. Preferably, the A subunit contains a detoxifying mutation; preferably the B subunit is not mutated. 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 incoφorated by reference herein in their entirety (Beignon, et al. Infection and Immunity (2002) 70(6):3012 - 3019; Pizza, et al., Vaccine (2001) 19:2534 - 2541; Pizza, et al, Int. J. Med. Microbiol (2000) 290(4- 5):455-461; Scharton-Kersten et al. Infection and Immunity (2000) 68(9):5306 - 5313; Ryan et al. Infection and Immunity (1999) 67(12):6270 - 6280; Partidos et al. Immunol. Lett. (1999) 67(3):209 - 216; Peppoloni et al. Vaccines (2003) 2(2):285 - 293; and Pine et al J. Control Release (2002) 85(l-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 incoφorated herein by reference in its entirety.
By way of further example, at least one of the entertoxin subunit coding regions may be genetically modified to detoxify the subunit peptide encoded thereby, for example wherein the truncated A subunit coding region has been genetically modified to disrupt or inactivate ADP-ribosyl transferase activity in the subunit peptide expression product (see, for example, WO 03/004055).
Thus, these results demonstrate that this genetic adjuvant is particularly desirable where an even more enhanced CMI response is desired. Other desirable genetic adjuvants include but are not limited to nucleotide sequences encoding IL-10, IL-12, IL-13, the interferons (IFNs) (for example, IFN-alpha, IFN-ss, and IFN-gamma), and preferred combinations thereof. Still other such biologically active factors that enhance the CMI response may be readily selected by one of skill in the art, and a suitable plasmid vector containing same constructed by known techniques.
Preferred further adjuvants include, but are not limited to, one or more of the following set forth below: Mineral Containing Compositions
Mineral containing compositions suitable for use as adjuvants in the invention include mineral salts, such as aluminium salts and calcium salts. The invention includes mineral salts such as hydroxides (e.g. oxyhydroxides), phosphates (e.g. hydroxyphoshpates, orthophosphates), sulphates, etc. {e.g. see chapters 8 & 9 of ref. Bush and Everett (2001) Int J Syst Evol Microbiol 51: 203-220), or mixtures of different mineral compounds, with the compounds taking any suitable form (e.g. gel, crystalline, amoφhous, etc.), and with adsoφtion being preferred. The mineral containing compositions may also be formulated as a particle of metal salt. See WO 00/23105.
Aluminum salts may be included in immunogenic compositions and/or vaccines of the invention such that the dose of Al3+ is between 0.2 and 1.0 mg per dose. Preferably the adjuvant is alum, preferably an aluminium salt such as aluminium hydroxide (AIOH) or aluminium phospate or aluminium sulfate. Still more preferably the adjuvant is aluminium hydroxide (AIOH). Preferably a mineral salt, such as an aluminium salt, is combined with and another adjuvant, such as an oligonucleotide containing a CpG motif or an ADP ribosylating toxin. Still more preferably, the mineral salt is combined with an oligonucleotide containing a CpG motif. 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, 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) 21:4234-4237. MF59 is used as the adjuvant in the FLUAD™ influenza virus bivalent subunit vaccine.
Particularly preferred adjuvants for use in the compositions are submicron oil-inwater 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%) Span 85™ (sorbitan trioleate), and, optionally, N-acetylmuramyl-L-alanyl-D-isogluatminyl-L- alanine-2-( -2,-dipalmitoyl-5,ra-glycero-3-huydroxyphosphophoryloxy)-ethylamine
(MTP-PE), for example, the submicron oil-in-water emulsion known as "MF59" (International Publication No. WO90/14837; US Patent Nos. 6,299,884 and 6,451,325, incoφorated 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 HOY microfluidizer (Microfluidics, Newton, MA). 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. WO90/14837 and US Patent Nos. 6,299,884 and 6,45 1,325, incoφorated herein by reference in their entireties. Complete Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IF A) may also be used as adjuvants in the invention.
Saponin Formulations Saponin formulations, may also be used as adjuvants in the invention. Saponins are a heterologous group of sterol glycosides and triteφenoid 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. Patent No. 5,057,540. Saponin formulations may also comprise a sterol, such as cholesterol (see WO 96/33739). Combinations of saponins and cholesterols can be used to form unique particles called ___nmunostim.ula.ing 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 EP 0 109 942, 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 in Barr et al (1998) Advanced Drug Delivery Reviews 32: 247-271 and Sjolander et al (1998) Advanced Drug Delivery Reviews (1998) 32: 321-338.
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 pi). VLPs are discussed further in WO 03/024480, WO 03/024481; Niikura et al Virology (2002) 293:273 - 280; Lenz et al Journal of Immunology (2001) 5246 - 5355; Pinto, et al Journal of Infectious Diseases (2003) 188:327 - 338; and Gerber et al Journal of Virology (2001) 75(10):4752 - 47601; Virosomes are discussed further in, for example, Gluck et al Vaccine (2002) 20:B10 -B16. Bacterial or Microbiol Derivatives
Adjuvants suitable for use in the invention include bacterial or microbial derivatives such as: 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 tlirough 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) BioorgMed Chem Lett 9:2273-2278.
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. Vaccine (2003) 21:2485 - 2491; Pajak, et al Vaccine (2003) 21:836 - 842.
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 Nucleic Acids Research (2003) 31(9): 2393 - 2400; WO 02/26757 and WO 99/62923 for examples of possible analog substitutions. The adjuvant effect of CpG oligonucleotides is further discussed in Krieg Nature Medicine (2003) 9(7): 831 - 835; McCluskie, et al FEMS Immunology and Medical Microbiology (2002) 32:179 - 185; WO 98/40100, U.S. Patent No. 6,207,646, U.S. Patent No. 6,239,116, and U.S. Patent No. 6,429,199.
The CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT. See Kalman et al (1999) (Nature Genetics 21: 385-389). The CpG sequence may be specific for inducing a Thl 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 J. Immunol. (2003) 170(8):4061 - 4068; Krieg BBRC (2003) 306:948 - 953; and WO 01/95935. 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 (2003) 31(part 3):664 - 658; Bhagat et al BBRC (2003) 300:853 - 861 and WO 03/035836.
Preferably the adjuvant is CpG. Even more preferably, the adjuvant is Alum and an oligonucleotide containg a CpG motif or AIOH and an oligonucleotide containing a CpG motif. Human Immunomodulators
Human immunomodulators suitable for use as adjuvants in the invention include cytokinfes, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g. interferon-γ), macrophage colony stimulating factor, and tumor necrosis factor.
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 W095/17211 and as parenteral adjuvants in W098/42375. 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 incoφorated 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 Thl 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 incoφorated herein by reference in its entirety. Preferably the adjuvant is an ADP-ribosylating toxin and an oligonucleotide containing a CpG motif (see for example, WO 01/34185)
Preferably the adjuvant is a detoxified ADP-ribosylating toxin and an oligonucleotide containing a CpG motif.
Preferably the detoxified ADP-ribosylating toxin is LTK63 or LTK72.
Preferably the adjuvant is LTK63. Preferably the adjuvant is LTK72. Preferably the adjuvant is LTK63 and an oligonucleotide containing a CpG motif. Preferably the adjuvant is LTK72 and an oligonucleotide containing a CpG motif.
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. See for example, WO99/27960.
Microparticles
Microparticles may also be used as adjuvants in the invention. Microparticles (i.e. a particle of -lOOnm to ~150μm in diameter, more preferably ~200nm to ~30μm in diameter, and most preferably ~500nm to ~10μm 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).
Liposomes
Examples of liposome formulations suitable for use as adjuvants are described in U.S.
Patent No. 6,090,406, U.S. Patent No. 5,916,588, andEP 0626 169. Polyoxyethylene ether and Polyoxyethylene Ester Formulations
Adjuvants suitable for use in the invention include polyoxyethylene ethers and polyoxyethylene esters (W099/52549). Such formulations further include polyoxyethylene sorbitan ester surfactants in combination with an octoxynol (WOO 1/21207) as well as polyoxyethylene alkyl ethers or ester surfactants in combination with at least one additional non-ionic surfactant such as an octoxynol (WO01/21152). 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.
Polyphosphazene (PCPP)
PCPP formulations are described, for example, in Andrianov et al Biomaterials
(1998) 19(1 - 3): 109 - 115; Payne et al Adv. Drug. Delivery Review (1998)
31(3):185 - 196.
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-L- alanyl-D-isoglutamine (nor-MDP), and N-acetyhnuramyl-L-alanyl-D-isoglutaminyl-L- alanine-2-(r-2,-dipalmitoyl-5ra-glycero-3-hydroxyphosphoryloxy)-ethylamine MTP- PE).
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 (W099/11241);
(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 (W098/57659); combinations of 3dMPL with, for example, QS21 and/or oil-in-water emulsions (European patent applications 0835318, 0735898 and 0761231). (5) 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. (6) Ribi adjuvant system (RAS), (Ribi Immunochem) containing 2%> Squalene, 5 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™); and (7) one or more mineral salts (such as an aluminum salt) + a non-toxic derivative of LPS (such as 3dPML). 10 (7) combinations of 3dMPL with, for example, QS21 and or oil-in-water emulsions (See European patent applications 0835318, 0735898 and 0761231); (8) one or more mineral salts (such as an aluminum salt) + a non-toxic derivative of LPS (such as 3dPML); and (9) one or more mineral salts (such as an aluminum salt) + an immunostimulatory 15 oligonucleotide (such as a nucleotide sequence including a CpG motif). Aluminium salts and MF59 are preferred adjuvants for parenteral immunisation. Mutant bacterial toxins are preferred mucosal adjuvants. Bacterial toxins and bioadhesives are preferred adjuvants for use with mucosally-delivered vaccines, such 20 as nasal vaccines. The composition may include an antibiotic. Preferably the compositions of the present invention are administered with alum and/or CpG sequences. 25 Nucleic Acid The antigens or epitopes of the present invention may be administered as nucleotide sequences encoding the antigens or epitopes. As used herein, the term nucleotide sequence refers to one of more nucleotide sequences which encode one or more 30 epitopes which are used in the compositions or combinations of the present invention. The term "nucleotide sequence (NOI)" is synonymous with the term "polynucleotide" or "nucleic acid". The NOI may be DNA or RNA of genomic or synthetic or of recombinant origin. The NOI may be double-stranded or single-stranded whether representing the sense or antisense strand or combinations thereof. For some 35 applications, preferably, the NOI is DNA. For some applications, preferably, the NOI is prepared by use of recombinant DNA techniques (e.g. recombinant DNA). For some applications, preferably, the NOI is cDNA. For some applications, preferably, the NOI may be the same as the naturally occurring form. 40 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 puφoses). 45 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 Chlamydial or host cell nucleic acids). 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.
VECTOR In one embodiment of the present invention, an antigen or antigenic combination or NOI encoding same is administered directly to a host subject. In another embodiment of the present invention, a vector comprising an NOI is administered to a host subject. Preferably the NOI is prepared and/or administered using a genetic vector. As it is well known in the art, a vector is a tool that allows or faciliates the transfer of an entity from one environment to another. In accordance with the present invention, and by way of example, some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a host and/or a target cell for the puφose of replicating the vectors comprising the NOI of the present invention and/or expressing the antigens or epitopes of the present invention encoded by the NOI. Examples of vectors used in recombinant DNA techniques include but are not limited to plasmids, chromosomes, artificial chromosomes or viruses. The term "vector" includes expression vectors and/or transformation vectors. The term "expression vector" means a construct capable of in vivo or in vitro/ex vivo expression. The term "transformation vector" means a construct capable of being transferred from one species to another.
NAKED DNA
The vectors comprising the NOI of the present invention may be administered directly as "a naked nucleic acid construct", preferably further comprising flanking sequences homologous to the host cell genome. As used herein, the term "naked DNA" refers to a plasmid comprising the NOI of the present invention together with a short promoter region to control its production. It is called "naked" DNA because the plasmids are not carried in any delivery vehicle. When such a DNA plasmid enters a host cell, such as a eukaryotic cell, the proteins it encodes are transcribed and translated within the cell.
VIRAL VECTORS
Alternatively, the vectors comprising the NOI of the present invention may be introduced into suitable host cells using a variety of viral techniques which are known in the art, such as for example infection with recombinant viral vectors such as retroviruses, heφes simplex viruses and adenoviruses. The vector may be a recombinant viral vectors. Suitable recombinant viral vectors include but are not limited to adeno virus vectors, adeno-associated viral (AAV) vectors, heφes-virus vectors, a retroviral vector, lentiviral vectors, baculoviral vectors, pox viral vectors or parvovirus vectors (see Kestler et al 1999 Human Gene Ther 10(10): 1619-32). In the case of viral vectors, administration of the NOI is mediated by viral infection of a target cell.
TARGETED VECTOR The term "targeted vector" refers to a vector whose ability to infect or transfect or transduce a cell or to be expressed in a host and/or target cell is restricted to certain cell types within the host subject, usually cells having a common or similar phenotype. EXPRESSION VECTOR
Preferably, the NOI of the present invention which is inserted into a vector is operably linked to a control sequence that is capable of providing for the expression of the antigens or epitopes by the host cell, i.e. the vector is an expression vector. The agent produced by a host cell may be secreted or may be contained intracellularly depending on the NOI and/or the vector used. As will be understood by those of skill in the art, expression vectors containing the NOI can be designed with signal sequences which direct secretion of the EOI through a particular prokaryotic or eukaryotic cell membrane.
FUSION PROTEINS The Chlamydia pneumoniae antigens 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, 18, 19 or 20) of the antigens are expressed as a single polypeptide chain (a 'hybrid' polypeptide). Hybrid 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 hybrid 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 hybrid polypeptide may comprise two or more polypeptide sequences from the first antigen group. Accordingly, the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a Chlamydia bactgerium, preferably a Chlamydia pneumoniae antigen or a fragment thereof of the first antigen group. Preferably, the first and second amino acid sequences in the hybrid polypeptide comprise different epitopes.
The hybrid polypeptide may comprise two or more polypeptide sequences from the second antigen group. Accordingly, the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, wherein said first and second amino acid sequences are selected from a Chlamydia pneumoniae antigen or a fragment thereof of the second antigen group. Preferably, the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes. The hybrid polypeptide may comprise one or more polypeptide sequences from the first antigen group and one or more polypeptide sequences from the second antigen group. Accordingly, the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group and said second amino acid sequence selected from a Chlamydia bactgerium, preferably a Chlamydia pneumoniae antigen or a fragment thereof from the second antigen group. Preferably, the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes. The hybrid polypeptide may comprise one or more polypeptide sequences from the first antigen group and one or more polypeptide sequences from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group. Accordingly, the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group and said second amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group. Preferably, the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes. The hybrid polypeptide may comprise one or more polypeptide sequences from the second antigen group and one or more polypeptide sequences from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group. Accordingly, the invention includes a composition comprising a first amino acid sequence and a second amino acid sequence, said first amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the second antigen group and said second amino acid sequence selected from a Chlamydia pneumoniae antigen or a fragment thereof from the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group. Preferably, the first and second amino acid sequences in the hybrid polypeptide comprise difference epitopes.
Hybrids consisting of amino acid sequences from two, tliree, four, five, six, seven, eight, nine, or ten Chlamydia pneumoniae antigens are preferred. In particular, hybrids consisting of amino acid sequences from two, three, four, or five Chlamydia pneumoniae antigens are preferred. Different hybrid polypeptides may be mixed together in a single formulation. Within such combinations, a Chlamydia pneumoniae 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.
Two-antigen hybrids for use in the invention may comprise any one of the combinations disclosed above.
Hybrid polypeptides can be represented by the formula NH2-A-{-X-L-}„-B-COOH, wherein: X is an amino acid sequence of a Chlamydia pneumoniae antigen or a fragment thereof from the first antigen group, the second antigen group or the third antigen group or the fourth antigen group or the fifth antigen group or the sixth antigen group or the seventh antigen group or the eight antigen group or the ninth antigen group or the tenth antigen group.; 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 ra 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 Xi 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 Xi as moiety -A-.
For each ra instances of {-X-L-}, linker amino acid sequence -L- may be present or absent. For instance, when ra=2 the hybrid may be NH2-Xι-Lι-X2-L2-COOH, NH2-Xj- X2-COOH, NH2-X!-Lι-X2-COOH, NH2-X!-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 Gly„ where ra = 2, 3, 4, 5, 6, 7, 8, 9, 10 or more), and histidine tags (i.e. His„ where ra = 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 (SEQ ID No 77), with the Gly-Ser dipeptide being formed from a BamYS. 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 ra = 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 Xi 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. His„ where ra = 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, ra is 2 or 3.
The invention also provides nucleic acid encoding hybrid polypeptides of the invention. Furthermore, the invention provides nucleic acid which can hybridise to this nucleic acid, preferably under "high stringency" conditions (e.g. 65°C in a O.lxSSC, 0.5%> SDS solution).
The NOI of the present invention may be expressed as a fusion protein comprising an adjuvant and/or a biological response modifier and/or immunomodulator fused to the antigens or epitopes of the present invention to further enhance and/or augment the CMI response obtained. The biological response modifier may act as an adjuvant in the sense of providing a generalised stimulation of the CMI response. The antigens or epitopes may be attached to either the amino or carboxy terminus of the biological response modifier. METHODS OF MAKING
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 Chlamydial or host cell proteins).
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 further provides a process for producing a composition according to the invention comprising the step of bringing one or more of SEQ IDs 1-86 into combination with one or more other of SEQ IDs 1-86
Strains
Preferred polypeptides of the invention comprise an amino acid sequence found in
C.pneumoniae serovars, or in one or more of an epidemiologically prevalent serotype. Where hybrid polypeptides are used, the individual antigens within the hybrid (i.e. individual -X- moieties) may be from one or more strains. Where ra=2, for instance, X2 may be from the same strain as Xj or from a different strain. Where ra=3, the strains might be (i) X!=X2=X3 (ii) Xι=X2 C3 (iii) Xι X2=X3 (iv) Xι*0t2 X3 or (v) Xι=X3£X2, etc.
Heterologous host
Whilst expression of the polypeptides of the invention may take place in Chlamydia, the invention preferably utilises 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), yeasts, etc.
Details as to how the molecules which make up the SEQ IDs 1-86 can be produced and used can be found from the relevant international applications such as WO 00/37494, WO 02/02606 and WO 03/049762 and WO 03/068811 and these details need not be repeated here. Where the composition includes a protein that exists in different nascent and mature forms, the mature form of the protein is preferably used. For example, the mature form of the Chlamydia pneumoniae protein lacking the signal peptide may be used
ADMINISTRATION
Compositions of the invention will generally be administered directly to a patient.
Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal {e.g. see
W099/27961) or transcutaneous {e.g. WO02/074244 and WO02/064162 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. The compositions of the present invention may be administered, either alone or as part of a composition, via a variety of different routes. Certain routes may be favoured for certain compositions, as resulting in the generation of a more effective immune response, prefereably a CMI response, or as being less likely to induce side effects, or as being easier for administration.
By way of example, the compositions of the present invention may be administered via a systemic route or a mucosal route or a transdermal route or it may be administrered directly into a specific tissue. As used herein, the term "systemic administration" includes but is not limited to any parenteral routes of administration. In particular, parenteral administration includes but is not limited to subcutaneous, intraperitoneal, intravenous, intraarterial, intramuscular, or intrasternal injection, intravenous, intraarterial, or kidney dialytic infusion techniques. Preferably, the systemic, parenteral administration is intramuscular injection. In one preferred embodiment of the method, the compositions of the present invention are administered via a transdermal route. While it is believed that any accepted mode and route of immunization can be employed and nevertheless achieve some advantages in accordance herewith, the examples below demonstrate particular advantages with transdermal NOI administration. In this regard, and without being bound by theory, it is believed that transdermal administration of a composition may be preferred because it more efficiently activates the cell mediated immune (CMI) arm of the immune system.
The term "transdermal" delivery intends intradermal (e.g., into the dermis or epidermis), transdermal (e.g.,"percutaneous") and transmucosal administration, i.e., delivery by passage of an agent into or through skin or mucosal tissue. See, e.g., Transdermal Drug Delivery: Developmental Issues and Research Initiatives, Hadgraft and Guy (eds.), Marcel Dekker, Inc., (1989); Controlled Drug Delivery: Fundamentals and Applications, Robinson and Lee (eds.), Marcel Dekker Inc.,(1987); and Transdermal Delivery of Drugs, Vols. 1-3, Kydonieus and Berner (eds.), CRC Press, (1987). Thus, the term encompasses delivery of an agent using a particle delivery device (e.g., a needleless syringe) such as those described in U.S. Patent No. 5,630,796, as well as delivery using particle-mediated delivery devices such as those described in U.S. Patent No. 5,865,796. As used herein, the term "mucosal administration" includes but is not limited to oral, intranasal, intravaginal, intrarectal, intratracheal, intestinal and ophthalmic administration.
Mucosal routes, particularly intranasal, intratracheal, and ophthalmic are preferred for protection against natural exposure to environmental pathogens such as RSV, flu virus and cold viruses or to allergens such as grass and ragweed pollens and house dust mites. The enhancement of the immune response, preferably the CMI response will enhance the protective effect against a subsequently encountered target antigen such as an allergen or microbial agent.
In another preferred embodiment of the present invention, the compositions of the present invention may be administered to cells which have been isolated from the host subject. In this preferred embodiment, preferably the composition is administered to professional antigen presenting cells (APCs), such as dendritic cells. APCs may be derived from a host subject and modified ex vivo to express an antigen of interest and then transferred back into the host subject to induce an enhanced CMI response. Dendritic cells are believed to be the most potent APCs for stimulating enhanced CMI responses because the expressed epitopes of the antigen of interest must be acquired, processed and presented by professional APCs to T cells (both Thl and Th2 helper cells as well as CD8+ T-cells) in order to induce an enhanced CMI response.
PARTICLE ADMINISTRATION
Particle-mediated methods for delivering the compositions of the present invention are known in the art. Thus, once prepared and suitably purified, the above-described antigens or NOI encoding same can be coated onto core carrier particles using a variety of techniques known in the art. Carrier particles are selected from materials which have a suitable density in the range of particle sizes typically used for intracellular delivery from a gene gun device. The optimum carrier particle size will, of course, depend on the diameter of the target cells.
By "core carrier"" is meant a carrier on which a guest antigen or guest nucleic acid (e.g., DNA, RNA) is coated in order to impart a defined particle size as well as a sufficiently high density to achieve the momentum required for cell membrane penetration, such that the guest molecule can be delivered using particle-mediated techniques (see, e.g., U.S. Patent No. 5,100,792). Core carriers typically include materials such as tungsten, gold, platinum, ferrite, polystyrene and latex. See e.g., Particle Bombardment Technology for Gene Transfer, (1994) Yang, N. ed., Oxford University Press, New York, NY pages 10-11. Tungsten and gold particles are preferred. Tungsten particles are readily available in average sizes of 0.5 to 2.0 microns in diameter. Gold particles or microcrystalline gold (e. g., gold powder A1570, available from Engelhard Coφ., East Newark, NJ) will also find use with the present invention. Gold particles provide uniformity in size (available from Alpha Chemicals in particle sizes of 1-3 microns, or available from Degussa, South Plainfield, NJ in a range of particle sizes including 0.95 microns). Microcrystalline gold provides a diverse particle size distribution, typically in the range of 0.5-5 microns. However, the irregular surface area of microcrystalline gold provides for highly efficient coating with nucleic acids. A number of methods are known and have been described for coating or precipitating NOIs onto gold or tungsten particles. Most such methods generally combine a predetermined amount of gold or tungsten with plasmid DNA, CaC12 and spermidine. The resulting solution is vortexed continually during the coating procedure to ensure uniformity of the reaction mixture. After precipitation of the NOI, the coated particles can be transferred to suitable membranes and allowed to dry prior to use, coated onto surfaces of a sample module or cassette, or loaded into a delivery cassette for use in particular gene gun instruments.
The particle compositions or coated particles are administered to the individual in a manner compatible with the dosage formulation, and in an amount that will be effective for the puφoses of the invention. The amoimt of the composition to be delivered (e. g., about 0.1 mg to 1 mg, more preferably 1 to 50 ug of the antigen or allergen, depends on the individual to be tested. The exact amount necessary will vary depending on the age and general condition of the individual to be treated, and an appropriate effective amount can be readily determined by one of skill in the art upon reading the instant specification. HOST MAMMALIAN SUBJECT As used herein, the term "host mammalian subject" means any member of the subphylum cordata, including, without limitation, humans and other primates, including non-human primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like. The terms do not denote a particular age. Thus, both adult and newborn individuals are intended to be covered. The methods described herein are intended for use in any of the above vertebrate species, since the immune systems of all of these vertebrates operate similarly. If a mammal, the subject will preferably be a human, but may also be a domestic livestock, laboratory subject or pet animal.
The mammal is preferably a human. Where the vaccine is for prophylactic use, the human is preferably a child (e.g. a toddler or infant) or a teenager; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult. A vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc.
PREVENT AND/OR TREAT 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 and to the preparation of a vaccine to prevent and/or treat an disorder associated with a Chlamydia bacterium. It is to be appreciated that all references herein to treatment include curative, palliative and prophylactic treatment.
The administration of antigenic combinations of the present invention or a composition comprising the NOI encoding the antigenic combinations may be for either "prophylactic" or "therapeutic" puφose. As used herein, the term "therapeutic" or "treatment" includes any of following: the prevention of infection or reinfection; the reduction or elimination of symptoms; and the reduction or complete elimination of a pathogen. Treatment may be effected prophylactically (prior to infection) or therapeutically (following infection). Prophylaxis or therapy includes but is not limited to eliciting an effective immune response, preferably a CMI immune response and/or alleviating, reducing, curing or at least partially arresting symptoms and/or complications resulting from a T cell mediated immune disorder. When provided prophylactically, the composition of the present invention is typically provided in advance of any symptom. The prophylactic administration of the composition of the present invention is to prevent or ameliorate any subsequent infection or disease. When provided therapeutically, the composition of the present invention is typically provided at (or shortly after) the onset of a symptom of infection or disease. Thus the composition of the present invention may be provided either prior to the anticipated exposure to a disease causing agent or disease state or after the initiation of an infection or disease.
Whether prophylactic or therapeutic administration (either alone or as part of a composition) is the more appropriate will usually depend upon the nature of the disease. By way of example, immunotherapeutic composition of the present invention could be used in immunotherapy protocols to actively inducing immunity by vaccination. This latter form of treatment is advantageous because the immunity is prolonged. On the other hand a vaccine composition will preferably, though not necessarily be used prophylactically to induce an effective CMI response against subsequently encountered antigens or portions thereof (such as epitopes) related to the target antigen.
These uses and methods are preferably for the prevention and/or treatment of a disease caused by a Chlamydia (e.g. trachoma, pelvic inflammatory disease, epididymitis, infant pneumonia, artherosclerosis, cardiovascular disease etc.). The compositions may also be effective against C.pneumoniae.
PROPHYLACTICALLY OR THERAPEUTICALLY OR IMMUNOLOGICALLY EFFECTIVE AMOUNT
The composition dose administrated to a host subject, in the context of the present invention, should be sufficient to effect a beneficial prophylactic or therapeutic immune response, preferably a CMI response in the subject over time.
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. The method may raise a booster response.
As used herein, the term ""prophylactically or therapeutically effective dose" means a dose in an amount sufficient to elicit an enhanced immune response, preferably a CMI response to one or more antigens or epitopes and/or to alleviate, reduce, cure or at least partially arrest symptoms and/or complications from a T cell mediated immune disorder.
Immunogenic compositions used as vaccines comprise an immunologically effective amount of antigen(s), as well as any other components, 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. 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.
The mammal is preferably a human. Where the vaccine is for prophylactic use, the human is preferably a child (e.g. a toddler or infant) or a teenager or an adult; where the vaccine is for therapeutic use, the human is preferably a teenager or an adult. A vaccine intended for children may also be administered to adults e.g. to assess safety, dosage, immunogenicity, etc. Preferably, the human is a teenager. More preferably, the human is a pre-adolescent teenager. Even more preferably, the human is a pre- adolescent female or male Preferably the pre-adolescent male or female is around 9- 12 years of age. One 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 or by protein or DNA microarray. 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.
One way of checking efficacy of therapeutic treatment involves monitoring Chlamydia infection after administration of the composition of the invention. One way of checking efficacy of prophylactic treatment involves monitoring immune responses against the Chlamydia antigen, such as the Chlamydia pneumoniae antigen in the compositions of the invention after administration of the composition. For example, checking efficacy of prophylactic treatment may involve monitoring immune responses both sys temically (such as monitoring the level of IgGl and IgG2a production) and mucosally (such as monitoring the level of IgA production) against the Chlamydia pneumoniae antigens in the compositions of the invention after administration of the composition. Typically, serum Chlamydia specific antibody responses are determined post-immunization but pre-challenge whereas mucosal Chlamydia specific antibody body responses are determined post-immunization and post-challenge.
These uses and methods are preferably for the prevention and/or treatment of a disease caused by Chlamydia pneumoniae (e.g. pneumonia, bronchitis, pharyngitis, sinusitis, erythema nodosum, asthma, atherosclerosis, stroke, myocardial infarctions, coronary artery disease, etc.).
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. For example, in vitro neutralization by Peterson et al (1988) is suitable for testing vaccine compositions directed toward Chlamydia, preferably Chlamydia pneumoniae.
One example of such an in vitro test is described as follows. Hyper-immune antisera is diluted in PBS containing 5%> guinea pig serum, as a complement source. Chlamydia pneumoniae (104 IFU; inclusion forming units) are added to the antisera dilutions. The antigen-antibody mixtures are incubated at 37°C for 45 minutes and inoculated into duplicate confluent Hep-2 or HeLa cell monolayers contained in glass vials (e.g., 15 by 45 mm), which have been washed twice with PBS prior to inoculation. The monolayer cells are infected by centrifugation at 1000X g for 1 hour followed by stationary incubation at 37°C for 1 hour. Infected monolayers are incubated for 48 or 72 hours, fixed and stained with Chlamydia specific antibody, such as anti-MOMP. Inclusion-bearing cells are counted in ten fields at a magnification of 200X. Neutralization titer is assigned on the dilution that gives 50%o inhibition as compared to control monolayers/IFU. The efficacy of immunogenic compositions can also be determined in vivo by challenging animal models of Chlamydia pneumoniae infection, e.g., guinea pigs or mice, with the immunogenic compositions. The immunogenic compositions may or may not be derived from the same serovars as the challenge serovars. Preferably the immunogenic compositions are derivable from the same serovars as the challenge serovars. More preferably, The serovars of the present invention are obtainable from clinical isolates or from culture collections such as the American Tissue Culture Collection (ATCC). In vivo efficacy models include but are not limited to: (i) A murine infection model using human Chlamydia pneumoniae serotypes; (ii) a murine disease model which is a murine model using a mouse-adapted Chlamydia pneumoniae strain, such as the Chlamydia pneumoniae mouse pneumonitis (MoPn) strain also known as Chlamydia muridarum; and (iii) a primate model using human Chlamydia pneumoniae isolates. The MoPn strain is a mouse pathogen while human Chlamydia pneumoniae serotypes are human pathogens (see for example, Brunham et al (2000) J Infect Dis 181 (Suppl 3) S538-S543; Murdin et al (2000) J Infect Dis 181 (Suppl 3) S544-S551 and Read et al (2000) NAR 28(6); 1397-1406). As the Examples demonstrate, human Chlamydia pneumoniae serotypes can be used in mouse models although they normally require high inocula or pretreatment with progesterone. Progesterone is generally used because it seems to render the epithelium more susceptible to chlamydial infection (see Pal et al 2003 Vaccine 21: 1455-1465). One the other hand, MoPn, which was originally isolated from mouse tissues, is thought to be a natural murine pathogen and thus offers an evolutionarily adapted pathogen for analysis of host-pathogen interactions. Although the MoPn serovar is thought to have a high degree of DNA homology to the human Chlamydia serovars, it may also have some unique properties (see for example, Pal et al (2002) Infection and Immunity 70(9); 4812-4817.
By way of example, in vivo vaccine compositions challenge studies can be performed in the murine model of Chlamydia pneumoniae (Morrison et al 1995). A description of one example of this type of approach is as follows. Female mice 7 to 12 weeks of age receive 2.5 mg of depoprovera subcutaneously at 10 and 3 days before vaginal infection. Post-vaccination, mice are infected in the genital tract with 1,500 inclusion- forming units of Chlamydia pneumoniae contained in 5ml of sucrose-phosphate- glutamate buffer, pH 7.4. The course of infection is monitored by determining the percentage of inclusion-bearing cells by indirect immunofluorescence with Chlamydia pneumoniae specific antisera, or by a Giemsa-stained smear from a scraping from the genital tract of an infected mouse. The presence of antibody titers in the serum of a mouse is determined by an enzyme-linked immunosorbent assay. The immunogenic compositions of the present invention can be administered using a number of different immunization routes such as but not limited to intra-muscularly (i.m.), intra- peritoneal (i.p.), intra-nasal (i.n.), sub-cutaneous (s.c) or transcutaneous (t.c) routes. Generally, any route of administration can be used provided that the desired immune response at the required mucosal surface or surfaces is achieved. Likewise, the challenge serovars may be administered by a number of different routes. Typically, the challenge serovars are administered mucosally, such as but not limited to an intranasal (i.n) challenge.
Alternative in-vivo efficacy models include guinea pig models. For example, in vivo vaccine composition challenge studies in the guinea pig model of Chlamydia pneumoniae infection can be performed. A description of one example of this type of approach follows. Female guinea pigs weighing 450 - 500 g are housed in an environmentally controlled room with a 12 hour light-dark cycle and immunized with vaccine compositions via a variety of immunization routes. Post-vaccination, guinea pigs are infected in the genital tract with the agent of guinea pig inclusion conjunctivitis (GPIC), which has been grown in HeLa or McCoy cells (Rank et al. (1988)). Each animal receives approximately 1.4xl07 inclusion forming units (IFU) contained in 0.05 ml of sucrose-phosphate-glutamate buffer, pH 7.4 (Schacter, 1980). The course of infection monitored by determining the percentage of inclusion-bearing cells by indirect immunofluorescence with GPIC specific antisera, or by Giemsa- stained smear from a scraping from the genital tract (Rank et al 1988). Antibody titers in the serum is determined by an enzyme-linked immunosorbent assay.
Compositions of the invention will generally be administered directly to a patient. Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or mucosally, such as by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (See e.g. W099/27961) or transcutaneous (See e.g. WO02/074244 and WO02/064162), intranasal (See e.g. WO03/028760), ocular, aural, pulmonary or other mucosal administration.
DOSAGE
Prophylaxis or therapy can be accomplished by a single direct administration at a single time point or multiple time points. Administration can also be delivered to a single or to multiple sites. Some routes of administration, such as mucosal administration via ophthalmic drops may require a higher dose. Those skilled in the art can adjust the dosage and concentration to suit the particular route of delivery.
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. HOMOLOGUES
SEQ IDs 1-86 in the compositions of the invention may be supplemented or substituted with molecules comprising sequences homologous (ie. sharing sequence identitv) to SEQ ID Nos 1-86. Proteins (including protein antigens) as used in the invention (as encoded by the NOI) may have homology and/or sequence identity with naturally occurring forms. Similarly coding sequences capable of expressing such proteins will generally have homology and/or sequence identity with naturally occurring sequences. Techniques for determining nucleic acid and amino acid "sequence identity" also are known in the art. Typically, such techniques include determining the nucleotide sequence of the mRNA for a gene and/or determining the amino acid sequence encoded thereby, and comparing these sequences to a second nucleotide or amino acid sequence.
In general, "identity" refers to an exact nucleotide-to-nucleotide or amino acid-to- amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more sequences (polynucleotide or amino acid) can be compared by determining their "percent identity." The percent identity of two sequences, whether nucleic acid or amino acid sequences, is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100.
An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2: 482-489 (1981). This algorithm can be applied to amino acid sequences by using the scoring matrix developed by Dayhoff, Atlas of Protein Sequences and Structure, M. O. Dayhoff ed., 5 suppl. 3: 353-358, National Biomedical ResearchFoundation, Washington, D. C, USA, and normalized by Gribskov, Nucl. AcidsRes. 14 (6): 6745- 6763 (1986). An exemplary implementation of this algorithm to determine percent identity of a sequence is provided by the Genetics Computer Group (Madison, WI) in the"BestFit"utility application. The default parameters for this method are described in the Wisconsin Sequence Analysis Package Program Manual, Version 8 (1995) (available from Genetics Computer Group,
Madison, WI). A preferred method of establishing percent identity in the context of the present invention is to use the MPSRCH package of programs copyrighted by the University of Edinburgh, developed by John F. Collins and Shane S.
Sturrok, and distributed by IntelliGenetics, Inc. (Mountain View, CA). From this suite of packages the Smith- Waterman algorithm can be employed where default parameters are used for the scoring table (for example, gap open penalty of 12, gap extension penalty of one, and a gap of six). From the data generated the "Match"value reflects"sequence identity." Other suitable programs for calculating the percent identity or similarity between sequences are generally known in the art, for example, another alignment program is BLAST, used with default parameters. For example, BLASTN and BLASTP can be used using the following default parameters: genetic code = standard; filter = none; strand = both; cutoff= 60; expect = 10; Matrix = BLOSUM62 ; Descriptions = 50 sequences; sort by = HIGH SCORE; Databases = non-redundant, GenBank +EMBL + DDBJ + PDB + GenBank CDS translations + Swiss protein + Spupdate + PIR. Details of these programs can be found at the following internet address: http://www. ncbi. nlm. gov/cgi-bin/BLAST.
Alternatively, homology can be determined by hybridization of polynucleotides under conditions which form stable duplexes between homologous regions, followed by digestion with single-stranded-specific nuclease (s), and size determination of the digested fragments. Two DNA, or two polypeptide sequences are "substantially homologous" to each other when the sequences exhibit at least about 80%>-85%>, preferably at least about 90%,, and most preferably at least about 95%-98%> sequence identity over a defined length of the molecules, as determined using the methods above. As used herein, substantially homologous or homologous also refers to sequences showing complete identity to the specified DNA or polypeptide sequence. DNA sequences that are substantially homologous or homologous can be identified in a Southern hybridization experiment under, for example, stringent conditions, as defined for that particular system. For example, stringent hybridization conditions can include 50%> formamide, 5x Denhardt's Solution, 5x SSC, 0.1% SDS and 100 pg/ml denatured salmon sperm DNA and the washing conditions can include 2x SSC, 0.1 %> SDS at 37 C followed by lx SSC, 0.1% SDS at 68 C. Defining appropriate hybridization conditions is within the skill of the art. Preferably the degree of identity is preferably greater than 50% (eg. 65%. 80%. 90%. or more) and include mutants and allelic variants. Sequence identitv between the proteins is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford. Molecular), using an affine gap search with parameters gap open penalty=12 and gap extension penalty=l.
SEQ IDs 1-86 in the compositions of the invention may be supplemented or substituted with nucleic acid which can hybridise to the Chlamydia nucleic acid, preferably underv"high stringency" conditionsv(c. 65 C in an 0.1 x SSC, 0.5%, SDS solution).
Hypothetical Protein
As used herein, the term "hypothetical protein" refers to a protein which lacks a known cellular location or a known cellular function. Typically, a hypothetical protein lacks significant homologies with known well characterised proteins.
COMPOSITIONS
The invention also provides the compositions of the invention for use as medicaments (eg. as immunogenic compositions or vaccines) or as diagnostic reagents for detecting a Chylamydia infectioin in a host subject. It also provides the use of the compositions in the manufacture of: (i) a medicament for treating or preventing infection due to Chlamydia pneumoniae bacteria: (ii) a diagnostic reagent for detecting the presence of Chlamydia Pneumonaie bacteria or of antibodies raised against Chlamydia Pneumonaie bacteria; and/or (iii) a reagent which can raise antibodies against Chlamydia pneumonaie bacteria.
The invention also provides a method of treating a patient, comprising administering to the patient a therapeutically effective amount of a composition according to the invention. The present invention provides compositions that are useful for preventing and/or treating T cell mediated immune disorders. In one embodiment, the composition is a pharmaceutical composition. In another preferred embodiment, the composition is an immunotherapeutic composition. In an even more preferred embodiment, the composition is a vaccine composition.. The composition may also comprise a carrier such as a pharmaceutically or immunologically acceptable carrier. Pharmaceutically acceptable carriers or immunologically acceptable carriers are determined in part by the particular composition being administered as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions or vaccine compositions or immunotherapeutic compositions of the present invention.
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 Chlamydia pneumoniae infection in an animal susceptible to Chlamydial infection comprising administering to said animal a therapeutic or prophylactic amount of the immunogenic compositions of the invention. Preferably, the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five or all six Chlamydia pneumoniae antigens of the first antigen group. Still more preferably, the combination consists of all six Chlamydia pneumoniae antigens of the first antigen group.
Alternatively, the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination selected from the group consisting of two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve Chlamydia pneumoniae antigens selected from the first antigen group and the second antigen group. Preferably, the combination is selected from the group consisting of three, four, or five Chlamydia pneumoniae antigens selected from the second antigen group. Still more preferably, the combination consists of five Chlamydia pneumoniae antigens selected from the second antigen group. Alternatively, the immunogenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, or five Chlamydia pneumoniae antigens of the first antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group. Preferably, the combination consists of three, four or five Chlamydia pneumoniae antigens of the first antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group.
Alternatively, the immunigenic composition comprises a combination of Chlamydia pneumoniae antigens, said combination consisting of two, three, four, five, six, seven, eight, nine, ten, eleven or twelve Chlamydia pneumoniae antigens of the first antigen group and the second antigen group and one, two, three, four, five or six Chlamydia pneumoniae antigens of the third antigen group. Preferably, the combination is selected from the group consisting of three, four, or five Chlamydia pneumoniae antigens from the second antigen group and three, four or five Chlamydia pneumoniae from the third antigen group. Still more preferably, the combination consists of five Chlamydia pneumoniae antigens from the second antigen group and three, four or five Chlamydia pneumoniae antigens of the third antigen group.
In certain embodiments, the composition comprises molecules from different Chlamydia species. In some embodiments, the composition may comprise molecules from different serogroups and/or strains of the same Chlamydia species. Further embodiments comprise mixtures of one or more Chlamydia molecules from different strains. Many proteins are relatively conserved between different species serogroups and strains of Chlamydia trachomatis and Chlamydia pneumoniae. To ensure maximum cross-strain recognition and reactivity, regions of proteins that are conserved between different Chlamydia species, serogroups and strains can be used in the compositions of the present invention. The invention therefore provides proteins which comprise stretches of amino acid sequence that are shared across the majority of Chlamydia strains. Preferablv, therefore, the composition comprises a protein comprising a fragment of a Chlamydia pneumoniae protein (preferably a protein from SEQ ID Nos 1-86 or more preferably SEQ ID Nos 1-41 wherein said fragment consists of n consecutive conserved amino acids.
Further antigens
The compositions of the invention may further comprise antigen derived from one or more sexually transmitted diseases in addition to Chlamydia trachomatis. Preferably the antigen is derived from one or more of the following sexually transmitted diseases: N.gonorrhoeae {e.g. i, ii, iii, iv}; human papiloma virus; Treponema pallidum; heφes simplex virus (HSV-1 or HSV-2); HIV (HIV-1 or HIV-2); and Haemophilus ducreyi.
A preferred composition comprises: (1) at least t of the Chlamydia pneumoniae antigens from either the first antigen group or the second antigen group, where t is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13, preferably t is five; (2) one or more antigens from another sexually transmitted disease. Preferably, the sexually transmitted disease is selected from the group consisting of heφes simplex virus, preferably HSV-1 and/or HSV-2; human papillomavirus; N.gonorrhoeae; Treponema pallidum; and Haemophilus ducreyi. These compositions can thus provide protection against the following sexually-transmitted diseases: Chlamydia, genital heφes, genital warts, gonorrhoea, syphilis and chancroid (see Stephens et al (1998) Science 282: 754-759).
Where a saccharide or carbohydrate antigen is used, it is preferably conjugated to a carrier /protein i order to enhance immunogenicity (For example, Ramsay et al.
(2001) Lancet 357(9251): 195-196; Lindberg (1999) Vaccine 17 Suppl 2:S28-36;
Buttery & Moxon (2000) JR Coll Physicians Lond 34:163-168; Ahmad & Chapnick
(1999) Infect Dis Clin North Am 13:113-133; Goldblatt (1998) J. Med. Microbiol.
47:563-567; European patent 0 477 508; US Patent No. 5,306,492; International patent application W098/42721; Conjugate Vaccines (eds. Cruse et al.) ISBN
3805549326, particularly vol. 10:48-114; and Hermanson (1996) Bioconjugate
Techniques ISBN: 0123423368 or 012342335).
Preferred carrier proteins are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids. The CRMι9 diphtheria toxoid is particularly preferred (Research Disclosure, 453077 (Jan 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 (W093/17712, WO94/03208) pertussis proteins (W098/58668, EP-A-0471177) protein D from H.influenzae (WO00/56360) cytokines (WO91/01146), 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 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 9:471-480; Apostolopoulos & Plebanski (2000) Curr Opin Mol Ther 2:441-447; Ilan (1999) 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. DISEASE STATES
The compositions of the present invention- may be used to prevent and/or treat disorders such as but not limited to: pneumonia, cardiovascular diseases, atherosclerosis, bronchitis, pharyngitis, laryngitis, sinusitis, obstructive lung diseases, asthma, chronic obstructive pulmonary disease, reactive arthritis, otitis media, abdominal aortic aneurysm, erythema nodosum, Reiter syndrome, sarcoidosis, Alzheimer's disease, multiple sclerosis, lymphogranuloma venereum, ocular trachoma, pelvic inflammatory disease, inclusion conjunctivitis, genital trachoma, infant pneumonitis, incipient trachoma, keratitis, papillary hypertrophy, corneal infiltration, vulvovaginitis, mucopurulent rhinitis, salpingitis, cervicitis, cervical follicles, prostatitis, proctitis, urethritis, lymphogranule inguinale, climatic bubo, tropical bubo, andVoresthiomene.
FORMULATIONS
Chlamydial infections affect various areas of the body and so 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.
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.
The biological molecules of the present invention be formulated into a pharmaceutical composition or an immunotherapeutic composition or a vaccine composition. Such formulations comprise biological molecules combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative. Formulations include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents. In one embodiment of a formulation for parenteral administration, the active ingredient is provided in dry (for eg, a powder or granules) form for reconstitution with a suitable vehicle (e. g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.The pharmaceutical compositions may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution. This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein. Such sterile injectable formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example. Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono-or di-glycerides. Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, in a liposomal preparation, or as a component of a biodegradable polymer systems. Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
KITS
Also included in the invention is a kit for enhancing a CMI response to the biological molecules of the present invention. Such a kit may comprise an antigenic composition or nucleotide sequence encoding same. The kit may also include an adjuvant, preferably a genetic adjuvant is administered with or as part of the biological molecule and instructions for administering the biological molecule. Other preferred components of the kit include an applicator for administering the biological molecule. As used herein, the term "applicator" refers to any device including but not limited to a hypodermic syringe, gene gun, particle acceleration device, nebulizer, dropper, bronchoscope, suppository, impregnated or coated vaginally-insertable material such as a tampon, douche preparation, solution for vaginal irrigation, retention enema preparation, suppository, or solution for rectal or colonic irrigation for applying the NOI either systemically or mucosally or transdermally to the host subject.
The invention also provides for a kit comprising comprising a combination of Chlamydia pneumoniae antigens. The combination of Chlamydia pneumoniae antigens may be one or more of the immunogenic compositions of the invention. The kit may further include a second component comprising one or more of the following: instructions, syringe or other delivery device, adjuvant, or pharmaceutically acceptable formulating solution. The invention also provides a delivery device pre- filled with the immunogenic compositions of the invention. EXAMPLES
The following invention will now be further described only by way of example in which reference is made to the following Figures. The following examples are presented only to illustrate the present invention and to assist one of ordinary skill in making and using the same. The examples are not intended in any way to otherwise limit the scope of the invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.
Figure 1A. Assay of in vitro neutralization of C.pneumoniae infectivity for LLC-MK2 cells by polyclonal mouse antisera to recombinant Chlamydial proteins. Results are shown as reduction in the number of inclusions obtained when monolayers were infected with antiserum-treated infectious EBs, as compared to inclusion numbers given by untreated EBs. Percent reduction values are plotted against the reciprocal of the corresponding serum dilution. For each dilution inclusion counts were corrected for background inhibition of infectivity observed with the corresponding dilution of the pre- immune serum. The figure shows results obtained with serial dilutions of antibodies raised against a 'neutralizing' antigen (v), a 'non-neutralizing' FACS- positive antigen (v), and against the GST polypeptide, used in the fusion constructs, alone (σ). Figure IB shows serum litres giving 50% neutralization of infectivity for the 10 C.pneumoniae recombinant antigens described in the text. Each titer was assessed in 3 separate experiments (SEM values shown).
Figure 2 shows immunoblot analysis of two dimensional electrophoretic maps of C.pneumoniae EBs using the imune sera described in the text. Immunoblots were obtained from either of two EB gels (panels A and B at the top) covering different pH intervals, according to which of the two allowed the best detection of a given antigen. The arrows in the HtrA immunoblot show which of the signals had a corresponding stained spot in the gel (arrows in panel A) which was subjected to MALDI-TOF identification. The two patterns in the HtrA blot are both suggestive of typical electrophoretic 'trains' composed of single charge variants of the same protein.
Figure 3 shows mean numbers of C.pneumoniae IFU recovered from equivalent spleen samples from immunized and mock-immunized hamsters following a systemic challenge. Standard deviation values are shown above the bars. Antigens which induced significant protection are highlighted with an asterisk above the corresponding bar. All antigens were were delivered in Freund's adjuvant, n.i. = non immunized controls
Figure 4 shows flow cytometric analysis of splenocytes from DNA-immunized HLA- A2 transgenic and non transgenic mice. Groups of 4 mice were immunized 3 times i.m. with 50μg of plasmid DNA expressing C. pneumoniae Low Calcium Response Protein H. IFN-γ production from splenocytes was monitored following either a 6h (ex-vivo) or a 6 day (restimulated) pulse with peptide CH-6 (lOμg/ml). Equal numbers of gated live lymphocyte cells were acquired with a LSRII FACS System (Becton Dickinson) and percentages of IFN-γ producing CD8+ T cells were calculated using DIVA Software (Becton Dickinson).
Figure 5 shows a flow cytometric analysis of splenocytes from transgenic and non transgenic mice infected with C. pneumoniae EBs. (A) HLA-A2 transgenic mice were intranasally infected twice with 5x105 C. pneumoniae FB/96 EBs and splenocytes were stimulated for 6 days in the presence of relevant peptides before determining IFN-γ production by CD8+ T cells as described in the legend of Figure 4. (B) HLA-A2 transgenic and non transgenic mice were infected together with the same EBs preparation and CD8+ T cells were subjected to FACS analysis as reported in (A). Table I shows a summary of data and properties of the C.pneumoniae antigens described in the text. The neutralization titer is reported is as the reciprocal of the antiserum dilution causing a 50% reduction in the number of inclusions in the in vitro infectivity assay. For the hamster model data the statistical significance of the results was evaluated by a two-tailed Student's t-test: significant data (p< 0.05) are highlighted with an asterisk. ND = not detected.
Table 2 shows results from hamster mouse model studies for hypothetical proteins. Table 3 shows expressed genes of CPn EB selected by microarray.
Table 4 shows C. pneumoniae selected peptides: protein sources and HLA-A2 stabilization assay.
Table 5 shows ELISPOT assay with CD8+ T cells from DNA immunised HLA-A2 transgenic mice.
Table 6 shows IFN-γ production from splenocytes of DNA immunized HLA-A2 transgenic and non transgenic mice. METHODS AND MATERIALS (Examples 1-4) (see Reference Section 1) C.pneumoniae EB purification
C.pneumoniae FB/96, a clinical isolate obtained from a patient with pneumonia at the Sant'Orsola Polyclinic, Bologna, Italy, was grown in LLC-MK2 cells seeded in individual wells of a six- well plastic plate (7). Cells were harvested 72 hr after infection with a sterile rubber, sonically disrupted and the elementary bodies (EB) purified by gradient centrifugation as described (26). Purified Chlamydiae were resuspended in sucrose-phosphate-glutamic acid (SPG) transport buffer, and stored in 0.5 ml aliquots, at -80°C until used. When required, prior to storage, EB infectivity was heat-inactivated by 3 hour incubation at 56°C.
Expression and purification of recombinant proteins
Open reading frames (ORFs), selected from the C. pneumoniae CWL029 genome sequence (16), were PCR-cloned into plasmid expression vectors and purified from E.coli cultures, essentially as previously described (25). Recombinant Chlamydial proteins were obtained as GST fusion proteins by using pGEX-KG derived vectors (12) in E. coli BL21 (Novagen). PCR primers were designed so as to amplify genes without the N-terminal signal peptide coding sequence. When a signal peptide or processing site was not clearly predictable, the ORF sequence was cloned as annotated by Kalman and coworkers (16). Recombinant E.coli cells were grown in LB medium (500 ml), containing 100 μg/ml Ampicillin, and grown at 37°C until ODδoo = 0.5 , and then induced with 1 mM IPTG. Cells were collected by centrifugation ' 3 hr after induction and broken in a French Press (SLM Aminco, Rochester, NY). After centrifugation at 30.000 g, the supernatants were loaded onto Glutathione Sepharose 4B columns (Amersham Pharmacia Biotech) and column bound proteins were eluted with 50 mM Tris-HCl, 10 mM reduced glutathione, pH 8.0. Protein concentrations in the samples were determined using the Bradford method.
Preparation of mouse antisera Groups of four 5/6-week old CD1 female mice (Charles River, Como, Italy) were immunized intraperitoneally at day 1 with 20ug of protein in Complete Freund's adjuvant (CFA) and boosted at day 15 and 28 with 20ug of recombinant protein in Incomplete Freund's adjuvant (IF A). Pre-immune and immune sera were prepared from blood samples collected on days 0, 27 and 42. In order to reduce the amount of antibodies possibly elicited by contaminating E. coli antigens, the immune sera were incubated overnight at 4°C with nitrocellulose strips adsorbed with a total protein extract from E . coli BL21.
Flow cytometry assays Analyses were performed essentially as previously described (25). Gradient purified, heat-inactivated EBs (2x105 cells) from C.pneumoniae FB/9, resuspended in phosphate-saline buffer (PBS), 0.1% bovine serum albumin (BSA), were incubated for 30 min. at 4°C with the specific mouse antisera (standard dilution 1:400). After centrifugation and washing with 200 μl of PBS-0.1%> BSA, the samples were incubated for 30 minutes at 4°C with Goat Anti-Mouse IgG, F(ab)'2-specific, conjugated with R-Phycoerythrin (Jackson Immunoresearch Laboratories Inc.). The samples were washed with PBS-0.1%BSA, resuspended in 150 μl of PBS-0.1%BSA and analysed by Flow Cytometry using a FACSCalibur apparatus (Becton Dickinson, Mountain View, CA). Control samples were similarly prepared. Positive control antibodies were: i), a commercial anti-C.pneumoniae specific monoclonal antibody (Argene Biosoft, Varilhes, France) and, ii), a mouse polyclonal serum prepared by immunizing mice with gradient purified C.pneumoniae EBs. Background control sera were obtained from mice immunized with the purified GST peptide used in the fusion constructs (GST-fusions control). FACS data were analysed using the Cell Quest Software (Becton Dickinson, Mountain View, CA). The shift between the background control histogram and the immune serum testing histogram was taken as a measure of antibody binding to the EB cell surface. The Kolmorov-Smirnov (K-S) two-sample test (44) was performed on the two overlapped histograms. The D/s(n) values (an index of dissimilarity between the two curves) are reported as "K-S score" in Table 1.
2D Western Blot analysis of immune sera, and Mass Spectrometry Gradient purified C. pneumoniae EBs were washed with 5 mM Tris-HCl pH 7.5, 0.1 mM EDTA, 10%> glycerol, centrifuged 15 min. at 13 000 x g and pellets were resuspended in reswelling solution (7 M urea, 2 M thiourea, 2%> (w/v) CHAPS, (2%w/v) ASB14, 2% (v/v) IPG buffer pH 3-10 NL, or pH 4-7, 2 mM TBP, 65 mM DTT). Protein samples (200 or 20 μg of protein for Coomassie Blue stained reference gels, or gels to be processed for immunoblotting, respectively) were adsorbed overnight on Immobiline DryStrips (7 cm, pH 3-10 NL, or pH 4-7). Electrofocusing was performed in an IPGphor Isoelectric Focusing Unit (Amersham Biosciences, Uppsala, Sweden). The focused strips were equilibrated as described (15) and loaded on linear 9-16.5 % acrylamide gradients (7x 4 cm, 1.5 mm thick), for SDS-PAGE separation in a Mini Protean III Cell (Bio-Rad, Hercules, CA). Gels were stained with colloidal Coomassie Blue (Novex, San Diego, CA) (4) and the protein maps so obtained were scanned with a Personal Densitometer SI (Molecular Dynamics) at 12 bits and 50 mm per pixel.
For Western Blot analyses, the proteins separated in the 2DE maps were transferred onto nitrocellulose membranes, overnight at 30 Volts, using a Protean III apparatus (BioRad, Hercules, CA). Membranes were stained with a 0.05% (w/v) CPTS (Copper(II) phthalocyanine-3,4',4",4'"-tetrasulfonic acid tetrasodium salt) in 12 mM HCl, and marked peripherally with 8 India-ink dots to provide anchors for subsequent image superimposition and matching. After scanning and image acquisition, the membranes were destained with 0,5 M NaHC03, incubated with the mouse sera to be analyzed (either pre-immune or specific immune sera, diluted 1:1000), and then with a peroxidase-conjugated anti-mouse antibody (Amersham Biosciences, Uppsala, Sweden). After washing with PBS, 0.1 %> Tween-20, blots were developed using the Opti-4CN Substrate Kit (Biorad, Hercules, CA), and the images of the immunostained blots again acquired as above. Images were analysed with the computer program Image Master 2D Elite, version 4.01 (Amersham Biosciences, Uppsala, Sweden). Superimposition and matches between Western-blot membranes and Coomassie stained gels were performed as follow. First, the CPTS-stained membrane image and the immunostained blot image were superimposed using the peripheral dot marks. Then, the sum image so obtained was superimposed to the Coomassie stained protein map using the CPTS stained CPn proteins as anchors. The areas on the Coommassie stained map corresponding to immunostained spots on the blot were excised from the preparative gel for protein identification. Protein sample were dried in a vacuum centrifuge, and in-gel digested, for 2h at 37°C, with an excess of porcine Trypsin (Promega, Madison, WI), in 100 mM ammonium bicarbonate. Tryptic peptides were desalted and concentrated using Zip-Tip (Millipore, Bedford, MA). Peptides were directly eluted and loaded onto a SCOUT 384 Anchor Chip multiprobe plate (400 μm, Bruker Daltonics, Bremen, Germany) with a solution of 2-5 dihydroxybenzoic acid (5g/l), in 50% acetonitrile, 0.1% trifluoroacetic acid. Spectra were acquired on a Bruker Biflex III matrix-assisted laser desoφtion ionization-time of flight (MALDI- TOF) apparatus. Resulting values for monoisotopic peaks were used for database searches using the Mascot software (32), as available at the website http://www.matrixscience.com/.
In vitro neutralization assays
In vitro neutralization assays were performed on LLC-MK2 (Rhesus monkey kidney) epithelial cell cultures. Serial four-fold dilutions of mouse immune and corresponding preimmune sera were prepared in sucrose-phosphate-glutamic acid buffer (SPG). Mouse polyclonal sera to whole EBs were used as positive control of neutralization, whereas SPG buffer alone was used as negative control of neutralization (control of infection). Purified infectious EBs from the C.pneumoniae FB/96 were diluted in SPG buffer to contain 2.5xl07 IFU/ml, and lOul of EBs suspension were added to each serum dilution in a final volume of lOOul. Antibody-EB interaction was allowed to proceed for 30 min at 37°C on a slowly rocking platform. The lOOul of reaction mix of each sample was used to inoculate PBS-washed LLC-MK2 confluent monolayers (in triplicate for each serum dilution), in a 24-well tissue culture plate, and centrifuged at 805 x g for 1 hour at 37°C. After centrifugation Eagle's minimal essential medium containing Earle's salts, 20% fetal bovine serum and lug/ml cycloheximide was added. Infected cultures were incubated at 37°C in 5%>C02 for 72 hours. The monolayers were fixed with methanol and the Chlamydial inclusions were detected by staining with mouse a ti-Chlamydia fluorescein-conjugated monoclonal antibody (Merifluor Chlamydia, Meridian Diagnostics, Inc.) and quantified by counting 10 fields per well at a magnification of 40X. The inhibition of infectivity due to EBs interaction with the immune sera was calculated as percentage reduction in mean IFU number as compared to the SPG (buffer only)/EBs control. In this calculation the IFU counts obtained with immune sera were corrected for background inhibition of infection due to the corresponding pre-immune mouse serum. According to common practice, the sera were considered as "neutralizing" if they could cause a 50%> or greater reduction in infectivity. The corresponding neutralizing titer was defined as the serum dilution at which a 50% reduction of infectivity was observed. Experimental variability was evaluated by calculating the standard error of measurement (SEM), from three titration experiments for each recombinant antigen, as shown in Fig. IB.
In vivo screening
In vivo evaluation was performed using a hamster model of systemic infection, as recently described (34). Essentially, adult (10-11 week old) Syrian hamsters (Morini, S. Polo D'Enza, Italy), previously immunized with the recombinant vaccine candidates were challenged systemically with infectious Cpn elementary bodies (EB). Protection was assessed by determining the levels of viable EB recovered from the spleen, as compared to non-immunized animals. Statistical significance of the results was evaluated by a two-tailed Student's t-test. Groups of 8 hamsters were immunized subcutaneously with recombinant antigens, or only injected with buffer for the control group, at days 0, 7, and 21. For each immunization 20 ug protein 1:1 diluted with Freund's complete adjuvant (first dose) and Freund's incomplete adjuvant (booster doses) was injected. At day 35 post- infection the hamsters were anaesthetised with Ketamine and inoculated intraperitoneally and intranasally with 0.1 ml of C.pneumoniae EB suspension (1.0x10s) at each site. Animals were sacrificed seven days after infection. The spleen was weighed, and homogenized in a mortar to obtain a 10% (wt/vol) suspension in cold SPG buffer. Tissue suspensions were centrifuged at 300 x g for 10 min at 4°C to remove coarse debris. The clarified homogenates (0.2 ml) were inoculated in duplicate onto LLC-MK2 cells seeded in plastic individual well of a 24 well plate, incubated at 37°C for 72 h and fixed in acetone before detection and counting of numbers of Chlamydial inclusions per well by immunofluorescence microscopy. The protocol was approved by the ethical committee of the University of Bologna.
Example 1 (in vitro studies)
Screening antisera for in vitro neutralizing properties
Following a genome-wide screening for proteins likely to be localized on the cell surface of C. pneumoniae, we recently reported (25) that antisera to 53 recombinant Chlamydial antigens were capable to bind in a FACS assay, the surface of Chlamydial cells. In order to check whether some of the FACS positive antigens were capable of interfering with EB in vitro infectivity, we raised mouse antisera against the recombinant FACS positive antigens and assessed the effect of each antiserum on the infectivity of purified EBs with respect to monolayers of LLC-MK2 cells. Infectious EB were first incubated with the antiserum and then used to infect cell monolayers in 24-well multititer plates. In parallel, control samples were similarly processed in which the EBs were: i), either treated with buffer only, or, ii), treated with the same dilutions of the corresponding preimmune mouse sera.
Results I
Using this assay, 10 sera have so far proved to effectively neutralize in vitro infectivity to an extent greater than 50%, a property that common practice qualifies such antigens as "neutralising" (Figure 1). These 10 sera were obtained by mouse immunization with recombinant proteins derived from the following C.pneumoniae genes:
• pmp 10 and pmp2, encoding two members of the heterogeneous Chlamydial PMP family of polymoφhic membrane proteins; • artl, encoding a putative extracellular solute (possibly Arginine) binding protein of an aminoacid transport system;
• eno, encoding a protein homologous to bacterial enolases, glycolytic enzymes which can be found also on the bacterial surface;
• htrA, encoding a putative chaperone with heat-shock inducible protease activity; • the Cpn0301 "hypothetical" gene, encoding a protein homologous to the ompH family of bacterial proteins, some members of which have been shown to be chaperones involved in outer membrane biosynthesis;
• two Cpn-specific "hypothetical" genes Cpn0795 and Cpn0042;
• omcA encoding a 7-9 kDa protein annotated as an outer membrane protein; and • αtoS a putative sensor member of a transport system.
As shown in Figure 1 and summarized in Table I, OmpH, enolase and Cpn0795 appeared to induce the highest neutralizing sera, with titers above 400. By contrast, Pmp2, ArtJ and Cpn0042 induced titers equal or less than 100, while the remaining 4 antigens, PmplO, HtrA, AtoS and OmcA showed intermediate titers. Example 2 (in vivo studies)
Evaluation of antisera specificity by 2D immunoblot analysis of Cpn protein extracts
In order to investigate if the neutralizing activity observed in the in vitro infection of LLC-MK2 monolayers was actually due to the binding of the antibodies to the selected C.pneumoniae proteins, rather than to possible cross-rections with other antigens, we assessed the specificity of the antisera by immunoblot analysis of two dimensional electrophoretic maps of EB proteins.
In particular, this analysis was carried out on six antigens (Pmp2, PmplO, Eno, ArtJ, HtrA and OmpH-like) known to be visible in the 2D maps of EB total proteins (Montigiani et al, 2002 Infection and Immunity 70: 368-379). Total EB proteins were resolved by 2D-electrophoresis using two different pH intervals (pH 3-10 non linear, and pH 4-7, respectively) since previous experiments had shown that some of the proteins under study were better detected using one rather that the other of the above pH intervals. For each pH interval four gels were run in parallel. One gel was stained with Coomassie Blue to visualize the protein spots, while the other gels were blotted on nitrocellulose filters and stained with one of the selected sera at 1000-fold dilution. Subsequently, the images of the immunostained blots (Fig.2. panels c to h) were superimposed to the corresponding Coomassie Blue-stained gel to identify the spots which had reacted with a given antiserum. The matching protein spots were excised and processed for peptide identification by MALDI-TOF analysis. Results 2
In all six maps the immunoreactive protein species in the excised gel area were found to contain peptides from the expected Chlamydial protein. Even when the serum reacted with more than one electrophoretic protein species, the mass spectra of all spots which could be detected in the Coomassie Blue stains 2DE map were always consistent with the same polypeptide being present as multiple electrophoretic species.
Interestingly, the immunoblot obtained with the HtrA antiserum showed two sets of 4 spots arranged as two typical electrophoretic 'trains' at two different molecular weights. On the Coomassie Blue stained gel it was possible to identify 4 corresponding spots, 3 in the upper train and 1 in the lower Mw set. MS analysis identified all of them as products of Cpn HtrA gene. Interestingly the lower Mwt species missed 3 N-terminal tryptic peptides, detected in the higher Mw spot series, and mapping within the first 100 aa of the ORF. These results suggest that HtrA was present in the EB protein sample both as a full htrA. product, and as a discrete species missing a short N-terminal peptide, possibly as a result of some post-translational processing.
Discussion of Results 2
In the analysis of data which are based on polyclonal antibody reactivity one should consider that cross-reactions due to epitope mimickry are always difficult to exclude. The problem of antisera specificity was addressed in this work by 2D immunoblotting and identification of the reacting electrophoretic species by mass spectrometry analysis. This approach was possible for 6 of the 10 antisera, i.e. those corresponding to proteins previously identified by mass spectrometry (MALDI-TOF) analysis on 2D electrophoretic maps of C.pneumoniae EB proteins (25, 42) (Table 1, and Figure 2). The probability of fortuitous cross-reactions between unrelated Chlamydial protein species was minimized by the results of the immunoblotting analyses which showed that out of ca 300 protein spots in a map, all those reacting with the tested antisera were consistent with the expected antiserum specificity. Obviously, since during 2-D electrophoresis conformational epitopes are generally lost, structure-dependent cross- reactions cannot be ruled out in this type of analysis.
Example 3
In vivo evaluation of the in vitro neutralizing antigens in a hamster model of systemic infection
We have recently described a new hamster model of systemic Chlamydia pneumoniae infection in which replicating Chlamydia disseminate through macrophages and accumulate in the spleen (34). We therefore asked the question whether the in vitro neutralizing antigens we identified would also have protective activity in vivo using this model. To this aim, the 10 in vitro neutralizing recombinant antigens were used to immunize 8 hamsters with 3 subcutaneous injections over a three-week period, and challenged with 2xl08 Cpn EBs two weeks later. Spleen infection was assessed 7 days after challenge. The difference between the mean number of infectious Chlamydiae recovered from control animals and the mean number of Chlamydiae recovered from animals immunized with the recombinant Chlamydial antigens, was taken as a measure of protection specifically induced by the putative vaccine candidate.
Results 3
The results of spleen protection observed for the various antigens in repeated experiments are shown in Figure 3 and reported as percentage values in Table 1. Six out often antigens, Pmp2, PmplO, Enolase, the OmpH-like protein, and the products of the C.pneumoniae-specific genes Cpn0759 and Cpn0042, showed a statistically significant protective activity, with a reduction in IFU recovered from the spleens of immunized animals higher than 80% with respect to mock-immunized controls.
A limit of the hamster model is that, because of the absence of immunological reagents, the relative contribution of humoral and cell-mediated immunity cannot be assessed. However, we asked the question whether recombinant antigens could elicit also in the hamster neutralizing antibodies with sufficiently high titers. Therefore we tested the sera from hamsters immunized with Pmp2 and enolase, two of the most protective antigens, in the in vitro neutralization assay. Both antigens had a neutralizing titer of approximately 100 (data not shown).
Summary of Results 3 In conclusion, a considerable proportion (60%,) of the in vitro neutralizing antigens were also protective in the hamster in vivo model and our data suggest that antibody- mediated neutralization could play a role at least in this model of systemic infection.
Discussion of Results 3 Beside assaying the in vitro neutralization properties of the selected subset of 10 FACS-positive antigens, we also assessed the performance of these antigens in protecting against C. pneumoniae infection in an animal model of systemic infection recently described in the hamster (34). This evaluation addressed the capability that the recombinant antigens would have of inducing a protective response against naturally replicating Chlamydiae (rather than EB's purified from in vitro cultures grown under artificial conditions) and in the context of a systemic infection. In fact the hamster model we used, while it does not model the typical respiratory infection considered to be the predominant route by which C. pneumoniae infects humans, it nevertheless simulates a situation of systemic invasion which could be preliminary to the establishment of C. pneumoniae chronic infection considered by several authors as being associated to the development or progression of cardiovascular disease, and other chronic degenerative diseases. Notably, a limit of any hamster model is the current lack of hamster-specific immunological reagents which would allow the analysis of cell mediated immune responses. However, in the case of systemic infections, by common wisdom, neutralizing antibodies are likely to have a protective action. The finding that 6 of the 10 'in vitro neutralizing' antigens had also a >80%> protective action in vivo, and that a measurable neutralizing activity was also found in the sera of immunized hamsters, suggests that a specific antibody mediated immunity could at least partially contribute to the animal protection here described. Example 4 Two 'hypothetical'proteins 6784 and 6814 (encoded by the ORFs Cpn0498 and Cpn0525) yielded FACS-positive sera which, however, were not able to neutralize host cell infection in vitro. However, these antigens performed remarkably well in the hamster-spleen test. Table 2
Figure imgf000100_0001
Discussion of Results 4
Interestingly, whilst antiserum against CPn0525 gave negative in vitro results (ie no neutralising activity), the CPn0525 protein gave 97 per cent protection from spleen infection in an in vivo hamster immunisation assay (ie a positive in vivo result). Likewise, whilst antiserum against Cpn0498 gave negative in vitro results (ie no neutralising activity), the CPn0498 protein gave 94 per cent protection from spleen infection in an in vivo hamster immunisation assay. Thus a positive signal obtained in the FACS assay does not guarantee a corresponding positive in vitro neutralization activity and conversely a negative neutralization activity does not mean that a positive in vivo result can be excluded.
General Discussion of Results 1-4
Strategy for identification of Chlamydia pneumoniae antigens of interest
Our strategy was based on the following experimental steps: 1) analysis of Chlamydia genome sequence to select putative membrane-associated antigens, 2) cloning, expression and purification selected antigens, 3) preparation of antigen- specific sera by mouse immunization with the purified antigens, 4) FACS analysis of Chlamydia EBs using the mouse sera to identified surface-exposed antigens, 5) "in vitro neutralization" assay to test whether antibodies elicited by a given antigen can interfere with the process of eukaryotic cell infection, and 6) use of appropriate animal model to test the capacity of selected antigens to confer protection against Chlamydia challenge.
As recently described by Montigiani et al ((2002) Infection and Immunity 70: 368- 379) from the initial screening of the C.pneumoniae genome, a panel of mouse sera was prepared against over 170 recombinant His-tagged or GST-fusion proteins encoded by genes or "open reading frames" somehow predicted to be peripherally located in the Chlamydial cell. When these antibodies were tested in a FACS assay for their ability to bind the surface of purified C.pneumoniae EBs, a list of 53 "FACS- positive" sera was obtained. The corresponding putative surface antigens were then further assessed for their capability of inducing neutralizing antibodies. This part of the work involved testing which of the sera contained antibodies capable of interfering with the process of in vitro infection of epithelial cell cultures. In the in vitro "neutralization" assay purified infectious EBs are incubated with progressive dilutions of the immune sera and, in parallel, dilutions of the corresponding pre- immune sera, and of sera against non Chlamydia control antigens.
Cell cultures are infected in the presence of cycloheximide, which inhibits host cell protein synthesis and favours Chlamydial intracellular growth with the consequent formation of typical cytoplasmic inclusions which can be stained with Chlamydia specific fluorescence-labeled monoclonal antibodies and counted with an UV light microscope. Working with appropriate pathogen-to-host cell ratios, it can be reasonably assumed that the number of detected cytoplasmic inclusion is proportional to the number of infectious Chlamydiae in the original sample. So a reduction in inclusion numbers caused by the presence of an antigen-specific antiserum, as compared to the numbers obtained with control sera, gives a measure of the capability of a given antigen to elicit antibodies which can inhibit some stage of the Chlamydial infection process. According to common convention, an anti-serum is labelled as 'neutralizing' when the reduction of infectivity is equal or greater than 50%>, and the serum dilution yielding a 50% reduction in infectivity is referred to as the 50% end- point neutralization titer.
Some of the results obtained by screening the panel of recombinant antigens with the C.pneumoniae in vitro neutralization assay confirm that some of the listed antigens, like the members of the family of heterogeneous polymoφhic membrane proteins (PMP), which, on the basis of published literature data, could be reasonably expected to be surface-exposed and possibly induce neutralizing antibodies. However, there are also proteins which could be considered so far only hypothetical, and proteins which just on the basis of their current functional annotation could not be at all expected to be found on the bacterial surface. Using an in vitro neutralising assay, it was found that sera to 10 CPn antigens have so far proved to effectively neutralize in vitro infectivity to an extent greater than 50%, a property that common practice qualifies such antigens as "neutralising" (Figure 1). These 10 sera were obtained by mouse immunization with recombinant proteins derived from the C.pneumoniae genes listed below.
Using a recently described in vivo model of systemic infection (hamster model), hamsters immunised with 6 of the in vitro neutralising antigens, when challenged with CPn EBs, showed a greater than 80%> reduction of spleen infection as compared with non-immunised controls.
Characterisation of 10 CPn proteins
The proteins identified by the present work can be divided in 3 groups: • proteins which have an annotation compatible with (could be reasonably expected to have) an expected/predicted exposure on the Chlamydial cell surface and with the possibility that antibodies binding to them may actually interfere with host cell attachment and entry (ie proteins which could possibly induce neutralising antibodies)
• proteins which by homology with other gram-negative bacteria could be expected to have a periplasmic exposure (ie would not be expected at all to be found on the bacterial cell surface); and
• proteins which are still labelled as 'hypothetical' (ie cellular location and/or cellular function not known) Group 1
(Pmp proteins (pmp2 and pmplO), OmcA and OmpH)
Pmp proteins (pmp2 and pmp 10)
The first group includes the 2 polymoφhic outer membrane proteins (Pmp's) Pmp2 and PmplO (10, 11, 14, 30), the outer membrane protein OmpH-like, and OmcA, which is annotated (Chlamydia Genome Project at htkp-J/Chlamydia- www.berkeley.edu:4231/) as "predicted 9-kD cysteine-rich, outer membrane protein, lipoprotein". The Pmp family of Chlamydia-specific proteins is generally thought to comprise probable pathogenicity factors, with an autonomous secretion capacity (autotransporters), important for adhesion to host cells and are generally considered as promising vaccine candidates. However, apart from very recent unpublished results on Pmp21, this is the first time that antisera to recombinant Pmp's are reported to have neutralizing properties. OmcA
OmcA is the product of a gene co-transcribed in the same operon with the 60 kDa OmcB cystein-rich protein which is a major structural component of the Chlamydial outer membrane and a major immunogen in human C. trachomatis infections. OmcB and OmcA are likely to interact in some as yet unknown outer membrane structure, so it is possible that antibodies to OmcA can interfere with EB infectivity.
OmpH
Finally, the Chlamydial OmpH is probably a member of the OmpH (Skp) family of proteins which have been reported to have chaperonin activities in other bacteria very important for the correct biosynthesis of the outer membrane. These proteins appear to cooperate in this task with HtrA (see below). In fact, in E.coli single KO mutants of either OmpH (Skp) or HtrA (DegP) are still viable, but double mutants do not grow
(37). It should be pointed out that even if the amino acid sequences of the ompH-like proteins of Chlamydia (all C.pneumoniae and C.trachomatis or C.caviae variants) line-up very well with the rest of the bacterial OmpH proteins, they are the only ones to be acidic, whereas the rest of the family comprises mostly very basic proteins
(including some with histone like behaviour, at least in vitro). One could speculate that if the chaperone activity is maintained also in the ompH like Chlamydial proteins, this may be related to some Chlamydial peculiarity.
Second Group of Selected Proteins (ArtJ, AtoS, HtrA and Enolase)
The second group, which represents a somehow smprising finding, includes ArtJ, AtoS, HtrA and Enolase. If the current annotation (justified by analogy with homologous genes in other bacteria) is correct, all these proteins would be expected to have a periplasmic location in gram-negative bacteria, and to be surface-exposed only in a gram-positive bacterium. It is possible that owing to their atypical life cycle, requiring an efficient passage from a dormant spore-like status (the EB) to an active form needing to adapt quickly to host-cell responses to invasion, Chlamydiae in fact display some sensors directly on the outer surface of their infectious form.
ArtJ
In the case ArtJ - for which we have data supporting both antigen expression and serum specificity - the hypothesis of an atypical situation peculiar to Chlamydia is supported by the anomalous gene set-up resulting from the present analysis of the Chlamydia genomes. ArtJ is so annotated by analogy with the ART transport systems of E.coli which has 5 genes organized in two operons (24) : artPIQM and artJ which are responsible for the arginine transport. In Cpn however the artPIQM genes are absent and therefore it appears that Chlamydial ArtJ operates in a molecular context which is different from its E.coli model and must be peculiar to this species.
HtrA
HtrA (DegP), which in other bacteria has a complex hexameric structure, has been described as having multiple functions (3, 5, 18, 19, 27, 38) : a chaperonin assisting a correct outer membrane biogenesis, inducible protease for the elimination of misfolded membrane proteins, and also a sensor of 'stress' conditions. In Chlamydia none of these properties has been demonstrated yet, however we find that in purified EB HtrA is present in two forms one of which appears to be processed by being deprived of the N-terminal fragment. This fragment, if aligned with the homologous HtrA sequence from Thermologa maritima (18), would comprise a predicted loop acting as a structural lid controlling the access to the protease active. So it appears tempting to speculate that HtrA could have a similar protease activity and the two forms identified on the 2-D map represent the active and inactive species. Interestingly, the C. trachomatis HtrA ortholog is recognized by human sera from patients who had a Chlamydial genital infection (35), and a similarly HtrA is one of the antigens in the immunoproteome of Helicobacter pylori (13). Furthermore the homologue protein in Haemophilus influenzae is a protective antigen in both a passive infant rat model of bacteremia and the active chinchilla model of otitis media (23) .
Enolase
Also in the second group of proteins expected to be located elsewhere than the cell surface, is Cpn enolase. This protein aligns with the well known family of conserved glycosylases, which are essentially cytoplasmic enzymes, but in Streptococci enolase has been shown to have also a cell surface location, and extracellular matrix binding properties (1, 28, 29)). Interestingly, Gaston and colleagues (8) also showed that in patients with reactive arthritis induced by C. trachomatis, enolase induces specific CD4+ T-cell responses. Furthermore, a clone responding to the enolase C.trachomatis ortholog, responded also to C.pneumoniae EBs, and, since no proliferative response could be observed using a fungal or a mammalian enolase, the authors of this study concluded that the CD4 T-cell stimulating epitope must be Chlamydia specific.
Third Group of Proteins
(unknown cellular location and/or cellular function, Cpn0795, CPn0042) The third of the 3 groups in which we propose to divide, just for the sake of discussion, the 10 neutralizing antigens above described, comprises two proteins which are still annotated in public Chlamydial databases as the hypothetical products of two CPn-specific genes: Cpn0759 and Cpn0042. The Cpn0759 gene is the second gene in a cluster of 6 Cpn-specific hypothetical genes (from Cpn0794 to Cpn0799) immediately upstream of the enolase gene. With the exception of Cpn0759 the products of all the other genes in the cluster share similarities of 30 to 40% over long stretches of amino acids. The Cpn0042 gene encodes a hypothetical protein, with 4 coiled-coil regions, which has been described as a member of a new family of hypervariable outer membrane proteins (33). Interestingly, the hypervariability of these proteins could be due to a strand-slippage mechanism induced by the presence of a poly(C) stretch within the coding region of the corresponding genes, a mechanism already described in the Pmp's family for the pmpl 0 gene (30). However, as indicated by their annotation, the function of these proteins is still unknown, and our observations provide the first experimental indication of a possible function related to the Chlamydial infection process.
Table 1 of this application demonstrates that Cpn0795 (SEQ ID NO: 6) a Cpn specific hypothetical protein is a FACS positive protein which demonstrates significant immunoprotective activity in a hamster spleen model of Chlamydia pneumoniae infection. We have found evidence to demonstrate that other Cpn proteins in this group of Cpn specific hypothetical proteins have now been found to have a secreted autotransporter function. These proteins, which are absent from Chlamydia trachomatis include: gi/4377105 (Cpn0794), gi/4377106 (Cpn0795), gi/4377107 (Cpn0796), gi 4377108 (Cpn0797), gi/4377109 (CPn0798), gi/4377110 (Cpn0799). Fig. 6 shows an alignment of the proteins in the 7105-7110 protein family. This
Alignment shows a new family of proteins expected to constitute a system of antigens probably delivered on the Cpn surface or secreted by a type V (autotransporter) secretion mechanism. This alignment was generated as follows:
Imperfect repeats were identified which allowed the alignment of the genes.
Molecular modelling has also demonstrated that the C-terminal ends of 7106 and
7107 can be predicted to fold in a beta-barrel structure which can form a translocation pore for secretion across the outer membrane. Cpn0794 = 7105 = FACS positive
Cpn0795 = 7106 = FACS positive
Cpn0796 = 7107 = FACS positive
Cpn0797 = 7108 = FACS positive
Cpn0798 = 7109 = No data available Cpn0799 = 7110 = No data available
(Reference for FACS positive data = Montigiani et al (2002) Infect Immun 70(1) 368-
79)
Operonl = 0794, 0795, 0796 Operon2 = 0797, 0798
Cpn0795 and Cpn0796 have C terminal ends that may form transmembrane pores (see alignment, FIG. 9). CPn0794, Cpn0797, Cpn0798, and Cpn0799 have N-terminal ends indicating that all proteins have N-terminal and C-terminal ends.
Fig. 7 shows alignment of Cpn0794 - Cpn 0799. Proteins encoded by the genes Cpn0794, Cpn0795, Cpn0796, and Cpn0797 have been identified as likely to be exposed on the surface of the chlamydia cell and as possible vaccine candidates. These proteins are shown to be actually expressed by Cpn in vivo (WB data and FACS data). In the case of Cpn0797 we also showed that the level of expression in CPn EBs is high enough to be detected by mass spectrometry analysis on 2DE maps of protein extracts (see Montigiani et al.)
Following these observations, it is seen that the proteins encoded by Cpn0794, Cpn0796 and Cpn0797 proteins can be aligned according to a set of imperfect repeats present within their aminoacid sequences (see FIG. 7) , whereas the putative product of CPn0795 can be mostly aligned to the C-terminal portion of the Cpn0796 protein.
Furthermore, proteins encoded by genes Cpn0798 or Cpn0799 can alse be aligned to the above proteins according to the above mentioned repeated sequence motifs (see FIG. 7). Overall alignment of the 6 genes demonstrates that the genes encode for a family of functionally-related proteins.
Furthermore, in silico analysis of the protein encoded by Cpn0796, which encompasses the entire alignment of all the proteins in this family demonstrates that a functional precursor with the aminoacid sequence reported below:
SEQ ID NO: 80
MKPMKVLTPWIYRKD VTAF LTAIPGSFAHT VDIAGEPRHAAQATGVSGDGKIVIGMKVPDDPFA ITVGFQYIDGH QPLEAVRPQCSVYPNGITPDGTVIVGTNYAIGMGSVAVKWVNGKVSE PMLPDTLD SVASAVSADGRVIGGNRNIN GASVAVK EDDVITQLPSLPDAMNACVNGISSDGSIIVGTMVDVS R NTAVQ IGDQLSVIGTLGGTTSVASAISTDGTVIVGGSENADSQTHAYAYKNGVMSDIGTLGGFYS A HAVSSDGSVIVGVSTNSEHRYHAFQYADGQMVDLGTLGGPESYAQGVSGDGKVIVGRAQVPSGD HAF LCPFQAPSPAPVHGGSTWTSQNPRGMVDINATYSSLKNSQQQLQR IQHSAKVESVSSGAPSFTSV KGAISKQSPAVQNDVQKGTFLSYRSQVHGNVQNQQ TGAFMD KLASAPKCGFKVALHYGSQDALVE RAA PYTEQGLGSSVLSGFGGQVQGRYDFNLGETW QPFMGIQVLH SREGYSEK VRFPVSYDSVA YSAATSF GAHVFASL.SPKMSTAAT GVERD NSHIDEFKGSVSAMGNFVLENSTVSVLRPFAS AMY YDVRQQQLVT SWMNQQPLTGT S VSQSSYNLSF Processing sites that assiste in the secretion of the polypeptide from the cytoplasm and its release into the periplasm are located after aminoacid 31 (based on PSORT prediction and/or after aminoacid 47 similar to experimentally determined processing sites in other bacterial autotransporter molecules (e.g. BrkA from B.pertussis). Hence, the mature form of the Cpn0796 product is as follows:
SEQ ID NO: 81
HTLVDIAGEPRHAAQATGVSGDGKIVIGMKVPDDPFAITVGFQYIDGH QPLEAVRPQCSVYPNGITP D GTVIVGTNYAIGMGSVAVK VNGKVSELPMLPDTLDSVASAVSADGRVIGGNRNINLGASVAVKWEDD VITQLPSLPDAMNACV GISSDGSIIVGTMVDVS RNTAVQWIGDQLSVIGTLGGTTSVASAISTDGT VIVGGSENADSQTHAYAYKNGVMSDIGT GGFYSLAHAVSSDGSVIVGVSTNSEHRYHAFQYADGQMV DLGTLGGPESYAQGVSGDGKVIVGRAQVPSGDWHAF CPFQAPSPAPVHGGSTWTSQNPRGMVDINA TYSS KNSQQQ QRLLIQHSAKVESVSSGAPSFTSVKGAISKQSPAVQNDVQKGTF SYRSQVHGNVQ NQQLLTGAFMDWKLASAPKCGFKVALHYGSQDALVERAALPYTEQGLGSSV SGFGGQVQGRYDFNLG ETVVLQPFMGIQV HLSREGYSEKNVRFPVSYDSVAYSAATSFMGAHVFASLSPKMSTAAT GVERD NSHIDEFKGSVSAMGNFV ENSTVSV RPFASLA YYDVRQQQLVT SWMNQQPLTGTLSLVSQSSY NLSF Or
SEQ ID NO: 82 TGVSGDGKIVIGMKVPDDPFAITVGFQYIDGHLQPLEAVRPQCSVYPNGITPDGTVIVGTNYAIG MGSVAVK VNGKVSELPMLPDT DSVASAVSADGRVIGGNRNIN GASVAVKWEDDV1TQLPSLP DAMNACVNGISSDGSIIVGT VDVS RNTAVQ IGDQLSVIGTLGGTTSVASAISTDGTVIVGGS ENADSQTHAYAYKNGVMSDIGTLGGFYS AHAVSSDGSVIVGVSTNSEHRYHAFQYADGQMVDLG TLGGPESYAQGVSGDGKVIVGRAQVPSGD HAFLCPFQAPSPAPVHGGSTWTSQNPRGMVD1NA TYSS KNSQQQ QRLLIQHSAKVESVSSGAPSFTSVKGAISKQSPAVQNDVQKGTFLSYRSQVHG VQNQQL TGAFMDWK ASAPKCGFKVA HYGSQDALVERAALPYTEQG GSSVLSGFGGQVQGR YDFNLGETWLQPFMGIQV H SREGYSEKNVRFPVSYDSVAYSAATSFMGAHVFAS SPKMSTA ATLGVERDLNSHIDEFKGSVSAMGNFVLENSTVSVLRPFASLA YYDVRQQQLVTLSWMNQQPL TGTLSLVSQSSYNLSF In silico analysis of the protein encoded by Cpn0796 also demonstrates a C-terminal domain comprising approximately residues from 1 to 648. FIG. 8 illustrates Cpn0796. As shown in FIG. 8, Cpn0796 forms a beta-barrel structure and is capable of forming a pore across the bacterial outer membrane (OM). As is typical of 'autotransporter' molecules, after being secreted across the bacterial inner membrane into the periplasm tlirough an N-terminal signal peptide mechanism, the molecule may form a pore in the OM through which the N-terminal domain may pass (the 'passenger' domain) to the outside of the bacterial cell. Also, these molecules may either remain anchored to the bacterial surface or undergo a proteolytic cut which releases the 'passenger domain' or a portion of it into the medium surrounding the bacterial cell an example of which is represented in the following sequence:
SEQ ID NO: 83
MKFMKV TP IYRKDL VTAF TAIPGSFAHTLVDIAGEPRHAAQATGV SGDGKIVIGMKVPDDPFAITVGFQYIDGHLQP EAVRPQCSVYPNGITPD GTVIVGTNYAIGMGSVAVKWVNGKVSE PM PDTLDSVASAVSADGRVIG GNRNINLGASVAVKWEDDVITQLPS PDAMNACV GISSDGSIIVGTMVD VS RNTAVQWIGDQLSVIGTLGGTTSVASAISTDGTVIVGGSENADSQTH AYAYKNGVMSDIGTLGGFYSLAHAVSSDGSVIVGVSTNSEHRYHAFQYAD GQMVDLGTLGGPESYAQGVSGDGKVIVGRAQVPSGD HAF CPFQAPSPA PVHGGSTWTSQNPRGMVDINATYSSLKNSQQQLQ RLLIQHSAKVESVSSGAPSFT5VKGAISKQSPAVQNDVQKGTFLSYRSQVHGNVQNQQ TGAFM D KASAPKCGFKVALHYGSQDALVERAALPYTEQGLGSSVLSGFGGQVQ GRYDFNLGETWLQPFMGIQV HLSREGYSEKNVRFPVSYDSVAYSAATS FMGAHVFASLSPKMSTAATLGVERD NSHIDEFKGSVSAMGNFV ENSTV
SVLRPFASLAMYYDVRQQQLVTLSWMNQQPLTGTLSLVSQSSYNLSF
Also shown in FIG. 8, amino acid residues 365-385 represent an alpha helix conformation that spans the beta barrel pore
The N-terminal passenger domain may be cleaved via a specific proteolytic action from the membrane-anchored pore structure. A linker domain comprising the peptide sequence PSPAPV (SEQ ID NO: 84) as shown in bold in the following sequence illustrates a site at which cleavage of the N-terminal passenger domain may occur:
SEQ ID NO: 85 HTLVDIAGEPRHAAQATGVSGDGKIVIGMKVPDDPFAITVGFQYIDGHLQPLEAVRPQCSVYPNG 1TPDGTVIVGTNYAIGMGSVAVKVNGKVSE PMLPDTLDSVASAVSADGRVIGGNRNINLGASV AVK EDDVITQLPS PDAMNACVNGISSDGSIIVGTMVDVS RNTAVQWIGDQLSVIGTLGGTTS VASAISTDGTVIVGGSENADSQTHAYAYKNGVMSDIGTLGGFYSLAHAVSSDGSVIVGVSTNSEH RYHAFQYADGQ VDLGT GGPESYAQGVSGDGKVIVGRAQVPSGD HAF CPFQAPSPAPVHGGS TWTSQNPRGMVDINATYSS KNSQQQLQRLLIQHSAKVESVSSGAPSFTSVKGAISKQSPAVQN DVQKGTFLSYRSQVHGNVQNQQ LTGAFMDWKLASAPKCGFKVALHYGSQDALVERAA PYTEQG LGSSVLSGFGGQVQGRYDFNLGETWLQPFMGIQVLHLSREGYSE NVRFPVSYDSVAYSAATSF MGAHVFASLSPKMSTAAT GVERDLNSHIDEFKGSVSAMGNFV ENSTVSVLRPFASLAMYYDVR QQQLVTLSWMNQQPLTGTLSLVSQSSYNLSF
The N-terminal peptide may be secreted to be exposed on the bacterial cell surface and can also become detached via the proteolytic event described above. The peptide may form a structural conformation known as beta-propellers indicated in the following sequence:
SEQ ID NO: 86 HTLVDIAGEPRHAAQATGVSGDGKIVIGMKVPDDPFAITVGFQYIDGH QPLEAVRPQCSVYPNG ITPDGTVIVGTNYAIGMGSVAVKVNGKVSELP PDT DSVASAVSADGRVIGGNRNIN GASV AVK EDDVITQLPSLPDAMNACV GISSDGSIIVGTMVDVS R TAVQ IGDQLSVIGT GGTTS VASAISTDGTVIVGGSENADSQTHAYAYKNGVMSDIGT GGFYSLAHAVSSDGSVIVGVSTNSEH RYHAFQYADGQMVD GTLGGPESYAQGVSGDGKVIVGRAQVPSGD HAF C Furthermore, the N-terminal passenger domain can also possess a specific protease activity, such as a serine protease-like activity. In addition to acting on a variety of substrates, the protease activity may act on the membrane anchored form of the molecule such that the N-terminal passenger domain is cleaved off form the surface of the chlamydial cell. The serine protease like activity is supported by the presence of a consensus serine protease triad of adequately spaced amino acid residues (namely H, D and S) which can be located on the virtual structure of the 'passenger' domain modelled on a set of experimentally-determined templates, e.g. InrO (PDB identification code)
Based on the above analysis, the gene Cpn0796 gene encodes for a protein which promotes its own secretion on the EB surface and may also mediate or promote its own release into the surrounding medium. The secreted passenger peptide has several activities, including: 1. actin binding peptide, part of a chlamydial surface layer, and instrumental to the process of establishing the host cell infection 2. specific protease activity within the host cell cytoplasm instrumental to the intracellular survival of infecting chlamydiae. 3. specific activity within the host cell cytoplasm to down regulate expression of selected genes, either by repressing their transcription and/or by repressing their translation (m-RNA degradation) 4. cooperation with the products of genes Cpn0794, 0795, 0797, 0798, 0799 5. another function of the above N-terminal beta propeller domain is the regulation modulation of the activity of a cytosolic protease of the host cell in order to alter host cell properties in favour of chlamydial development, survival or persistence. See Fulop V, Bocskei Z, Polgar L. in "Prolyl oligopeptidase: an unusual beta-propeller domain regulates proteolysis." Cell. 1998 Jul 24;94(2): 161-70.
The proteins encoded by Cpn0794, Cpn0797, Cpn0798, Cpn0799 - all comprising variants of the above described Cpn0796 structure - also provide beta propeller structures with activities similar and/or complementary to the ones described above. Thus, a family of proteins cooperating to a common function either by generating - through events of site specific recombination - new molecules with structures and activities similar to the above described Cpn0796 product, OR by independently contributing to a multi-protein structure requiring a coordinated action of several related components.
FIG. 9 illustrates an alignment of the C-terminal domains of the proteins encoded by C.pneumoniae genes Cpn0795 and Cpn0796. As seen in FIG. 9, beta barrel domains of Cpn0795 or Cpn0796 include MKDLGTLGG (SEQ ID NO: 87), SXDGK (SEQ ID NO: 88) VIVG (SEQ ID NO: 89), VIXG (SEQ ID NO: 90) or HAF (SEQ ID NO: 91).
Fourth Group of Proteins Cpn0498
So in this case the triple parallel-screening evaluation, with two positive and one negative result, brought to the identification of a previously unknown antigen (ie an antigen with unknown biological function) having, according to current views, just the desirable basic properties in terms of antigenic function of a vaccine candidate. Further characterization of Cpn antigen data is included in Finco et al, "Identification of New Potential Vaccine Candidates Against Chlamydia pneumoniae by Multiple Screenings," Vaccine, 23 (2005) 1178-1188, incoφorated herein in its entirety.
Example 5 Background
The main stages in the Chlamydial life cycle are: (i) the binding to the host cell surface and entry into the cytoplasm through a specialised vacuole (the Chlamydial inclusion) by an extracellular sporelike infective form, called the elementary body (EB); and (ii) the conversion of the EB to a non-infective replicative form called a reticulate body (RB) that replicates by binary fission a number of times within the inclusion to form a microcolony.
The sets of genes which are expressed in the various stages of the Chlamydial life cycle and the signals that trigger the passage from one stage to the next are largely unknown and still need investigation. Protein microarrays are used for high throughput protein analysis by detecting proteins and monitoring their expression levels. Through use of protein microarrays, complex screening of thousands of proteins and interactions with proteins may be performed in parallel. A protein array typically includes a surface, such as glass, membrane, microtiter wells, mass spectrometer plates, beads or other particles, for binding ligands, proteins, or antibodies. For example, antibodies may be bound to the microarray to form a capture array. The capture array may be contacted with a biological sample to quantify the proteins in the biological sample. Also, proteins may be bound to the microarray and contacted with a biological sample to quantify protein-protein or protein-ligand interactions. Thus, protein microarrays may also be used in diagnostics in which multiple immunoassays may be conducted in parallel such that levels of proteins in different samples may be quantified and compared for applications in the treatment or diagnosis of disease.
For example, in a capture array, antibodies are bound to the microarray and exposed to a biological sample. Proteins and ligands that bind to the antibody array may be detected by direct labelling of the bound proteins. If a higher sensitivity or specificity is desired, a sandwich technique may be employed in which pairs of antibodies are directed to the same protein ligand. This technique is particularly useful if the amount of protein to be detected is low or if there are modifications to the protein. In addition, the use of sandwich assays minimizes the risk of cross-reactivity in highly multiplexed assays by providing dual level target recognition, i.e. two levels of specificity for each locus in the array. Alternatively, the bound proteins may be detected via label-free detection methods such as including mass spectrometry, surface plasmon resonance and atomic force microscopy. This technique is useful if modification or alteration of the protein is to be avoided.
Also, Large-scale functional chips containing large numbers of immobilized purified proteins may be used to assay a wide range of biochemical functions, such as protein interactions with other proteins, drug-target interactions, enzyme-substrates, etc. Such proteins may be purified from an expression library, for example, and the protein array can be used to screen libraries to select specific binding partners, including antibodies, synthetic scaffolds, peptides and aptamers. In this way, 'library against library' screening can be carried out, such as screening of drug candidates in combinatorial chemical libraries against an array of protein targets identified from genome projects.
Protein microarray technology permits analysis of the proteins themselves rather than inferring protein function, interactions and characteristics through mRNA expression. In many cases, mRNA expression does not correlate accurately with protein abundance. Furthermore, mRNA expression analysis does not provide sufficient information on protein-protein interaction or post-translational modifications. Thus, direct analysis of proteins via protein microarrays provides an advantage by providing more accurate information of proteins and protein-protein interactions that may not be readily available through measurment of mRNA expression.
Current DNA microarray techniques permit profiling of gene expression at the mRNA level as a function of the cellular state. This can lead to the identification of genes or clusters of genes whose up- or down-regulation is associated to a particular state of the cell and to the identification of therapeutically relevant targets. Using this technology, DNA fragments representing specific portions of all genes belonging to a given organism (the fragments can be derived from cDNA libraries or can be obtained by PCR amplification and chemical synthesis) are chemically bound to the surfaces of solid supports (chips) at high densities and in an ordered manner. Currently up to 10, 000 DNA fragments or 250, 000 oligonucleotides can be spotted onto a single squared centimetre of chip surface. The DNA chips are then utilised to define which of the spotted genes are transcriptionally active in a particular cellular population. To do so, RNA is prepared, labelled with fluorescent dyes and finally hybridised to the DNA fragments fixed to the surface of the chip. By using an appropriate computer-assisted fluorescence detector, the fluorescence signals emitted by each spot upon excitation with a laser beam is carefully quantified to define the transcription activity of all the arrayed genes.
CPn DNA microarrays have been developed to look at the transcriptional events which occur when a given CPn pathogen gets into contact with the host cells, both in in vivo and in vitro settings. DNA chips carrying the entire genome of a particular bacterium, such as the CPn bacterium can be prepared in a very short period of time so that whole genome expression analysis can be determined.
Experimental Methodology Specifically, a genomic DNA (open reading frame probes) microarray approach for gene expression in CPn bacteria was adopted. In this respect, an array was prepared for the analysis of the CPn life cycle on the basis of the published annotation of the complete genome. The Chlamydia DNA chips carry about 1000 PCR-derived DNA fragments, which have an average size of 400-700bp and correspond to internal portions of all CPn annotated genes.
Results 5
Table 3(i)-(xi) shows transcriptional activity for expressed genes for CPn EB selected by microarray. The data in Tables 3(i)-(iv) shows different rnRNAs in order of abundance from cells in their infectious "spore-like" (EB) form. Data in Tables 3(v)- (xi) correlates and summarizes mRNA expression levels of genes for CPn. The cells were used at the end of their cycle where gene expression is likely to be at its highest. As values less than approximately 10000 is likely to be background, the top set of proteins (approx top 30) with more intense signals are likely to be the most interesting proteins.
A review of the values for the expressed genes indicates that three of the FACS positive CPn antigens (CPn0331 (hypothetical), CPn0234 (hypothetical) and CPn0572 (hypothetical) are all highly expressed genes. Table 3(v)-(xi) shows the transcriptional activity for expressed genes for CPn EB selected by microarray. The Table shows different mRNA in order of abundance from cells in their infectious "spore-like" (EB) form. The cells were used at the end of the cycle where gene expression is likely to be at its highest. A review of Table 3(i)-(iv) and (v)-(xi) indicates that CPn antigens CPn0558 (OmcA), CPn0331 (hypothetical), CPn0539 (Pmpl9), CPn0234 (Hypothetical) and CPn0572 (Hypothetical) are all relatively highly expressed genes.
Where possible, attempts were made to place the transcriptional activities disclosed in Table 3(v)-(xi) in the context of the Chlamydia developmental cycle In this respect, Chlamdydia late gene products have been described more frequently than early gene products. This is primarily because of the presence of late gene products in EBs but not RBs and that it is easier to study EBs rather than RBs.
In addition, late gene functions appear to be predominantly those associated with the terminal differentiation of RBs back to EBs (Shaw et al., Mol Microbiology 37(4), 2000, 913-925). Late gene products appear to function in the termination of bacterial cell division and constitute structural components and remodelling activities involved in the formation of the cross-linked outer membrane complex that functions in the attachment and invasion of new host cells. By way of example, an important aspect of the secondary differentiation process (RB to infectious EB) is the expression of genes that encode proteins that form the highly disulfide cross-linked bacterial outer membrane (OM) complex. It is thought that several late cycle genes encode proteins with potential roles in the formation and maturation of the OM complex, a crucial step in the development of infectious EBs (see Belland et al., PNAS (USA) 100(14), 2003, 8478-83). The late genes omcA and omcB encode two cysteine-rich OM proteins that interact with the major OM protein (OmpA) to form this complex. A key protein component of the OM complex, the OmcB protein, has been found to undergo post- translational proteolytic processing. We have found that OmcB and OmcA show high levels of transcriptional activity (see top of Table 3(ii)). Cpn 0384 whose CT equivalent is CT046 (hctB) has been shown to be associated with differentiation from RB to EB (see Belland et al, PNAS (USA) 100(14), 2003, 8478-83). We also found Cpn0384 to have relatively high levels of transcriptional activity (again see top of Table 3(v)-(xi)). Other Cpn antigens thought to be involved in the Type III secretion system were found to have moderate expression levels in terms of transcriptional activity. This finding appears to be in line with published commentary where it is thought that while transcription of the two putative structural components of the Type III secretion system (yscJ and yscN (Cpn669)) begins at mid-cycle, export of effector molecules may be at a different stage of the developmental cycle. Table 3(v)-(χi) indicates that high transcriptional activity was observed for Cpn0539 (CT412) which corresponds with a 98Kda protein known either as PmpA or Pmpl9. Even though the Pmp 19 protein demonstrates relatively "high" levels of transcriptional activity, this result is interesting because mRNA abundance for pmp 19 does not seem to correlate with protein abundance. In this respect, results from our laboratory have shown that (i) Pmp 19 was not detected in either 2D maps, Western Blots or FACS analysis studies which suggests that the pmp 19 protein either is not surface exposed even though high levels of mRNA are expressed or that (ii) Pmp 19 protein is expressed but processed or degraded by proteolytic digestion rendering it undectable by immunoblot analysis. The results in our laboratory are confirmed by others. In this respect, Grimwood et al (2001) Infection and Immunity 69(4) 2383- 2389 have shown that transcriptional profiles were detected for each of the Chlamydia pneumoniae 21 Pmp genes demonstrating that all pmp genes are transcribed during infection. Since each of the Pmp genes was transcribed, Grimwood et al (2001) evaluated protein expression by immunoblotting of Chlamydia pneumoniae CWL029 EB lysates using peptide-specific antisera. Interestingly, no Pmp-specific reactivity was detected for sera from either PmpA (Pmp 19) or PmpB/C and PmpD gene by immunoblot analysis regardless of high antipeptide reactivity. This result suggested that these proteins either are not stable or not translated. These findings demonstrate that there appears to be a variation in Pmp expression for the Chlamydia pneumoniae family of 21 polymoφhic membrane proteins (Pmps) which are predicted to be localised to the bacterial outer membrane. The function of Chlamydial Pmps remains unknown, although based on sequence prediction and experimental testing, these Pmps are regarded as surface proteins and thus, likely to be critical for Chlamydial virulence. Like the Inclusion (Inc) Membrane proteins, the Pmp proteins are regarded, at present, as unique to the Chlamydiae family (see Rockey et al (2000) Infection and Immunity 69(10) 5473-5479). The findings disclosed here and by others, such as Grimwood et al, demonstrates that the Chlamydia organism appears to expend a considerable metabolic cost in Pmp transcription, such as Pmp 19 transcription, despite the potential lack of production of a functional Pmp proteins, such as the Pmp 19 protein.
Materials and Methods (Examples 6-8) (Reference Section II) T cell Epitope prediction and peptide synthesis
T cell epitope prediction was carried out on the genomic sequence of C. pneumoniae CWL029 strain (Accession numbers NC 000922 or AE001363. using the BIMAS algorithm [24]. Synthetic peptides (purity > 80%o) were synthesized by Primm Sri (Milan, Italy), suspended in 100% DMSO and kept at -20° C before use.
RMA-S/A2 cell line and HLA-A2 transgenic and non transgenic mice
The T cell lymphoma murine cell line RMA-S stably transfected with HLA-A2 (RMA-S/A2, H-2b , TAP2"), was kindly provided by Dr. Barnaba, Universita degli Studi "La Sapienza", Rome, Italy, and cultured at 37° C in RPMI-1640 (GIBCO) supplemented with heat inactivated 10%) FCS, 100 RJ/ml penicillin/streptomycin, 2 mM Lglutamine (GIBCO) and 5x10-5 M 2-ME (Sigma). H2-b HLA-A2 transgenic mice [35] were housed in a pathogen-free environment and screened for HLA-A2 expression by FCM carried out on total blood samples using the BB7.2 anti-A2 mAb [48]. Only mice with percentages of A2 expressing cells higher than 70-80 %> were used for DNA immunization and C. pneumoniae infection experiments. Animals which showed no HLA-A2 expression were mated in order to obtain an HLA-A2 non transgenic population, to be used as a control in the experiments. Epitope stabilization assay
RMA-S/A2 cells (3-5 x 105/well) were seeded in serum-free RPMI medium, supplemented with human β2 microglobulin (3 μg/ml, Sigma), without or with the test peptide (lOμM). Following overnight incubation at 26°C in humidified 5% C02 atmosphere, cells were shifted to 37° C for 2 h before determining the HLA-A2 expression level at the cell surface using the BB7.2 anti-A2 mAb and a PE-conjugated anti-mouse IgG (Jackson ImmunoResearch). Fluorescence intensity on living cells, which did not incoφorate propidium iodide, was analyzed by FCM. As controls, corresponding samples without peptide and samples with peptide but treated only with the anti-mouse secondary antibody, were used.
Infection and DNA immunization of HLA-A2 transgenic and non transgenic mice
Transgenic mice were intranasally infected twice with a month interval, using 5xl05
C. pneumoniae FB/96 EBs [4] diluted in 50 μl of PBS. C. pneumoniae antigen coding genes were amplified by PCR using FB/96 genomic DNA, cloned into plasmid pcmvKaSF2120 [49] and verified by DNA sequence analysis. Fifty μg of endotoxin free recombinant plasmid DNA, diluted in Dulbecco's phosphate buffer (GIBCO), were injected into the tibialis muscle of mice at days 0, 21 and 35. CD8+ T cells isolation and IFN-γ determination by ELISpot assay
Splenocytes from DNA immunized mice were prepared one week after the third immunization using Cell Strainer (Falcon) filters. Following red blood cells lysis, CD8+ T cells from spleen cells suspensions were enriched by positive selection using magnetic activated cell sorting (MACS-Miltenyi Biotec) with CD8a (Ly-2) microbeads. CD8+ T cells purity was higher than 90%>, as determined by FMC. Multiscreen 96-well nitrocellulose plates (Millipore) were coated with 5 μg/ml of the anti-mouse IFN-γ antibody (R4-6A2, PharMingen) in 100 μl of carbonate buffer, pH 9.2. After overnight incubation at 4°C, plates were saturated at 37°C with 200 μl of BSA (1%) in PBS for 2 h. Purified CD8+ (5xl04) were added in a total volume of 200 μl/well in the presence of irradiated (3,000 rad) spleen cells from non immunized HLA-A2 transgenic mice as a source of antigen-presenting cells (2xl05/well), 10 μg/ml of peptide and lOU/ml of human r-IL-2 (Chiron Coφoration). After incubation for 20 h at 37° C, 5%> C02, plates were washed and developed for bound IFN-γ using sequential incubations with biotinylated antimouse IFN-γ (XMB1.2, PharMingen), ExtrAvidin-alkaline phosphatase and substrate BCIP/NBT (Sigma) dissolved in water. Spots were enumerated in each well using a dissecting microscope. Medium containing an irrelevant peptide or without peptide was used as negative controls, while positive controls were represented by CD8+ T cells treated with ConA (5 μg/ml).
In vitro cultures and flow cytometric analysis of splenocytes from transgenic and non transgenic mice infected with C. pneumoniae
Splenocytes from infected mice were isolated one week after the second infection with C. pneumoniae Ebs. For ex vivo analysis of IFN-γ production, 2xl06 splenocytes were seeded in the presence of the test peptide (lOμg/ml) and anti-mouse CD28 antibody (1 μg/ml, PharMingen) as co-stimulus. After a two h incubation at 37° C, 5 % C02, Brefeldin A (10 μg/ml, Sigma) was added and the incubation was extended for 4 additional hours. Following two washes with PBS, cells were penneabilized, fixed and stained both with anti-murine-IFN-γ-(PE), anti-murine CD8 (APC) and anti-murine-CD69 (FITC) and the corresponding isotypes. Positive controls for cytokine production were performed on cells stimulated with anti-mouse CD3 and CD28 antibodies (1 μg/ml, PharMingen) . Cells cultured either in the absence of peptide or pulsed with the HepB negative control peptide were used as negative controls. All samples were analyzed using a FACS LSRII flow cytometer (Becton Dickinson). For analysis of IFN-γ production by short term T cell lines, 5-10xl06 splenocytes from infected mice were cultured for 6 days in the presence of the test peptide (20 μg/ml), with rIL-2 (10 μg/ml) being added after the first two days. At the end of the incubation period, cells were washed twice in RPMI, pulsed again for 6 h in the presence of the test peptide (lOμg/ml), lxlO5 freshly prepared CD8 depleted antigen presenting cells from HLA-A2 transgenic mice (irradiated at 3000 rad) and anti-mouse CD28 antibody (1 μg/ml, PharMingen) as co-stimulus. After a two h incubation at 37° C, 5 % C02, Brefeldin A (10 μg/ml, Sigma) was added, the incubation was extended for 4 additional hours and IFN-γ production was analyzed by FCM.
Example 6
In silico analysis of Chlamydia pneumoniae genome and prediction of HLA-A2 T cell epitopes The genome of the Chlamydia pneumoniae CWL029 strain was used to predict 9mer peptide sequences with high probability to bind class I HLA-A2 molecules. The analysis was carried out using the predictive algorithm available at the NIH Bioinformatics & Molecular Analysis Section Web server (http://bimas.cit.nih.gov/), which ranks potential MHC binders according to the predictive half-time dissociation of peptide/MHC complexes [24]. Since some Chlamydial proteins have been reported to induce autoimmune responses [25-28], we restricted our search to a subset of proteins, distinctive of the Chlamydia genus, consisting of 13 protein identified as members of the type III secretion system, 17 Polymoφhic Membrane Proteins (PMP) and 19 additional proteins, 5 of which already identified as EB surface antigens [4]. The predicted binding score of 157.22, obtained for the well characterized HIV-1 pl7 gag epitope 77SLYNTVATL85 [29], was taken as an arbitrary cut-off for peptide selection. A total of 55 potential C. /.raew/raora/αe-derived T cell epitopes, belonging to 31 different proteins, were identified (Table I), which had predicted binding scores ranging from 156.77 to 42,485.263
In vitro binding of peptides to HLA-A2
The capacity of the selected peptides to bind to HLA-A2 was assessed using an in vitro MHC class I stabilization assay, carried out with the murine transporter associated with antigen processing (TAP)-deficient cell line RMA-S/A2, stably transfected with the human class I A2 gene. MHC class I molecules, cultured at 37° C, are unstably expressed on the cell surface of TAP-deficient cells [30-32]. Culturing the cells at 37° C in the presence of binding peptides, results in formation of a more stable MHC/peptide complex which can be monitored by flow cytometric analysis. RMA-S/A2 cells were therefore cultured overnight at 26° C in the presence of the test peptides, shifted to 37° C for 2 hours and the surface level of stabilized A2 molecules was quantified by direct staining with an anti-HLA-A2 specific mAb. Two known HLA-A2 restricted CTL epitopes were used as positive controls for binding to A2, the HIN-1 pl7 gag peptide [29] and the influenza matrix Ml protein peptide FluMP58 [33], while the Hepatitis B virus envelope antigen peptide HbenvAgl25 (HepB) was used as a negative control [34].
Results 6
The binding results obtained are shown in Table 4 and allowed the identification of 15 peptides with a net mean fluorescence intensity (Net MFI) higher than 92.3, corresponding to the value obtained with the HIV-1 pl7 gag positive control peptide, 8 peptides with a Net MFI intermediate between the values 92.3 and 63.1, obtained with the two positive control peptides, and 12 peptides with an Net MFI ranging between 29.6 and 63. Fifteen of the in silico predicted peptides (27.2 %) did not confer stabilization to the A2 molecules, showing a Net MFI lower than 14, obtained with the HepB negative control peptide.
Example 7
Some HLA-A2 binders are recognized by CD8+ T cells from DNA-immunized transgenic mice
The in vitro assay with RMA-S/A2 cells allowed the definition of a set of peptides which were able to bind and stabilize the HLA-A2 molecules on the cell surface. To gain information about the possibility that the predicted epitopes could indeed be generated by in vivo processing of the antigens from which they were derived, peptide recognition by CD8+ T cells was studied under conditions in which the related complete antigen was intracellularly expressed and presented in vivo. The full-length ORF sequences coding for 13 Chlamydial proteins, including a total of 24 predicted peptides, were cloned into a suitable DNA expression vector and each recombinant plasmids was used to immunize distinct groups of transgenic mice expressing a chimeric class I molecule composed of the αl and α2 domains of HLA- 0201 and the α.3 domains, transmembrane and cytoplasmic, of H-2Kb [35].
The ORF sequences were selected among those containing either one or more epitopes positive in the in vitro assay or a combination of positive and negative epitopes. The ORF sequence corresponding to the outer membrane protein A (OMPA, CPn 0695) was included in this analysis, since human MHC-I-restricted epitopes have already been reported for this protein in C. trachomatis [18;36]. One coding sequence, related to gene CPn 0131 was chosen, which included four epitopes, all negative in the in vitro stabilization assay. After three immunization cycles, transgenic mice were sacrificed, spleen CD8+ T cells were isolated, stimulated for 20 hour with the corresponding peptide and ex vivo IFN-γ production was assessed using an enzyme- linked immunospot (ELISpot) assay,
Results 7 DNA-mediated expression of the ORFs including peptides CH-6 (CPn 0811), CH-7 (CPn 0623), CH-10 (CPn 0828), CH-13 (CPn 0695, OMPA) and CH-37 (CPn 0210) were associated with numbers of spot forming cells (SFC) significantly higher than those obtained with the HepB unrelated peptide, whereas some peptides related to antigens coded by genes CPn 0131, CPn 0323 and CPn 0062 induced SFC values only 2-3 times higher than the HepB control peptide (Table 5). Peptides related to antigens coded by genes CPn 0132, CPn 0322, CPn 0325, CPn 0415 and CPn 0728 did not induce any IFN-γ production (data not shown).
Example 8 To test the capacity of peptides to amplify specific CD8+ T cell populations in vitro, some of these plasmids were used to repeat the DNA immunization experiment and to determine by flow cytometry the intracellular IFN-γ production by CD8+ T cells, both ex vivo and after a 6 day stimulation in the presence of the relevant peptides. In the attempt to establish a direct correlation between IFN-γ production by CD8+ T cells and HLA-A2 specific restriction, the experiment was carried out with both transgenic and non transgenic syngenic mice. The plasmids used contained genes CPn 0695, CPn 0811 and CPn 0823, including peptides CH-13, CH-6 and CH-7 respectively, which were highly positive in the in vitro binding and in the ELISpot assays and gene CPn 0323, including six different peptides, all of them with ELISpot values slightly higher than background
Results 8
The results of the experiment are summarized in Table 6, while representative dot plots from flow cytometric analysis of splenocytes stimulated with peptide CH-6 are shown in Fig. 4. When fresh spleen cells of DNA-immunized transgenic mice were pulsed with the tested peptides, only CH-6 or CH-7 induced relative fold increase (RFI) values about 5 times higher than those obtained pulsing the same cells with the HepB negative control peptide (Table 6, 4.58 and 5.2 RFI respectively). When short term T cell lines (TCLs) instead of fresh splenocytes were used, a larger panel of peptides were able to trigger a significantly higher IFN-γ production by CD8+ T cells (Table 6). In fact, in addition to peptides CH-6 and CH-7, also peptides CH- 13, CH-44, CH-45 and CH-46 were recognized by CD8+ T cell populations significantly larger than those induced by pulsing the same cells with the HepB peptide (RFI > 5). Importantly, peptide-induced IFN-γ production, appeared to be largely HLA-A2-dependent, since when the same experiments were carried out with non transgenic mice, the RFI values obtained were reliably lower (Table 6). The fact that non transgenic and transgenic spleen cells were both efficiently activated using the polyclonal stimulus (anti-CD3/anti-CD28), reinforced the hypothesis that the lower CD8+ T cells induction in non transgenic mice was due to the absence of specific interactions between the peptides and the human HLA-A2 molecules.
CD8+ T cells of transgenic mice infected with C. pneumoniae recognize HLA-A2 binders in vivo It has been recently shown that infection of mice with C. pneumoniae elicits a pathogen-specific murine class I-restricted immune response [22]. Therefore, we asked whether any of the A2 in vitro binders could be recognized by specific CD8 T cells that are clonally selected during the immune response raised against the corresponding native antigen in C. pneumoniae infected cells.
To address this issue, HLA-A2 transgenic mice were intranasally infected with a non lethal dose of C. pneumoniae EBs and challenged with an equal dose of bacteria one month later, before being sacrificed to obtain splenocytes that were used to measure IFN-γ production by CD8+ T cells. Since no appreciable IFN-γ-production could be observed if splenocytes from infected mice were tested directly ex vivo (data not shown), spleen cells were cultured with each individual peptide or with the HepB irrelevant peptide for 6 days. The resulting short-term TCLs were then pulsed again for 6 hours with the same peptides and intracellular IFN-γ production by CD8+ T cells was assessed. The results obtained with 40 tested peptides are shown in Fig. 5A. Sixteen peptides (CH-2, CH-7, CH-8, CH-10, CH-13, CH-15, CH-20, CH-21, CH-28, CH-35, CH-37, CH-45, CH-46, CH-47, CH-50 and CH-55) elicited the strongest CD8+ responses (1 to 7.1 % of IFN-γ-producing CD8+ T cells), while 19 peptides elicited low but consistent responses (percentages of CD8+/IFN-γ+ T cells between 0.3 and 0.9). Five peptides did not induce percentages of IFN-γ-producing CD8+ T cells significantly higher than those observed in response to the HepB control peptide.
When eight among the most reactive peptides were used again to pulse splenocytes of both transgenic and non transgenic mice infected with C. pneumoniae, seven of them were recognized by specific CD8+ IFN-γ+ T cell populations present only in the transgenic mice, while peptide CH-7 was recognized by CD8+ T cells present in both mice groups (Fig. 5B).
General Discussion of Results in Examples 6-8
In this work we have described peptides derived from C. pneumoniae antigens identified as putative T cell epitopes recognized by the common human class I MHC A2 haplotype.
Understanding C. pneumoniae-specific CD8 T cell-mediated immune response and designing protective vaccines rely on the possibility of identifying bacterial antigens or epitopes recognized by CD8+ T cells. Whereas the induction of a CTL-dependent immune response is predictable in response to pathogens which replicate in the cellular cytosol, providing antigens which can enter the cellular MHC-I presentation pathway, in the case of Chlamydiae it is not immediately obvious which antigens are made available to the proteasome and how they reach the cytosol, since these bacteria have a stringent intravacuolar localization inside the infected cell.
We have chosen an in vivo system based on HLA-A2 transgenic mice to test which of the predicted peptides could be recognized by specific CD8+ T cells following either DNA immunization with individual antigen coding genes or infection with C. pneumoniae. Our choice of a murine model is supported by previously published data. Wizel et al. [22], recently reported the first evidence that CD8+ T cells specific for C. pneumoniae antigens are induced in infected mice, and identified bacterial-derived murine MHC-I-restricted T cell epitopes able to trigger either lysis of C. pneumoniae infected cells or in vitro inhibition of the pathogen intracellular growth. These findings seem to confirm that some C. pneumoniae antigens can indeed reach the cytosol of infected cells and enter the MHC-I presentation pathway, i.e. during remodeling that occurs during Chlamydia replication or following autolysis of developing bacterial particles [22]. Furthermore, Kuon et al. [42] recently reported the identification of 11 C. trachomatis-deήved HLA-B27-restricted peptides, capable of stimulating CD8+ T cells obtained from patients with Chlamydia-mάuced reactive arthritis. Importantly, 8 of them overlapped those selected by analyzing splenocytes of HLA-B27 transgenic mice infected with C. trachomatis, indicating that antigen processing can be closely reproduced using the murine animal model, although differences between murine and human antigen processing and T cell repertoires have been hypothesized [43].
The experiment which we have performed with C. pneumoniae infected A2 transgenic mice revealed that at least 16 peptides were recognized by IFN-γ-positive CD8+ T cell populations, which were actually expanded as a consequence of bacterial infection, since we could not detect IFN-γ production pulsing spleen cells from non infected transgenic mice with the same peptides (data not shown). These results suggest that the corresponding Chlamydial antigen may be able to enter the MHC-I presentation pathway. The finding that a number of these peptides can also be recognized by specific CD8+ T cells when the corresponding protein is separately expressed by DNA immunization, strongly reinforces the hypothesis that the responses observed with the infected mice are indeed specific for the in silico predicted peptide epitopes and their corresponding antigens. Importantly, the comparisons of peptide-induced IFN-γ- positive CD8+ T cells in A2 transgenic and non transgenic mice, either immunized with DNA or infected with C. pneumoniae, indicate that this recognition event is largely A2-specific.
Though, we cannot rule out the possibility that some of the tested peptides are also able to interact with the murine class-I MHC molecules, as suggested by the result obtained with CH-7 in infected non transgenic mice (Fig. 5) and by the RFI values obtained with CH-7, CH-8 and CH-13 in DNA-immunized non transgenic mice (Table 6).
Both with DNA immunization and bacterial infection, we were able to show that the OMPA-derived CH-13 peptide induces a specific CD8+ T cell response in A2 transgenic mice. These results appear to validate the choice of this animal model, since our observation that OMPA can enter the MHC-I presentation pathway correlates with the previous identification of HLA-A2-restricted and of murine MHC-I-restricted epitopes in OMPA proteins of C. trachomatis [18] and of C. pneumoniae [23] respectively. With the exception of CH-13 and CH-17, all the other peptides recognized by CD8+ T cells of infected mice belong to C. pneumoniae antigens for which neither human nor murine T cell epitopes have been identified [22;23]. Interestingly, a couple of positively reacting peptides (CH-50 and CH-55) belong to the group of polymoφhic outer membrane proteins [44;45], while most of the others are part of the group of Type III secretion system-related proteins [45;46]. Peptides CH-7 and CH-8, both included in protein T of the Yersinia outer protein (Yop) system [47] and CH-10, included in protein J, which is part of the same translocation system, appear to be particularly reactive in the assay with the infected mice (Fig. 5 A).
This is also true for other peptides included in antigens which are again related to the type III secretion system, such as CH-45, CH-46, and CH-47, all present in the low calcium response protein D. Intriguingly, CH-8, which is the most reactive peptide in the assay with the infected mice, does not seem to be recognized by a specific T cell population when the corresponding antigen is expressed by DNA immunization (Tables 5 and 6). This may be due to different factors, i.e. low in vivo expression level of the injected DNA or altered protein conformation.
On the other hand, we should also consider the possibility that, following infection of mice with C. pneumoniae, this peptide is recognized by a CD8+ T cell population which is instead specific for an epitope derived from an unidentified C. pneumoniae antigen having a closely related sequence. Contrarily to CH-8, stimulation of spleen cells from infected transgenic mice with peptide CH-6 did not allow the detection of IFN-γ+/CD8+ T cells (Fig. 5A), but the same peptide was clearly reactive in the DNA immunization experiments (Tables 5 and 6). This may suggest that Low Calcium Response Protein H is not available for the cellular proteasome, but we could also assume either that the amount of peptide available to the MHC-presenting machinery is not sufficient to induce a cell response which is detectable with our assay, or that the reacting CD8+ T cell population does not expand over the detection limit of our assay. On the whole, the results presented here allowed the identification of a number of antigens which may be available for proteasome-mediated processing in the course of C. pneumoniae infections, proposing them as possible targets for a HLA-A2- dependent cellular immune response. Further analysis will be required to verify if the specifically induced CD8+ T cells are able to recognize and induce lysis of peptide pulsed or C. pneumoniae infected mammalian cells and, possibly, to correlate the identified T cell epitopes with CD8+ T cell populations naturally induced in C. pneumoniae infected patients. Importantly, the results obtained with DNA-mediated expression of distinct antigens, can represent an initial step towards the definition of a significant set of C. pneumoniae HLA-A2-restricted epitopes, which could eventually be combined in DNA minigenes in the attempt to induce a CTL-dependent anti- Chlamydia protective immune response Example 9 Immunizations with Combinations of the First Antigen Group The five antigens of the first antigen group (OmpH-like protein, pmp 10, pmp2, Enolase, OmpH-like, CPn0042 and CPn00795 were prepared as described in the Materials and Methods Section above for Examples 1-4. The antigens are expressed and purified. Compositions of antigen combinations are then prepared comprising five antigens per composition (and containing 15 μg of each antigen per composition). CD1 mice are divided into seven groups (5-6 mice per group for groups 1 through 4; 3 to 4 mice for groups 5, 6 and 7), and immunized as follows:
Figure imgf000119_0001
Mice are immunized at two week intervals. Two weeks after the last immunization, all mice are challenged by intravaginal infection with Chlamydia pneumoniae serovars. Experiment 9 was repeated with another group of CPn antigens. These were: CPn0385 (PepA), CPn0324 (LcrE), CPn0503 (DnaK), CPn0525 (Hypothetical) and CPn0482 (ArtJ). These antigens are combined and administered with and without alum and CpG as described in Experiment 9. Summary
Applicants have identified a number of CPn proteins with desirable immunological and or biological properties. Specifically, at least twelve CPn proteins have been identified which are capable of inducing the production of antibodies, which can neutralise, in a dose-dependent manner, the infectivity of C. pneumoniae in in vitro cell cultures. The induction of neutralising antibodies is important because it prevents infectious EBs from invading human tissues. Furthermore, at least six of these CPn proteins were also capable of attenuating Chlamydial (C. pneumoniae) infection in a in vivo hamster model. In addition, some of these CPn proteins were also capable of inducing not only adequate T-cell responses but also high serum levels of neutralising antibodies.
Apart from very recent unpublished results on pmp21, this is the first time that antisera to recombinant pmps (pmp2 and pmplO) are reported to have neutralising properties. Interestingly, whilst antiserum against CPn0525 gave negative in vitro results (ie no neutralising activity), the CPn0525 protein gave 97 per cent protection from spleen infection in an in vivo hamster immunisation assay (see Table 2) (ie a positive in vivo result). Likewise, whilst antiserum against Cpn0498 gave negative in vitro results (ie no neutralising activity), the CPn0498 protein gave 94 per cent protection from spleen infection in an in vivo hamster immunisation assay (ie a positive in vivo result). Thus a positive signal obtained in the FACS assay does not guarantee a corresponding positive in vitro neutralization activity and conversely a negative neutralization activity does not mean that a positive in vivo result can be excluded.
Some of the results obtained by screening the panel of recombinant antigens with the C.pneumoniae in vitro neutralization assay are shown in Table 2. Just by a cursory look at the 'current annotation' column it can be seen that both in Table 1 and 2 are listed antigens, like the members of the family of heterogeneous polymoφhic membrane proteins (PMP), which, on the basis of published literature data, could be reasonably expected to be surface-exposed and possibly induce neutralizing antibodies, but there are also proteins which could be considered so far only hypothetical, and proteins which just on the basis of their current functional annotation could not be at all expected to be found on the bacterial surface.
The characterisation for the first time of some of these CPn proteins in terms of not only neutralising properties but also different score profiles in a panel of screening tests is an important contribution to the art because it facilitates the selective combination of CPn antigens with particular immunological and biological properties.
In conclusion, this paper describes a group of recombinant antigens which can induce antibodies inhibiting the infectivity of C. pneumoniae in vitro and have protective effects in vivo.
All publications mentioned in the above specification are herein incoφorated by reference. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be covered by the present invention.
REFERENCE SECTION I (Examples 1-4)
I. Bergmann, S., M. Rohde, G. S. Chhatwal, and S. Hammerschmidt. 2001. alpha-Enolase of Streptococcus pneumoniae is a plasmin(ogen)-binding protein displayed on the bacterial cell surface. Mol Microbiol 40:1273-87. 2. Bush, R. M., and K. D. Everett. 2001. Molecular evolution of the Chlamydiaceae. Int J Syst Evol Microbiol 51:203-20. 3. Clausen, T., C. Southan, and M. Ehrmann. 2002. The HtrA family of proteases: implications for protein composition and cell fate. Mol Cell 10:443- 55. 4. Doherty, N. S., B. H. Littman, K. Reilly, A. C. Swindell, J. M. Buss, and N. L. Anderson. 1998. Analysis of changes in acute-phase plasma proteins in an acute inflammatory response and in rheumatoid arthritis using two- dimensional gel electrophoresis. Electrophoresis 19:355-63.
5. Elzer, P. H., R. W. Phillips, G. T. Robertson, and R. M. Roop, 2nd. 1996. The HtrA stress response protease contributes to resistance of Brucella abortus to killing by murine phagocytes. Infect Immun 64:4838-41.
6. Everett, K. D., R. M. Bush, and A. A. Andersen. 1999. Emended description of the order Chlαmydiαles, proposal of Ϋa Chlαmydiαceae fam. nov. and Simkaniaceae fam. nov., each containing one monotypic genus, revised taxonomy of the family Chlamydiaceae, including a new genus and five new species, and standards for the identification of organisms. Int J Syst Bacteriol 49 Pt 2:415-40.
7. Farencena, A., M. Comanducci, M. Donati, G. Ratti, and R. Cevenini. 1997. Characterization of a new isolate of Chlamydia trachomatis which lacks the common plasmid and has properties of biovar trachoma. Infect Immun 65:2965-9.
8. Goodall, J. C, G. Yeo, M. Huang, R. Raggiaschi, and J. S. Gaston. 2001. Identification of Chlamydia trachomatis antigens recognized by human CD4+ T lymphocytes by screening an expression library. Eur J Immunol 31:1513-22. 9. Granoff, D. M., G. R. Moe, M. M. Giuliani, J. Adu-Bobie, L. Santini, B. Brunelli, F. Piccinetti, P. Zuno-Mitchell, S. S. Lee, P. Neri, L. Bracci, L. Lozzi, and R. Rappuoli. 2001. A novel mimetic antigen eliciting protective antibody to Neisseria meningitidis. J Immunol 167:6487-96. 10. Grimwood, J., L. Olinger, and R. S. Stephens. 2001. Expression of Chlamydia pneumoniae polymoφhic membrane protein family genes. Infect Immun 69:2383-9.
II. Grimwood, J., and R. S. Stephens. 1999. Computational analysis of the polymoφhic membrane protein superfamily of Chlamydia trachomatis and Chlamydia pneumoniae. Microb Comp Genomics 4:187-201. 12. Guan, K. L., and J. E. Dixon. 1991. Eukaryotic proteins expressed in Escherichia coli: an improved thrombin cleavage and purification procedure of fusion proteins with glutathione S-transferase. Anal Biochem 192:262-7.
13. Haas, G., G. Karaali, K. Ebermayer, W. G. Metzger, S. Lamer, U. Zimny- Arndt, S. Diescher, U. B. Goebel, K. Vogt, A. B. Roznowski, B. J. Wiedenmann, T. F. Meyer, T. Aebischer, and P. R. Jungblut. 2002. Immunoproteomics of Helicobacter pylori infection and relation to gastric disease. Proteomics 2:313-24.
14. Henderson, I. R., and A. C. Lam. 2001. Polymoφhic proteins of Chlamydia spp.—autotransporters beyond the Proteobacteria. Trends Microbiol 9:573-8. 15. Herbert, B. R., M. P. Molloy, A. A. Gooley, B. J. Walsh, W. G. Bryson, and K. L. Williams. 1998. Improved protein solubility in two-dimensional electrophoresis using tributyl phosphine as reducing agent. Electrophoresis 19:845-51. 16. alman, S., W. Mitchell, R. Marathe, C. Lammel, J. Fan, R. W. Hyman, L. Olinger, J. Grimwood, R. W. Davis, and R. S. Stephens. 1999. Comparative genomes of Chlamydia pneumoniae and C. trachomatis. Nat Genet 21:385-9.
17. Kawa, D. E., and R. S. Stephens. 2002. Antigenic topology of Chlamydial PorB protein and identification of targets for immune neutralization of infectivity. J Immunol 168:5184-91.
18. Kim, D. Y., D. R. Kim, S. C. Ha, N. K. Lokanath, C. J. Lee, H. Y. Hwang, and K. K. Kim. 2002. Crystal structure of the protease domain of a heat- shock protein HtrA from Thermotoga maritima. J Biol Chem 27:27. 19. Krojer, T., M. Garrido-Franco, R. Huber, M. Ehrmann, and T. Clausen. 2002. Crystal structure of DegP (HtrA) reveals a new protease-chaperone machine. Nature 416:455-9. 20. Kubo, A., and R. S. Stephens. 2000. Characterization and functional analysis of PorB, a Chlamydia porin and neutralizing target. Mol Microbiol 38:772-80. 21. Kuo, C. C, J. T. Grayston, L. A. Campbell, Y. A. Goo, R. W. Wissler, and E. P. Benditt. 1995. Chlamydia pneumoniae (TWAR) in coronary arteries of young adults (15-34 years old). Proc Natl Acad Sci U S A 92:6911-4.
22. Kuo, C. C, A. Shor, L. A. Campbell, H. Fukushi, D. L. Patton, and J. T. Grayston. 1993. Demonstration of Chlamydia pneumoniae in atherosclerotic lesions of coronary arteries. J Infect Dis 167:841-9.
23. Loosmore, S. M., Y. P. Yang, R. Oomen, J. M. Shortreed, D. C. Coleman, and M. H. Klein. 1998. The Haemophilus influenzae HtrA protein is a protective antigen. Infect Immun 66:899-906.
24. Makarova, K. S., A. A. Mironov, and M. S. Gelfand. 2001. Conservation of the binding site for the arginine repressor in all bacterial lineages. Genome Biol 2:RESEARCH0013.
25. Montigiani, S., F. Falugi, M. Scarselli, O. Finco, R. Petracca, G. Galli, M. Mariani, R. Manetti, M. Agnusdei, R. Cevenini, M. Donati, R. Nogarotto, N. Norais, I. Garaguso, S. Nuti, G. Saletti, D. Rosa, G. Ratti, and G. Grandi. 2002. Genomic approach for analysis of surface proteins in Chlamydia pneumoniae. Infect Immun 70:368-79.
26. Moroni, A., G. Pavan, M. Donati, and R. Cevenini. 1996. Differences in the envelope proteins of Chlamydia pneumoniae, Chlamydia trachomatis, and Chlamydia psittaci shown by two-dimensional gel electrophoresis. Arch Microbiol 165:164-8.
27. Pallen, M. J., and B. W. Wren. 1997. The HtrA family of serine proteases. Mol Microbiol 26:209-21.
28. Pancholi, V., and V. A. Fischetti. 1998. alpha-enolase, a novel strong plasmin(ogen) binding protein on the surface of pathogenic streptococci. J Biol Chem 273:14503-15.
29. Pancholi, V., and V. A. Fischetti. 1997. A novel plasminogen/plasmin binding protein on the surface of group A streptococci. Adv Exp Med Biol 418:597-9. 30. Pedersen, A. S., G. Christiansen, and S. Birkelund. 2001. Differential expression of PmplO in cell culture infected with Chlamydia pneumoniae CWL029. FEMS Microbiol Lett 203:153-9.
31. Perez Melgosa, M., C. C. Kuo, and L. A. Campbell. 1994. Isolation and characterization of a gene encoding a Chlamydia pneumoniae 76-kilodalton protein containing a species-specific epitope. Infect Immun 62:880-6.
32. Perkins, D. N., D. J. Pappin, D. M. Creasy, and J. S. Cottrell. 1999. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20:3551-67. 33. Rocha, E. P., O. Pradillon, H. Bui, C. Sayada, and E. Denamur. 2002. A new family of highly variable proteins in the Chlamydophila pneumoniae genome. Nucleic Acids Res 30:4351-60.
34. Sambri, V., M. Donati, E. Storni, K. Di Leo, M. Agnusdei, R. Petracca, O. Finco, G. Grandi, G. Ratti, and R. Cevenini. 2004. Experimental infection by Chlamydia pneumoniae in the hamster. Vaccine in press.
35. Sanchez-Campillo, M., L. Bini, M. Comanducci, R. Raggiaschi, B. Marzocchi, V. Pallini, and G. Ratti. 1999. Identification of immunoreactive proteins of Chlamydia trachomatis by Western blot analysis of a two- dimensional electrophoresis map with patient sera. Electrophoresis 20:2269- 79.
36. Schachter, J., R. S. Stephens, P. Timms, C. Kuo, P. M. Bavoil, S. Birkelund, J. Boman, H. Caldwell, L. A. Campbell, M. Chernesky, G. Christiansen, I. N. Clarke, C. Gaydos, J. T. Grayston, T. Hackstadt, R. Hsia, B. Kaltenboeck, M. Leinonnen, D. Ocjius, G. McClarty, J. Orfila, R. Peeling, M. Puolakkainen, T. C. Quinn, R. G. Rank, J. Raulston, G. L. Ridgeway, P. Saikku, W. E. Stamm, D. T. Taylor-Robinson, S. P. Wang, and P. B. Wyrick. 2001. Radical changes to Chlamydial taxonomy are not necessary just yet. Int J Syst Evol Microbiol 51:249, 251-3.
37. Schafer, U., K. Beck, and M. Muller. 1999. Skp, a molecular chaperone of gram-negative bacteria, is required for the formation of soluble periplasmic intermediates of outer membrane proteins. J Biol Chem 274:24567-74.
38. Sebert, M. E., L. M. Palmer, M. Rosenberg, and J. N. Weiser. 2002. Microarray-based identification of htrA, a Streptococcus pneumoniae gene that is regulated by the CiaRH two-component system and contributes to nasopharyngeal colonization. Infect Immun 70:4059-67.
39. Siscovick, D., R. Alexander, M. Davidson, M. Leinonen, S. O'Connor, P. Ewald, C. Meier, M. Puolakkainen, J. Hughes, and J. Nieto. 2000. Collaborative multidisciplinary workshop report: the role of epidemiology studies in determining a possible relationship between Chlamydia pneumoniae infection and atherothrombotic diseases. J Infect Dis 181:S430-1.
40. Siscovick, D. S., S. M. Schwartz, M. Caps, S. P. Wang, and J. T. Grayston. 2000. Chlamydia pneumoniae and atherosclerotic risk in populations: the role of seroepidemiology. J Infect Dis 181:S417-20.
41. Stephens, R. S. (ed.). 1999. Chlamydia: Intracellular Biology, Pathogenesis and Immunology. American Society for Microbology Press, Washington.
42. Vandahl, B. B., S. Birkelund, H. Demol, B. Hoorelbeke, G. Christiansen, J. Vandekerckhove, and K. Gevaert. 2001. Proteome analysis of the Chlamydia pneumoniae elementary body. Electrophoresis 22:1204-23.
43. Wolf, K., E. Fischer, D. Mead, G. Zhong, R. Peeling, B. Whitmire, and H. D. Caldwell. 2001. Chlamydia pneumoniae major outer membrane protein is a surface-exposed antigen that elicits antibodies primarily directed against conformation-dependent determinants. Infect Immun 69:3082-91. 44. Young, I. T. 1977. Proof without prejudice: use of the Kolmogorov-Smimov test for the analysis of histograms from flow systems and other sources. J Histochem Cytochem 25:935-41.
REFERENCE SECTION II (EXAMPLES 6-8)
1. Pizza,M., ScarlatoN., MasignaniN., Giuliani,M.M., Aric&ograve, ,B., Comanducci,M., Jennings,G.T., Baldi,L., Bartolini,E., Capecchi,B., GaleottijC.L., Luzzi,E., Manetti,R., Marchetti,E., Mora,M., Nuti,S., Ratti,G., Santini,L., Savino,S., Scarselli,M., Storni,E., Zuo,P., Broeker,M., Hundt,E., Knapp,B., Blair,E., Mason,T., Tettelin,H., Hood,D.W., Jeffries, A. C, Saunders,N.J., Granoff,D.M., Venter,J.C, Moxon,E.R., Grandi,G., and Rappuoli,R., Identification of Vaccine Candidates Against Serogroup B Meningococcus by Whole-Genome Sequencing. Science 2000. 287: 1816-1820.
2. Rappuoli,R., Reverse vaccinology. Current Opinion in Microbiology 2000. 3: 445- 450. 3. Rappuoli,R., Reverse vaccinology, a genome-based approach to vaccine development. Vaccine 2001. 19: 2688-2691.
4. Montigiani,S., Falugi,F., Scarselli,M., Finco,0., Petracca,R., Galli,G., Mariani,M., Manetti,R., Agnusdei,M., Cevenini,R., Donati,M., Nogarotto,R., Norais,N., Garaguso,I., Nuti,S., Saletti,G., Rosa,D., Ratti,G., and Grandi,G.,
Genomic Approach for Analysis of Surface Proteins in Chlamydia pneumoniae. Infect mmun. 2002. 70: 368-379.
5. RosSjB.C, Czajkowski,L., Hocking,D., Margetts,M., Webb,E., Rothel,L., Patterson,M., Agius,C, Camuglia,S., and Reynolds,E., Identification of vaccine candidate antigens from a genomic analysis of Poφhyromonas gingivalis. Vaccine 2001. 19: 4135-4142.
6. Wizemann,T.M., Heinrichs,J.H., Adamou,J.E., Erwin,A.L., Kunsch,C, Choi,G.H., Barash,S.C, Rosen,C.A., Masure,H.R., Tuomanen,E., Gayle,A.,
Brewah,Y.A., Walsh, W., Barren,P., Lathigra,R., Hanson,M., Langermann,S., Johnson,S., and Koenig,S., Use of a Whole Genome Approach To Identify Vaccine Molecules Affording Protection against Streptococcus pneumoniae Infection. InfectJmmun. 2001. 69: 1593-1598.
7. De Groot,A.S., Bosma,A., Chinai,N., Frost,J., Jesdale,B.M., Gonzalez,M.A., Martin, W., and Saint-Aubin,C, From genome to vaccine: in silico predictions, ex vivo verification. Vaccine 2001. 19: 4385-4395. 8. Martin, W., Sbai,H., and De Groot,A.S., Bioinformatics tools for identifying class I-restricted epitopes. Methods 2003. 29: 289-298.
9. Hill, A. V. and Davenport, M. P., Reverse Immunogenetics: from HLA-disease associations to vaccine candidates. Mol.Med.Today 1996. 2: 38-45.
10. Pamer, E. G., Harry, J. T., and Bevan, M. J., Precise prediction of a dominant class I MHC-restricted epitope of Listeria monocytogenes. Nature 1991. 353 (6347): 852-855. 11. KuOjC.C, Jackson,L.A., Campbell,L.A., and Grayston, J.T., Chlamydia pneumoniae (TWAR). Clin.Microbiol.Rev. 1995. 8: 451-461.
12. Grayston, J. T., Background and current knowledge of Chlamydia pneumoniae and atherosclerosis. The Journal of Infectious Diseases 2000. 181: S402-S410.
13. CampbelljL. and Kuo,C.C, Chlamydia pneumoniae and atherosclerosis. Seminars in Respiratory Infections 2003. 18: 48-54. 14. Hammerschlag, M. R., The intracellular life of chlamidiae. Semin.Pediatr.Infect.Dis. 2002. 13 (4): 239-248.
15. Rottenberg,M.E., Gigliotti Rothfuchs,A.C, Gigliotti,D., Svanholm,C, Bandholtz,L., and Wigzell,H., Role of Innate and Adaptive Immunity in the Outcome of Primary Infection with Chlamydia pneumoniae, as Analyzed in Genetically Modified Mice. J Immunol 1999. 162: 2829-2836.
16. Penttila, Anttila, Varkila, Puolakkainen, Sarvas, Makela, and Rautonen,
Depletion of CD8 cells abolishes memory in acquired immunity against Chlamydia pneumoniae in BALB/c mice. Immunology 1999. 97: 490-496.
17. Halme,S., Latvala,J., Karttunen,R., Palatsi,L, Saikku,P., and Surcel,H.M.,
Cell-Mediated Immune Response during Primary Chlamydia pneumoniae Infection. InfectJmmun. 2000. 68: 7156-7158.
18. Kim,S.K., Angevine,M., Demick,K., Ortiz,L., Rudersdorf,R., Watkins,D., and DeMars,R., Induction of HLA Class I-Restricted CD8+ CTLs Specific for the Major Outer Membrane Protein of Chlamydia trachomatis in Human Genital Tract Infections. J Immunol 1999. 162: 6855-6866.
19. Read,T.D., Brunham,R.C, Shen,C, Gill,S.R., Heidelberg, J.F., White,0., Hickey,E.K, Peterson,J., Utterback,T., Berry,K., Bass,S., Linher,K., Weidman,J., Khouri,H., Craven,B., Bowman,C, Dodson,R., Gwinn,M., Nelson, W., DeBoy,R., Kolonay,J., McClarty,G., Salzberg,S.L., Eisen,J., and Fraser,C.M., Genome sequences of Chlamydia trachomatis MoPn and Chlamydia pneumoniae AR39. Nucl.Acids.Res. 2000. 28: 1397-1406.
20. Shirai,M., Hirakawa,H., Kimoto,M., Tabuchi,M., Kishi,F., Ouchi,K., Shiba,T., Ishii,K., Hattori,M., Kuhara,S., and Nakazawa,T., Comparison of whole genome sequences of Chlamydia pneumoniae J138 from Japan and CWL029 from USA. Nucl.Acids.Res. 2000. 28: 2311-2314.
21. Kalman, S., Mitchell, W., Maranthe, R., Lammel, C, Fan, J., Hyman, R. W., Olinger, R., Grimwood, J., Davis, R. W., and Stephens, R. S., Comparative genomes of Chlamydia pneumoniae and C. trachomatis. Nat.Genet. 1999. 21 (4), 385- 389.
22. Wizel,B., Starcher,B.C, Samten,B., Chroneos,Z., Barnes,P.F., Dzuris,J., HigashimotOjY., Appella,E., and Sette,A., Multiple Chlamydiapne mo iae Antigens Prime CD8+ Tel Responses That Inhibit Intracellular Growth of This Nacuolar Pathogen. J Immunol 2002. 169: 2524-2535.
23. Saren,A., Pascolo,S., Stevanovic,S., Dumrese,T., PuoIakkainen,M., Sarvas,M., Rammensee,H.G., and Nuola,J.M., Identification of Chlamydia pneumoniae-Deύved Mouse CD8 Epitopes. InfectJmmun. 2002. 70: 3336-3343.
24. Parker, K. C, Bednarek, M. A., and Coligan, J. E., Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains. J.Immunol. 1994. 152: 163-175.
25. Yi,Y., Yang,X., and Brunham,R.C, Autoimmunity to heat shock protein 60 and antigen-specific production of interleukin- 10. InfectJmmun. 1997. 65: 1669-1674. 26. Lamb,D.J., El Sankary,W., and Ferns,G.A.A., Molecular mimicry in atherosclerosis: a role for heat shock proteins in immunisation. Atherosclerosis 2003. 167: 177-185.
27. Stephens, R. S., The cellular paradigm of Chlamydial pathogenesis. Trends Microbiol. 2003. 11 (1), 44-51.
28. Morrison, RP., Chlamydial hsp60 and the immunopathogenesis of Chlamydial disease. Semin.hnmunol. 2003. 3 (1): 25-33. 29. Νixon, DF and McMichael, A. J., Cytotoxic T-cell recognition of HIN proteins and peptides. AIDS 1991. 5 (9): 1049-1059.
30. Anderson,K.S., Alexander,.!., Wei,M., and Cresswell,P., Intracellular transport of class I MHC molecules in antigen processing mutant cell lines. JJmmunol. 1993. 151: 3407-3419.
31. Ljunggren,H.G., Stam,Ν.J., Ohlen,C, Neefjes,J.J., Hoglund,P., Heemels,M.T., Bastin,J., Schumacher,T.N., Townsend,A., Karre,K., and ., Empty MHC class I molecules come out in the cold. Nature 1990. 346: 476-480.
32. Ljunggren,H.G., Ohlen,C, Hoglund,P., Franksson,L., and Karre,K., The RMA-S lymphoma mutant; consequences of a peptide loading defect on immunological recognition and graft rejection. Int. J. Cancer Suppl 1991. 6: 38-44. 33. Bednarek,M.A., Sauma,S.Y., Ganimon,M.C, Porter, G., Tamhankar,S., Williamson,A.R., and Zweerink,H.J., The minimum peptide epitope from the influenza virus matrix protein. Extra and intracellular loading of HLA-A2. J Immunol 1991. 147: 4047-4053. 34. Gagliardi,M.C, De Petrillo,G., Salemi,S., Boffa,L., Longobardi,M.G., Dellabona,P., Casorati,G., Tanigaki,N., Harris,R., and Lanzavecchia,A.,
Presentation of peptides by cultured monocytes or activated T cells allows specific priming of human cytotoxic T lymphocytes in vitro. Int.Immunol. 1995. 7: 1741-1752. 35. Vitiello, A., Marchesini, D., Furze, J., Sherman, L. A., and Chesnut, R. W.,
Analysis of HLA-restricted influenza-specific cytotoxic T lymphocyte response in transgenic mice carrying a chimeric human-mouse class I major histocpmpatibility complex. J.Exp.Med. 1991. 173 (4): 1007-1015.
36. Kim,S.K, Devine,L., Angevine,M., DeMars,R., and Kavathas,P.B., Direct Detection and Magnetic Isolation of Chlamydia trachomatis Major Outer Membrane Protein-Specific CD8+ CTLs with HLA Class I Tetramers. J Immunol 2000. 165: 7285-7292.
37. Meister,G.E., Roberts,C.G.P., Berzofsky,J.A., and De Groot,A.S., Two novel T cell epitope prediction algorithms based on MHC-binding motifs; comparison of predicted and published epitopes from Mycobacterium tuberculosis and HIV protein sequences. Vaccine 1995. 13: 581-591.
38. Schafer,J.R., Jesdale,B.M., George,J.A., Kouttab,N.M., and De Groot,A.S.,
Prediction of well-conserved HIN-1 ligands using a matrix-based algorithm, EpiMatrix. Vaccine 1998. 16: 1880-1884. 39. Reche,P.A., Glutting,J.P., and Reinherz,E.L., Prediction of MHC class I binding peptides using profile motifs. Human Immunology 2002. 63: 701-709.
40. Rammensee, H. G., Bachmann, J., Emmerich, Ν. P., Bachor, O. A., and Stevanovic, S., SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics 1999. 50 (3-4); 213-219.
41. CerundoloN., Alexander ,J., Anderson,K., Lamb,C, Cresswell,P., McMichael,A., Gotch,F., and Townsend,A., Presentation of viral antigen controlled by a gene in the major histocompatibility complex. Nature 1990. 345: 449-452.
42. Kuon,W., Holzhutter,H.G., Appel,H., Grolms,M., Kollnberger,S., Traeder,A., Henklein,P., Weiss,E., Thiel,A., Lauster,R., Bowness,P., Radbruch,A., Kloetzel,P.M., and Sieper,J., Identification of HLA-B27-Restricted Peptides from the Chlamydia trachomatis Proteome with Possible Relevance to HLA- B27-Associated Diseases. J Immunol 2001. 167: 4738-4746.
43. Endert van, P. M., Riganelli, D., Greco, G., Fleischhauer, K., Sidney, J., Sette, A., and Bach, J. F., The peptide-binding motif for the human transporter associated with antigen processing. J.Exp.Med. 1995. 182 (6): 1883-1895.
44. Grimwood, J. and Stephens, R. S., Computational analysis of the polymoφhic membrane protein superfamily of Chlamydia trachomatis and Chlamydia pneumoniae. Microb.Comp.Genomics 1999. 4: 187-201. 45. Rockey, D. D., Lenart, J., and Stephens, R. S., Genome sequencing and our understanding of Chlamydiae. Infection and Immunity 2000. 68 (10): 5473-5479.
46. Kim,J.F., Revisiting the Chlamydial type III protein secretion system: clues to the origin of type III protein secretion. Trends in Genetics 2001. 17: 65-69. 47. Cornelis, G. R., The Yersinia Ysc-Yop virulence apparatus. Int.J.Med.Microbiol. 2002. 291 (6-7): 455-462.
48. Parham, P. and Brodsky, FM., Partial purification and some properties of BB7.2. A cytotoxic monoclonal antibody with specificity for HLA-A2 and a variant of HLA-A28. Hum.Immunol. 2003. 3 (4): 277-299.
49. Grifantini, R., Finco, O., Bartolini, E., Draghi, M., Del Giudice, G., Kocken, C, Thomas, A., Abrignani, S., and Grandi, G., Multi-plasmid DNA vaccination avoids antigenic competition and enhances immunogenicity of a poorly immunogenic plasmid. Eur.J.Immunol. 1998. 28 (4): 1225-1232.
Figure imgf000131_0001
TABLE 1
~ϊ iιe iC)
Figure imgf000132_0001
■pπipy. .p'mjQ. • PjcOJm jpltct.Out^ M^pb'ι^i)e. t<jιι... iirt y
Figure imgf000133_0001
Cft«.$74 C Q5C 1ιj-p0tili(_ pul protein Itøie 2 i))
Figure imgf000134_0001
Figure imgf000135_0001
Chlamydia Further Filing', j Microarray Expressed Oene able 15-9-04 and 17-12-04 TABLE 3v- _i
Figure imgf000136_0001
Chlamydia Further Filing - -J Microarray Expressed Oene Table 15-9-04 and 17-12-04 TABLE 3 v
Figure imgf000137_0001
Chlamydia Further Filing/- J Microarray Expressed Oene Table 15-9-04 and 17-12-04 TABLE 3v-xi
Figure imgf000138_0001
Chlamydia Further Filing^ r Microarray Expressed Oene r_ble 15-9-04 and 17-12-04
Figure imgf000139_0001
Chlamydia Further Filing/ _ J Microarray Expressed Gene rsble 15-9-04 and 17-12-04 TABLE 3v- d
Figure imgf000140_0001
Chlamydia Further Filing , } Microarray Expressed Gene Table 15-9-04 and 17-12-04
Figure imgf000141_0001
Chlamydia Further Filing/ j Microarra Ex ressed Gene table
Figure imgf000142_0001
Table 4. C. pneumoniae selected peptides: protein sources and HLA-A2 stabilization assay
Peptide Sequence CPn a> Protein Group "> Score 01 Net MFI ">
HepB 126TAFHQTLQD133 Hepatitis B virus envelope antigen sss- 14.0*24.4
GAG "SLYNTVATL85 HIV-1 gag 157.22 92.4s23.8
IMa ^GILGFVFTL66 Influenza virus matrix M1 550.92 63.1*18.1
CH1 3,5QLLDEGKEL323 0322 Yop proteins translocation protein U Type ill 324.06 74.0dt22.6
CH2 "^ILLNEVPYV441 0323 Low calcium response protein D Type III 5534.14 140.5*36.1
CH3 ^VLNLFFSAL351 0324 Low calcium response protein E Type III 262.20 40.1±23.1
CH4 7QLLESLAPL16 0325 Secretion chaperone Type III 745.35 120.1±25.2
CH5 a71SILELLQFV273 0702 Probable Yop proteins translocation protein C Type III 1835.22 85.5±34.4
CH6 ^LLEEIYTV87 0811 Low calcium response protein H Type III 11162.99 148.5*38.9
CH7 ∞YMDNNLFYV91 0823 Yop proteins translocation protein T Type III 6781.36 164.1*24.3
CH8 254FLT__AWWFIse2 0823 Yop proteins translocation protein T Type III 3365.36 144.1*22.1
CH10 ^GLTEEIDYV222 0828 Yop proteins translocation protein J Type III 1767.58 144.0±37.9
CH12 ^LVFFNPFV87 1021 Low calcium response locus protein H Type III 6686.72 50.1*22.2
CH13 ^YVFDRILKV77 0695 Outer membrane protein A Ch spec 976.76 139.0*36.7
CH14 ""VMUFEKKV"4 0415 CT266 hypothetical protein Cpn spec 1200.64 74.1±20.2 i2roγLTSγspγvi278
CH15 0444 Polymorphic outer membrane protein G/l family Pmp 1759.66 138.1*23.5
CH16 1530VQ YVFDV 538 0963 Putative outer membrane protein D family Pmp 591.70 48.1*19.1
CH17 ^ILQEAEQMV316 0728 76 Da homolog_1 Ch spec 484.77 202.1*24.2
CH18 71IALLVIFFV79 0186 Similar to CT119 IncA Ch spec 445.80 46.1*22.3
CH19 13arLαTLGYAV<335 0444 Polymorphic outer membrane protein G/l family Pmp 437.48 56.1*21.6
CH20 146ALMLLNNYV154 0005 Polymorphic outer membrane protein G family Pmp 1415.38 142.5*38.6
CH21 6 4TLWGSFVDV622 0447 Polymorphic outer membrane protein G/l family Pmp 1096.83 121.1*18.0
CH22 ,sδ<WLFDLRFSVW74 0540 Polymorphic membrane protein B family Pmp 28150.17 68.5±11.0
CH24 ^UQETLLFV498 0021 Predicted OMP Ch spec 843.21 105.1*20.8
CH28 ^RLLEII GV53 0062 CHLPS 43 kDa protein homo!og_1 Cpn spec 18200.54 99.5*15.0
CH29 288YLMQ LQNV296 0791 CT 590 hypothetical protein Ch spec 2722.68 108.5*12.1
CH30 ^FLQRGESFV523 0792 CT 589 hypothetical protein Ch spec 759.66 105.1*8.1
CH31 ^'WLLRDDWLL409 0009 hypothetical Cpn spec 2726.91 101.1*16.3
CH32 87KLWEWLGYL195 0041 hypothetical Cpn spec 4184.21 72.1*12.0
CH33 LLMLAISLV76 0131 hypothetical Cpn spec 1006.20 18.0*1.4
CH34 ^KLLKDHFDL209 0132 hypothetical Cpn spec 1604.53 85.1*4.9
CH35 ILSFLPWLV84 0169 hypothetical Cpn spec 886.78 90.6*5.7
CH36 "9LLLIFNNYL157 0170 hypothetical Ch spec 2808.32 41.0*19.8
CH37 126YLLDFRWPL134 0210 hypothetical Cpn spec 42485.26 97.1*17.7
CH38 ^NLLKRWQFV382 0352 hypothetical Cpn spec 2406.15 64.1*13.4
CH39 ^FLLRHLSSV386 0355 hypothetical Cpn spec 2722.68 88.1*6.4
CH41 162KLSEQLEAL170 0186 Similar to CT1 9 tncA Ch spec 345.48 51.6*27.6
CH42 ^"KVLLGQEWV222 0186 Similar to CT119 IncA Ch spec 212.39 16.0*37.5
CH43 5I .LAEQvTAL323 0186 Similar . O CT119 IncA Ch spec 201.44 71.6*34.6
CH44 123YWGFIIFL131 0323 Low calcium response protein D Type III 413.32 25.0*16.3
CH45 ^MMGWLMI40 0323 Low calcium response protein D Type III 294.95 8.1*18.4
CH46 ∞NLSISVFLL64 0323 Low calcium response protein D Type 111 284.97 18.0*26.2
CH47 ""VIQAFGDFV118 0323 Low calcium response protein D Type 111 166.49 23.0*32.5
CH4S ^YLALDPDSV*43 0323 Low calcium response protein D Type III 156.77 74.6*34.4
CH49 "9KMSHFQQAL157 0415 CT266 hypothetical protein Cpn spec 205.19 29.0*33.9
CH50 11β7SLCAQSSYV"95 0444 Polymorphic outer membrane protein G/l family Pmp 382.53 45.1*22.6
CH51 1360NLSRQAFFA1368 0444 Polymorphic outer membrane protein G/l family Pmp 158.47 25.0*34.5
CH52 ^SLLEEHPW686 0963 Putative outer membrane protein D family Pmp 432.59 43.6*21.9
CH53 1302NLWSHYTDL1310 0963 Putative outer membrane protein D family Pmp 265.96 1.6*24.7
CH54 377AL KENQAL385 0963 Putative outer membrane protein D family Pmp 177.30 45.6*21.9
CH55 S68ALWGHNVLL578 0963 Putative outer membrane protein D family Pmp 177.30 47.6*29.0
CH56 ^NLAGGILSV341 0963 Putative outer membrane protein D family Pmp 159.97 29.6*29.0
CH57 ^FVSKF FS 34 1021 Low calcium response locus protein H Type III 322.16 16.0*0.1
CH58 73SITVFRWLVW 1021 Low calcium response locus protein H Type III 272.55 30.0*20.5
CH5Θ ^UVFVLTI66 0131 hypothetical Cpn spec 419.44 29,0*10.6
CH60 "'VMLFIGLGV50 0131 hypothetical Cpn spec 315.95 28.0*2.8
CH61 ^VLFLLIRSV84 0131 hypothetical Cpn spec 201.24 14.0*2.8
CH62 ^FLFQLGMQI405 0415 CT266 hypothetical protein Ch spec 177.56 30.0*12.0 a) Gene sequence designation as annotated from the genome sequence of Cpn strain C L029 (http://chlamydia- www.berkeley.edu.4231) b)Type III: type III secretion system; Ch and Cpn spec: Chlamydia and C. pneumoniae specific; Pmp: Polymoφhic membrane protein c) Calculated using the BIMAS algorithm d) Mean Fluorescence Intensity of cells with peptide - M Mean Fluorescence Intensity of cells without peptide ± Standard Deviation calculated on three experiments Table 5. ELISpot assay with CD8+ T cells from DNA immunized HLA-A2 transgenic mice Protein Gene Peptide SFC8' Hypothetical CPn 0131 medium 13 HepB 47 CH33 33 CH59 53 CH60 80 CH61 40 Hypothetical CPn 0210 medium 7 HepB 13 CH37 120 LCR Protein D CPn 0323 medium 27 HepB 27 CH2 93 CH44 80 CH45 87 CH46 40 CH47 80 CH48 60 CHLPS 43 kDa CPn 0062 medium 33 HepB 27 CH28 93 OMPA CPn 0695 medium 13 HepB 33 CH13 727
LCR Protein H CPn 0811 medium 13 HepB 27 CH6 213 Yop pt protein T CPn 0823 medium 7 HepB 47 CH7 493 CH8 53 Yop pt protein J CPn 0828 medium 20 HepB 60 CH10 247
SFC = Spot Forming Colonies/106 CD8 cells Table 6. IFN-γ production from splenocytes of DNA immunized HLA-A2 transgenic and non transgenic mice Ex Vivo RFI a) StTCLsb> RFI a) Protein Gene Peptide AST* A2+c) A2-°> A2«> LCR Protein D CPn 0323 CH 2 0,05 1,11 0,79 2,57 CH 44 3,30 1,78 0,73 6,86 CH 45 1,00 1,56 0,47 4,71 CH 46 0,90 1,44 0,41 9,00 CH 47 1,00 1,78 1,17 1,14 CH 48 1,30 1,67 0,11 1,29 CD3+CD28 134,00 90,55
OMP A CPn 0695 CH 13 3,29 2,54 23,42 209,81 CD3 + CD28 248,71 73,23
LCR Protein H CPn 0811 CH 6 1,00 4,58 1,53 31,56 CD34 CD28 290,83 96,10
Yop pt Protein T CPn 0823 CH 7 1,20 5,20 11,69' 94,57 CH 8 2,00 1,60 16,81 28,21 CD3 + CD28 247,60 91 ,00 a) Relative Fold Increase: ratio between the percentage of IFN-γ7CD8+ cells obtained with the tested peptide (or the CD3/CD28 co-stimulus) and the HepB negative control peptide ) Short term T cell lines c) HLA-A2 non transgenic and transgenic mice

Claims

CLAIMS: 1. A polypeptide for use as an autotransporter antigen, the polypeptide comprising: (a) an amino acid sequence selected from the group consisting of SEQ ID NO:
54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79, (b)an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a); or (c) an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids from an amino acid sequence of (a) or combinations thereof. 2. The polypeptide of claim 1 where use is as an antigen for raising a Chlamydia pneumoniae specific immune response 3. The polypeptide of claim 2 wherein the use is for raising a systemic immune response in an individual infected with Chlamydia pneumoniae. 4. The polypeptide of any one of claims 1-3 which is secreted into the cytoplasm of the host cell through a Type V autotransporter secretion system mechanism. 5. The polypeptide of any one of claims 1-3 wherein the polypeptide is selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79 and share one or more common N-terminal sequence motifs selected from the group consisting of G, DG, VG, G, AN G, IVG, GTLGG, S, IVG, and M. 6. The polypeptide of claim 5 wherein the common Ν-terminal sequence motif is selected from the group consisting of GTLGG, S, IVG and M. 7. The polypeptide of any one of claims 1-3 for use in diagnosis. 8. The use of a polypeptide according to any one of claims 1-3 in the preparation of a medicament for the prevention or treatment of a Chlamydia pneumoniae infection in an individual. 9. The use according to claim 8 wherein the use is of an autotransporter protein which immunoreacts with seropositive serum of an individual infected with Chlamydia pneumoniae. 10. The use of a polypeptide according to any one of claims 1-3 in the preparation of an assay for the diagnosis of a Chlamydia pneumoniae infection in an individual. 11. A method of eliciting an immune response in an individual comprising administering to the individual a polypeptide comprising: (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, and SEQ ID NO: 79, (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a), or (c) an amino acid sequence comprising one or more fragment of at least 1, 2, 3, 4, 5, 6, or 7 amino acids from an amino acid sequence of (a) or mixtures thereof. 12. A method of diagnosing an immune response in an individual comprising: (a) contacting a biological sample obtained from the individual with a binding agent that binds to a polypeptide according to any one of claims 1-3; (b) detecting in the biological sample the amount of the polypeptide that binds to the binding agent; and (c) comparing the amount of the polypeptide to a predetermined cut-off value and thereby determining the presence of an immune response in the individual. 13. The method of claim 11 or claim 12 wherein the polypeptide is defined according to any one of claims 1 -3. 14. The method of claim 11 or the use of according to any one of claims 2-6 or 8-9 wherein the polypeptide. 15. A composition for eliciting an immune response comprising one or more
Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof and one or more immunostimulants. 16. The composition according to claim 15 wherein the Chlamydia pneumoniae autotransporter protein or the immunogenic fragment thereof comprises: (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, SEQ ID NO: 86 and SEQ ID NO: 79; (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a); or (c) an amino acid sequence comprising one or more fragments of at least 1, 2, 3, 4, 5, 6 or 7 amino acids from an amino acid sequences of (a) or combinations thereof. 17. The composition according to claim 15 or 16 wherein the protein or immunogenic fragment thereof is defined according to any one of claims 1-3. 18. A composition for eliciting an immune response in a subject comprising two or more Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof. 19. The composition according to claim 18 wherein the Chlamydia pneumoniae autotransporter protein or the immunogenic fragment thereof comprises: (a) an amino acid sequence selected from the group consisting of SEQ ID NO: 54, SEQ ID NO: 6, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 78, SEQ ID NO:
86 and SEQ ID NO: 79; (b) an amino acid sequence having at least 50% sequence identity to an amino acid sequence of (a); or (c) an amino acid sequence comprising one or more fragments of at least 1, 2, 3, 4, 5, 6 or 7 amino acids from an amino acid sequences of (a) or combinations thereof. 20. The composition according to claim 18 or 19 wherein the composition further comprises one or more immunostimulants. 21. A method of making a composition according to any one of claims 15 or 16 wherein the method comprises combining one or more Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof with one or more immunostimulants. 22. A method of making a composition according to claim 18 or 19 wherein the method comprises combining two or more Chlamydia pneunoniae autotransporter proteins or immunogenic fragments thereof. 23. The method according to claim 22 wherein the method comprises adding one or more immunostimulants to the Chlamydia pneumoniae autotransporter proteins or immunogenic fragments thereof. 24. A Chlamydia pneumoniae autotransporter protein selected from the group consisting of Cpn0794, Cpn0795, Cpn0796, Cpn0797, CPn0798 and Cpn0799 or an immunogenic fragment thereof wherein the autotransporter protein an amino acid motif comprising IVG, A, LGG and S. 25. The_autotransporter protein according to claim 24 wherein the repeat amino acid motif comprises IVG, A, LGG and S. 26. A polypeptide for use as an autotransporter antigen comprising an amino acid sequence corresponding to SEQ ID NO: 86, an amino acid sequence having at least 50% sequence identity to SEQ ID NO: 86, or an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids of SEQ ID NO: 86. 27. The polypeptide of claim 26 where use is as an antigen for raising a
Chlamydia pneumoniae specific immune response 28. The polypeptide of claim 2 wherein the use is for raising a systemic immune response in an individual infected with Chlamydia pneumoniae. 29. The use of a polypeptide according to any one of claims 26-28 in the preparation of a medicament for the prevention or treatment of a Chlamydia pneumoniae infection in an individual. 30. A method of raising an immune response in an individual, the method comprising administering to the individual a polypeptide comprising an amino acid sequence corresponding to SEQ ID NO: 86, an amino acid sequence having at least 50% sequence identity to SEQ ID NO: 86, or an amino acid sequence comprising one or more fragments of at least 7 consecutive amino acids of SEQ ID NO: 86.
31. A method of diagnosing an immune response in an individual, the method comprising: (a) contacting a biological sample obtained from an individual with a binding agent that binds to a polypeptide defined in any one of claims 26-28; (b) detecting in the sample the amount of the polypeptide that binds to the binding agent; and (c) comparing the amount of polypeptide to a predetermined cut-off value and thereby determining the presence of an immune response in the individual.
PCT/US2005/006588 2004-03-02 2005-03-02 Immunogenic compositions for chlamydia pneunomiae WO2005084306A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP05724183A EP1729800A4 (en) 2004-03-02 2005-03-02 Immunogenic compositions for chlamydia pneunomiae
BRPI0508365-6A BRPI0508365A (en) 2004-03-02 2005-03-02 immunogenic compositions for chlamydia pneumoniae
RU2006134631/13A RU2006134631A (en) 2004-03-02 2005-03-02 IMMUNOGENIC COMPOSITIONS AGAINST CHLAMIDIA PNEUMONIAE
JP2007501899A JP2007526318A (en) 2004-03-02 2005-03-02 Immunogenic composition against Chlamydiapneumoniae
CA002557353A CA2557353A1 (en) 2004-03-02 2005-03-02 Immunogenic compositions for chlamydia pneunomiae

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US54983204P 2004-03-02 2004-03-02
US60/549,832 2004-03-02
US64311005P 2005-01-12 2005-01-12
US60/643,110 2005-01-12
US64455205P 2005-01-19 2005-01-19
US60/644,552 2005-01-19

Publications (2)

Publication Number Publication Date
WO2005084306A2 true WO2005084306A2 (en) 2005-09-15
WO2005084306A3 WO2005084306A3 (en) 2006-05-26

Family

ID=34923269

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/006588 WO2005084306A2 (en) 2004-03-02 2005-03-02 Immunogenic compositions for chlamydia pneunomiae

Country Status (6)

Country Link
EP (1) EP1729800A4 (en)
JP (1) JP2007526318A (en)
BR (1) BRPI0508365A (en)
CA (1) CA2557353A1 (en)
RU (1) RU2006134631A (en)
WO (1) WO2005084306A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010032138A2 (en) 2008-09-18 2010-03-25 Novartis Ag Vaccine adjuvant combinations
WO2010136897A2 (en) 2009-05-28 2010-12-02 Novartis Ag Expression of recombinant proteins
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
EP2298795A1 (en) 2005-02-18 2011-03-23 Novartis Vaccines and Diagnostics, Inc. Immunogens from uropathogenic escherichia coli
EP2351772A1 (en) 2005-02-18 2011-08-03 Novartis Vaccines and Diagnostics, Inc. Proteins and nucleic acids from meningitis/sepsis-associated Escherichia coli
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
WO2012031140A1 (en) 2010-09-01 2012-03-08 Novartis Ag Adsorption of immunopotentiators to insoluble metal salts
WO2012082914A1 (en) 2010-12-14 2012-06-21 Novartis Ag Flow cytometry analysis of materials adsorbed to metal salts
EP2586790A2 (en) 2006-08-16 2013-05-01 Novartis AG Immunogens from uropathogenic Escherichia coli
WO2013131985A1 (en) 2012-03-07 2013-09-12 Novartis Ag Immunologically useful arginine salts
WO2013131983A1 (en) 2012-03-07 2013-09-12 Novartis Ag Adjuvanted formulations of streptococcus pneumoniae antigens
JP2014170003A (en) * 2008-07-25 2014-09-18 Cellestis Ltd Diagnostic method
US9375471B2 (en) 2012-03-08 2016-06-28 Glaxosmithkline Biologicals Sa Adjuvanted formulations of booster vaccines
US9950062B2 (en) 2009-09-02 2018-04-24 Glaxosmithkline Biologicals Sa Compounds and compositions as TLR activity modulators
CN108761054A (en) * 2017-04-11 2018-11-06 希森美康株式会社 The measurement of immunocyte immunostimulatory response, the judgement of immunocyte immunological synapse Forming ability and cell analysis apparatus
US10603369B2 (en) 2011-03-02 2020-03-31 Glaxosmithkline Biologicals Sa Combination vaccines with lower doses of antigen and/or adjuvant

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6822071B1 (en) * 1998-11-12 2004-11-23 The Regents Of The University Of California Polypeptides from Chlamydia pneumoniae and their use in the diagnosis, prevention and treatment of disease
AU2001276619A1 (en) * 2000-07-03 2002-01-14 Chiron S.P.A. Immunisation against chlamydia pneumoniae

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1729800A4 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2298795A1 (en) 2005-02-18 2011-03-23 Novartis Vaccines and Diagnostics, Inc. Immunogens from uropathogenic escherichia coli
EP2351772A1 (en) 2005-02-18 2011-08-03 Novartis Vaccines and Diagnostics, Inc. Proteins and nucleic acids from meningitis/sepsis-associated Escherichia coli
EP2586790A2 (en) 2006-08-16 2013-05-01 Novartis AG Immunogens from uropathogenic Escherichia coli
JP2014170003A (en) * 2008-07-25 2014-09-18 Cellestis Ltd Diagnostic method
WO2010032138A2 (en) 2008-09-18 2010-03-25 Novartis Ag Vaccine adjuvant combinations
WO2010136897A2 (en) 2009-05-28 2010-12-02 Novartis Ag Expression of recombinant proteins
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
US9950062B2 (en) 2009-09-02 2018-04-24 Glaxosmithkline Biologicals Sa Compounds and compositions as TLR activity modulators
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
US9315530B2 (en) 2010-09-01 2016-04-19 Novartis Ag Adsorption of immunopotentiators to insoluble metal salts
US10098949B2 (en) 2010-09-01 2018-10-16 Glaxosmithkline Biologicals S.A. Adsorption of immunopotentiators to insoluble metal salts
EP2719395A1 (en) 2010-09-01 2014-04-16 Novartis AG Adsorption of immunopotentiators to insoluble metal salts
WO2012031140A1 (en) 2010-09-01 2012-03-08 Novartis Ag Adsorption of immunopotentiators to insoluble metal salts
US9618508B2 (en) 2010-12-14 2017-04-11 Glaxosmithkline Biologicals Sa Flow cytometry analysis of materials adsorbed to metal salts
WO2012082914A1 (en) 2010-12-14 2012-06-21 Novartis Ag Flow cytometry analysis of materials adsorbed to metal salts
US10603369B2 (en) 2011-03-02 2020-03-31 Glaxosmithkline Biologicals Sa Combination vaccines with lower doses of antigen and/or adjuvant
US9320748B2 (en) 2012-03-07 2016-04-26 Novartis Ag Immunologically useful arginine salts
WO2013131983A1 (en) 2012-03-07 2013-09-12 Novartis Ag Adjuvanted formulations of streptococcus pneumoniae antigens
WO2013131985A1 (en) 2012-03-07 2013-09-12 Novartis Ag Immunologically useful arginine salts
US9375471B2 (en) 2012-03-08 2016-06-28 Glaxosmithkline Biologicals Sa Adjuvanted formulations of booster vaccines
US9931399B2 (en) 2012-03-08 2018-04-03 Glaxosmithkline Biologicals Sa Adjuvanted formulations of booster vaccines
US10842868B2 (en) 2012-03-08 2020-11-24 Glaxosmithkline Biologicals Sa Adjuvanted formulations of booster vaccines
CN108761054A (en) * 2017-04-11 2018-11-06 希森美康株式会社 The measurement of immunocyte immunostimulatory response, the judgement of immunocyte immunological synapse Forming ability and cell analysis apparatus
US11808761B2 (en) 2017-04-11 2023-11-07 Sysmex Corporation Method for measuring immunostimulatory response of immune cell, method for determining ability to form immune synapse in immune cell, and cell analyzer

Also Published As

Publication number Publication date
EP1729800A4 (en) 2008-06-11
WO2005084306A3 (en) 2006-05-26
RU2006134631A (en) 2008-04-10
BRPI0508365A (en) 2007-07-24
JP2007526318A (en) 2007-09-13
CA2557353A1 (en) 2005-09-15
EP1729800A2 (en) 2006-12-13

Similar Documents

Publication Publication Date Title
EP1729800A2 (en) Immunogenic compositions for chlamydia pneunomiae
US8133973B2 (en) Immunogenic compositions for Chlamydia trachomatis
JP4896715B2 (en) Immunogenic composition against Chlamydiatrachomatis
US7776336B2 (en) Compositions comprising Yersinia pestis antigens
US8481057B2 (en) Chlamydial antigens
US20100183674A1 (en) Compositions comprising yersinia pestis antigens
EP2448592A1 (en) Vaccines and compositions against streptococcus pneumoniae
US20190290748A1 (en) Antigens and antigen combinations
SK34699A3 (en) Nucleic acid and amino acid sequences relating to helicobacter pylori and vaccine compositions thereof
WO2013033092A2 (en) Streptococcus suis pilus antigens
MXPA06010041A (en) Immunogenic compositions for chlamydia pneunomiae
CN1984678A (en) Immunogenic compositions for pneumonia chlamydia
CZ148399A3 (en) Sequence of nucleic acids and amino acids connected with Helicobacter pylori and vaccine preparations
AU2012261545A1 (en) Immunogenic compositions for Chlamydia trachomatis
AU2013202316A1 (en) Compositions comprising Yersinia pestis antigens
MXPA01006576A (en) Chlamydia

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2557353

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2007501899

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/010041

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 2005724183

Country of ref document: EP

Ref document number: 2006134631

Country of ref document: RU

WWE Wipo information: entry into national phase

Ref document number: 200580013852.3

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005724183

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0508365

Country of ref document: BR