AU2007231768A1 - Immunisation against Chlamydia trachomatis - Google Patents

Immunisation against Chlamydia trachomatis Download PDF

Info

Publication number
AU2007231768A1
AU2007231768A1 AU2007231768A AU2007231768A AU2007231768A1 AU 2007231768 A1 AU2007231768 A1 AU 2007231768A1 AU 2007231768 A AU2007231768 A AU 2007231768A AU 2007231768 A AU2007231768 A AU 2007231768A AU 2007231768 A1 AU2007231768 A1 AU 2007231768A1
Authority
AU
Australia
Prior art keywords
protein
sequence
nucleic acid
seq
gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2007231768A
Inventor
Guido Grandi
Giulio Ratti
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GSK Vaccines SRL
Original Assignee
NOVARTIS VACCINES and DIAGNOSTIC
Novartis Vaccines and Diagnostics SRL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2002366592A external-priority patent/AU2002366592B2/en
Application filed by NOVARTIS VACCINES and DIAGNOSTIC, Novartis Vaccines and Diagnostics SRL filed Critical NOVARTIS VACCINES and DIAGNOSTIC
Priority to AU2007231768A priority Critical patent/AU2007231768A1/en
Publication of AU2007231768A1 publication Critical patent/AU2007231768A1/en
Abandoned legal-status Critical Current

Links

Description

AUSTRALIA
gill FB RICE CO Patent and Trade Mark Attorneys Patents Act 1990 NOVARTIS VACCINES AND DIAGNOSTICS S.R.L COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Immunisation against Chlamydia trachomatis The following statement is a full description of this invention including the best method of performing it known to us:- O IMMUNISATION AGAINST CHLAMYDIA TRACHOMATIS
(N
The present application is a divisional application of Australian Patent Application No.
O 2002366592 filed on December 12, 2002 which is the national phase application of International Application No. PCT/IB02/05761 which claims priority from the British c 5 applications GB 0129732.4 filed December 12, 2001, GB 0128233.5 filed August 6, 2002, and GB 0218924.9 filed August 12, 2002, each of which is herein specifically incorporated 00 by reference in their entirety.
r TECHNICAL FIELD This invention is in the field of immunisation against chlamydial infection, in particular 0 against infection by Chlamydia trachomatis.
BACKGROUND ART Chlamydiae are obligate intracellular parasites of eukaryotic cells which are responsible for endemic sexually transmitted infections and various other disease syndromes. They occupy an exclusive eubacterial phylogenic branch, having no close relationship to any other known organisms they are classified in their own order (Chlamydiales) which contains a single family (Chlamydiaceae) which in turn contains a single genus (Chlamydia, also referred to as Chlamydophila). A particular characteristic of the Chlamydiae is their unique life cycle, in which the bacterium alternates between two morphologically distinct forms: an extracellular infective form (elementary bodies, EB) and an intracellular non-infective form (reticulate bodies, RB). The life cycle is completed with the re-organization of RB into EB, which leave the disrupted host cell ready to infect further cells.
Four chlamydial species are currently known C.trachomatis, C.pneumoniae, C.pecorum and C.psittaci refs. 1, 2} and genome sequences are available {refs. 3 to 9}.
The human serovariants ("serovars") of C.trachomatis are divided into two biovariants ("biovars"). Serovars A-K elicit epithelial infections primarily in the ocular tissue or urogenital tract Serovars LI, L2 and L3 are the agents of invasive lymphogranuloma venereum (LGV).
Although chlamydial infection itself causes disease, it is thought that, in some patients, the severity of symptoms is due, in fact, to an aberrant host immune response. Failure to clear the infection results in persistent immune stimulation and, rather than helping the host, this results in chronic infection with severe consequences, including sterility and blindness addition, the protection conferred by natural chlamydial infection, is usually incomplete, transient, and strain-specific.
Due to the serious nature of the disease, there is a desire to provide suitable vaccines. These may be useful for immunisation against chlamydial infection or against chlamydia-induced disease (prophylactic vaccination) or for the eradication of an O established chronic chlamydial infection (therapeutic vaccination). Being an intracellular C parasite, however, the bacterium can generally evade antibody-mediated immune responses.
O Various antigenic proteins have been described for C.trachomatis, and the cell surface in particular has been the target of detailed research {eg. 1, 11}. These include, for instance, m 5 pgp3 {12, 13, 14}, MOMP Hsp60 (GroEL) {16} and Hsp70 (DnaK-like) Not all of these have proved to be effective vaccines, however, so it is an object of the invention 00 to identify C.trachomatis antigens which elicit an immune response during natural infection, in order to provide antigens and immunogens suitable for use in vaccine development. It is a further object to identify antigens useful for diagnosis (e.g.
¢C 10 immunodiagnosis) of C.trachomatis.
SDISCLOSURE OF THE INVENTION Reference 18 discloses various proteins from C.pneumoniae which were empirically verified as being immunoreactive, immunoaccessible and/or present in elementary bodies.
These properties of the proteins were not derivable from the genomic sequence information.
Reference 18 discloses that these proteins can be used in the treatment or prevention of infection due to Chlamydia bacteria, with C.pneumoniae being the main focus. The C.pneumoniae proteins can also be used for treating or preventing infection by other species of Chlamydia, due to inter-species cross-reactivity.
C.pneumoniae is closely related to C.trachomatis, as shown by whole genome comparisons The present invention relates to C.trachomatis proteins (odd numbered SEQ IDs 1-261) which correspond to the C.pneumoniae proteins disclosed in reference 18. These proteins can be used in the treatment or prevention of infection due to Chlamydia bacteria, and in particular C.trachomatis. Particularly preferred proteins are those previously annotated as 'hypothetical protein' (see Table I herein) or those which were previously thought to have a cytoplasmic location.
C.trachomatis proteins The invention provides proteins comprising one or more of the odd-numbered amino acid sequences SEQ IDs 1-261.
It also provides proteins comprising sequences which share at least x% sequence identity with one or more of the odd-numbered amino acid sequences SEQ IDs 1-261. Depending on the particular sequence, x is preferably 50% or more 60%, 70%, 80%, 90%, 99% or more). These include mutants and allelic variants. Typically, 50% identity or more between two proteins is considered to be an indication of functional equivalence. Identity between proteins is preferably determined by the Smith-Waterman homology search 0 algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affine C1 gap search with parameters gap open penalty=12 and gap extension penalty=l.
The invention further provides proteins comprising fragments of the odd-numbered amino _acid sequences SEQ IDs 1-261. The fragments should comprise at least n consecutive C 5 amino acids from the sequences and, depending on the particular sequence, n is 7 or more 8, 10, 12, 14, 16, 18, 20, 30, 40, 50, 75, 100, 150, 200 or more). Preferably the 00 \O fragments comprise one or more epitope(s) from the sequence. Other preferred fragments I omit a signal peptide.
C The proteins of the invention can be prepared by various means e.g. by chemical synthesis (at least in part), by digesting longer polypeptides using proteases, by translation from RNA, by purification from cell culture from recombinant expression or from C.trachomatis culture) etc. Heterologous expression in E.coli is a preferred preparative route.
The proteins of the invention can take various forms e.g. native, fusions, glycosylated, non-glycosylated, lipidated etc.).
Proteins of the invention are preferably prepared in substantially pure form (ie. substantially free from other C.trachomatis or host cell proteins).
Proteins of the invention may be attached to a solid support. They may comprise a detectable label a radioactive or fluorescent label, or a biotin label).
Proteins of the invention are preferably Chlamydial proteins.
This invention further provides the use of a protein in the manufacture of a medicament for the treatment or prevention of infection due to Chlamydia trachomatis, wherein the protein is a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123, 125, 127, 129, 131, 133, 135, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229, 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 257, 259 and 261, a protein comprising an amino acid sequence having 50% or greater sequence identity to the amino acid sequence of or a protein comprising a fragment of an amino acid sequence of This invention further provides the use of a protein in the manufacture of a medicament for the treatment or prevention of infection due to Chlamydia trachomatis, wherein the protein is a protein comprising an amino acid sequence according to SEQ ID NO: 125, or SEQ O ID NO: 155, or SEQ ID NO: 157, or SEQ ID NO: 229, or SEQ ID NO: 247, a protein rC comprising an amino acid sequence having 50% or greater sequence identity to the amino 0 acid sequence of or a protein comprising a fragment of an amino acid sequence of 0 C.trachomatis nucleic acids The invention provides proteins comprising one or more of the even-numbered nucleotide IN sequences SEQ IDs 2-262.
c The invention also provides nucleic acid comprising sequences which share at least x% sequence identity with the even-numbered nucleotide sequences SEQ IDs 2-262. Depending on the particular sequence, x is preferably 50% or more 60%, 70%, 80%, 90%, Ci 99% or more).
Furthermore, the invention provides nucleic acid which can hybridise to nucleic acid comprising the even-numbered nucleotide sequences SEQ IDs 2-262. Hybridisation reactions can be performed under conditions of different "stringency". Conditions that increase stringency of a hybridisation reaction of widely known and published in the art.
Examples of relevant conditions include (in order of increasing stringency): incubation temperatures of 25 0 C, 37°C, 50 0 C, 55°C and 68 0 C; buffer concentrations of 10 X SSC, 6 X SSC, 1 X SSC, 0.1 X SSC and their equivalents using other buffer systems; formamide concentrations of 25%, 50%, and 75%; incubation times from 5 minutes to 24 hours; 1, 2, or more washing steps; wash incubation times of 1, 2, or 15 minutes; and wash solutions of 6 x SSC, 1 x SSC, 0.1 x SSC, or de-ionized water. In some embodiments, the isolated nucleic acid of the invention selectively hybridises under low stringency conditions; in other embodiments it selectively hybridises under intermediate stringency conditions; in other embodiments, it selectively hybridises under high stringency conditions. An exemplary set of low stringency hybridisation conditions is 50 0 C and 10xSSC. An exemplary set of intermediate stringency hybridisation conditions is 55°C and IxSSC. An exemplary set of high stringent hybridisation conditions is 68 0 C and 0.1 x SSC.
Nucleic acid comprising fragments of the even-numbered nucleotide sequences SEQ IDs 2- 262 are also provided. These should comprise at least n consecutive nucleotides from the C.trachomatis sequences and, depending on the particular sequence, n is 7 or more 12, 14, 15, 18, 20, 25, 30, 35, 40, 50, 75, 100, 200, 300 or more).
According to a further aspect, the invention provides nucleic acid encoding the proteins and protein fragments of the invention.
The invention provides nucleic acid comprising sequences complementary to those described above for antisense or probing purposes).
1^ O Nucleic acid of the invention can, of course, be prepared in many ways e.g. by chemical CN synthesis (at least in part), by digesting longer polynucleotides using restriction enzymes, 0 from genomic or cDNA libraries, from the organism itself etc.
O
_Nucleic acid of the invention can take various forms single-stranded, double-stranded, C 5 linear, circular, vectors, primers, probes etc.).
00 This invention provides the use of a nucleic acid in the manufacture of a medicament for the I treatment or prevention of infection due to Chlamydia trachomatis, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence selected from the group consisting of SSEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, (N 96,98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260 and 262, a nucleic acid comprising a nucleotide sequence having 50% or greater sequence identity to the nucleotide sequence of or a nucleic acid comprising a fragment of an nucleotide sequence of(a).
This invention further preferably provides the use of a nucleic acid in the manufacture of a medicament for the treatment or prevention of infection due to Chlamydia trachomatis, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence according to SEQ ID NO: 126, or SEQ ID NO: 156, or SEQ ID NO: 188, or SEQ ID NO:230; SEQ ID NO: 248.
Nucleic acids of the invention may be attached to a solid support a bead, plate, filter, film, slide, resin, etc.). Nucleic acids of the invention may include a detectable label a radioactive or fluorescent label, or a biotin label). This is particularly useful where the polynucleotide is to be used in nucleic acid detection techniques e.g. where the nucleic acid is a primer or as a probe for use in techniques such as PCR, LCR, TMA, NASBA, bDNA etc.
Nucleic acids of the invention are preferably Chlamydial nucleic acids.
The term "nucleic acid" includes DNA, RNA, DNA/RNA hybrids, and DNA or RNA analogs, such as those containing modified backbones or bases, and also peptide nucleic acids (PNA) etc.
Nucleic acids of the invention may be isolated and obtained in substantial purity, generally as other than an intact chromosome. Usually, the polynucleotides will be obtained O substantially free of other naturally-occurring nucleic acid sequences, generally being at CN least about 50% (by weight) pure, usually at least about 90% pure.
-4-4 U Nucleic acids can be used, for example: to produce polypeptides; as probes for the detection _of nucleic acid in biological samples; to generate additional copies of the polynucleotides; ¢C 5 to generate ribozymes or antisense oligonucleotides; and as single-stranded DNA probes or as triple-strand forming oligonucleotides etc.
00 N0 The invention provides vectors comprising nucleotide sequences of the invention (e.g.
Scloning or expression vectors) and host cells transformed therewith.
Compositions According to a further aspect, the invention provides compositions comprising protein and/or nucleic acid according to the invention. These compositions are preferably immunogenic compositions, such as vaccines, and are suitable for immunisation and vaccination purposes. Vaccines of the invention may be prophylactic or therapeutic, and will typically comprise an antigen which can induce antibodies capable of inhibiting (a) chlamydial adhesion, chlamydial entry, and/or successful replication within the host cell. The vaccines preferably induce any cell-mediated T-cell responses which are necessary for chlamydial clearance from the host.
The invention also provides nucleic acid or protein according to the invention for use as medicaments as vaccines).
The invention also provides the use of nucleic acid or protein according to the invention in the manufacture of a medicament a vaccine or an immunogenic composition) for treating or preventing infection due to a Chlamydia. This will generally be C.trachomatis but, due to inter-species cross-reactivity, it may also be C.pneumoniae, C.pecorum or C.psittaci. For prevention, the medicament preferably elicits an immune response which is specific to the EB form of Chlamydia; for treatment, the medicament preferably elicits an immune response which is specific to the RB form of Chlamydia.
The invention also provides the use of nucleic acid or protein according to the invention in the manufacture of a medicament a vaccine or an immunogenic composition) for neutralizing Chlamydia trachomatis elementary bodies.
The invention further provides an immunogenic composition comprising a protein and an adjuvant, wherein the protein is a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81,83,85,87,89,91,93,95,97,99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123, 125, 127, 129, 131, 133, 135, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, S159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229, O 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 257, 259 and 261, a C protein comprising an amino acid sequence having 50% or greater sequence identity to the amino acid sequence of or a protein comprising a fragment of an amino acid sequence of 00 00 Preferably, an immunogenic composition of the invention comprises a protein and an adjuvant, wherein the protein is a protein comprising an amino acid sequence according to SEQ ID NO: 125, or SEQ ID NO: 155, or SEQ ID NO: 157, or SEQ ID NO: 229, or SEQ (N 10 ID NO: 247, a protein comprising an amino acid sequence having 50% or greater Ssequence identity to the amino acid sequence of or a protein comprising a fragment of an amino acid sequence of(a).
The invention further provides an immunogenic composition comprising a nucleic acid and an adjuvant, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260 and 262, a nucleic acid comprising a nucleotide sequence having 50% or greater sequence identity to the nucleotide sequence of or a nucleic acid comprising a fragment of an nucleotide sequence of Preferably, an immunogenic composition of the invention comprises a nucleic acid and an adjuvant, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence according to SEQ ID NO: 126, or SEQ ID NO: 156, or SEQ ID NO: 188, or SEQ ID NO:230; SEQ ID NO: 248.
The invention also provides a method of treating immunising) a patient a human), comprising administering to the patient a therapeutically effective amount of nucleic acid or protein according to the invention.
The invention also provides a method of raising an immune response in a patient, comprising administering to the patient an immunologically effective amount of nucleic acid or protein according to the invention. The immune response may involve raising antibodies in the patient and/or raising a cellular immune response a CTL response).
The immune response may be specific for an EB or a RB protein, or to a protein which is O expressed in the host cytoplasm. An antibody response is preferably specific to an EB, C"1 whereas a cellular immune response is preferably specific to a cytoplasmic protein or, 0 preferably, to an RB protein.
O
_The invention also provides a method of raising antibodies which recognise a protein of the c 5 invention, comprising the step of administering to a patient a Chlamydia elementary body or reticulate body. The antibodies are preferably specific to an EB.
00 D The invention also provides a method of neutralizing C.trachomatis infectivity, comprising Sthe step of administering to a patient a protein, nucleic acid or antibody of the invention.
(C The method preferably neutralizes EB infectivity.
The invention also provides a method for detecting a Chlamydia EB or RB in a biological i1 sample, comprising the step of contacting an antibody of the invention with the sample. The sample could be a blood sample, another bodily fluid, or a tissue sample. The method may be used to diagnose chlamydial infection.
Immunogenic compositions of the invention may also include one or more of the following antigens: a protein antigen from Helicobacter pylori such as VacA, CagA, NAP, HopX, HopY {e.g.
W098/04702} and/or urease.
a protein antigen from N.meningitidis serogroup B, such as those in W099/24578, W099/36544, W099/57280, WO00/22430, Tettelin et al. (2000) Science 287:1809-1815, Pizza et al. (2000) Science 287:1816-1820 and W096/29412, with protein '287' and derivatives being particularly preferred.
an outer-membrane vesicle (OMV) preparation from N.meningitidis serogroup B, such as those disclosed in WO01/52885; Bjune et al. (1991) Lancet 338(8775):1093-1096; Fukasawa et al. (1999) Vaccine 17:2951-2958; Rosenqvist et al. (1998) Dev. Biol. Stand.
92:323-333 etc.
a saccharide antigen from N.meningitidis serogroup A, C, W135 and/or Y, such as the oligosaccharide disclosed in Costantino et al. (1992) Vaccine 10:691-698 from serogroup C {see also Costantino et al. (1999) Vaccine 17:1251-1263}.
a saccharide antigen from Streptococcus pneumoniae Watson (2000) Pediatr Infect Dis J 19:331-332; Rubin (2000) Pediatr Clin North Am 47:269-285, v; Jedrzejas (2001) Microbiol Mol Biol Rev 65:187-207}.
an antigen from hepatitis A virus, such as inactivated virus Bell (2000) Pediatr Infect DisJ 19:1187-1188; Iwarson (1995) APMIS 103:321-326}.
an antigen from hepatitis B virus, such as the surface and/or core antigens Gerlich et al. (1990) Vaccine 8 Suppl:S63-68 79-80}.
O an antigen from hepatitis C virus Hsu et al. (1999) Clin Liver Dis 3:901-915}.
c an antigen from Bordetella pertussis, such as pertussis holotoxin (PT) and filamentous O haemagglutinin (FHA) from B.pertussis, optionally also in combination with pertactin
O
and/or agglutinogens 2 and 3 Gustafsson et al. (1996) N. Engl. J. Med. 334:349-355; m 5 Rappuoli et al. (1991) TIBTECH 9:232-238}.
a diphtheria antigen, such as a diphtheria toxoid chapter 3 of Vaccines (1988) eds.
00 00 Plotkin Mortimer. ISBN 0-7216-1946-0} e.g. the CRM 1 97 mutant Del Guidice et al. (1998) Molecular Aspects of Medicine 19:1-70}.
a tetanus antigen, such as a tetanus toxoid chapter 4 of Plotkin Mortimer}.
a saccharide antigen from Haemophilus influenzae B.
C an antigen from N.gonorrhoeae W099/24578, W099/36544, W099/57280}.
an antigen from Chlamydia pneumoniae PCT/IB01/01445; Kalman et al. (1999) Nature Genetics 21:385-389; Read et al. (2000) Nucleic Acids Res 28:1397-406; Shirai et al. (2000) J. Infect. Dis. 181(Suppl 3):S524-S527; W099/27105; WO00/27994; WO00/37494}.
an antigen from Chlamydia trachomatis W099/28475}.
an antigen from Porphyromonas gingivalis Ross et al. (2001) Vaccine 19:4135- 4142).
polio antigen(s) Sutter et al. (2000) Pediatr Clin North Am 47:287-308; Zimmerman Spann (1999) Am Fam Physician 59:113-118, 125-126} such as IPV or OPV.
rabies antigen(s) Dreesen (1997) Vaccine 15 Suppl:S2-6} such as lyophilised inactivated virus MMWR Morb Mortal Wkly Rep 1998 Jan 16;47(1):12, 19; RabAvertTM}.
measles, mumps and/or rubella antigens chapters 9, 10 11 of Plotkin Mortimer}.
influenza antigen(s) chapter 19 of Plotkin Mortimer), such as the haemagglutinin and/or neuraminidase surface proteins.
an antigen from Moraxella catarrhalis McMichael (2000) Vaccine 19 Suppl 1:S101- 107}.
an antigen from Staphylococcus aureus Kuroda et al. (2001) Lancet 357(9264):1225- 1240; see also pages 1218-1219}.
an antigen from Streptococcus agalactiae see W002/34771 an antigen from Streptococcus pyogenes see W002/34771 Where a saccharide or carbohydrate antigen is included, it is preferably conjugated to a carrier protein in order to enhance immunogenicity Ramsay et al. (2001) Lancet 357(9251):195-196; Lindberg (1999) Vaccine 17 Suppl 2:S28-36; Conjugate Vaccines (eds.
O Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114 etc.}. Preferred carrier proteins C are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids. The CRM 97 diphtheria 0 toxoid is particularly preferred. Other suitable carrier proteins include the N.meningitidis O outer membrane protein EP-0372501}, synthetic peptides EP-0378881, EP- 5 0427347}, heat shock proteins WO93/17712}, pertussis proteins W098/58668; EP-0471177}, protein D from H.influenzae WO00/56360}, toxin A or B from 00 C.difficile WO00/61761}, etc. Any suitable conjugation reaction can be used, with any suitable linker where necessary.
n Toxic protein antigens may be detoxified where necessary detoxification of pertussis r 10 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 are preferably adsorbed to an aluminium salt.
Antigens in the composition will typically be present at a concentration of at least 1 lg/ml each. In general, the concentration of any given antigen will be sufficient to elicit an immune response against that antigen.
The invention also provides compositions comprising two or more proteins of the present invention.
Processes The invention provides a process for producing proteins of the invention, comprising the step of culturing a host cell according to the invention under conditions which induce protein expression.
The invention provides a process for producing protein or nucleic acid of the invention, wherein the protein or nucleic acid is synthesised in part or in whole using chemical means.
The invention provides a process for detecting C.trachomatis in a sample, wherein the sample is contacted with an antibody which binds to a protein of the invention A summary of standard techniques and procedures which may be employed in order to perform the invention to utilise the disclosed sequences for immunisation) follows.
This summary is not a limitation on the invention but, rather, gives examples that may be used, but are not required.
-12- 0 General The practice of the present invention will employ, unless otherwise indicated, conventional techniques 0 of molecular biology, microbiology, recombinant DNA, and immunology, which are within the skill C of the art. Such techniques are explained fully in the literature e.g. Sambrook Molecular Cloning; A Laboratory Manual, Second Edition (1989) and Third Edition (2001); DNA Cloning, Volumes I and ii (D.N Glover ed. 1985); Oligonucleotide Synthesis Gait ed, 1984); Nucleic Acid Hybridization oO Hames S.J. Higgins eds. 1984); Transcription and Translation Hames S.J. Higgins I\O eds. 1984); Animal Cell Culture Freshney ed. 1986); Immobilized Cells and Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide to Molecular Cloning (1984); the Methods in Enzymology series S 10 (Academic Press, Inc.), especially volumes 154 155; Gene Transfer Vectors for Mammalian Cells Miller and M.P. Calos eds. 1987, Cold Spring Harbor Laboratory); Mayer and Walker, eds.
(1987), Immunochemical Methods in Cell and Molecular Biology (Academic Press, London); Scopes, C1 (1987) Protein Purification: Principles and Practice, Second Edition (Springer-Verlag, and Handbook of Experimental Immunology, Volumes I-IV Weir and C. C. Blackwell eds 1986).
Standard abbreviations for nucleotides and amino acids are used in this specification.
Definitions A composition containing X is "substantially free of' Y when at least 85% by weight of the total X+Y in the composition is X. Preferably, X comprises at least about 90% by weight of the total of X+Y in the composition, more preferably at least about 95% or even 99% by weight.
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 to X, such as X+Y.
The term "heterologous" refers to two biological components that are not found together in nature.
The components may be host cells, genes, or regulatory regions, such as promoters. Although the heterologous components are not found together in nature, they can function together, as when a promoter heterologous to a gene is operably linked to the gene. Another example is where a Chlamydial sequence is heterologous to a mouse host cell. A further examples would be two epitopes from the same or different proteins which have been assembled in a single protein in an arrangement not found in nature.
An "origin of replication" is a polynucleotide sequence that initiates and regulates replication of polynucleotides, such as an expression vector. The origin of replication behaves as an autonomous unit of polynucleotide replication within a cell, capable of replication under its own control. An origin of replication may be needed for a vector to replicate in a particular host cell. With certain origins of replication, an expression vector can be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells.
A "mutant" sequence is defined as DNA, RNA or amino acid sequence differing from but having sequence identity with the native or disclosed sequence. Depending on the particular sequence, the degree of sequence identity between the native or disclosed sequence and the mutant sequence is preferably greater than 50% 60%, 70%, 80%, 90%, 95%, 99% or more, calculated using the -13- Smith-Waterman algorithm as described above). As used herein, an "allelic variant" of a nucleic acid molecule, or region, for which nucleic acid sequence is provided herein is a nucleic acid molecule, or region, that occurs essentially at the same locus in the genome of another or second isolate, and that, O due to natural variation caused by, for example, mutation or recombination, has a similar but not identical nucleic acid sequence. A coding region allelic variant typically encodes a protein having similar activity to that of the protein encoded by the gene to which it is being compared. An allelic variant can also comprise an alteration in the 5' or 3' untranslated regions of the gene, such as in 0 0 regulatory control regions see US patent 5,753,235).
Expression systems 10 The Chlamydial nucleotide sequences can be expressed in a variety of different expression systems; for example those used with mammalian cells, baculoviruses, plants, bacteria, and yeast.
i. Mammalian Systems Mammalian expression systems are known in the art. A mammalian promoter is any DNA sequence capable of binding mammalian RNA polymerase and initiating the downstream transcription of a coding sequence structural gene) into mRNA. A promoter will have a transcription initiating region, which is usually placed proximal to the 5' end of the coding sequence, and a TATA box, usually located 25-30 base pairs (bp) upstream of the transcription initiation site. The TATA box is thought to direct RNA polymerase II to begin RNA synthesis at the correct site. A mammalian promoter will also contain an upstream promoter element, usually located within 100 to 200 bp upstream of the TATA box. An upstream promoter element determines the rate at which transcription is initiated and can act in either orientation {Sambrook et al. (1989) "Expression of Cloned Genes in Mammalian Cells." In Molecular Cloning: A Laboratory Manual, 2nd ed.).
Mammalian viral genes are often highly expressed and have a broad host range; therefore sequences encoding mammalian viral genes provide particularly useful promoter sequences. Examples include the SV40 early promoter, mouse mammary tumor virus LTR promoter, adenovirus major late promoter (Ad MLP), and herpes simplex virus promoter. In addition, sequences derived from nonviral genes, such as the murine metallotheionein gene, also provide useful promoter sequences.
Expression may be either constitutive or regulated (inducible), depending on the promoter can be induced with glucocorticoid in hormone-responsive cells.
The presence of an enhancer element (enhancer), combined with the promoter elements described above, will usually increase expression levels. An enhancer is a regulatory DNA sequence that can stimulate transcription up to 1000-fold when linked to homologous or heterologous promoters, with synthesis beginning at the normal RNA start site. Enhancers are also active when they are placed upstream or downstream from the transcription initiation site, in either normal or flipped orientation, or at a distance of more than 1000 nucleotides from the promoter (Maniatis et al. (1987) Science 236.1237; Alberts et al. (1989) Molecular Biology of the Cell, 2nd Enhancer elements derived from viruses may be particularly useful, because they usually have a broader host range. Examples include the SV40 early gene enhancer {Dijkema et al (1985) EMBO J. 4:761} and the enhancer/promoters derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus {Gorman et al. (1982) PNAS USA 79:6777} and from human cytomegalovirus {Boshart et al. (1985) Cell O 41:521}. Additionally, some enhancers are regulatable and become active only in the presence of an I inducer, such as a hormone or metal ion {Sassone-Corsi and Borelli (1986) Trends Genet. 2:215; Maniatis et al. (1987) Science 236:1237}.
0 A DNA molecule may be expressed intracellularly in mammalian cells. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If o0 desired, the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen O bromide.
SAlternatively, foreign proteins can also be secreted from the cell into the growth media by creating CK1 10 chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in mammalian cells. Preferably, there are processing sites encoded between the leader fragment and the foreign gene that can be cleaved either in vivo or in vitro. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell. The adenovirus triparite leader is an example of a leader sequence that provides for secretion of a foreign protein in mammalian cells.
Usually, transcription termination and polyadenylation sequences recognized by mammalian cells are regulatory regions located 3' to the translation stop codon and thus, together with the promoter elements, flank the coding sequence. The 3' terminus of the mature mRNA is formed by site-specific post-transcriptional cleavage and polyadenylation {Birnstiel et al. (1985) Cell 41:349; Proudfoot and Whitelaw (1988) "Termination and 3' end processing of eukaryotic RNA. In Transcription and splicing (ed. B.D. Hames and D.M. Glover); Proudfoot (1989) Trends Biochem. Sci. 14:105}. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Examples of transcription terminater/polyadenylation signals include those derived from {Sambrook et al (1989) "Expression of cloned genes in cultured mammalian cells." In Molecular Cloning: A Laboratory Manual}.
Usually, the above described components, comprising a promoter, polyadenylation signal, and transcription termination sequence are put together into expression constructs. Enhancers, introns with functional splice donor and acceptor sites, and leader sequences may also be included in an expression construct, if desired. Expression constructs are often maintained in a replicon, such as an extrachromosomal element plasmids) capable of stable maintenance in a host, such as mammalian cells or bacteria. Mammalian replication systems include those derived from animal viruses, which require trans-acting factors to replicate. For example, plasmids containing the replication systems of papovaviruses, such as SV40 {Gluzman (1981) Cell 23:175} or polyomavirus, replicate to extremely high copy number in the presence of the appropriate viral T antigen. Additional examples of mammalian replicons include those derived from bovine papillomavirus and Epstein-Barr virus. Additionally, the replicon may have two replicaton systems, thus allowing it to be maintained, for example, in mammalian cells for expression and in a prokaryotic host for cloning and amplification. Examples of such mammalian-bacteria shuttle vectors include pMT2 {Kaufman et al.
(1989) Mol. Cell. Biol. 9:946} and pHEBO {Shimizu et al. (1986) Mol. Cell. Biol. 6:1074}.
O The transformation procedure used depends upon the host to be transformed. Methods for introduction CN of heterologous polynucleotides into mammalian cells are known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, O protoplast fusion, electroporation, encapsulation of polynucleotide(s) in liposomes, direct microinjection of the DNA into nuclei.
Cn, Mammalian cell lines available as hosts for expression are known in the art and include many 0 immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to, Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells Hep G2), and a number of S 10 other cell lines.
ii. Baculovirus Systems C= The polynucleotide encoding the protein can also be inserted into a suitable insect expression vector, and is operably linked to the control elements within that vector. Vector construction employs techniques which are known in the art. Generally, the components of the expression system include a transfer vector, usually a bacterial plasmid, which contains both a fragment of the baculovirus genome, and a convenient restriction site for insertion of the heterologous gene or genes to be expressed; a wild type baculovirus with a sequence homologous to the baculovirus-specific fragment in the transfer vector (this allows for the homologous recombination of the heterologous gene in to the baculovirus genome); and appropriate insect host cells and growth media.
After inserting the DNA sequence encoding the protein into the transfer vector, the vector and the wild type viral genome are transfected into an insect host cell where the vector and viral genome are allowed to recombine. The packaged recombinant virus is expressed and recombinant plaques are identified and purified. Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA ("MaxBac" kit). These techniques are generally known to those skilled in the art and fully described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987) (hereinafter "Summers and Smith").
Prior to inserting the DNA sequence encoding the protein into the baculovirus genome, the above described components, comprising a promoter, leader (if desired), coding sequence of interest, and transcription termination sequence, are usually assembled into an intermediate transplacement construct (transfer vector). This construct may contain a single gene and operably linked regulatory elements; multiple genes, each with its owned set of operably linked regulatory elements; or multiple genes, regulated by the same set of regulatory elements. Intermediate transplacement constructs are often maintained in a replicon, such as an extrachromosomal element plasmids) capable of stable maintenance in a host, such as a bacterium. The replicon will have a replication system, thus allowing it to be maintained in a suitable host for cloning and amplification.
Currently, the most commonly used transfer vector for introducing foreign genes into AcNPV is pAc373. Many other vectors, known to those of skill in the art, have also been designed. These include, for example, pVL985 (which alters the polyhedrin start codon from ATG to ATT, and which introduces a BamHI cloning site 32 basepairs downstream from the ATT; see Luckow and Summers, Virology (1989) 17:31.
-16- O The plasmid usually also contains the polyhedrin polyadenylation signal (Miller et al. (1988) Ann.
Rev. Microbiol., 42:177) and a prokaryotic ampicillin-resistance (amp) gene and origin of replication for selection and propagation in E.coli.
O Baculovirus transfer vectors usually contain a baculovirus promoter. A baculovirus promoter is any DNA sequence capable of binding a baculovirus RNA polymerase and initiating the downstream to transcription of a coding sequence structural gene) into mRNA. A promoter will have a o0 transcription initiation region which is usually placed proximal to the 5' end of the coding sequence.
O This transcription initiation region usually includes an RNA polymerase binding site and a transcription initiation site. A baculovirus transfer vector may also have a second domain called an enhancer, which, if present, is usually distal to the structural gene. Expression may be either regulated or constitutive.
0 Structural genes, abundantly transcribed at late times in a viral infection cycle, provide particularly useful promoter sequences. Examples include sequences derived from the gene encoding the viral polyhedron protein, Friesen et al., (1986) "The Regulation of Baculovirus Gene Expression," in: The Molecular Biology ofBaculoviruses (ed. Walter Doerfler); EPO Publ. Nos. 127 839 and 155 476; and the gene encoding the pl0 protein, Vlak et al., (1988), J. Gen. Virol. 69:765.
DNA encoding suitable signal sequences can be derived from genes for secreted insect or baculovirus proteins, such as the baculovirus polyhedrin gene (Carbonell et al. (1988) Gene, 73:409).
Alternatively, since the signals for mammalian cell posttranslational modifications (such as signal peptide cleavage, proteolytic cleavage, and phosphorylation) appear to be recognized by insect cells, and the signals required for secretion and nuclear accumulation also appear to be conserved between the invertebrate cells and vertebrate cells, leaders of non-insect origin, such as those derived from genes encoding human a-interferon, Maeda et al., (1985), Nature 315:592; human gastrin-releasing peptide, Lebacq-Verheyden et al., (1988), Molec. Cell. Biol. 8:3129; human IL-2, Smith et al., (1985) Proc. Nat'l Acad. Sci. USA, 82:8404; mouse IL-3, (Miyajima et al., (1987) Gene 58:273; and human glucocerebrosidase, Martin et al. (1988) DNA, 7:99, can also be used to provide for secretion in insects.
A recombinant polypeptide or polyprotein may be expressed intracellularly or, if it is expressed with the proper regulatory sequences, it can be secreted. Good intracellular expression of nonfused foreign proteins usually requires heterologous genes that ideally have a short leader sequence containing suitable translation initiation signals preceding an ATG start signal. If desired, methionine at the Nterminus may be cleaved from the mature protein by in vitro incubation with cyanogen bromide.
Alternatively, recombinant polyproteins or proteins which are not naturally secreted can be secreted from the insect cell by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in insects. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the translocation of the protein into the endoplasmic reticulum.
After insertion of the DNA sequence and/or the gene encoding the expression product precursor of the protein, an insect cell host is co-transformed with the heterologous DNA of the transfer vector and the genomic DNA of wild type baculovirus usually by co-transfection. The promoter and transcription O termination sequence of the construct will usually comprise a 2-5kb section of the baculovirus C-1 genome. Methods for introducing heterologous DNA into the desired site in the baculovirus virus are known in the art. (See Summers and Smith supra; Ju et al. (1987); Smith et al., Mol. Cell. Biol. (1983) O 3:2156; and Luckow and Summers (1989)). For example, the insertion can be into a gene such as the polyhedrin gene, by homologous double crossover recombination; insertion can also be into a restriction enzyme site engineered into the desired baculovirus gene. Miller et al., (1989), Bioessays 4:91.The DNA sequence, when cloned in place of the polyhedrin gene in the expression vector, is O flanked both 5' and 3' by polyhedrin-specific sequences and is positioned downstream of the polyhedrin promoter.
r 10 The newly formed baculovirus expression vector is subsequently packaged into an infectious r recombinant baculovirus. Homologous recombination occurs at low frequency (between and thus, the majority of the virus produced after cotransfection is still wild-type virus. Therefore, a c method is necessary to identify recombinant viruses. An advantage of the expression system is a visual screen allowing recombinant viruses to be distinguished. The polyhedrin protein, which is produced by the native virus, is produced at very high levels in the nuclei of infected cells at late times after viral infection. Accumulated polyhedrin protein forms occlusion bodies that also contain embedded particles. These occlusion bodies, up to 15pm in size, are highly refractile, giving them a bright shiny appearance that is readily visualized under the light microscope. Cells infected with recombinant viruses lack occlusion bodies. To distinguish recombinant virus from wild-type virus, the transfection supernatant is plaqued onto a monolayer of insect cells by techniques known to those skilled in the art.
Namely, the plaques are screened under the light microscope for the presence (indicative of wild-type virus) or absence (indicative of recombinant virus) of occlusion bodies. "Current Protocols in Microbiology" Vol. 2 (Ausubel et al. eds) at 16.8 (Supp. 10, 1990); Summers Smith, supra; Miller et al. (1989).
Recombinant baculovirus expression vectors have been developed for infection into several insect cells. For example, recombinant baculoviruses have been developed for, inter alia: Aedes aegypti, Autographa californica, Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda, and Trichoplusia ni (WO 89/046699; Carbonell et al., (1985) J. Virol. 56:153; Wright (1986) Nature 321:718; Smith et al., (1983) Mol. Cell. Biol. 3:2156; and see generally, Fraser, et al. (1989) In Vitro Cell. Dev. Biol. 25:225).
Cells and cell culture media are commercially available for both direct and fusion expression of heterologous polypeptides in a baculovirus/expression system; cell culture technology is generally known to those skilled in the art. See, e.g. Summers and Smith supra.
The modified insect cells may then be grown in an appropriate nutrient medium, which allows for stable maintenance of the plasmid(s) present in the modified insect host. Where the expression product gene is under inducible control, the host may be grown to high density, and expression induced.
Alternatively, where expression is constitutive, the product will be continuously expressed into the medium and the nutrient medium must be continuously circulated, while removing the product of interest and augmenting depleted nutrients. The product may be purified by such techniques as chromatography, e.g. HPLC, affinity chromatography, ion exchange chromatography, etc.; electrophoresis; density gradient centrifugation; solvent extraction, or the like. As appropriate, the 0 product may be further purified, as required, so as to remove substantially any insect proteins which C are also secreted in the medium or result from lysis of insect cells, so as to provide a product which is at least substantially free of host debris, e.g. proteins, lipids and polysaccharides.
O In order to obtain protein expression, recombinant host cells derived from the transformants are incubated under conditions which allow expression of the recombinant protein encoding sequence.
These conditions will vary, dependent upon the host cell selected. However, the conditions are readily ascertainable to those of ordinary skill in the art, based upon what is known in the art.
\O
iii. Plant Systems C There are many plant cell culture and whole plant genetic expression systems known in the art.
S 10 Exemplary plant cellular genetic expression systems include those described in patents, such as: US S5,693,506; US 5,659,122; and US 5,608,143. Additional examples of genetic expression in plant cell culture has been described by Zenk, Phytochemistry 30:3861-3863 (1991). Descriptions of plant protein signal peptides may be found in addition to the references described above in Vaulcombe et al., Mol. Gen. Genet. 209:33-40 (1987); Chandler et al., Plant Molecular Biology 3:407-418 (1984); Rogers, J. Biol. Chem. 260:3731-3738 (1985); Rothstein et al., Gene 55:353-356 (1987); Whittier et al., Nucleic Acids Research 15:2515-2535 (1987); Wirsel et al., Molecular Microbiology 3:3-14 (1989); Yu et al., Gene 122:247-253 (1992). A description of the regulation of plant gene expression by the phytohormone, gibberellic acid and secreted enzymes induced by gibberellic acid can be found in R.L. Jones and J. MacMillin, Gibberellins: in: Advanced Plant Physiology,. Malcolm B. Wilkins, ed., 1984 Pitman Publishing Limited, London, pp. 21-52. References that describe other metabolically-regulated genes: Sheen, Plant Cell, 2:1027-1038(1990); Maas et al., EMBO J. 9:3447- 3452 (1990); Benkel and Hickey, Proc. Natl. Acad. Sci. 84:1337-1339 (1987) Typically, using techniques known in the art, a desired polynucleotide sequence is inserted into an expression cassette comprising genetic regulatory elements designed for operation in plants. The expression cassette is inserted into a desired expression vector with companion sequences upstream and downstream from the expression cassette suitable for expression in a plant host. The companion sequences will be of plasmid or viral origin and provide necessary characteristics to the vector to permit the vectors to move DNA from an original cloning host, such as bacteria, to the desired plant host. The basic bacterial/plant vector construct will preferably provide a broad host range prokaryote replication origin; a prokaryote selectable marker; and, for Agrobacterium transformations, T DNA sequences for Agrobacterium-mediated transfer to plant chromosomes. Where the heterologous gene is not readily amenable to detection, the construct will preferably also have a selectable marker gene suitable for determining if a plant cell has been transformed. A general review of suitable markers, for example for the members of the grass family, is found in Wilmink and Dons, 1993, Plant Mol. Biol.
Reptr, 11(2):165-185.
Sequences suitable for permitting integration of the heterologous sequence into the plant genome are also recommended. These might include transposon sequences and the like for homologous recombination as well as Ti sequences which permit random insertion of a heterologous expression cassette into a plant genome. Suitable prokaryote selectable markers include resistance toward O antibiotics such as ampicillin or tetracycline. Other DNA sequences encoding additional functions C may also be present in the vector, as is known in the art.
O The nucleic acid molecules of the subject invention may be included into an expression cassette for C expression of the protein(s) of interest. Usually, there will be only one expression cassette, although 5 two or more are feasible. The recombinant expression cassette will contain in addition to the heterologous protein encoding sequence the following elements, a promoter region, plant 0 untranslated sequences, initiation codon depending upon whether or not the structural gene comes IN equipped with one, and a transcription and translation termination sequence. Unique restriction enzyme sites at the 5' and 3' ends of the cassette allow for easy insertion into a pre-existing vector.
Cl 10 A heterologous coding sequence may be for any protein relating to the present invention. The Ssequence encoding the protein of interest will encode a signal peptide which allows processing and 0 translocation of the protein, as appropriate, and will usually lack any sequence which might result in the binding of the desired protein of the invention to a membrane. Since, for the most part, the transcriptional initiation region will be for a gene which is expressed and translocated during germination, by employing the signal peptide which provides for translocation, one may also provide for translocation of the protein of interest. In this way, the protein(s) of interest will be translocated from the cells in which they are expressed and may be efficiently harvested. Typically secretion in seeds are across the aleurone or scutellar epithelium layer into the endosperm of the seed. While it is not required that the protein be secreted from the cells in which the protein is produced, this facilitates the isolation and purification of the recombinant protein.
Since the ultimate expression of the desired gene product will be in a eucaryotic cell it is desirable to determine whether any portion of the cloned gene contains sequences which will be processed out as introns by the host's splicosome machinery. If so, site-directed mutagenesis of the "intron" region may be conducted to prevent losing a portion of the genetic message as a false intron code, Reed and Maniatis, Cell41:95-105, 1985.
The vector can be microinjected directly into plant cells by use of micropipettes to mechanically transfer the recombinant DNA. Crossway, Mol. Gen. Genet, 202:179-185, 1985. The genetic material may also be transferred into the plant cell by using polyethylene glycol, Krens, et al., Nature, 296, 72- 74, 1982. Another method of introduction of nucleic acid segments is high velocity ballistic penetration by small particles with the nucleic acid either within the matrix of small beads or particles, or on the surface, Klein, et al., Nature, 327, 70-73, 1987 and Knudsen and Muller, 1991, Planta, 185:330-336 teaching particle bombardment of barley endosperm to create transgenic barley. Yet another method of introduction would be fusion of protoplasts with other entities, either minicells, cells, lysosomes or other fusible lipid-surfaced bodies, Fraley, et al., Proc. Natl. Acad. Sci. USA, 79, 1859-1863, 1982.
The vector may also be introduced into the plant cells by electroporation. (Fromm et al., Proc. Natl Acad. Sci. USA 82:5824, 1985). In this technique, plant protoplasts are electroporated in the presence of plasmids containing the gene construct. Electrical impulses of high field strength reversibly permeabilize biomembranes allowing the introduction of the plasmids. Electroporated plant protoplasts reform the cell wall, divide, and form plant callus.
O All plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be C transformed by the present invention so that whole plants are recovered which contain the transferred gene. It is known that practically all plants can be regenerated from cultured cells or tissues, including O but not limited to all major species of sugarcane, sugar beet, cotton, fruit and other trees, legumes and vegetables. Some suitable plants include, for example, species from the genera Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersion, 00 00 Nicotiana, Solanum, Petunia, Digitalis, Majorana, Cichorium, Helianthus, Lactuca, Bromus, SAsparagus, Antirrhinum, Hererocallis, Nemesia, Pelargonium, Panicum, Pennisetum, Ranunculus, c 10 Senecio, Salpiglossis, Cucumis, Browaalia, Glycine, Lolium, Zea, Triticum, Sorghum, and Datura.
SMeans for regeneration vary from species to species of plants, but generally a suspension of Stransformed protoplasts containing copies of the heterologous gene is first provided. Callus tissue is formed and shoots may be induced from callus and subsequently rooted. Alternatively, embryo formation can be induced from the protoplast suspension. These embryos germinate as natural embryos to form plants. The culture media will generally contain various amino acids and hormones, such as auxin and cytokinins. It is also advantageous to add glutamic acid and proline to the medium, especially for such species as corn and alfalfa. Shoots and roots normally develop simultaneously.
Efficient regeneration will depend on the medium, on the genotype, and on the history of the culture.
If these three variables are controlled, then regeneration is fully reproducible and repeatable.
In some plant cell culture systems, the desired protein of the invention may be excreted or alternatively, the protein may be extracted from the whole plant. Where the desired protein of the invention is secreted into the medium, it may be collected. Alternatively, the embryos and embryolesshalf seeds or other plant tissue may be mechanically disrupted to release any secreted protein between cells and tissues. The mixture may be suspended in a buffer solution to retrieve soluble proteins.
Conventional protein isolation and purification methods will be then used to purify the recombinant protein. Parameters of time, temperature pH, oxygen, and volumes will be adjusted through routine methods to optimize expression and recovery of heterologous protein.
iv. Bacterial Systems Bacterial expression techniques are known in the art. A bacterial promoter is any DNA sequence capable of binding bacterial RNA polymerase and initiating the downstream transcription of a coding sequence structural gene) into mRNA. A promoter will have a transcription initiation region which is usually placed proximal to the 5' end of the coding sequence. This transcription initiation region usually includes an RNA polymerase binding site and a transcription initiation site. A bacterial promoter may also have a second domain called an operator, that may overlap an adjacent RNA polymerase binding site at which RNA synthesis begins. The operator permits negative regulated (inducible) transcription, as a gene repressor protein may bind the operator and thereby inhibit transcription of a specific gene. Constitutive expression may occur in the absence of negative regulatory elements, such as the operator. In addition, positive regulation may be achieved by a gene activator protein binding sequence, which, if present is usually proximal to the RNA polymerase binding sequence. An example of a gene activator protein is the catabolite activator protein (CAP), which helps initiate transcription of the lac operon in Escherichia coli (E.coli) {Raibaud et al. (1984) -21- 0 Annu. Rev. Genet. 18:173). Regulated expression may therefore be either positive or negative, thereby r<l either enhancing or reducing transcription.
O Sequences encoding metabolic pathway enzymes provide particularly useful promoter sequences.
Examples include promoter sequences derived from sugar metabolizing enzymes, such as galactose, 5 lactose (lac) {Chang et al. (1977) Nature 198:1056}, and maltose. Additional examples include promoter sequences derived from biosynthetic enzymes such as tryptophan (trp) {Goeddel et al.
o0 (1980) Nuc. Acids Res. 8:4057; Yelverton et al. (1981) Nucl. Acids Res. 9:731; US patent 4,738,921; kDO EP-A-0036776 and EP-A-0121775}. The g-laotamase (bla) promoter system {Weissmann (1981) "The cloning of interferon and other mistakes." In Interferon 3 (ed. I. Gresser)}, bacteriophage lambda r n 10 PL {Shimatake et al. (1981) Nature 292:128} and T5 {US patent 4,689,406} promoter systems also provide useful promoter sequences.
0In addition, synthetic promoters which do not occur in nature also function as bacterial promoters. For example, transcription activation sequences of one bacterial or bacteriophage promoter may be joined with the operon sequences of another bacterial or bacteriophage promoter, creating a synthetic hybrid promoter {US patent 4,551,433}. For example, the tac promoter is a hybrid trp-lac promoter comprised of both trp promoter and lac operon sequences that is regulated by the lac repressor {Amann et al. (1983) Gene 25:167; de Boer et al. (1983) Proc. Natl. Acad. Sci. 80:21}. Furthermore, a bacterial promoter can include naturally occurring promoters of non-bacterial origin that have the ability to bind bacterial RNA polymerase and initiate transcription. A naturally occurring promoter of non-bacterial origin can also be coupled with a compatible RNA polymerase to produce high levels of expression of some genes in prokaryotes. The bacteriophage T7 RNA polymerase/promoter system is an example of a coupled promoter system {Studier et al. (1986) J. Mol. Biol. 189:113; Tabor et al.
(1985) Proc Natl. Acad. Sci. 82:1074}. In addition, a hybrid promoter can also be comprised of a bacteriophage promoter and an E.coli operator region (EPO-A-0 267 851).
In addition to a functioning promoter sequence, an efficient ribosome binding site is also useful for the expression of foreign genes in prokaryotes. In E.coli, the ribosome binding site is called the Shine- Dalgarno (SD) sequence and includes an initiation codon (ATG) and a sequence 3-9 nucleotides in length located 3-11 nucleotides upstream of the initiation codon {Shine et al. (1975) Nature 254:34}.
The SD sequence is thought to promote binding of mRNA to the ribosome by the pairing of bases between the SD sequence and the 3' and of E.coli 16S rRNA {Steitz et al. (1979) "Genetic signals and nucleotide sequences in messenger RNA." In Biological Regulation and Development: Gene Expression (ed. R.F. Goldberger)}. To express eukaryotic genes and prokaryotic genes with weak ribosome-binding site {Sambrook et al. (1989) "Expression of cloned genes in Escherichia coli." In Molecular Cloning: A Laboratory Manual}.
A DNA molecule may be expressed intracellularly. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus will always be a methionine, which is encoded by the ATG start codon. If desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide or by either in vivo on in vitro incubation with a bacterial methionine N-terminal peptidase (EPO-A-0 219 237).
-22- O Fusion proteins provide an alternative to direct expression. Usually, a DNA sequence encoding the N- C terminal portion of an endogenous bacterial protein, or other stable protein, is fused to the 5' end of heterologous coding sequences. Upon expression, this construct will provide a fusion of the two amino O acid sequences. For example, the bacteriophage lambda cell gene can be linked at the 5' terminus of a foreign gene and expressed in bacteria. The resulting fusion protein preferably retains a site for a processing enzyme (factor Xa) to cleave the bacteriophage protein from the foreign gene {Nagai et al.
(1984) Nature 309:810}. Fusion proteins can also be made with sequences from the lacZ {Jia et al.
O (1987) Gene 60:197}, trpE {Allen et al. (1987) J. Biotechnol. 5:93; Makoff et al. (1989) J. Gen.
Microbiol. 135:11 and Chey {EP-A-0 324 647} genes. The DNA sequence at the junction of the two 10 amino acid sequences may or may not encode a cleavable site. Another example is a ubiquitin fusion protein. Such a fusion protein is made with the ubiquitin region that preferably retains a site for a O processing enzyme ubiquitin specific processing-protease) to cleave the ubiquitin from the Cg foreign protein. Through this method, native foreign protein can be isolated {Miller et al. (1989) Bio/Technology 7:698 Alternatively, foreign proteins can also be secreted from the cell by creating chimeric DNA molecules that encode a fusion protein comprised of a signal peptide sequence fragment that provides for secretion of the foreign protein in bacteria {US patent 4,336,336}. The signal sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell. The protein is either secreted into the growth media (gram-positive bacteria) or into the periplasmic space, located between the inner and outer membrane of the cell (gramnegative bacteria). Preferably there are processing sites, which can be cleaved either in vivo or in vitro encoded between the signal peptide fragment and the foreign gene.
DNA encoding suitable signal sequences can be derived from genes for secreted bacterial proteins, such as the E.coli outer membrane protein gene (ompA) {Masui et al. (1983), in: Experimental Manipulation of Gene Expression; Ghrayeb et al. (1984) EMBO J. 3:2437} and the E.coli alkaline phosphatase signal sequence (phoA) {Oka et al. (1985) Proc. Natl. Acad. Sci. 82:7212}. As an additional example, the signal sequence of the alpha-amylase gene from various Bacillus strains can be used to secrete heterologous proteins from B. subtilis {Palva et al. (1982) Proc. Natl. Acad. Sci.
USA 79:5582; EP-A-0 244 042}.
Usually, transcription termination sequences recognized by bacteria are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Transcription termination sequences frequently include DNA sequences of about nucleotides capable of forming stem loop structures that aid in terminating transcription. Examples include transcription termination sequences derived from genes with strong promoters, such as the trp gene in E.coli as well as other biosynthetic genes.
Usually, the above described components, comprising a promoter, signal sequence (if desired), coding sequence of interest, and transcription termination sequence, are put together into expression constructs. Expression constructs are often maintained in a replicon, such as an extrachromosomal element plasmids) capable of stable maintenance in a host, such as bacteria. The replicon will have a replication system, thus allowing it to be maintained in a prokaryotic host either for expression O or for cloning and amplification. In addition, a replicon may be either a high or low copy number plasmid. A high copy number plasmid will generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150. A host containing a high copy number plasmid will O preferably contain at least about 10, and more preferably at least about 20 plasmids. Either a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host.
o Alternatively, the expression constructs can be integrated into the bacterial genome with an integrating O vector. Integrating vectors usually contain at least one sequence homologous to the bacterial chromosome that allows the vector to integrate. Integrations appear to result from recombinations n 10 between homologous DNA in the vector and the bacterial chromosome. For example, integrating vectors constructed with DNA from various Bacillus strains integrate into the Bacillus chromosome (EP-A- 0 127 328). Integrating vectors may also be comprised of bacteriophage or transposon sequences.
Usually, extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of bacterial strains that have been transformed. Selectable markers can be expressed in the bacterial host and may include genes which render bacteria resistant to drugs such as ampicillin, chloramphenicol, erythromycin, kanamycin (neomycin), and tetracycline {Davies et al.
(1978) Annu. Rev. Microbiol. 32:469}. Selectable markers may also include biosynthetic genes, such as those in the histidine, tryptophan, and leucine biosynthetic pathways.
Alternatively, some of the above described components can be put together in transformation vectors.
Transformation vectors are usually comprised of a selectable market that is either maintained in a replicon or developed into an integrating vector, as described above.
Expression and transformation vectors, either extra-chromosomal replicons or integrating vectors, have been developed for transformation into many bacteria. For example, expression vectors have been developed for, inter alia, the following bacteria: Bacillus subtilis {Palva et al. (1982) Proc. Natl.
Acad. Sci. USA 79:5582; EP-A-0 036 259 and EP-A-0 063 953; WO 84/04541}, Escherichia coli {Shimatake et al. (1981) Nature 292:128; Amann et al. (1985) Gene 40:183; Studier et al. (1986) J.
Mol. Biol. 189:113; EP-A-0 036 776,EP-A-0 136 829 and EP-A-0 136 907}, Streptococcus cremoris {Powell et al. (1988) Appl. Environ. Microbiol. 54:655); Streptococcus lividans {Powell et al. (1988) Appl. Environ. Microbiol. 54:655}, Streptomyces lividans {US patent 4,745,056}.
Methods of introducing exogenous DNA into bacterial hosts are well-known in the art, and usually include either the transformation of bacteria treated with CaCI 2 or other agents, such as divalent cations and DMSO. DNA can also be introduced into bacterial cells by electroporation.
Transformation procedures usually vary with the bacterial species to be transformed. See e.g. {Masson et al. (1989) FEMS Microbiol. Lett. 60:273; Palva et al. (1982) Proc. Natl. Acad. Sci. USA 79:5582; EP-A-0 036 259 and EP-A-0 063 953; WO 84/04541, Bacillus}, {Miller et al. (1988) Proc. Natl.
Acad. Sci. 85:856; Wang et al. (1990) J. Bacteriol. 172:949, Campylobacter}, {Cohen et al. (1973) Proc. Natl. Acad. Sci. 69:2110; Dower et al. (1988) Nucleic Acids Res. 16:6127; Kushner (1978) "An improved method for transformation of Escherichia coli with ColEl-derived plasmids. In Genetic Engineering: Proceedings of the International Symposium on Genetic Engineering (eds. H.W. Boyer 0 and S. Nicosia); Mandel et al. (1970) J. Mol. Biol. 53:159; Taketo (1988) Biochim. Biophys. Acta 949:318; Escherichia}, {Chassy et al. (1987) FEMS Microbiol. Lett. 44:173 Lactobacillus}; {Fiedler et al. (1988) Anal. Biochem 170:38, Pseudomonas}; {Augustin et al. (1990) FEMS Microbiol. Lett.
O 66:203, Staphylococcus}, {Barany et al. (1980) J. Bacteriol. 144:698; Harlander (1987) "Transformation of Streptococcus lactis by electroporation, in: Streptococcal Genetics (ed. J. Ferretti and R. Curtiss III); Perry et al. (1981) Infect. Immun. 32:1295; Powell et al. (1988) Appl. Environ.
Microbiol. 54:655; Somkuti etal. (1987) Proc. 4th Evr. Cong. Biotechnology 1:412, Streptococcus}.
00 I v. Yeast Expression SYeast expression systems are also known to one of ordinary skill in the art. A yeast promoter is any C1 10 DNA sequence capable of binding yeast RNA polymerase and initiating the downstream transcription of a coding sequence structural gene) into mRNA. A promoter will have a Stranscription initiation region which is usually placed proximal to the 5' end of the coding sequence.
This transcription initiation region usually includes an RNA polymerase binding site (the "TATA Box") and a transcription initiation site. A yeast promoter may also have a second domain called an upstream activator sequence (UAS), which, if present, is usually distal to the structural gene. The UAS permits regulated (inducible) expression. Constitutive expression occurs in the absence of a UAS.
Regulated expression may be either positive or negative, thereby either enhancing or reducing transcription.
Yeast is a fermenting organism with an active metabolic pathway, therefore sequences encoding enzymes in the metabolic pathway provide particularly useful promoter sequences. Examples include alcohol dehydrogenase (ADH) (EP-A-0 284 044), enolase, glucokinase, glucose-6-phosphate isomerase, glyceraldehyde-3-phosphate-dehydrogenase (GAP or GAPDH), hexokinase, phosphofructokinase, 3-phosphoglycerate mutase, and pyruvate kinase (PyK) (EPO-A-0 329 203).
The yeast PH05 gene, encoding acid phosphatase, also provides useful promoter sequences {Myanohara etal. (1983) Proc. Natl. Acad. Sci. USA 80:1}.
In addition, synthetic promoters which do not occur in nature also function as yeast promoters. For example, UAS sequences of one yeast promoter may be joined with the transcription activation region of another yeast promoter, creating a synthetic hybrid promoter. Examples of such hybrid promoters include the ADH regulatory sequence linked to the GAP transcription activation region (US Patent Nos. 4,876,197 and 4,880,734). Other examples of hybrid promoters include promoters which consist of the regulatory sequences of either the ADH2, GAL4, GALIO, OR PH05 genes, combined with the transcriptional activation region of a glycolytic enzyme gene such as GAP or PyK (EP-A-0 164 556).
Furthermore, a yeast promoter can include naturally occurring promoters of non-yeast origin that have the ability to bind yeast RNA polymerase and initiate transcription. Examples of such promoters include, inter alia, {Cohen et al. (1980) Proc. Natl. Acad. Sci. USA 77:1078; Henikoff et al. (1981) Nature 283:835; Hollenberg et al. (1981) Curr. Topics Microbiol. Immunol. 96:119; Hollenberg et al.
(1979) "The Expression of Bacterial Antibiotic Resistance Genes in the Yeast Saccharomyces cerevisiae," in: Plasmids of Medical, Environmental and Commercial Importance (eds. K.N. Timmis and A. Puhler); Mercerau-Puigalon et al. (1980) Gene 11:163; Panthier et al. (1980) Curr. Genet.
2:109;}.
O A DNA molecule may be expressed intracellularly in yeast. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If O desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide.
Fusion proteins provide an alternative for yeast expression systems, as well as in mammalian, oo baculovirus, and bacterial expression systems. Usually, a DNA sequence encoding the N-terminal IND portion of an endogenous yeast protein, or other stable protein, is fused to the 5' end of heterologous coding sequences. Upon expression, this construct will provide a fusion of the two amino acid 10 sequences. For example, the yeast or human superoxide dismutase (SOD) gene, can be linked at the terminus of a foreign gene and expressed in yeast. The DNA sequence at the junction of the two amino acid sequences may or may not encode a cleavable site. See e.g. EP-A-0 196 056. Another C1 example is a ubiquitin fusion protein. Such a fusion protein is made with the ubiquitin region that preferably retains a site for a processing enzyme ubiquitin-specific processing protease) to cleave the ubiquitin from the foreign protein. Through this method, therefore, native foreign protein can be isolated W088/024066).
Alternatively, foreign proteins can also be secreted from the cell into the growth media by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provide for secretion in yeast of the foreign protein. Preferably, there are processing sites encoded between the leader fragment and the foreign gene that can be cleaved either in vivo or in vitro. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell.
DNA encoding suitable signal sequences can be derived from genes for secreted yeast proteins, such as the genes for invertase (EP-A-0012873; JPO 62,096,086) and A-factor (US patent 4,588,684).
Alternatively, leaders of non-yeast origin exit, such as an interferon leader, that also provide for secretion in yeast (EP-A-0060057).
A preferred class of secretion leaders are those that employ a fragment of the yeast alpha-factor gene, which contains both a "pre" signal sequence, and a "pro" region. The types of alpha-factor fragments that can be employed include the full-length pre-pro alpha factor leader (about 83 amino acid residues) as well as truncated alpha-factor leaders (usually about 25 to about 50 amino acid residues) (US Patents 4,546,083 and 4,870,008; EP-A-0 324 274). Additional leaders employing an alpha-factor leader fragment that provides for secretion include hybrid alpha-factor leaders made with a presequence of a first yeast, but a pro-region from a second yeast alphafactor. see WO 89/02463.) Usually, transcription termination sequences recognized by yeast are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Examples of transcription terminator sequence and other yeast-recognized termination sequences, such as those coding for glycolytic enzymes.
Usually, the above described components, comprising a promoter, leader (if desired), coding sequence of interest, and transcription termination sequence, are put together into expression constructs.
-26- O Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g.
N, plasmids) capable of stable maintenance in a host, such as yeast or bacteria. The replicon may have two replication systems, thus allowing it to be maintained, for example, in yeast for expression and in O a prokaryotic host for cloning and amplification. Examples of such yeast-bacteria shuttle vectors include YEp24 {Botstein et al. (1979) Gene 8:17-24}, pCl/1 {Brake et al. (1984) Proc. Natl. Acad. Sci USA 81:4642-4646}, and YRpl7 {Stinchcomb et al. (1982) J. Mol. Biol. 158:157}. In addition, a replicon may be either a high or low copy number plasmid. A high copy number plasmid will 0 0 generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150.
-A host containing a high copy number plasmid will preferably have at least about 10, and more S 10 preferably at least about 20. Enter a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host. See e.g. Brake et al., supra.
Alternatively, the expression constructs can be integrated into the yeast genome with an integrating c I vector. Integrating vectors usually contain at least one sequence homologous to a yeast chromosome that allows the vector to integrate, and preferably contain two homologous sequences flanking the expression construct. Integrations appear to result from recombinations between homologous DNA in the vector and the yeast chromosome {Orr-Weaver et al. (1983) Methods in Enzymol. 101:228-245}.
An integrating vector may be directed to a specific locus in yeast by selecting the appropriate homologous sequence for inclusion in the vector. See Orr-Weaver et al., supra. One or more expression construct may integrate, possibly affecting levels of recombinant protein produced {Rine et al. (1983) Proc. Natl. Acad. Sci. USA 80:6750). The chromosomal sequences included in the vector can occur either as a single segment in the vector, which results in the integration of the entire vector, or two segments homologous to adjacent segments in the chromosome and flanking the expression construct in the vector, which can result in the stable integration of only the expression construct.
Usually, extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of yeast strains that have been transformed. Selectable markers may include biosynthetic genes that can be expressed in the yeast host, such as ADE2, HIS4, LEU2, TRPI, and ALG7, and the G418 resistance gene, which confer resistance in yeast cells to tunicamycin and G418, respectively. In addition, a suitable selectable marker may also provide yeast with the ability to grow in the presence of toxic compounds, such as metal. For example, the presence of CUP1 allows yeast to grow in the presence of copper ions {Butt et al. (1987) Microbiol, Rev. 51:351 Alternatively, some of the above described components can be put together into transformation vectors. Transformation vectors are usually comprised of a selectable marker that is either maintained in a replicon or developed into an integrating vector, as described above.
Expression and transformation vectors, either extrachromosomal replicons or integrating vectors, have been developed for transformation into many yeasts. For example, expression vectors have been developed for, inter alia, the following yeasts:Candida albicans {Kurtz, et al. (1986) Mol. Cell. Biol.
6:142}, Candida maltosa {Kunze, et al. (1985) J. Basic Microbiol. 25:141}. Hansenula polymorpha {Gleeson, et al. (1986) J. Gen. Microbiol. 132:3459; Roggenkamp et al. (1986) Mol. Gen. Genet.
202:302}, Kluyveromyces fragilis {Das, et al. (1984) J. Bacteriol. 158:1165}, Kluyveromyces lactis {De Louvencourt et al. (1983) J. Bacteriol. 154:737; Van den Berg et al. (1990) Bio/Technology 8:135}, Pichia guillerimondii {Kunze et al. (1985) J. Basic Microbiol. 25:141}, Pichia pastoris -27- O {Cregg, et al. (1985) Mol. Cell. Biol. 5:3376; US Patent Nos. 4,837,148 and 4,929,555}, C, Saccharomyces cerevisiae {Hinnen et al. (1978) Proc. Natl. Acad. Sci. USA 75:1929; Ito et al. (1983) J. Bacteriol. 153:163}, Schizosaccharomyces pombe {Beach and Nurse (1981) Nature 300:706}, and O Yarrowia lipolytica {Davidow, et al. (1985) Curr. Genet. 10:380471 Gaillardin, et al. (1985) Curr.
Genet. 10:49}.
Methods of introducing exogenous DNA into yeast hosts are well-known in the art, and usually include either the transformation of spheroplasts or of intact yeast cells treated with alkali cations.
Transformation procedures usually vary with the yeast species to be transformed. See e.g. {Kurtz et al.
(1986) Mol. Cell. Biol. 6:142; Kunze et al. (1985) J. Basic Microbiol. 25:141; Candida}; {Gleeson et S 10 al. (1986) J. Gen. Microbiol. 132:3459; Roggenkamp et al. (1986) Mol. Gen. Genet. 202:302; Hansenula}; {Das et al. (1984) J. Bacteriol. 158:1165; De Louvencourt et al. (1983) J. Bacteriol.
154:1165; Van den Berg et al. (1990) Bio/Technology 8:135; Kluyveromyces}; {Cregg et al. (1985) r Mol. Cell. Biol. 5:3376; Kunze et al. (1985) J. Basic Microbiol. 25:141; US Patents 4,837,148 4,929,555; Pichia}; {Hinnen et al. (1978) Proc. Natl. Acad. Sci. USA 75;1929; Ito et al. (1983) J.
Bacteriol. 153:163 Saccharomyces}; {Beach Nurse (1981) Nature 300:706; Schizosaccharomyces}; {Davidow et al. (1985) Curr. Genet. 10:39; Gaillardin et al. (1985) Curr. Genet. 10:49; Yarrowia}.
Pharmaceutical Compositions Pharmaceutical compositions can comprise polypeptides and/or nucleic acid of the invention. The pharmaceutical compositions will comprise a therapeutically effective amount of either polypeptides, antibodies, or polynucleotides of the claimed invention.
The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect. The effect can be detected by, for example, chemical markers or antigen levels.
Therapeutic effects also include reduction in physical symptoms, such as decreased body temperature.
The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by routine experimentation and is within the judgement of the clinician.
For purposes of the present invention, an effective dose will be from about 0.01 mg/ kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the DNA constructs in the individual to which it is administered.
A pharmaceutical composition can also contain a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents. The term refers to any pharmaceutical carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Such carriers are well known to those of ordinary skill in the art.
O Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of O pharmaceutically acceptable excipients is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
¢€3 Pharmaceutically acceptable carriers in therapeutic compositions may contain liquids such as water, oo saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, 11pH buffering substances, and the like, may be present in such vehicles. Typically, the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable 10 for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. Liposomes are included within the definition of a pharmaceutically acceptable carrier.
Delivery Methods Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be animals; in particular, human subjects can be treated.
Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion. Other modes of administration include oral and pulmonary administration, suppositories, and transdermal or transcutaneous applications see WO98/20734), needles, and gene guns or hyposprays. Dosage treatment may be a single dose schedule or a multiple dose schedule.
Vaccines Vaccines according to the invention may either be prophylactic (ie. to prevent infection) or therapeutic (ie. to treat disease after infection).
Such vaccines comprise immunising antigen(s), immunogen(s), polypeptide(s), protein(s) or nucleic acid, usually in combination with "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 metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Additionally, these carriers may function as immunostimulating agents ("adjuvants"). Furthermore, the antigen or immunogen may be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, etc.
pathogens.
Preferred adjuvants to enhance effectiveness of the composition include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc; (2) oil-in-water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components), such as for example MF59 T M (WO 90/14837; Chapter 10 in Vaccine design. the subunit and adjuvant approach, eds. Powell Newman, Plenum Press 1995), containing 5% Squalene, 0.5% Tween 80, and 0.5% Span (optionally containing various amounts of MTP-PE (see below), although not required) formulated -29- O into submicron particles using a microfluidizer such as Model 1 OY microfluidizer (Microfluidics, Newton, MA), SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer LI21, and thr-MDP (see below) either microfluidized into a submicron emulsion or vortexed to O generate a larger particle size emulsion, and RibiTM adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL CWS (Detox
TM
saponin adjuvants, such O as StimulonTM (Cambridge Bioscience, Worcester, MA) may be used or particles generated therefrom r_ such as ISCOMs (immunostimulating complexes); Complete Freund's Adjuvant (CFA) and 10 Incomplete Freund's Adjuvant (IFA); cytokines, such as interleukins IL-1, IL-2, IL-4, IL-6, IL-7, IL-12, etc.), interferons gamma interferon), macrophage colony stimulating factor (M- O CSF), tumor necrosis factor (TNF), etc; and other substances that act as immunostimulating agents to enhance the effectiveness of the composition. Alum and MF59TM are preferred.
As mentioned above, muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl- D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), Nacetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-( 1 '-2'-dipalmitoyl-sn-glycero-3hydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
The immunogenic compositions the immunising antigen/immunogen/polypeptide/protein/ nucleic acid, pharmaceutically acceptable carrier, and adjuvant) typically will contain diluents, such as water, saline, glycerol, ethanol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
Typically, the immunogenic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation also may be emulsified or encapsulated in liposomes for enhanced adjuvant effect, as discussed above under pharmaceutically acceptable carriers.
Immunogenic compositions used as vaccines comprise an immunologically effective amount of the antigenic or immunogenic polypeptides, as well as any other of the above-mentioned 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, the taxonomic group of individual to be treated nonhuman primate, primate, etc.), the capacity of the individual's immune system to synthesize 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 immunogenic compositions are conventionally administered parenterally, e.g. by injection, either subcutaneously, intramuscularly, or transdermally/transcutaneously W098/20734). Additional formulations suitable for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Dosage treatment may be a single dose schedule or a multiple dose schedule. The vaccine may be administered in conjunction with other immunoregulatory agents.
1^ O As an alternative to protein-based vaccines, DNA vaccination may be employed Robinson Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al. (1997) Annu Rev Immunol 15:617- 648; see later herein}.
O Gene Delivery Vehicles 5 Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic of the invention, to be delivered to the mammal for expression in the mammal, can be administered either 00 locally or systemically. These constructs can utilize viral or non-viral vector approaches in in vivo or I ex vivo modality. Expression of such coding sequence can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence in vivo can be either constitutive or S 10 regulated.
The invention includes gene delivery vehicles capable of expressing the contemplated nucleic acid Ssequences. The gene delivery vehicle is preferably a viral vector and, more preferably, a retroviral, adenoviral, adeno-associated viral (AAV), herpes viral, or alphavirus vector. The viral vector can also be an astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picornavirus, poxvirus, or togavirus viral vector. See generally, Jolly (1994) Cancer Gene Therapy 1:51-64; Kimura (1994) Human Gene Therapy 5:845-852; Connelly (1995) Human Gene Therapy 6:185-193; and Kaplitt (1994) Nature Genetics 6:148-153.
Retroviral vectors are well known in the art and we contemplate that any retroviral gene therapy vector is employable in the invention, including B, C and D type retroviruses, xenotropic retroviruses (for example, NZB-X1, NZB-X2 and NZB9-1 (see O'Neill (1985) J. Virol. 53:160) polytropic retroviruses e.g. MCF and MCF-MLV (see Kelly (1983) J. Virol. 45:291), spumaviruses and lentiviruses. See RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985.
Portions of the retroviral gene therapy vector may be derived from different retroviruses. For example, retrovector LTRs may be derived from a Murine Sarcoma Virus, a tRNA binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus.
These recombinant retroviral vectors may be used to generate transduction competent retroviral vector particles by introducing them into appropriate packaging cell lines (see US patent 5,591,624).
Retrovirus vectors can be constructed for site-specific integration into host cell DNA by incorporation of a chimeric integrase enzyme into the retroviral particle (see W096/37626). It is preferable that the recombinant viral vector is a replication defective recombinant virus.
Packaging cell lines suitable for use with the above-described retrovirus vectors are well known in the art, are readily prepared (see W095/30763 and W092/05266), and can be used to create producer cell lines (also termed vector cell lines or "VCLs") for the production of recombinant vector particles.
Preferably, the packaging cell lines are made from human parent cells HT1080 cells) or mink parent cell lines, which eliminates inactivation in human serum.
Preferred retroviruses for the construction of retroviral gene therapy vectors include Avian Leukosis Virus, Bovine Leukemia, Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis Virus and Rous Sarcoma Virus. Particularly preferred Murine O Leukemia Viruses include 4070A and 1504A (Hartley and Rowe (1976) J Virol 19:19-25), Abelson C(ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi, Gross (ATCC Nol VR-590), Kirsten, Harvey Sarcoma Virus and Rauscher (ATCC No. VR-998) and Moloney Murine Leukemia Virus O (ATCC No. VR-190). Such retroviruses may be obtained from depositories or collections such as the American Type Culture Collection ("ATCC") in Rockville, Maryland or isolated from known sources using commonly available techniques.
o0 Exemplary known retroviral gene therapy vectors employable in this invention include those described 11 in patent applications GB2200651, EP0415731, EP0345242, EP0334301, W089/02468; W089/05349, W089/09271, W090/02806, W090/07936, W094/03622, W093/25698, 10 W093/25234, W093/11230, W093/10218, W091/02805, W091/02825, W095/07994, US 5,219,740, US 4,405,712, US 4,861,719, US 4,980,289, US 4,777,127, US 5,591,624. See also Vile (1993) Cancer Res 53:3860-3864; Vile (1993) Cancer Res 53:962-967; Ram (1993) Cancer Res 53 N (1993) 83-88; Takamiya (1992) J Neurosci Res 33:493-503; Baba (1993) J Neurosurg 79:729-735; Mann (1983) Cell 33:153; Cane (1984) Proc Natl Acad Sci 81:6349; and Miller (1990) Human Gene Therapy 1.
Human adenoviral gene therapy vectors are also known in the art and employable in this invention.
See, for example, Berkner (1988) Biotechniques 6:616 and Rosenfeld (1991) Science 252:431, and W093/07283, W093/06223, and W093/07282. Exemplary known adenoviral gene therapy vectors employable in this invention include those described in the above referenced documents and in W094/12649, W093/03769, W093/19191, W094/28938, W095/11984, W095/00655, W095/27071, W095/29993, W095/34671, W096/05320, W094/08026, W094/11506, W093/06223, W094/24299, W095/14102, W095/24297, W095/02697, W094/28152, W094/24299, W095/09241, W095/25807, W095/05835, W094/18922 and W095/09654.
Alternatively, administration of DNA linked to killed adenovirus as described in Curiel (1992) Hum.
Gene Ther. 3:147-154 may be employed. The gene delivery vehicles of the invention also include adenovirus associated virus (AAV) vectors. Leading and preferred examples of such vectors for use in this invention are the AAV-2 based vectors disclosed in Srivastava, W093/09239. Most preferred AAV vectors comprise the two AAV inverted terminal repeats in which the native D-sequences are modified by substitution of nucleotides, such that at least 5 native nucleotides and up to 18 native nucleotides, preferably at least 10 native nucleotides up to 18 native nucleotides, most preferably native nucleotides are retained and the remaining nucleotides of the D-sequence are deleted or replaced with non-native nucleotides. The native D-sequences of the AAV inverted terminal repeats are sequences of 20 consecutive nucleotides in each AAV inverted terminal repeat (ie. there is one sequence at each end) which are not involved in HP formation. The non-native replacement nucleotide may be any nucleotide other than the nucleotide found in the native D-sequence in the same position.
Other employable exemplary AAV vectors are pWP-19, pWN-1, both of which are disclosed in Nahreini (1993) Gene 124:257-262. Another example of such an AAV vector is psub201 (see Samulski (1987) J. Virol. 61:3096). Another exemplary AAV vector is the Double-D ITR vector.
Construction of the Double-D ITR vector is disclosed in US Patent 5,478,745. Still other vectors are those disclosed in Carter US Patent 4,797,368 and Muzyczka US Patent 5,139,941, Chartejee US Patent 5,474,935, and Kotin W094/288157. Yet a further example of an AAV vector employable in -32- O this invention is SSV9AFABTKneo, which contains the AFP enhancer and albumin promoter and N, directs expression predominantly in the liver. Its structure and construction are disclosed in Su (1996) Human Gene Therapy 7:463-470. Additional AAV gene therapy vectors are described in US O 5,354,678, US 5,173,414, US 5,139,941, and US 5,252,479.
The gene therapy vectors of the invention also include herpes vectors. Leading and preferred examples are herpes simplex virus vectors containing a sequence encoding a thymidine kinase polypeptide such as those disclosed in US 5,288,641 and EP0176170 (Roizman). Additional exemplary herpes simplex IsN virus vectors include HFEM/ICP6-LacZ disclosed in WO95/04139 (Wistar), pHSVlac described in SGeller (1988) Science 241:1667-1669 and in W090/09441 WO92/07945, HSV Us3::pgC-lacZ S 10 described in Fink (1992) Human Gene Therapy 3:11-19 and HSV 7134, 2 RH 105 and GAL4 described in EP 0453242 (Breakefield), and those deposited with ATCC as accession numbers ATCC VR-977 and ATCC VR-260.
Also contemplated are alpha virus gene therapy vectors that can be employed in this invention.
Preferred alpha virus vectors are Sindbis viruses vectors. Togaviruses, Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Middleberg virus (ATCC VR-370), Ross River virus (ATCC VR-373; ATCC VR-1246), Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532), and those described in US patents 5,091,309, 5,217,879, and W092/10578. More particularly, those alpha virus vectors described in US Serial No. 08/405,627, filed March 15, 1995,W094/21792, W092/10578, WO95/07994, US 5,091,309 and US 5,217,879 are employable. Such alpha viruses may be obtained from depositories or collections such as the ATCC in Rockville, Maryland or isolated from known sources using commonly available techniques.
Preferably, alphavirus vectors with reduced cytotoxicity are used (see USSN 08/679640).
DNA vector systems such as eukaryotic layered expression systems are also useful for expressing the nucleic acids of the invention. See W095/07994 for a detailed description of eukaryotic layered expression systems. Preferably, the eukaryotic layered expression systems of the invention are derived from alphavirus vectors and most preferably from Sindbis viral vectors.
Other viral vectors suitable for use in the present invention include those derived from poliovirus, for example ATCC VR-58 and those described in Evans, Nature 339 (1989) 385 and Sabin (1973) J. Biol.
Standardization 1:115; rhinovirus, for example ATCC VR-1110 and those described in Arnold (1990) J Cell Biochem L401; pox viruses such as canary pox virus or vaccinia virus, for example ATCC VR- 11 and ATCC VR-2010 and those described in Fisher-Hoch (1989) Proc Natl Acad Sci 86:317; Flexner (1989) Ann NY Acad Sci 569:86, Flexner (1990) Vaccine 8:17; in US 4,603,112 and US 4,769,330 and W089/01973; SV40 virus, for example ATCC VR-305 and those described in Mulligan (1979) Nature 277:108 and Madzak (1992) J Gen Virol 73:1533; influenza virus, for example ATCC VR-797 and recombinant influenza viruses made employing reverse genetics techniques as described in US 5,166,057 and in Enami (1990) Proc Natl Acad Sci 87:3802-3805; Enami Palese (1991) J Virol 65:2711-2713 and Luytjes (1989) Cell 59:110, (see also McMichael (1983) NEJ Med 309:13, and Yap (1978) Nature 273:238 and Nature (1979) 277:108); human immunodeficiency virus as described in EP-0386882 and in Buchschacher (1992) J. Virol. 66:2731; measles virus, for example ATCC VR-67 and VR-1247 and those described in EP-0440219; Aura virus, for example ATCC VR-368; Bebaru virus, for example ATCC VR-600 and ATCC VR-1240; OCabassou virus, for example ATCC VR-922; Chikungunya virus, for example ATCC VR-64 and ATCC VR-1241; Fort Morgan Virus, for example ATCC VR-924; Getah virus, for example ATCC VR-369 and ATCC VR-1243; Kyzylagach virus, for example ATCC VR-927; Mayaro virus, for O example ATCC VR-66; Mucambo virus, for example ATCC VR-580 and ATCC VR-1244; Ndumu virus, for example ATCC VR-371; Pixuna virus, for example ATCC VR-372 and ATCC VR-1245; Tonate virus, for example ATCC VR-925; Triniti virus, for example ATCC VR-469; Una virus, for example ATCC VR-374; Whataroa virus, for example ATCC VR-926; Y-62-33 virus, for example 0 0 ATCC VR-375; O'Nyong virus, Eastern encephalitis virus, for example ATCC VR-65 and ATCC SVR-1242; Western encephalitis virus, for example ATCC VR-70, ATCC VR-1251, ATCC VR-622 p. 10 and ATCC VR-1252; and coronavirus, for example ATCC VR-740 and those described in Hamre S(1966) Proc Soc Exp Biol Med 121:190.
Delivery of the compositions of this invention into cells is not limited to the above mentioned viral vectors. Other delivery methods and media may be employed such as, for example, nucleic acid expression vectors, polycationic condensed DNA linked or unlinked to killed adenovirus alone, for example see US Serial No. 08/366,787, filed December 30, 1994 and Curiel (1992) Hum Gene Ther 3:147-154 ligand linked DNA, for example see Wu (1989)JBiol Chem 264:16985-16987, eucaryotic cell delivery vehicles cells, for example see US Serial No.08/240,030, filed May 9, 1994, and US Serial No. 08/404,796, deposition of photopolymerized hydrogel materials, hand-held gene transfer particle gun, as described in US Patent 5,149,655, ionizing radiation as described in US5,206,152 and in WO92/11033, nucleic charge neutralization or fusion with cell membranes. Additional approaches are described in Philip (1994) Mol Cell Biol 14:2411-2618 and in Woffendin (1994) Proc Natl Acad Sci 91:1581-1585.
Particle mediated gene transfer may be employed, for example see US Serial No. 60/023,867. Briefly, the sequence can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, as described in Wu Wu (1987) J. Biol. Chem. 262:4429-4432, insulin as described in Hucked (1990) Biochem Pharmacol 40:253-263, galactose as described in Plank (1992) Bioconjugate Chem 3:533-539, lactose or transferrin.
Naked DNA may also be employed. Exemplary naked DNA introduction methods are described in W090/11092 and US 5,580,859. Uptake efficiency may be improved using biodegradable latex beads.
DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads.
The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption of the endosome and release of the DNA into the cytoplasm.
Liposomes that can act as gene delivery vehicles are described in US 5,422,120, WO95/13796, W094/23697, W091/14445 and EP-524,968. As described in USSN. 60/023,867, on non-viral delivery, the nucleic acid sequences encoding a polypeptide can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then be incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin. Other delivery systems include the use of liposomes to encapsulate DNA comprising the -34- O gene under the control of a variety of tissue-specific or ubiquitously-active promoters. Further C non-viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al (1994) Proc. Natl. Acad. Sci. USA 91(24):11581-11585. Moreover, the O coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials. Other conventional methods for gene delivery that can be used for delivery of the coding sequence include, for example, use of hand-held gene transfer particle gun, as described in US 5,149,655; use of ionizing radiation for activating transferred gene, as described in 0 0 US 5,206,152 and WO92/11033 Exemplary liposome and polycationic gene delivery vehicles are those described in US 5,422,120 and 10 4,762,915; in WO 95/13796; WO94/23697; and W091/14445; in EP-0524968; and in Stryer, SBiochemistry, pages 236-240 (1975) W.H. Freeman, San Francisco; Szoka (1980) Biochem Biophys Acta 600:1; Bayer (1979) Biochem Biophys Acta 550:464; Rivnay (1987) Meth Enzymol 149:119; c Nl Wang (1987) Proc Natl Acad Sci 84:7851; Plant (1989) Anal Biochem 176:420.
A polynucleotide composition can comprises therapeutically effective amount of a gene therapy vehicle, as the term is defined above. For purposes of the present invention, an effective dose will be from about 0.01 mg/ kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the DNA constructs in the individual to which it is administered.
Delivery Methods Once formulated, the polynucleotide compositions of the invention can be administered directly to the subject; delivered ex vivo, to cells derived from the subject; or in vitro for recombinant protein expression. The subjects to be treated can be mammals or birds. Also, human subjects can be treated.
Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion. Other modes of administration include oral and pulmonary administration, suppositories, and transdermal or transcutaneous applications see W098/20734), needles, and gene guns or hyposprays. Dosage treatment may be a single dose schedule or a multiple dose schedule.
Methods for the ex vivo delivery and reimplantation of transformed cells into a subject are known in the art and described in e.g. WO93/14778. Examples of cells useful in ex vivo applications include, for example, stem cells, particularly hematopoetic, lymph cells, macrophages, dendritic cells, or tumor cells.
Generally, delivery of nucleic acids for both ex vivo and in vitro applications can be accomplished by the following procedures, for example, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei, all well known in the art.
0 Polynucleotide and polopeptide pharmaceutical compositions In addition to the pharmaceutically acceptable carriers and salts described above, the following 0 additional agents can be used with polynucleotide and/or polypeptide compositions.
A.Polypeptides One example are polypeptides which include, without limitation: asioloorosomucoid (ASOR); transferrin; asialoglycoproteins; antibodies; antibody fragments; ferritin; interleukins; interferons, 00 0\0 granulocyte, macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor S(G-CSF), macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin. Viral Cantigens, such as envelope proteins, can also be used. Also, proteins from other invasive organisms, such as the 17 amino acid peptide from the circumsporozoite protein of plasmodium falciparum known as RII.
B.Hormones, Vitamins, etc.
Other groups that can be included are, for example: hormones, steroids, androgens, estrogens, thyroid hormone, or vitamins, folic acid.
C.Polvalkylenes. Polysaccharides, etc.
Also, polyalkylene glycol can be included with the desired polynucleotides/polypeptides. In a preferred embodiment, the polyalkylene glycol is polyethlylene glycol. In addition, mono-, di-, or polysaccharides can be included. In a preferred embodiment of this aspect, the polysaccharide is dextran or DEAE-dextran. Also, chitosan and poly(lactide-co-glycolide) D.Lipids. and Liposomes The desired polynucleotide/polypeptide can also be encapsulated in lipids or packaged in liposomes prior to delivery to the subject or to cells derived therefrom.
Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid. The ratio of condensed polynucleotide to lipid preparation can vary but will generally be around 1:1 (mg DNA:micromoles lipid), or more of lipid. For a review of the use of liposomes as carriers for delivery of nucleic acids, see, Hug and Sleight (1991) Biochim. Biophys.
Acta. 1097:1-17; Straubinger (1983) Meth. Enzymol. 101:512-527.
Liposomal preparations for use in the present invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner (1987) Proc. Natl. Acad. Sci. USA 84:7413-7416); mRNA (Malone (1989) Proc. Natl. Acad. Sci. USA 86:6077-6081); and purified transcription factors (Debs (1990) J. Biol. Chem. 265:10189-10192), in functional form.
Cationic liposomes are readily available. For example, N{l-2,3-dioleyloxy)propyl}-N,N,Ntriethylammonium (DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, NY. (See, also, Felgner supra). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger). Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. Szoka 0 (1978) Proc. Natl. Acad. Sci. USA 75:4194-4198; W090/11092 for a description of the synthesis of DOTAP (1, 2 -bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes.
0 Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids C (Birmingham, AL), or can be easily prepared using readily available materials. Such materials include M 5 phosphatidyl choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others.
o0 These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate sNO ratios. Methods for making liposomes using these materials are well known in the art.
SThe liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUVs), or C=I 10 large unilamellar vesicles (LUVs). The various liposome-nucleic acid complexes are prepared using methods known in the art. See e.g. Straubinger (1983) Meth. Immunol. 101:512-527; Szoka (1978) SProc. Natl. Acad. Sci. USA 75:4194-4198; Papahadjopoulos (1975) Biochim. Biophys. Acta 394:483; Wilson (1979) Cell 17:77); Deamer Bangham (1976) Biochim. Biophys. Acta 443:629; Ostro (1977) Biochem. Biophys. Res. Commun. 76:836; Fraley (1979) Proc. Natl. Acad. Sci. USA 76:3348); Enoch Strittmatter (1979) Proc. Natl. Acad. Sci. USA 76:145; Fraley (1980) J. Biol. Chem. (1980) 255:10431; Szoka Papahadjopoulos (1978) Proc. Natl. Acad. Sci. USA 75:145; and Schaefer-Ridder (1982) Science 215:166.
E.Lipoproteins In addition, lipoproteins can be included with the polynucleotide/polypeptide to be delivered.
Examples of lipoproteins to be utilized include: chylomicrons, HDL, IDL, LDL, and VLDL. Mutants, fragments, or fusions of these proteins can also be used. Also, modifications of naturally occurring lipoproteins can be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors. Preferably, if lipoproteins are including with the polynucleotide to be delivered, no other targeting ligand is included in the composition.
Naturally occurring lipoproteins comprise a lipid and a protein portion. The protein portion are known as apoproteins. At the present, apoproteins A, B, C, D, and E have been isolated and identified. At least two of these contain several proteins, designated by Roman numerals, AI, AII, AIV; CI, CII,
CIII.
A lipoprotein can comprise more than one apoprotein. For example, naturally occurring chylomicrons comprises of A, B, C, E, over time these lipoproteins lose A and acquire C and E apoproteins.
VLDL comprises A, B, C, E apoproteins, LDL comprises apoprotein B; HDL comprises apoproteins A, C, E.
The amino acid of these apoproteins are known and are described in, for example, Breslow (1985) Annu Rev. Biochem 54:699; Law (1986) Adv. Exp Med. Biol. 151:162; Chen (1986) J Biol Chem 261:12918; Kane (1980) Proc Natl Acad Sci USA 77:2465; and Utermann (1984) Hum Genet 65:232.
Lipoproteins contain a variety of lipids including, triglycerides, cholesterol (free and esters), and phospholipids. The composition of the lipids varies in naturally occurring lipoproteins. For example, chylomicrons comprise mainly triglycerides. A more detailed description of the lipid content of naturally occurring lipoproteins can be found, for example, in Meth. Enzymol. 128 (1986). The 1^ O composition of the lipids are chosen to aid in conformation of the apoprotein for receptor binding N activity. The composition of lipids can also be chosen to facilitate hydrophobic interaction and association with the polynucleotide binding molecule.
O Naturally occurring lipoproteins can be isolated from serum by ultracentrifugation, for instance. Such methods are described in Meth. Enzymol. (supra); Pitas (1980) J. Biochem. 255:5454-5460 and Mahey (1979) J Clin. Invest 64:743-750. Lipoproteins can also be produced by in vitro or recombinant 00 methods by expression of the apoprotein genes in a desired host cell. See, for example, Atkinson I (1986) Annu Rev Biophys Chem 15:403 and Radding (1958) Biochim Biophys Acta 30: 443.
Lipoproteins can also be purchased from commercial suppliers, such as Biomedical Techniologies, Inc., Stoughton, Massachusetts, USA. Further description of lipoproteins can be found in Zuckermann et al. PCT/US97/14465.
F.Polycationic Agents Polycationic agents can be included, with or without lipoprotein, in a composition with the desired polynucleotide/polypeptide to be delivered.
Polycationic agents, typically, exhibit a net positive charge at physiological relevant pH and are capable of neutralizing the electrical charge of nucleic acids to facilitate delivery to a desired location.
These agents have both in vitro, ex vivo, and in vivo applications. Polycationic agents can be used to deliver nucleic acids to a living subject either intramuscularly, subcutaneously, etc.
The following are examples of useful polypeptides as polycationic agents: polylysine, polyarginine, polyornithine, and protamine. Other examples include histones, protamines, human serum albumin, DNA binding proteins, non-histone chromosomal proteins, coat proteins from DNA viruses, such as (X174, transcriptional factors also contain domains that bind DNA and therefore may be useful as nucleic aid condensing agents. Briefly, transcriptional factors such as C/CEBP, c-jun, c-fos, AP-I, AP-2, AP-3, CPF, Prot-1, Sp-1, Oct-1, Oct-2, CREP, and TFIID contain basic domains that bind DNA sequences.
Organic polycationic agents include: spermine, spermidine, and purtrescine.
The dimensions and of the physical properties of a polycationic agent can be extrapolated from the list above, to construct other polypeptide polycationic agents or to produce synthetic polycationic agents.
Synthetic polycationic agents which are useful include, for example, DEAE-dextran, polybrene.
Lipofectin and lipofectAMINE T are monomers that form polycationic complexes when combined with polynucleotides/polypeptides.
Nucleic Acid Hybridisation "Hybridization" refers to the association of two nucleic acid sequences to one another by hydrogen bonding. Typically, one sequence will be fixed to a solid support and the other will be free in solution.
Then, the two sequences will be placed in contact with one another under conditions that favor hydrogen bonding. Factors that affect this bonding include: the type and volume of solvent; reaction temperature; time of hybridization; agitation; agents to block the non-specific attachment of the liquid phase sequence to the solid support (Denhardt's reagent or BLOTTO); concentration of the sequences; -38- O use of compounds to increase the rate of association of sequences (dextran sulfate or polyethylene C"i glycol); and the stringency of the washing conditions following hybridization. See Sambrook et al.
{supra} vol.2, chapt.9, pp.9.47 to 9.57.
O "Stringency" refers to conditions in a hybridization reaction that favor association of very similar sequences over sequences that differ. For example, the combination of temperature and salt concentration should be chosen that is approximately 120 to 200 0 C below the calculated Tm of the 00 hybrid under study. The temperature and salt conditions can often be determined empirically in preliminary experiments in which samples of genomic DNA immobilized on filters are hybridized to the sequence of interest and then washed under conditions of different stringencies. See Sambrook et 10 al. at page 9.50.
(Ni Variables to consider when performing, for example, a Southern blot are the complexity of the SDNA being blotted and the homology between the probe and the sequences being detected. The total amount of the fragment(s) to be studied can vary a magnitude of 10, from 0.1 to l g for a plasmid or phage digest to 10 9 to 10 8 g for a single copy gene in a highly complex eukaryotic genome. For lower complexity polynucleotides, substantially shorter blotting, hybridization, and exposure times, a smaller amount of starting polynucleotides, and lower specific activity of probes can be used. For example, a single-copy yeast gene can be detected with an exposure time of only 1 hour starting with 1 ig of yeast DNA, blotting for two hours, and hybridizing for 4-8 hours with a probe of 108 cpm/lg. For a single-copy mammalian gene a conservative approach would start with 10 jg of DNA, blot overnight, and hybridize overnight in the presence of 10% dextran sulfate using a probe of greater than 108 cpm/pg, resulting in an exposure time of ~24 hours.
Several factors can affect the melting temperature (Tm) of a DNA-DNA hybrid between the probe and the fragment of interest, and consequently, the appropriate conditions for hybridization and washing.
In many cases the probe is not 100% homologous to the fragment. Other commonly encountered variables include the length and total G+C content of the hybridizing sequences and the ionic strength and formamide content of the hybridization buffer. The effects of all of these factors can be approximated by a single equation: Tm= 81 16.6(logloCi) 0.4 C)}-0.6(%formamide) 600/n-1.5(%mismatch).
where Ci is the salt concentration (monovalent ions) and n is the length of the hybrid in base pairs (slightly modified from Meinkoth Wahl (1984) Anal. Biochem. 138: 267-284).
In designing a hybridization experiment, some factors affecting nucleic acid hybridization can be conveniently altered. The temperature of the hybridization and washes and the salt concentration during the washes are the simplest to adjust. As the temperature of the hybridization increases (ie.
stringency), it becomes less likely for hybridization to occur between strands that are nonhomologous, and as a result, background decreases. If the radiolabelled probe is not completely homologous with the immobilized fragment (as is frequently the case in gene family and interspecies hybridization experiments), the hybridization temperature must be reduced, and background will increase. The temperature of the washes affects the intensity of the hybridizing band and the degree of background in a similar manner. The stringency of the washes is also increased with decreasing salt concentrations.
O In general, convenient hybridization temperatures in the presence of 50% formamide are 42 0 C for a probe with is 95% to 100% homologous to the target fragment, 37'C for 90% to 95% homology, and O 32°C for 85% to 90% homology. For lower homologies, formamide content should be lowered and O temperature adjusted accordingly, using the equation above. If the homology between the probe and the target fragment are not known, the simplest approach is to start with both hybridization and wash conditions which are nonstringent. If non-specific bands or high background are observed after autoradiography, the filter can be washed at high stringency and reexposed. If the time required for 00 Sexposure makes this approach impractical, several hybridization and/or washing stringencies should be tested in parallel.
Nucleic Acid Probe Assays Methods such as PCR, branched DNA probe assays, or blotting techniques utilizing nucleic acid Sprobes according to the invention can determine the presence of cDNA or mRNA. A probe is said to "hybridize" with a sequence of the invention if it can form a duplex or double stranded complex, which is stable enough to be detected.
The nucleic acid probes will hybridize to the Chlamydial nucleotide sequences of the invention (including both sense and antisense strands). Though many different nucleotide sequences will encode the amino acid sequence, the native Chlamydial sequence is preferred because it is the actual sequence present in cells. mRNA represents a coding sequence and so a probe should be complementary to the coding sequence; single-stranded cDNA is complementary to mRNA, and so a cDNA probe should be complementary to the non-coding sequence.
The probe sequence need not be identical to the Chlamydial sequence (or its complement) some variation in the sequence and length can lead to increased assay sensitivity if the nucleic acid probe can form a duplex with target nucleotides, which can be detected. Also, the nucleic acid probe can include additional nucleotides to stabilize the formed duplex. Additional Chlamydial sequence may also be helpful as a label to detect the formed duplex. For example, a non-complementary nucleotide sequence may be attached to the 5' end of the probe, with the remainder of the probe sequence being complementary to a Chlamydial sequence. Alternatively, non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the a Chlamydial sequence in order to hybridize therewith and thereby form a duplex which can be detected.
The exact length and sequence of the probe will depend on the hybridization conditions, such as temperature, salt condition and the like. For example, for diagnostic applications, depending on the complexity of the analyte sequence, the nucleic acid probe typically contains at least 10-20 nucleotides, preferably 15-25, and more preferably >30 nucleotides, although it may be shorter than this. Short primers generally require cooler temperatures to form sufficiently stable hybrid complexes with the template.
Probes may be produced by synthetic procedures, such as the triester method of Matteucci et al. {J.
Am. Chem. Soc. (1981) 103:3185), or according to Urdea et al. {Proc. Natl. Acad. Sci. USA (1983) 7461 or using commercially available automated oligonucleotide synthesizers.
O The chemical nature of the probe can be selected according to preference. For certain applications, DNA or RNA are appropriate. For other applications, modifications may be incorporated e.g.
backbone modifications, such as phosphorothioates or methylphosphonates, can be used to increase in O vivo half-life, alter RNA affinity, increase nuclease resistance etc. see Agrawal Iyer (1995) Curr Opin Biotechnol 6:12-19; Agrawal (1996) TIBTECH 14:376-387}; analogues such as peptide nucleic acids may also be used see Corey (1997) TIBTECH 15:224-229; Buchardt et al. (1993) TIBTECH 11:384-386}.
00 0 Alternatively, the polymerase chain reaction (PCR) is another well-known means for detecting small Samounts of target nucleic acids. The assay is described in: Mullis et al. {Meth. Enzymol. (1987) 155: 335-350}; US patents 4,683,195 4,683,202. Two 'primers' hybridize with the target nucleic acids and are used to prime the reaction. The primers can comprise sequence that does not hybridize to the sequence of the amplification target (or its complement) to aid with duplex stability or, for example, to C 1 incorporate a convenient restriction site. Typically, such sequence will flank the desired Chlamydial sequence.
A thermostable polymerase creates copies of target nucleic acids from the primers using the original target nucleic acids as a template. After a threshold amount of target nucleic acids are generated by the polymerase, they can be detected by more traditional methods, such as Southern blots. When using the Southern blot method, the labelled probe will hybridize to the Chlamydial sequence (or its complement).
Also, mRNA or cDNA can be detected by traditional blotting techniques described in Sambrook et al {supra}. mRNA, or cDNA generated from mRNA using a polymerase enzyme, can be purified and separated using gel electrophoresis. The nucleic acids on the gel are then blotted onto a solid support, such as nitrocellulose. The solid support is exposed to a labelled probe and then washed to remove any unhybridized probe. Next, the duplexes containing the labelled probe are detected. Typically, the probe is labelled with a radioactive moiety.
BRIEF DESCRIPTION OF THE DRAWINGS Figures 1 to 44 show data from examples 1 to 44. Where a figure is of a gel, lane 1 is at the left of the figure.
For Western Blots, two samples were tested for each protein. The left lane in a pair used membrane strips stained with pre-immune sera whilst the right lane used membrane strips stained with immune sera. In the Western blots in figures 1 to 5, 35B, 37B, 38B and 39, markers are at 66, 45, 30, 20.1 and 14.4 kDa. In the Western blots in figures 6 to 16, 23C, 24D, 27E, 38A, 40, 41, 42 and 43 markers are at 172.6, 111.4, 79.6, 61.3, 49.0, 36.4, 24.7, 19.2 and 13.1 kDa.
In the Western Blots in figures 1 to 5, lanes 2 and 3 show control sera raised against GSTfusion control antigens. In the Western blots in figures 1 to 5, lanes 4 and 5 contain control sera raised against His-tagged control antigens.
Low molecular weight markers are run in lane 1 of the purification gels.
O MODES FOR CARRYING OUT THE INVENTION Table I gives the names of C.pneumoniae proteins from reference 18, the GenBank 0 accession numbers and titles for those proteins, the GenBank accession numbers and titles
O
for the corresponding C.trachomatis proteins of the invention, and SEQ ID numbers (SEQ C 5 IDs 1-262, with odd numbers being amino acid sequences and even numbers being nucleotide sequences) for these C.trachomatis proteins. These can be expressed and used in 00 the same ways as described in reference 18 for the corresponding C.pneumoniae proteins.
The C.trachomatis proteins are useful for diagnostic and immunogenic purposes. These C properties are not evident from the sequence alone.
Various tests can be used to assess the in vivo immunogenicity of the proteins of the invention. For example, the proteins can be expressed recombinantly and used to screen patient sera by immunoblot. A positive reaction between the protein and patient serum indicates that the patient has previously mounted an immune response to the protein in question i.e. the protein is an immunogen. This method can also be used to identify immunodominant proteins.
The recombinant protein can also be conveniently used to prepare antibodies e.g. in a mouse. These can be used for direct confirmation that a protein is located on the cell-surface of C.trachomatis by using the antibodies in a Western Blot against intact Chlamydia).
Labelled antibody fluorescent labelling for FACS) can be incubated with intact bacteria and the presence of label on the bacterial surface confirms the location of the protein. FACS figures show a scatter profile of the Chlamydia preparation used in the assay, the peak shift obtained when antibodies against the recombinant antigen bind to the Chlamydial cells (open area control sample; filled area antibody-reacted sample), quantitative Kolmogorov-Smimov statistical analysis, and output of the FACS analysis software.
Example 1 CT242 (SEQ ID 57 and SEQ ID 58) was expressed in E.coli. The recombinant product was purified both as a GST-fusion protein (Figure 1A; lanes 4 and 5, chromatography fractions 1 and 2, expected molecular weight 42.4 kDa) and as a His-tagged fusion protein (Figure 1B; lanes 2-4, chromatography fractions 1, 2 and 3, expected molecular weight 16.4 kDa).
The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 1C: His-tagged: lanes 12 and 13; GST-fusion: lanes 20 and 21). Lane 12 shows membrane strips stained with pre-immune sera for His-tagged CT242 whilst lane 13 shows membrane strips stained with immune sera for His-tagged CT242. Lane 20 shows -42- O membrane strips stained with preimmune sera for GST-fusion CT242 whilst lane 21 shows C1 membrane strips stained with immune sera for GST-fusion CT242.
O These experiments show that CT242 is a surface-exposed and immunoaccessible protein, _and that it is a useful immunogen. These properties are not evident from the sequence c 5 alone.
00 Example 2 SCT045 (SEQ ID 71 and SEQ ID 72) was expressed in E.coli. The recombinant product was CM purified as a His-tagged fusion protein (Figure 2A; lanes 4-6, chromatography fractions 1, 2 and 3, expected molecular weight 55.8 kDa). The recombinant protein was used to
O
immunise mice, whose sera were used in a Western blot (Figure 2B, lanes 8 and 9) and for FACS analysis (Figure 2C, K-S value 16.81).
These experiments show that CT045 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 3 CT381 (SEQ ID 105 and SEQ ID 106) was expressed in E.coli. The recombinant product was purified both as a GST-fusion protein (Figure 3A; lanes 2 and 3, chromatography fractions 1 and 2, expected molecular weight 52.7 kDa) and as a His-tagged fusion protein (Figure 3A; lanes 7-9, chromatography fractions 1, 2 and 3, expected molecular weight 26.7 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 3B: His-tagged: lanes 6 and 7; GST-fusion: lanes 16 and 17) and for FACS analysis (Figure 3C: GST-tagged, K-S value 35.98; Figure 3D: His-tagged, K-S value 32.54).
These experiments show that CT381 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 4 CT396 (SEQ ID 107 and SEQ ID 108) was expressed in E.coli. The recombinant product was purified both as a GST-fusion protein (Figure 4A; lanes 6 and 7, chromatography fractions 1 and 2, expected molecular weight 99.5 kDa) and as a His-tagged fusion protein (Figure 4B; lanes 5-7, chromatography fractions 1, 2 and 3, expected molecular weight 73.5 kDa). The recombinant His-tagged protein was used to immunise mice, whose sera were used in a Western blot (Figure 4C, lanes 14 and 15). The recombinant His-tagged protein and GST-fusion protein were also used for FACS analysis (Figure 4D: His-tagged, K-S value 34.50; Figure 4E: GST-fusion, K-S value 32.76).
-43- SThese experiments show that CT396 is a surface-exposed and immunoaccessible protein, N and that it is a useful immunogen. These properties are not evident from the sequence alone.
O Example Cc CT398 (SEQ ID 111 and SEQ ID 112) was expressed in E.coli. The recombinant product was purified both as a GST-fusion protein (Figure 5A; lanes 8 and 9, chromatography 00 fractions 1 and 2, expected molecular weight 54.8 kDa) and as a His-tagged fusion protein S(Figure 5B; lanes 8-10, chromatography fractions 1, 2 and 3, expected molecular weight ,28.8 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 5C: His-tagged: lanes 10 and 11; GST-fusion: lanes 18 and 19) and for FACS analysis (Figure 5D: GST-fusion, K-S value 31.24; Figure 5E: His-tagged, K-S value (N 26.10).
These experiments show that CT398 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 6 CT089 (SEQ ID 61 and SEQ ID 62) was expressed in E.coli. The recombinant product was purified both as a GST-fusion protein (Figure 6C: lane 2, chromatography fraction 1, expected molecular weight 70.8 kDa) and as a His-tagged fusion protein (Figure 6C: lanes 3, 4 and 5, chromatography fractions 1, 2 and 3, expected molecular weight 44.8 kDa). The recombinant proteins were used to immunise mice, whose sera were used in a Western blot (Figure 6A: GST-fusion: lanes 14 and 15; His-tagged: lanes 16 and 17) and for FACS analysis (Figure 6B: His-tagged, K-S value 26.59).
These experiments show that CT089 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 7 CT443 (SEQ ID 125 and SEQ ID 126) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein. The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 7A: lanes 10 and 11) and for FACS analysis (Figure 7B: K-S value 21.28).
These experiments show that CT443 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 8 CT541 (SEQ ID 149 and SEQ ID 150) was expressed in E.coli. The recombinant product was purified as a GST-fusion protein (Figure 8C: lanes 2 and 3, chromatography fractions 1 -44- O and 2, expected molecular weight 51.6 kDa). The recombinant protein was used to (C immunise mice, whose sera were used in a Western blot (Figure 8A: lanes 6 and 7) and for U FACS analysis (Figure 8B: K-S value 9.94).
O
_These experiments show that CT541 is a surface-exposed and immunoaccessible protein, c 5 and that it is a useful immunogen. These properties are not evident from the sequence alone.
00 Example 9 CT547 (SEQ ID 151 and SEQ ID 152) was expressed in E.coli. The recombinant product c was purified both as a GST-fusion protein (Figure 9D: lanes 4 and 5, chromatography fractions I and 2, expected molecular weight 58.3 kDa) and as a His-tagged fusion protein.
O
10 The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 9A: His-tagged: lanes 20 and 21) and for FACS analysis (Figure 9B: GSTfusion, K-S values 14.60 and 15.57; Figure 9C: His-tagged, K-S value 28.21).
These experiments show that CT547 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example CT587 (SEQ ID 189 and SEQ ID 190) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 10C: lanes 5, 6 and 7, chromatography fractions 1, 2 and 3, expected molecular weight 47.5 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 10A: lanes 12 and 13) and for FACS analysis (Figure 10B: His-tagged, K-S value 20.85).
These experiments show that CT587 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 11 CT266 (SEQ ID 77 and SEQ ID 78) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 11 C: lanes 11 and 12, chromatography fractions 1 and 2, expected molecular weight 44.1 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 11A: lanes 4 and 5) and for FACS analysis (Figure 11B: K-S value 21.29).
These experiments show that CT266 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 12 CT444 (SEQ ID 127 and SEQ ID 128) was expressed in E.coli. The recombinant product was purified as a GST-fusion protein (Figure 12B: lanes 2 and 3, chromatography fractions O 1 and 2, expected molecular weight 87.3 kDa) and as a His-tagged fusion protein (Figure "1 12C: lanes 3 and 4, chromatography fractions 2 and 3, expected molecular weight 9.0 kDa).
O The recombinant protein was used to immunise mice, whose sera were used in a Western o blot (Figure 12A: lanes 16 and 17) and for FACS analysis (Figure 12D: GST-tagged: K-S m 5 value 14.98; Figure 12E: His-tagged: K-S value 13.28).
These experiments show that CT444 is a surface-exposed and immunoaccessible protein, 00 0 and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 13 CT559 (SEQ ID 199 and SEQ ID 200) was expressed in E.coli. The recombinant product was purified as a His-tagged protein (Figure 13C: lanes 2, 3 and 4, chromatography fractions 1, 2 and 3. expected molecular weight 34.9 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 13A: lanes 6 and 7) and for FACS analysis (Figure 13B: K-S value 23.21).
These experiments show that CT559 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 14 CT681 (SEQ ID 155 and SEQ ID 156) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 14C: lanes 5 and 6, chromatography fractions 1 and 2, expected molecular weight 41.8 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 14A: lanes 10 and 11) and for FACS analysis (Figure 14B: K-S value 34.66).
These experiments show that CT681 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example CT713 (SEQ ID 201 and SEQ ID 202) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 15B: lanes 4, 5 and 6; chromatography fractions 1, 2 and 3, expected molecular weight 35.4 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 15A: lanes 12 and 13) and for FACS analysis (Figure 15C: K-S value 25.82).
These experiments show that CT713 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
O Example 16 CT823 (SEQ ID 229 and SEQ ID 230) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 16C: lanes 7, 8 and 9, chromatography Sfractions 1, 2 and 3, expected molecular weight 53.9 kDa). The recombinant protein was C 5 used to immunise mice, whose sera were used in a Western blot (Figure 16A: lanes 14 and and for FACS analysis (Figure 16B: K-S value 26.62).
00 0 These experiments show that CT823 is a surface-exposed and immunoaccessible protein, Sand that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 17 CT114 (SEQ ID 243 and SEQ ID 244) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 17; lanes 6 and 7, chromatography fractions 1 and 2, expected molecular weight 48.5 kDa).
Example 18 CT198 (SEQ ID 43 and SEQ ID 44) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 18A; lane 6, chromatography fraction 1, expected molecular weight 56.3 kDa). The His-tagged recombinant protein was used to immunise mice, whose sera were used for FACS analysis (Figure 18B).
These experiments show that CT198 is present in only part of an EB heterogeneous population (as chlamydial preparations usually are). Where it is present, it is a surfaceexposed and immunoaccessible protein. These properties are not evident from the sequence alone.
Example 19 CT241 (SEQ ID 55 and SEQ ID 56) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 19: lane 4, chromatography fraction 3, expected molecular weight 85.3 kDa).
Example CT350 (SEQ ID 27 and SEQ ID 28) was expressed in E.coli. The recombinant product was purified both as a His-tagged fusion protein (Figure 20A: lanes 2, 3 and 4, chromatography fractions 1, 2 and 3, expected molecular weight 61.3 kDa) and as a GST-tagged fusion protein. (Figure 20A: lanes 7, 8 and 9, chromatography fractions 1, 2 and 3, expected molecular weight 87.3 kDa). The recombinant proteins were used to immunise mice, whose sera were used in a Western blot (Figure 20B: His-tagged, lanes 4 and 5; GST-tagged, lanes 8 and 9).
O Example 21 CT351 (SEQ ID 25 and SEQ ID 26) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 21: lanes 2 and 3, chromatography fractions I1 and 2, expected molecular weight 76.1 kDa) Example 22 0 0 CT391 (SEQ ID 251 and SEQ ID 252) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 22: lanes 8 and 9, chromatography ¢C fractions 1 and 2, expected molecular weight 32.6 kDa).
Example 23
C
N
1 10 CT077 (SEQ ID 65 and SEQ ID 66) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein (Figure 23: lanes 2 and 3, chromatography fractions 1 and 2, expected molecular weight 59.7 kDa) and as a His-tagged fusion protein. The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 23C: lanes 6 and 7) and for FACS analysis (Figure 23B, His-tagged: K-S value 9.17).
These experiments show that CT077 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 24 CT181 (SEQ ID 245 and SEQ ID 246) was expressed in E.coli. The recombinant product was purified both as a GST-tagged fusion protein (Figure 24A: lane 4, chromatography fraction 1, expected molecular weight 50.1 kDa) and a His-tagged fusion protein (Figure 24B: lanes 2, 3 and 4, chromatography fractions 1, 2 and 3, expected molecular weight 32.0 kDa). The GST-tagged recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 24D, lanes 4 and 5 (indicated by arrow)) and for FACS analysis (Figure 24C, K-S value 7.62).
These experiments show that CT181 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example CT589 (SEQ ID 185 and SEQ ID 186) was expressed in E.coli. The recombinant product was purified both as a GST-tagged fusion protein (Figure 25A: lanes 4 and chromatography fractions 1 and 2, expected molecular weight 89.4 kDa) and as a Histagged fusion protein (Figure 25B: lanes 2 and 3, chromatography fractions 1 and 2, O expected molecular weight 63.4 kDa). The His-tagged recombinant protein was used to Nl immunise mice, whose sera were used for FACS analysis (Figure -4-4 These experiments show that CT589 is present in only part of an EB heterogeneous population (as chlamydial preparations usually are). Where it is present, it is a surface- C 5 exposed and immunoaccessible protein. These properties are not evident from the sequence alone.
00 OO Example 26 MC CT597 (SEQ ID 179 and SEQ ID 180) was expressed in E.coli. The recombinant product
(N
Swas purified both as a GST-tagged fusion protein (Figure 26A: lanes 5 and 6, chromatography fractions 1 and 2, expected molecular weight 36.0 kDa) and as a Histagged fusion protein (Figure 26B: lanes 2, 3 and 4, chromatography fractions 1, 2 and 3, expected molecular weight 10.3 kDa).
Example 27 CT623 (SEQ ID 163 and SEQ ID 164) was expressed in E.coli. The recombinant product was purified both as a GST-tagged fusion protein (Figure 27A: lanes 3 and 4, chromatography fractions 1 and 2, expected molecular weight 71.8 kDa) and as a Histagged fusion protein (Figure 27B: lanes 2, 3 and 4, chromatography fractions 1, 2 and 3, expected molecular weight 45.8 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western Blot (Figure 27E: GST-tagged, lane 4 (indicated by arrow); His-tagged, lane 13 (indicated by arrow)) and for FACS analysis (Figure 27C: GST-tagged: K-S value 15.89; Figure 27D: His-tagged: K-S value 20.27).
These experiments show that CT623 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 28 CT700 (SEQ ID 261 and SEQ ID 262) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein (Figure 28A: lanes 5, 6 and 7, chromatography fractions 1, 2 and 3, expected molecular weight 73.7 kDa). The recombinant protein was used to immunise mice, whose sera were used for FACS analysis (Figure 28B: K-S value 8.72).
These experiments show that CT700 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
-49- 0 Example 29 CT761 (SEQ ID 217 and SEQ ID 218) was expressed in E.coli. The recombinant product O was purified as a GST-tagged fusion protein (Figure 29A: lanes 6 and 7, chromatography _fractions 1 and 2, expected molecular weight 63.9 kDa). The recombinant protein was used to immunise mice, whose sera were used for FACS analysis (Figure 29B, K-S value 11.45).
00 These experiments show that CT761 is a surface-exposed and immunoaccessible protein, -1 and that it is a useful immunogen. These properties are not evident from the sequence alone.
SExample
(N
CT415 (SEQ ID 117 and SEQ ID 118) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein (Figure 30: lanes 3 and 4, chromatography fractions I and 2, expected molecular weight 55.4 kDa).
Example 31 CT454 (SEQ ID 253 and SEQ ID 254) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 31: lanes 2 and 3, chromatography fractions 1 and 2, expected molecular weight 56.2 kDa).
Example 32 CT467 (SEQ ID 129 and SEQ ID 130) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein (Figure 32: lanes 3 and 4, chromatography fractions 1 and 2, expected molecular weight 65.6 kDa).
Example 33 CT551 (SEQ ID 257 and SEQ ID 258) was expressed in E.coli. The recombinant product was purified both as a His-tagged fusion protein (Figure 33A: lanes 5, 6 and 7, chromatography fractions 1, 2 and 3, expected molecular weight 34.1 kDa) and as a GSTtagged fusion protein (Figure 33B: lanes 4 and 5, chromatography fractions 1 and 2, expected molecular weight 60.1 kDa).
Example 34 CT567 (SEQ ID 195 and SEQ ID 196) was expressed in E.coli. The recombinant product was purified both as a GST-tagged fusion protein (Figure 34A: lanes 8 and 9, chromatography fractions 1 and 2, expected molecular weight 44.0 kDa) and as a Histagged fusion protein (Figure 34B: lanes 7 and 8, chromatography fractions 1 and 2, expected molecular weight 18.3 kDa).
0 Example CT569 (SEQ ID 193 and SEQ ID 194) was expressed in E.coli. The recombinant product O was purified as a His-tagged fusion protein (Figure 35A: lanes 2, 3 and 4, chromatography _fractions 1, 2 and 3, expected molecular weight 11.2 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 35B: lanes 8 and 9, indicated with an arrow).
00 Example 36 CT647 (SEQ ID 169 and SEQ ID 170) was expressed in E.coli. The recombinant product r- was purified as a GST-tagged fusion protein (Figure 36: lanes 6 and 7, chromatography fractions 1 and 2, expected molecular weight 45.7 kDa).
Example 37 CT600 (SEQ ID 173 and SEQ ID 174) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein (Figure 37A: lanes 5, 6 and 7, chromatography fractions 1, 2 and 3, expected molecular weight 19.5 kDa). The recombinant protein was used to immunise mice, whose sera were used in a Western Blot (Figure 37B, lanes 10 and 11, indicated by arrow) and for FACS analysis (Figure 37C, K-S value 10.46).
These experiments show that CT600 is a surface-exposed and immunoaccessible protein, and that it is a useful immunogen. These properties are not evident from the sequence alone.
Example 38 CT279 (SEQ ID 247 and SEQ ID 248) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein and as a His-tagged fusion protein. The recombinant His-tagged protein and the recombinant GST-tagged protein were used to immunise mice, whose sera were used in Western blots (Figure 38A: His-tagged: lane (indicated by an arrow); Figure 38B: GST-tagged: lanes 12 and 13 (indicated by an arrow)).
Example 39 CT560 (SEQ ID 259 and SEQ ID 260) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein. The recombinant His-tagged protein was used to immunise mice, whose sera were used in a Western blot (Figure 39: lanes 6 and 7 (indicated by an arrow)).
Example CT389 (SEQ ID 249 and SEQ ID 250) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein. The recombinant protein was used to -51- 0 immunise mice, whose sera were used in a Western blot (Figure 40: lanes 16 and 17 C (indicated by an arrow)).
O Example 41 SCT456 (SEQ ID 255 and SEQ ID 256) was expressed in E.coli. The recombinant product was purified as a GST-tagged fusion protein. The recombinant protein was used to 00 immunise mice, whose sera were used in a Western blot (Figure 41: lanes 2 and 3 (indicated by an arrow)).
SExample 42 CT622 (SEQ ID 161 and SEQ ID 162) was expressed in E.coli. The recombinant product C 10 was purified as a His-tagged fusion protein. The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 42: lane 9 (indicated by an arrow)).
Example 43 CT759 (SEQ ID 213 and SEQ ID 214) was expressed in E.coli. The recombinant product was purified as a His-tagged fusion protein. The recombinant protein was used to immunise mice, whose sera were used in a Western blot (Figure 43: lanes 8 and 9 (indicated by an arrow)).
Example 44 In vitro neutralization assays, which show the ability of sera obtained from mice that have been immunised with the different recombinant proteins of the present invention to inhibit C. trachomatis infectivity for eukaryotic cells in culture, were performed using LLCMK2 (Rhesus monkey kidney epithelial) cells. Serial four-fold dilutions of mouse polyclonal sera were prepared in SP (Sucrose-Phosphate) buffer. Mouse antisera to whole EBs were used as a positive control, and preimmune sera and SP buffer alone were used as negative controls. Purified EBs from C. trachomatis (serovar D) were diluted in SP buffer to contain 3x10 5 IFU/ml, and 10pl of this suspension were added to each serum dilution in a final volume of 100pl. Antibody-EB interaction was allowed to proceed for 30 min at 37 0
C.
Then 100tl of reaction mix from each sample were added on top of PBS-washed LLCMK2 cell monolayers, in a 96-well microtiter plate, and centrifuged at 805xg for 1 hour at 37 0
C.
All sera and controls were examined in duplicated samples. After removal of the excess inoculum, the cells were rinsed once with PBS, replenished with 2004l of DMEM medium supplemented with 20%FCS and lgg/ml cycloheximide, and incubated at 37 0 C for 48 hours. The cells were fixed with methanol and the typical cytoplasmic inclusions generated by the ongoing intracellular chlamydial infection were stained with an anti-Chlamydia fluorescein-conjugated monoclonal antibody (Meridian Diagnostics). At adequate dilutions -52- O and EB to host cell ratios, the number of inclusions observed is considered to be equal to the CN number of viable chlamydiae which were initially capable of successfully establishing a 0 host cell infection (these are named Inclusion Forming Units, IFU). Fluorescein-labelled C inclusions were counted in four microscopical fields per well at a magnification of M 5 The inhibition of infectivity due to antibody interaction was calculated as percentage reduction in mean IFU as compared to the SP control (buffer only). According to common 00 practice, the sera were labelled as "neutralizing" if they could cause a 50% or greater
\O
reduction in infectivity, however, considering the complexity of the whole screening assay c (for instance, a change of host cell, or chlamydial isolate, or a variation in the environmental conditions in the preparation of the infectious inoculum), sera capable of inhibiting EB Sinfectivity to a lower extent should also be considered as vaccine candidates for further "1 study. Figure 44A shows an example of a result obtained from a neutralisation-positive serum whilst Figure 44B shows an example of a result obtained from a neutralisationnegative serum.
In vitro neutralization assays were carried out using sera obtained from mice immunised with the recombinant proteins mentioned in Example I to 10, 13-22, 24-26 and 29-37. The results are presented in Table II. These results indicate that CT045, CT242, CT381, CT396, CT398, CT467, CT547, CT587 and CT681 are all particularly good candidates for vaccines to prevent infection by C. trachomatis. These properties are not evident from the sequences alone.
In further experiments, the sera raised against C. trachomatis were tested against C.
pneumoniae EBs for cross-neutralization activity. The procedure was as described above, but purified EBs from C. pneumoniae were diluted in SP buffer to contain 3x10 6 IFU/ml, and 10til of this suspension were added to each serum dilution in a final volume of 100pl.
Sera obtained using CT242 and CT467 were able to cross-neutralise C. pneumoniae EBs.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application.
-53- SIt will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments 0 without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
00 IN1 -54- REFERENCES (the contents of which are hereby incorporated in full) (I1I Raulston (1995) Mci Microbiol 15:607-616 Everett (2000) Vet Microbiol 75:109-126 Kalman et al. (1999) Nature Genetics 21:385-389 Read et a. (2000) Nucleic Acids Res 28:1397-1406 00 Shirai el (2000) Nucleic Acids Res 28:2311-2314 Stephens et (1998) Science 282:754-759 (7 nentoalptn plcainW9/70 International patent application W099/27 105 International patent application W099/2875 10) Ward (1995) Apmis. 103:769-96.
f{I1I} Moulder (199 1) Microbiol Rev 55(l):143-190.
12) Comanducci et a. (1994) Infect Immun 62(12):5491-5497.
(13) EP-A-0499681 14) International patent application W095/28487 15)} Murdin et (1993) Infect Immun 61:4406A4414 f1I6} Cerrone et (199 1) Infect Immun 59(1l):79-90.
f17} Raulston et (1993)]. Bio. Chem. 268:23139-23147.
f 18 International patent application W002/02606.
2007231768 31 Oct 2007 TABLE I Ref. 18 Cpneumoniae accession number annotation Ctirachomalis accession number annotation SEQ S[IDs giJ43767291gblAAD18590.11 Polymorphic Outer Membrane Protein G Family gil33293461gbMAC68469.11 Putative Outer Membrane Protein G 1, 2 cpOO14 gij43767291gblAAD18590.1 I Polymorphic Outer Membrane Protein G Family gil33293461gblAAC68469.1 I Putative Outer Membrane Protein G 3, 4 cp0015 gii43767311gblAAD18591.11 Polymorphic Outer Membrane Protein Gi Family gil33293461gblAA068469.11 Putative Outer Membrane Protein G 5,6 cpO0116 Igil43767311gbAAD18591.11 Polymorphic Outer Membrane Protein G/l Family giI3329350IgblAAC68472.11 Putative Outer Membrane Protein 1 7, 8 cpOOl 7 Igil4376731 igbiMADl8591.11 Polymorphic Outer Membrane Protein G/l Family gil33293461gblAAC68469.1 I Putative Outer Membrane Protein G 9,10 cpOO18 Igil43767331gblAAD18593.11 Polymorphic Outer Membrane Protein G Family gil3328840igblAAC68009.11 Putative outer membrane protein A 11, 12 c00 19 IgiI43767311gblAAD18591.11 Polymorphic Outer Membrane Protein G/l Family gi133293461gblAAC68469.11 Putative Outer Membrane Protein G 13,14 cp0468 giJ43767541gbIAAD1861 1.11 Polymorphic Outer Membrane Protein (Frame-shift gi133293441gbjAAC68467.li Putative Outer Membrane Protein E 15, 16 Iwith C cp6260 IgiJ437626OigbIAAD18163.11 Polymorphic Outer Membrane Protein G Family giI33293461gbIAAC68469.11 Putative Outer Membrane Protein G 17, 18 cp6262 Igil43762621gblAA018165.11 hypothetical protein gil3328765gbAAC6794O.1[ hypothetical protein 19, cp6269 Igil43762691gblAD1 8171.11 hypothetical protein gil33288251gbIAA067995.11 [hypothetical protein 21, 22 cp6270 gil43762701gblAAD18172.11 Polymorphic Outer Membrane Protein G Family giJ33293501gbIAAC68472.1[ Putative Outer Membrane Protein 1 23, 24 cp6272 gil43762721gblAAD181 73.11 Predicted OMP (leader peptide: outer membrane) gil33287721gblAAC67946.1 [hypothetical protein 25, 26 cp6273 gil43762731gblAAD18l 74.1j Predicted OMP (leader peptide) gi 3328771 IgblAAC67945.1 j hypothetical protein 27, 28 cp6296 giI43762961gblAAD18195.11 hypothetical protein gi 3328520IgblAAC67712.11 Ribulose-P Epimerase 29, cp6362 gil43763621gbAM018254.i 1 YbbP family hypothetical protein giJ3328401 IgbIAAC67602.111 hypothetical protein 31,32 cp6372 Igil43763721gblAADl 8263.1 [Signal Peptidase I gil332841 OigbAAC676l 0.11 Signal Peptidlase 1 33,34 cp6397 giJ437 3971gblAA018286.i CHLPS hypothetical protein gil33285061gblAAC67700.1 I CHLPS hypothetical protein 35, 36 cp6402 gil43764021gblAAD18290.1 I ACR family giJ33285051gb1M0C67699.1 I ACR family 37,38 cp6419 giI43764191gblAAD18305.11 CT149 hypothetical protein gil33285511gblAAC67740.11 possible hydrolase 39,40 cp6446 gi143764461gblAAD18330.11 hypothetical protein giJ3329261 jg iAAC68390.11i hypothetical protein 41,42 cp6466 gil43764661gblAAD18348.11 Oligopeptide Binding Protein gil3328604igblAAC67790.11 Oligopeptide Binding Protein 43,44 cp6467 giJ43764671gblAAD18349.11 Oligopeptide Binding Protein gii33286041gblAAC67790.11 Oligopeptide Binding Protein 45,46 cp6468 gil43764681gb1M0D18350.11 Oligopeptide Binding Protein giI33285391gblAAC67730.11 Oligopeptide Binding Protein 47,48 cp6469 gil43764691gblAA018351 .11 Oligopeptide Binding Protein giJ33285791gb AAC67766.1 I Oligopeptide binding protein permease 49, cp6520 gil43765201gblAAD18398.11 Polysacchande Hydrolase-Invasin Repeat Family gil3328526jgb1MAC67718.11 predicted polysaccharide hydrolase-invasin repeat 51, 52 family p6567 gil43765671gbIAAD18441.11 Inclusion Membrane Protein C gil33286421gblAAC67825.11 Inclusion Membrane Protein C 53,54 cp6576 gi[43765761gblAD18449.11 Omp85 Analo giJ3328651 IblAMC67834.11 Omp85 Analo 55, 56 cp6577 gii43765771gblAAD18450.11 (0mpH-Like Outer Membrane Protein) gil33286521gblAAC67835.11 (0mpH-Like Outer Membrane Protein) 57, 58 cp6601 giI4376601IgbIAD 18472.11 Low Calcium Response D i3286gblAAC67681.11 Low Calcium Response 0 59,60 cp6602 gil43766021gblAAD18473.11 Low Calcium Response E gij33284851gblAAC67680.lj Low Calcium Response E 161, 62 2007231768 31 Oct 2007 cp6607 giJ437 6071gbAAD18478.11 Phopholipase D Superfamily gil33284791gblAAC67675.11 Phopholipase 0 Superfamily (leader (33) peptide) 63, 64 cp6615 gil4376615IgblAAO18485.11 YojL hypothetical protein gil33284721gblAAC67668.11 hypothetical protein 65,66 cp6624 gil43766241gblAAD18493.11 Solute Protein Binding Family gij33284611gblAAC67658.11 Solute Protein Binding Family 67, 68 cp6639 gi143766391gb1AA018507.11 Flagellar Secretion Protein gi 3328453igblAA067651.11 Flagellar Secretion Protein 69, cp6664 gil43766641gblAAD18529.1j Leucyl Aminopeptidase A giI3328437IgblAAC67636.11 Leucyl Aminopeptidase A 71, 72 cp6672 giJ437 6721gbIAAD1 8537.11 CBS Domain protein (Hemolysin Homolog) giI33286671gblAAC67849.11 Hypothetical protein containing CBS domains 73, 74 cp6679 gil43766791gblAAD18543.11 CT253 hypothetical protein giJ3328664 gblAAC67846.11 hypothetical protein 75, 76 cp6696 gii43766961gbjAAD18559.11 CT266 hypothetical protein gil3328678 gblAA067859.11 hypothetical protein 77, 78 cp6717 gil43767171gblAAD18579.11 Phospholipase 0 superfamily gil33286981gblAA067877.11 Phospholipase 0 superfamily 79,80 cp6727 giJ43767271gbIAAD1 8588.11 Polymorphic Outer Membrane Protein Gi Family gil33293461gblAA068469.1J Putative Outer Membrane Protein G 81,82 cp6728 gij43767281gblAAD18589.11 Polymorphic Outer Membrane Protein G Family gii33293461gblAA068469.11 Putative Outer Membrane Protein G 83, 84 cp6729 gil43767291gblAA018590.11 Polymorphic Outer Membrane Protein G Family gi[33293501gblAAC68472.11 Putative Outer Membrane Protein 1 85, 86 cp6731 gi1437673ligb]AAD18591.11 Polymorphic Outer Membrane Protein G/l Family giJ33293501gbIAA068472.11 Putative Outer Membrane Protein I 87, 88 cp6733 giJ43767331gbIAAD1 8593.11 Polymorphic Outer Membrane Protein G Family gij3328840igb1MAC68009.1 I Putative outer membrane protein A 89, cp6735 gil43767351gb1M0D18594.1l Polymorphic Outer Membrane Protein (truncated) All giJ33288401gbIAM068009.11 Putative outer membrane protein A 91, 92 Fain cp6736 Igil43767361gblAAD18595.11 Polymorphic Outer Membrane Protein G Family gil33293461gbjM0C68469.11 Putative Outer Membrane Protein G 93, 94 cp6737 IgiI43767371gblAAD18596.11 Polymorphic Outer Membrane Protein H Family gij3329347igb1AA068470.11 Putative Outer Membrane Protein H 95,96 cp6751 IgiJ4376751 IgbJAAD1 8608.11 Polymorphic Outer Membrane Protein E Family gil33293441gblAAC68467.11 Putative Outer Membrane Protein E 97, 98 cp6752 Igii43767521gblAAD18609.11Polymorphic Outer Membrane Protein E Family gii33293441gblAA068467.11 Putative Outer Membrane Protein E 99,100 cp6753 Igil43767531gblAAD18610.11 Polymorphic Outer Membrane Protein E/F Family gil33293441gblAA068467.11 Putative Outer Membrane Protein E 101,102 cp6757 Igil43767571gblAAD18613.11 hypothetical protein qil3328701 IgbAAC67880.lj PP-loop superfamily ATPase 103, 104 cp6767 Igil43767671gblAAD18622.11 Arginine Periplasmic Binding Protein gil3328806jgblAAC67977.11 Arginine Binding Protein 105, 106 cp6790 Igi143767901gbIAAD18643.11 Heat Shock Protein-70 giJ33288221gbAAC67993.11 HSP-70 107,108c6802 gi1437 8021gbIAAD1 8654.11 CT427 hypothetical protein gil33288571gblAAC68024.11 hypothetical protein 109,110 c681 4 gil437681 41gblAADl8665.11 CT398 hypothetical protein gil33288251gblAAC67995.11 [hypothetical protein 111,112 c6829 gil43768291qblAAnl18679.1Iv oyopi membrne protein A Family gi133288401gbIAA068009.1[ Putative outer membrane protein A 113, 114 c6830 gil4376830[gb1M0D18680.11 polymorphic membrane protein B Family gil3328841 1gbAA06801 0.11[Putative outer membrane protein B 115, 116 cp6832 gil43768321gblAA1 8681.11 Solute binding protein gi133288441gblAAC6801 2.11 Solute-binding protein 117, 118 cp6834 Igil43768341gblAAD18683.11 (Metal Transport Protein) gil33288461gblAA068014.lj (Metal Transport Protein) 119, 120 cp6847 Igil43768471gblAAD18695.11 Tail-Specific Protease gi33288721gblAAC68040.11 Tail-Specific Protease 121, 122 cp6848 Igil43768481gblAAD18696.1I 15 kDa Cysteine-Rich Protein gi133288731gb1AA068041.1I l5kDa Cysteine-Rich Protein 123, 124 cp6849 Igil43768491gblAAD18697.11 60 kDa Cysteine-Rich OMP giJ33288741gbIAAC68042.li 6OkDa Cysteine-Rich OMP 125, 126 cp6850 giJ43768501gbIAAD18698.11 9 kDa-Cysteine-Rich Lipoprotein gil33288761gblAAC68043.11 9kDa-Cysteine-Rich Lipoprotein 127, 128 cp6878 gil43768781gblAAD18723.11 2-Component Sensor gi 33289011gblAAC68067.li 2-component regulatory system-sensor histidine kinase 129,130 cp6879 gil43768791gblAA018724.11 similarity to CHLPS IncA g3285gblAAC67649. iI hypothetical protein 131,132 cp6884 giJ43768841gbIAAD1 8729.11 CT471 hypothetical protein gil33289051gblAM068071.lj hypothetical protein 133, 134 2007231768 31 Oct 2007 c6886 giJ437 8861gb1M01 8731.1] YidD family gij33289081gblAAC68073.11 hypoth~etical protein 135, 136 cp6890 gil4376890jgblAM018734.11 CT476 hypothetical protein gi1332891 I IgbjAAC68076.11 hypothetical protein 137,138 cp6892 gil43768921gbAA18736.11 Oligopeptide Permease gil33289131gblAAC68078.11 Oligopepfide Permease 139, 140 cp6894 gil43768941gbAAD18738.lj Oligopeptide Binding Lipoprotein gil33289151gb AAC68080.11 oligopeptide Binding Lipoprotein 141,142 cp6900 gil43769001gblAAD18743.11 Glutamnine Binding Protein gi133289221gbjAAC68086.11 Glutamnine Binding Protein 143,144 cp6909 giJ437 9091gblAADl 8752.11 Protease gil65781 07igblAAC68094.21 Protease 145, 146 cp6952 gil43769521gblAAD1 8792.11 Apolipoprotein N-Acetyltransferase gil33289721gblAA0681 36.11 Apolipoprotein N-Acetyltransferase 147,148 cp6960 gil43769601gblAAD18800.11 FKBP-type peptidyl-prolyl cis-trans isomerase gil33289791gbAA068143.11 FKBP-type peptdyl-prolyl cis-trans isomerase 149,150 cp6968 gil43769681gb1M0D18807.11 CT547 hypothefical protein gil33289861gblAAC68149.11 hypothetical protein 151, 152 cp6969 gil43769691gb1AAD18808.11 CT548 hypothefical protein gil33289871gblAAC68150.11 hypothefical protein 153, 154 cp6998 giJ437 9981gblAAD18834.11 Major Outer Membrane Protein giI33291331gblAAC68276.11 Major Outer Membrane Protein 155,156 cp7005 gij43770051gblAM018841.11 YopCIGen Secretion Protein D gil33291251gblAAC68269.11 probable Yop proteins translocation protein 157,158 cpl7l 5 gi1437701 SigbIMAD1 8851.11 FHA domain; (homology to adenylate cydase) g!iN3291 1 5gb1AAC68259.i (FHA domain; homology to adenylate cyclase) 159,160 cp7033 gil43770331gblAM018867.11 CHLPN 76 kDa Homolog-1 (CT622) gij33290691gblAAC68226.11 CHLPN 76kDa Homolog 161,162 cp7034 gil43770341gblAAD18868.11 CHLPN 76 kDa Homnolog-2 (CT623) gi165781091gbjMAC68227.21 CHLPN 76k~a Homolog 163, 164 cp7035 gil43770351gblAAD18869.11 Integral Membrane Protein giJ3329071 IgblAAC68228.11 Integral Membrane Protein 165, 166 cp7072 gil43770721gblA01 8902.11 CT648 hypothetical protein gil33290971gblAAC68825.1 I hypothetical protein 167, 168 cp7073 gil43770731gb1M0D18903.1[0CT647 hypothetical protein gi133290961gblAAC68824.11 hypothetical protein 169,170 cp7085 gi143770851gblAAD18914.11[CT605 hypothetical protein gil3329050IgblAAC68208.11 hypothetical protein 171,172 cp709O Igil437709OjgblAAD18919.11 Peptidoglycan-Associated Lipoprotein gil3329044igb1M0C68202.11 Peptidoglycan-Associated Lipoprotein 173,174 cp7091 giJ4377091 IgbAA018920.1 I macromolecule transporter gij33290431gblAA068201 .11 component of a macromolecule transport system 175,176 cp7092 gil43770921gblAMD18921.11 CT598 hypothetical protein gil33290421gblAAC68200.11 hypothetical protein 177, 178 cp7093 giJ43770931gbIAAD18922.11 Biopolymer Transport Protein giI3329041 IgbIAAC68199.11 Biopolymer Transport Protein 179,180 cp7094 gil43770941gblAM018923.11 Macromolecule transporter gil3329040igbAAC68198.11 polysaccharide transporter 181,182 cp7l 01 giJ43771 01 IgblAAD1 8929.11 CT590 hypothetical protein gil33290331gblAA0681 92.11 hypothetical protein 183, 184 cp7102 giJ43771 O21gblAADI 8930.11 CT589 hypothetical protein giI33290321gbjM0C681 91 .11hypothetical protein 185, 186 cp7106 gil43771 O61gblAADl 8933.11 hypotheical protein gil33287961gblAAC67968.11 hypothetical protein 187,188 cp7l 11 giJ43771 1 IgbjAADl 8938.11 Enolase 189_190 cp7127 gil4377127igblAAD18953.11 General Secreion Protein D gil33290131gblAAC68174.11 Gen. Secretion Protein 0 191,192_ cp7130 gil43771301gb1M0D18956.11 predicted OMP (leader peptide} gi 3329010 gblAC68171.1 predicted OMP 193,194_ cp7132 gil43771321gb1MAD18958.11 CT567 hypothetical protein gil33290081gbjAAC68169.11 hypothetcal protein 195,196_ Cp7133 gil43771 331gbIMADl8959.11 CT566 hypothetcal protein gil33290071gblAAC681 68.11 hypotheical protein 197,198_ 0p7140 gil4377140IgblAAD18965.11 Yop Translocation J gil3329000igblAAC68161.11 Yop proteins translocaion lipoprotein J19,0 Cp7170 gil4377170jgblAM018992.11 Outer Membrane Protein B gil33291691gblAAC68308.11 Outer Membrane Protein Analog 201,202_ p 7177 gil43771771gblAAD18998.11 Flagellar M-Ring Protein gil33291751gblAAC68314.11 Flagellar M-Ring Protein 1203, 204 0p7182 gi[4377182igblAAD19003.11 01724 hypothetcal protein gil3329181 1gbjAAC68319.11 hypothetical protein 1205,206 0p7184 gi143771841gblAAD19005.11 Rod Shape Protein Igil33291831gblAAC68321.11 Rod Shape Protein 1207,208 2007231768 31 Oct 2007 Cp7193 giJ437 1931gb1MAD1 9013.11 CT734 hypothetical protein gil33291921gblAAC68329.11 hypothetical protein 209,210 Cp7206 gil43772061gblAA019025.11 CHLTR possible phosphoprotein gil33292041gblAAC68339.11 CHLTR possible phosphoprotein 211, 212 Cp7222 gil43772221gblAA019040.11 Muramidase (invasin repeat family) giJ3329221 Igb1M068354.11 Muramidase (invasin repeat family) 213, 214 Cp7223 gij43772231gb1M0D19041.11 Cell Division Protein FtsW gi 3329222lgblAAC68355.11 Cell Division Protein FtsW 215, 216 Cp7224 gil43772241gblAA019042.lj Peptidoglycan Transferase gil33292231gblAAC68356.11 Peptidoglycan Transferase 217, 218 Cp7225 giJ437 2251gblAA019043.11 Muramate-Ala Ligase D-Ala-D-Ala Ligase gil33292241gblAA068357.11 UDP-N-acetymuramate-alanine ligase 219, 220 Cp7248 giI43772481gbIMAD19064.11 Thioredoxin Disulfide Isomerase gil33292441gblAAC68375.11 Thioredoxin Disulfide Isomerase 221, 222 Cp7261 giI4377261 IgbIAAD1 9076.11 CT788 hypothetical protein -{leader pepfide-periplasmi gil33292531gblAAC68383.11 (leader (60) peptide-periplasmic} 223, 224 Cp7280 gil4377280IgblAAD19093.11 Insulinase family/Protease Ill gil3329273lgb1MAC68402.11 Insulinase family/Protease 111 225, 226 Cp7287 giI43772871gblAAD19099.11 Putative Outer Membrane Protein D Family gil33292791gblAAC68408.11 Putative Outer Membrane Protein 0 227, 228 Cp7306 gi 437 3061gb1M0119116.11 DO Serine Protease gi133292931gb1AAC68420.11 DO Serine Protease 229, 230 Cp7342 gil43773421gblAMD19149.11 ABC transporter permease gil33293271gblAMC68451.11 ABC transporter permease pyrimidine biosynthesis 231, 232 protein Cp7347 gij43773471gblAAD19153.11 CT858 hypothetical protein giJ65781 1 8IgblAAC68456.21 predicted Protease containing IRBP and DHR domains 233, 234 Cp7353 gil43773531gb AAD19159.11 CT863 hypothetical protein gil33293371gbAAC68461 .lj hypothetical protein 235, 236 Cp7367 gil43773671gbAAD1 9171.11 Predicted OMP gil33287951gblAAC67967.11 hypothetical protein 237, 238 Cp7408 gil43774081gblAADl9209.11 hypothetical protein gil3328795lgbiAAC67967.11 hypothetical protein 239, 240 Cp7409 gil43774091gblAADi9210.11 Predicted Outer Membrane Protein (CT371) gil33287951gblAAC67967.11 hypothetical protein 241, 242 iJ437641 1 Igbl gil33285121gblAAC67705.11 hypothetical protein 243, 244 il43765081gb1 gil33285851gblAAC67772.11 hypothetical protein 245, 246 gii437671 Oigbj gi133286921gbAAC67872.11 NADH (Ubiguinone) Oxidoreductase, Gamma 247, 248 ____gil43767771gbl gil3328815igblAAC67986.11 hypothetical protein 249, 250 gil43767821gbl gil33288171gblAAC67988.11 hypothetical protein 251, 252 giJ43768631gbI gil33288871gbjAAC68054.1 I Arginyl tRNA transferase 253, 254 giJ43768661gbI gil33288891gblAAC68056.11 hypothetical protein 255, 256 gil43769721gbi gij3328991 IgbiMAC68l 53.11 D-Ala-D-Ala Carboxypeptidase 257, 258 ____gij43771 391gbl giJ3329001 IgblAAC68l 62.11 hypothetical protein 259, 260 ____gil43771 S41gbl Igil33291541gblAAC68295.11 hypothetical protein 261, 262 -59- TABLE 11 neutralization of EB infectivity for LLCMK2 cell cultures Fusion neutralization at specified serum dilutions CT type titer 1/40 1/160 1/640 1/2560 CT045 HIS 1:160 32 50 18 CT089 HIS 44 37 0 GST 25 37 CT 114 HIS 0 18 CT181 GST 19 0 CT198 HIS 19 0 13 CT241 HIS 42 CT242 HIS 1:100 58 45 0 GST 46 24 40 CT350 OST 0 39 CT351 HIS 5 CT381 HIS 1:450 67 56 48 GST 24 0 0 CT391 HIS 0 14 CT396 HIS 1:300 55 62 GST 8 33 CT398 HIS 1:640 57 45 GST 1 :>640 68 60 60 CT415 GST 21 21 CT427 HIS 25 13 13 CT443 HIS 25 25 CT454 HIS 16 4 CT467 GST 1:1100 65 67 62 48 CT541 GST 10 24 13 CT547 HIS 1:40 50 13 18 GST 0 18 CT551 HIS 11 CT559 HIS 20 23 CT567 26 CT569 HIS 0 5 CT587 HIS 1:1200 51 61 56 42 CT589 HIS 37 21 GST _0 33 CT597 GST 0 4 CT600 HIS 0 11 CT647 GST 15 0 CT681 HIS 1:160 95 53 CT713 HIS 10 10 CT761 GST 16 CT823 HIS 5 23 NN-GST 0 0 0 NN-HIS 0 0

Claims (10)

  1. 2. The use of a nucleic acid in the manufacture of a medicament for the treatment or prevention of infection due to Chiamydia trachomatis, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260 and 262 242, a nucleic acid comprising a nucleotide sequence having 50% or greater sequence identity to the nucleotide sequence of or a nucleic acid comprising a fragment of an nucleotide sequence of
  2. 3. The use of claim 1 or claim 2, wherein infection is treated or prevented by the medicament eliciting an immune response which is specific to a Chiamydia elementary body.
  3. 4. The use of a protein as defined in claim 1, or a nucleic acid as defined in claim 2, in the manufacture of a medicament for neutralizing Chiamydia trachomatis elementary bodies. An immunogenic composition comprising a protein and an adjuvant, wherein the protein is a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, rKI 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, C0 123, 125, 127, 129, 131, 133, 135, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 00 225, 227, 229, 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 257, 259 and 261, a protein comprising an amino acid sequence having 50% or greater sequence identity to the amino acid sequence of or a protein comprising a fragment of an amino acid sequence of
  4. 6. An immunogenic composition comprising a nucleic acid and an adjuvant, wherein the nucleic acid is a nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260 and 262, a nucleic acid comprising a nucleotide sequence having 50% or greater sequence identity to the nucleotide sequence of or a nucleic acid comprising a fragment of an nucleotide sequence of
  5. 7. The composition of claim 5 or claim 6, for use as a pharmaceutical.
  6. 8. A method of neutralizing Ctrachomatis infectivity in a patient, comprising the step of administering to the patient the composition of claim 5 or claim 6, or an antibody which recognises a protein as defined in claim 1.
  7. 9. A method of immunising a patient against Chiamydia trachomatis, comprising administering to the patient the composition of claim 5 or claim 6. A method of raising antibodies specific for Chiamydia trachomatis elementary bodies, comprising administering to the patient the composition of claim 5 or claim 6.
  8. 11. A method of raising antibodies which recognise a protein as defined in claim 1, comprising the step of administering to a patient a Chiamydia trachomatis elementary body. O 12. A method for detecting a Chlamydia trachomatis elementary body in a biological C1 sample, comprising the step of contacting the sample with an antibody which recognizes 0 a protein as defined in claim 1. O
  9. 13. The use according to any one of claims 1 to 4 substantially as hereinbefore described with reference to the accompanying Figures and/or Examples. 00 14. The composition according to any one of claims 5 to 7 substantially as hereinbefore \N described with reference to the accompanying Figures and/or Examples.
  10. 15. The method according to any one of claims 8 to 12 substantially as hereinbefore described with reference to the accompanying Figures and/or Examples. O (N DATED this THIRTIETH day of OCTOBER 2007 Novartis Vaccines and Diagnostics S.r.l. Patent Attorneys for the Applicant: F.B. RICE CO.
AU2007231768A 2001-12-12 2007-10-31 Immunisation against Chlamydia trachomatis Abandoned AU2007231768A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2007231768A AU2007231768A1 (en) 2001-12-12 2007-10-31 Immunisation against Chlamydia trachomatis

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB0129732.4 2001-12-12
GB0218233.5 2002-08-06
GB0218924.9 2002-08-14
AU2002366592A AU2002366592B2 (en) 2001-12-12 2002-12-12 Immunisation against Chlamydia trachomatis
AU2007231768A AU2007231768A1 (en) 2001-12-12 2007-10-31 Immunisation against Chlamydia trachomatis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2002366592A Division AU2002366592B2 (en) 2001-12-12 2002-12-12 Immunisation against Chlamydia trachomatis

Publications (1)

Publication Number Publication Date
AU2007231768A1 true AU2007231768A1 (en) 2007-11-29

Family

ID=38787487

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007231768A Abandoned AU2007231768A1 (en) 2001-12-12 2007-10-31 Immunisation against Chlamydia trachomatis

Country Status (1)

Country Link
AU (1) AU2007231768A1 (en)

Similar Documents

Publication Publication Date Title
US7361353B2 (en) Immunisation against Chlamydia trachomatis
US10226524B2 (en) Staphylococcus aureus proteins and nucleic acids
EP1385876B1 (en) Gonococcal proteins and nucleic acids
EP2275552A2 (en) Neisserial antigenic peptides
EP2270175A1 (en) Streptococcus pneumoniae proteins and nucleic acids
AU2007231768A1 (en) Immunisation against Chlamydia trachomatis

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted