WO2010084350A1 - Mutant pathogenic bacterial and live attenuated vaccine compositions - Google Patents

Mutant pathogenic bacterial and live attenuated vaccine compositions Download PDF

Info

Publication number
WO2010084350A1
WO2010084350A1 PCT/GB2010/050086 GB2010050086W WO2010084350A1 WO 2010084350 A1 WO2010084350 A1 WO 2010084350A1 GB 2010050086 W GB2010050086 W GB 2010050086W WO 2010084350 A1 WO2010084350 A1 WO 2010084350A1
Authority
WO
WIPO (PCT)
Prior art keywords
pathogenic bacterium
mutant
ptshi
pfkb
genes
Prior art date
Application number
PCT/GB2010/050086
Other languages
French (fr)
Inventor
Arthur Thompson
Steven D. Bowden
Original Assignee
Plant Bioscience Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Plant Bioscience Limited filed Critical Plant Bioscience Limited
Publication of WO2010084350A1 publication Critical patent/WO2010084350A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/36Adaptation or attenuation of cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/025Enterobacteriales, e.g. Enterobacter
    • A61K39/0275Salmonella
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/255Salmonella (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1085Transferases (2.) transferring alkyl or aryl groups other than methyl groups (2.5)
    • C12N9/10923-Phosphoshikimate 1-carboxyvinyltransferase (2.5.1.19), i.e. 5-enolpyruvylshikimate-3-phosphate synthase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1223Phosphotransferases with a nitrogenous group as acceptor (2.7.3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1247DNA-directed RNA polymerase (2.7.7.6)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/522Bacterial cells; Fungal cells; Protozoal cells avirulent or attenuated
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to mutant pathogenic bacteria, particularly mutant intracellular pathogenic bacteria, useful in the preparation of attenuated pathogenic bacteria, to attenuated pathogenic bacteria, to such bacteria carrying a heterologous gene, to vaccines comprising live attenuated pathogenic bacteria, optionally carrying a heterologous gene, useful for the prevention of microbial pathogenesis, and to methods for the preparation of such vaccines and bacteria.
  • the mammalian cell provides a uniquely stable environment for the replication and survival of intracellular bacteria.
  • the host cell provides constant temperature and pH regulation, a ready supply of nutrients for growth, and protection against antimicrobial agents.
  • Intracellular bacteria can be divided into strict and facultative bacteria (1). Strict intracellular bacteria cannot be cultivated in broth medium and can replicate only in vivo or in tissue-cultured cells in vitro. Strict intracellular bacterial pathogens include Chlamydia (responsible for genital and ophthalmic infections in humans), Coxiella (the etiological agent of Q fever), Ehrlichia (headache, fever and chills in humans) and the Rikettsias.
  • facultative intracellular bacteria are able to replicate in broth medium as well as within mammalian cells.
  • Certain facultative intracellular pathogens are able to enter and multiply in phagocytic cells such as neutrophils or macrophages.
  • the intracellular environment of a phagocyte may protect the bacteria during the early stages of infection or until they develop a full complement of virulence factors.
  • the intracellular environment also guards the bacteria against the activities of extracellular bactericides, antibodies, drugs, etc.
  • Facultative intracellular parasites that are able to replicate in phagocytic cells include Yersinia, Legionella pneumophila, Salmonella, Mycobacterium tuberculosis and Mycobacerium leprae, the agents of tuberculosis and leprosy respectively.
  • Other facultative intracellular pathogens are able to replicate in nonphagocytic cells and include Bartonella, associated with several human diseases, Brucella, Franciscella tularensis, Listeria, Salmonella and Tropheryma whippelii (associated with Whipple's disease).
  • bacterial intracellular pathogens (1) and the diseases that they cause are:
  • Intracellular bacteria have developed an array of weaponry that enables them to evade host cell immune mechanisms. These include slow growth phenotypes in phagocytic cells, and the ability to shield themselves from host cell defence mechanisms.
  • the invasion and growth of intracellular pathogens in host cells also involves many virulence factors acting in concert. Therefore, unlike extracellular pathogens, it is unusual to identify a single gene product such as a toxin, which provides an overriding role in the pathogenesis of the disease and may provide a target for antimicrobials.
  • antibodies have been developed that provide protection against some intracellular pathogens, the level of protection that they offer is relatively low.
  • antibodies can protect against low but not high virulence strains of F. tularensis (3), or can provide protection against low challenge doses of B. pseudomallei administered by the intraperitoneal route, but not administered by the aerosol route of infection (4,5).
  • Salmonella is a facultative intracellular pathogen that causes gastroenteritis and fatal systemic disease in mammals including humans, cattle and pigs.
  • Typhoidal Salmonella serovars such as Salmonella enterica serovars Typhi and Paratyphi cause an estimated 20 million cases of salmonellosis and 200,000 human deaths worldwide per annum (6).
  • Salmonella infections occur as a result of ingestion of contaminated food and water.
  • An important source of Salmonella infections in humans arises from contaminated poultry, pig, and cattle products.
  • a randomised UK National Survey carried out in 2004 for faecal carriage of Salmonella in slaughter pigs, cattle and sheep identified S. Typhimurium in 11.1 % of caecal samples and 2.1 % of carcass swabs of 2509 pigs (7).
  • Salmonella In systemic infections, Salmonella penetrates the small intestinal barrier by invading gut epithelial cells. From there Salmonella can gain access to the mesenteric lymph nodes where the bacteria are engulfed by phagocytic cells such as macrophages. Once inside macrophages, Salmonella is compartmentalised into an intracellular phagosome, which is modified to become the "Salmonella containing vacuole” (SCV).
  • SCV Stalmonella containing vacuole
  • the SCV acts as a shield that prevents lysosomal fusion and exposure to host cell antimicrobial agents (13,14). Salmonella also resides within an SCV during invasion of epithelial cells (15). The Salmonella must acquire nutrients for replication in epithelial cells and macrophages.
  • the invention provides a mutant pathogenic bacterium, preferably a mutant intracellular pathogenic bacterium, that is incapable of expressing one or more metabolic genes.
  • Such a mutation can be a deletion, an insertion, a substitution or a combination thereof, provided that the mutation leads to the failure to express a functional protein (functional silencing).
  • the mutation is a deletion.
  • the metabolic gene or genes are deleted, however other mutations, for example partial deletion of the gene or mutation in sequences that regulate expression of the metabolic gene or genes may be used to achieve functional silencing.
  • the mutant pathogenic bacterium is attenuated as a result of functional silencing of one or more metabolic genes.
  • the invention also relates to mutant pathogenic bacteria that are incapable of expressing one or more metabolic genes, but are not attenuated, such mutants can be subjected to further mutation in metabolic or other virulence genes to provide a multiple mutant attenuated form of the pathogenic bacteria.
  • mutation in one or multiple genes that involve proteins implicated in metabolism enables the development of novel antimicrobial live attenuated vaccine strains.
  • a mutant pathogenic bacterium of the invention which may or may not be attenuated, can be incapable of expressing two or more metabolic genes.
  • the two or more metabolic genes can encode proteins that are involved in the same metabolic pathway, or in different metabolic pathways.
  • the metabolic gene or genes may be involved in glycolysis and/or sugar transport.
  • a metabolic gene or genes involved in glycolysis can encode a phosphofructokinase, examples of such genes are pfkA and pfkB.
  • Metabolic genes involved in sugar transport include those of sugar phosphotransferase systems, such as one or more metabolic genes that encode components of a glucose, mannose and/or galactose transport system, for example ptstil, err, glk, ptsG, manXYZ, mglABC and galP.
  • S. Typhimurium causes a self-limited gastroenteritis in humans and results in a systemic typhoid-like disease in mice. Infected mice are frequently used as an experimental model for human typhoid diseases (19).
  • a problem in the development of Salmonella vaccines is the variety of typhoidal and non-typhoidal salmonelloses caused by various serovars and strains. Since the central metabolic pathways of sugar transport and glycolysis are likely to be found in all Salmonella serovars, introduction of mutations into metabolic gene or genes of any disease causing strain of Salmonella may provide potential candidate strains for development as live attenuated vaccines.
  • the invention relates to mutant pathogenic bacteria, preferably mutant intracellular pathogenic bacteria, incapable of expressing:
  • Preferred attenuated strains include mutant pathogenic bacteria, preferably mutant intracellular pathogenic bacteria, incapable of expressing: (a) pfkA and pfkB; (b) ptsHI, err, and/or glk;
  • Particularly preferred attenuated strains are glycolysis mutant strains in which phosphofructokinase is functionally silenced, such as ApfkAB strains in which phosphofructokinase genes pfkA and pfkB are functionally silenced; sugar transport mutant strains in which sugar transport genes are functionally silenced, such as glucose transport mutant strains AptsHlcrrAglk in which ptsHI, err, and glk are functionally silenced; or glucose transport mutant strains ⁇ ptsHI in which ptsHI is functionally silenced.
  • strains in which both glycolysis and sugar transport genes are functionally silenced for example mutant strains in which ApfkAB is combined with the AptsHlcrr and AgIk mutations, or in which ApfkAB is combined with AptsHI; to minimise the risk of reversion of an attenuated strain to virulence it may be advantageous for two independent metabolic pathways to be rendered inoperable, e.g. both glycolysis (e.g., ApfkAB) and sugar transport (e.g., AptsHI, or AptsHlcrr ⁇ glk).
  • the attenuated strain is selected from AptsHI, AptsHlcrr, AptsGAmanXYZAglk, AptsHlcrrAglk or ApfkAB.
  • S. Typhimurium phosphofructokinase is encoded by two genes pfkA and pfkB; both the pfkA and pfkB genes must be mutated, preferably deleted, in order to functionally silence phosphofructokinase and thereby effect attenuation in virulence of S. Typhimurium in the infection models described herein.
  • the S. Typhimurium ⁇ pfkAB mutant was observed to be avirulent in infected mice. The fur of infected mice was found to be slightly ruffled, suggesting that the strain provokes an immunogenic response protective against future infections by S. Typhimurium. This indicates that S. Typhimurium ApfkAB mutant is a promising candidate for a successful live attenuated vaccine for animals and humans.
  • a mutant pathogenic bacterium of the invention which is attenuated is also incapable of expressing one or more other virulence genes, such as aroA, aroC, aroD, crp, cya, htrA, nirB, rpoS, ssaV, surA, Salmonella pathogenicity island 2 (SPI2) or combinations thereof.
  • virulence genes such as aroA, aroC, aroD, crp, cya, htrA, nirB, rpoS, ssaV, surA, Salmonella pathogenicity island 2 (SPI2) or combinations thereof.
  • the attenuated metabolic mutant bacteria of the invention provide a highly immunogenic live oral vaccine, suitable as a carrier of a gene or genes that express protective antigens cloned from other pathogens (20-22).
  • a pathogenic bacterium of the invention may carry a heterologous gene or genes. Given the number of different immunisations that can be administered to humans, pets and farm animals, combined administration of several immunogens is desirable.
  • a live attenuated pathogenic bacterium of the invention can be used as a recombinant carrier for a heterologous gene or genes, encoding antigens selected from other pathogenic microorganisms or viruses. Administration of such a recombinant carrier has the advantage that protective immunity can be induced against two or more diseases at the same time.
  • the live attenuated mutant pathogenic bacteria according to the present invention provide suitable carriers for heterologous genes.
  • recombinant bacteria mutant pathogenic bacteria of the invention, into which a heterologous gene has been inserted
  • a heterologous gene can be obtained from pathogenic bacteria; suitable bacterial heterologous genes include a gene encoding a bacterial toxin, such as an Actinobacillus pleuropneumoniae toxin, Clostridium toxin, outer membrane protein and the like.
  • a heterologous gene can, be derived from pathogenic herpesviruses (e.g., the genes encoding the structural proteins of herpesviruses), retroviruses (e.g., the gp160 envelope protein), adenoviruses and the like. Another possibility is insertion of a heterologous a gene encoding a protein involved in triggering the immune system, such as an interleukin or an interferon, or another gene involved in immune regulation.
  • a pathogenic bacterium of the invention can be a bacterial intracellular pathogen, such as a virulent strain of Salmonella, Bartonella, Burkholderia, Brucella, Chlamydia, Coxiella, Ehrlichia, Francisella, Legionella, Listeria, Mycobacterium, Nocardia, Rickettsia, Shigella, Tropheryma or Yersinia.
  • the pathogenic bacterium is a typhoidal Salmonella serovar, such as a Salmonella enterica Typhi or Paratyphi.
  • the Salmonella enterica Typhi can be a Salmonella Typhimurium.
  • the invention further provides a live attenuated vaccine for protection against infection with a pathogenic bacterium or the pathogenic effects thereof, comprising a live attenuated mutant pathogenic bacterium of the invention and a pharmaceutically acceptable carrier.
  • a vaccine according to the present invention also contains a pharmaceutically acceptable carrier, such as a vaccine-compatible pharmaceutically acceptable (i.e., sterile and non-toxic) liquid, semisolid, or solid diluent.
  • a pharmaceutically acceptable carrier such as a vaccine-compatible pharmaceutically acceptable (i.e., sterile and non-toxic) liquid, semisolid, or solid diluent.
  • Liquid diluents include water, but may, for example also comprise culture fluid in which the bacteria were cultured.
  • Another suitable liquid carrier is a solution of physiological salt concentration. Any carrier known in the art may be used.
  • Exemplary carriers include, but are not limited to, polyoxyethylene sorbitan monolaurate, magnesium stearate, methyl- and propylhydroxybenzoate, talc, alginates, starches, lactose, sucrose, dextrose, sorbitol, mannitol, gum acacia, calcium phosphate, mineral oil, cocoa butter, and oil of theobroma.
  • Pharmaceutically acceptable carriers useful in the present invention may comprise stabilisers such as SPGA, carbohydrates (e.g. sorbitol, mannitol, starch, sucrose, glucose, dextrin), proteins for example albumin or casein, protein-containing agents such as bovine serum or skimmed milk and buffers (e.g., phosphate buffer). When such stabilisers are present in the vaccine, the vaccine can be freeze-dried. Thus, a live attenuated vaccine of the invention can be provided in freeze-dried form.
  • the vaccine compositions can be packaged in forms convenient for delivery.
  • the compositions can be enclosed within a capsule, caplet, sachet, cachet, gelatin, paper, or other container. These delivery forms are preferred when compatible with entry of the immunogenic composition into the recipient organism and, particularly, when the immunogenic composition is being delivered in unit dose form.
  • the dosage units can be packaged, e.g., in tablets, capsules, suppositories or cachets.
  • the vaccine compositions may be introduced into the subject to be immunized by any conventional method including, e.g., by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, or subcutaneous injection; by oral, transdermal, sublingual, intranasal, anal, or vaginal, delivery. In a preferred embodiment, the vaccine is delivered intraperitoneal.
  • the useful dosage to be administered will vary depending on the age, weight and animal vaccinated, the mode of administration and the type of pathogen against which immunisation is sought.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • the vaccine may comprise any dose of bacteria, sufficient to evoke an immune response.
  • the dose can be 10 9 bacteria or less.
  • a live attenuated vaccine of the invention may comprise an adjuvant.
  • one or more compounds having adjuvant activity may be included in the vaccine.
  • Adjuvants are non-specific stimulators of the immune system. They enhance the immune response of the host to the vaccine. Examples of adjuvants known in the art are Freund's Complete and Incomplete adjuvant, vitamin E, non-ionic block polymers, muramyldipeptides, ISCOMs (immune stimulating complexes), saponins, mineral oil, vegetable oil, and Carbopol.
  • Adjuvants, especially suitable for mucosal application are, for example, E. coli heat-labile toxin (LT) or Cholera toxin (CT).
  • Suitable adjuvants are for example aluminium hydroxide, aluminium phosphate or aluminium oxide, oil-emulsions (e.g., of Bayol F ® or Marcol 52 ® , saponins or vitamin-E solubilisate).
  • the vaccine according to the present invention can be given orally, intranasally, intradermal ⁇ , subcutaneously, by aerosol or intramuscularly.
  • the live attenuated vaccine is formulated for oral administration.
  • the invention yet further provides for the use of a live attenuated pathogenic bacterium of the invention for the manufacture of a vaccine for the protection of humans or animals against infection with a pathogenic bacterium or against the pathogenic effects of infection.
  • a method for the preparation of a vaccine comprising admixing a live attenuated pathogenic bacterium according to the invention with a pharmaceutically acceptable carrier.
  • the invention provides a method for immunizing a human or animal against infection with a pathogenic bacterium, comprising administering to the human or animal a vaccine of the invention.
  • the invention relates to a method of confirming that a bacterial strain is a mutant, potentially attenuated, pathogenic bacterial strain by virtue of not expressing one or more metabolic genes, which comprises confirming that one or more metabolic genes are functionally silenced. Additionally, the invention relates to a method of confirming that a bacterial strain is a mutant attenuated pathogenic bacterial strain by virtue of not expressing one, two or more metabolic genes. Such methods may comprise identifying a mutation in the metabolic gene or genes, and/or a mutation in sequences that regulate expression of the metabolic gene or genes. The mutation identified can be a deletion, an insertion, a substitution or a combination thereof, provided that the mutation leads to the failure to express a functional protein (functional silencing).
  • the mutation identified is a deletion.
  • the mutation identified is deletion of the entire coding sequence of the metabolic gene or genes; however, the mutation identified can be, for example, a partial deletion of the gene or a mutation in sequences that regulate expression of the metabolic gene or genes provided that the mutation results in functional silencing.
  • Methods for identifying such mutations include sequencing, restriction enzyme digestion, PCR or other hybridisation- based methods such as in situ hybridisation, northern or southern blotting, and the like. Analysis of proteins expressed may be performed to confirm functional silencing of the metabolic gene or genes; protein analysis methods may involve detection of absence of expression of protein from the metabolic gene or genes, or detection of expression of a non-functional mutant form of the protein.
  • Analysis of proteins expressed may comprise use of a ligand or ligands specific for the protein encoded by the metabolic gene(s) to identify absence of functional protein, or to identify presence of a mutated, non-functional form of protein encoded by the metabolic gene(s).
  • Methods that employ specific ligands include immunological methods based on use of specific antibodies, such as ELISA, immunostaining and western blotting methods.
  • Analysis of protein may involve testing to determine if protein encoded by the metabolic gene(s) is active.
  • Fig. 1 Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and ⁇ pfkAB strains in RAW 264.7 macrophages. The data are the statistical mean from three biological replicate experiments.
  • Fig. 2 Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and ApfkAB strains in HeLa epithelial cells. The data are the statistical mean from three biological replicate experiments.
  • Fig. 3. Salmonella cfu's from BALB/c mouse livers and spleens (n 5) after 72 hours infection.
  • Fig. 4 Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and AptsHlcrr ⁇ glk strains in RAW macrophages. The data are the statistical mean from three biological replicate experiments.
  • Fig. ⁇ Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and AptsHlcrr ⁇ glk strains in cultured human HeLa epithelial cells. The data are the statistical mean from three biological replicate experiments.
  • Example 1 Mutant construction and testing
  • oligonucleotides were designed that contain flanking DNA sequences of the target genes to be deleted (Table 1).
  • the oligonucleotides were used to amplify, by polymerase chain reaction (PCR), a cassette containing the kanamycin or the chloramphenicol resistance gene carried by plasmids pKD4 and pKD3 respectively (Table 2; (23)).
  • PCR polymerase chain reaction
  • the resulting PCR product was then transformed by electroporation into Salmonella Typhimurium 4/74 (24) carrying plasmid pKD46 (Table 2).
  • Plasmid pKD46 encodes a recombinase enzyme that facilitates homologous recombination of the target gene with the kanamycin or the chloramphenicol resistance cassette resulting in complete replacement of the deleted gene.
  • the kanamycin/chloramphenicol resistance region of the deleted gene was transferred by P22 transduction into S. Typhimurium wild type strain 4/74 (25) to avoid unwanted genetic recombination events that may have occurred during the original transformation.
  • the transductants were screened on green agar plates to obtain lysogen free colonies (26).
  • the kanamycin or chloramphenicol resistance gene was then removed via a further recombination event by transforming the cells with the FLP-recombinase expression plasmid, pCP20 (27).
  • the FLP- recombinase removed the kanamycin or chloramphenicol resistance gene and the strains were cured of the pCP20 plasmid by growing the strain at 42 0 C.
  • Primer Name Primer DNA sequence pfkaredf CAATAGATTTCATTTTGCATTCCAAAGTTCAG
  • TTACTGCCATATGAATATCCTCCTTAG manxredf TGTCAAGTTGATGTGTTGACAATAATAAAG GAGGTAGCAAGTGTAGGCTGGAGCTGCTTC manzredr AAAAAACGGGGCCGTTTGGCCCCGGTAGT GTACAACAGCCCATATGAATATCCTCCTTAG
  • murine macrophages (RAW264.7; obtained from American Type Culture Collection, Rockville, MD) were grown in MEM medium (Sigma) supplemented with 10% fetal bovine serum (FBS; Sigma), L-glutamine (2 mM final concentration; Sigma) and 1x non-essential amino acids (Sigma) as described (28).
  • FBS fetal bovine serum
  • L-glutamine (2 mM final concentration; Sigma)
  • 1x non-essential amino acids Sigma
  • 1x10 5 macrophage cells were seeded into each well of a 12-well cell culture plate and infected with complement-opsonized S.
  • MOI multiplicity of infection
  • bacteria were grown overnight on Luria broth plates at 37 0 C and suspended in phosphate buffered saline (PBS) before opsonization.
  • PBS phosphate buffere
  • Phosphofructokinase is required for intracellular replication and survival of S. Typhimurium in cultured RAW macrophages.
  • Glycolysis is the sequence of catabolic reactions that converts sugars into pyruvate with the concomitant synthesis of ATP and NADH. It is the foundation of both aerobic and anaerobic respiration and is found in nearly all organisms (33).
  • the enzyme phosphofructokinase irreversibly converts ⁇ -D-fructose 6-phosphate into ⁇ -D-fructose1 , 6-bisphosphate and is encoded by two genes in most bacteria (pfkA and pfkB) (34).
  • An S. Typhimurium mutant strain in which the pfkAB genes were completely deleted was constructed according to procedures described above.
  • the marker antibiotic resistance genes (kanamycin and chloramphenicol) were completely removed from the chromosome of the S. Typhimurium ApfkAB strain. The growth of the marker-free S. Typhimurium ApfkAB strain was inhibited by kanamycin and chloramphenicol.
  • Phosphofructokinase is required for intracellular replication of S. Typhimurium in cultured HeLa epithelial cells.
  • the intracellular replication rates of the S. Typhimurium 4/74 wild type and ApfkAB mutant strains were determined in cultured human HeLa epithelial cells. A comparison of the intracellular replication rate of S. Typhimurium ApfkAB bacteria between 2 and 6 hours showed that the mutant strain replicated less than 2-fold in HeLa cells (Fig. 2, Table 3B).
  • Phosphofructokinase is avirulent in the mouse typhoid infection model.
  • mice were infected (IP) with the S. Typhimurium ⁇ pfkAB mutant and the wild type strain. Salmonella were recovered and enumerated from the spleen and liver of mice after 72 hours infection. The results demonstrated that the S. Typhimurium ApfkAB mutant was severely attenuated by approximately 10 2 fold compared to the wild type strain in the spleen and liver of infected mice. (Fig. 3, Table 4). Mice infected with the S. Typhimurium wild type strain showed severe symptoms of systemic disease after 72 hours. The fur of mice infected with the S. Typhimurium ApfkAB strain was slightly ruffled after 72 hours.
  • S. Typhimurium mutant strain containing complete deletions of the ptsHI, err and glk genes was constructed according to the procedures described above.
  • the ptsHI and err genes encode components of a phosphotransferase (PTS) system, which detects, transports, and phosphorylates several sugars and sugar derivatives into the bacterial cell (35).
  • the glk gene encodes glucose kinase, which specifically phosphorylates intracellular glucose (36).
  • In vitro growth experiments on M9 minimal salts media showed that, as expected, the S. Typhimurium AptsHlcrr ⁇ glk mutant was unable to utilize glucose for growth (37). Infection assays of the S.
  • the intracellular phenotype of the S. Typhimurium ApfkAB mutant demonstrated that phosphofructokinase and glycolysis are required for intracellular replication and survival of S. Typhimurium in RAW macrophages.
  • the S. Typhimurium ApfkAB mutant was unable to replicate within human HeLa epithelial cells suggesting phosphofructokinase and glycolysis are necessary for growth within this cell type.
  • the S. Typhimurium pfkAB mutant is severely attenuated during intraperitoneal infection of BALB/c mice demonstrating that phosphofructokinase and glycolysis are required for Salmonella survival and replication within mice.
  • Example 2 Oral live vaccine strain verification
  • Experiment 1 Attenuation of potential vaccine strains
  • mice with three or more of these symptoms scored as highly developed will constitute the end point of the experiment and animals will be humanely killed. Any dyspnoeic animals will be killed at first sight.
  • the results from this experiment are shown in Table 5. According to the above scoring procedure, after 4 days, mice infected with the wild-type strain displayed all the symptoms of a severe systemic typhoid infection and were humanely killed.
  • the S. Typhimurium AptsGAmanXYZAglk strain displayed attenuated upon infection and mice were killed after 9 days (Table 5).
  • the S. Typhimurium ApfkAB, AptsHlcrrAglk, AptsHlcrr, AptsHI strains were avirulent upon infection and continued to survive past 3 weeks post infection (Table 5).
  • Experiment 1 is repeated and bacterial cfu's enumerated from spleen, liver, mesenteric lymph nodes and Peyer's patches every 14 days for a total of 50 days as described in Experiment 1.
  • a positive outcome with respect to strain validation as potential vaccines would be no cfu's are recovered from the organs of mice infected with attenuated metabolism mutant strain(s), a positive control aroA strain, or LB control (sterility).
  • Experiment 2 is repeated after 30 days infection using potential vaccine strain(s), S. Typhimurium ⁇ aroA strain and an LB control. Faecal and blood samples are collected after day 30. Faecal samples are weighed, and 1.0 ml of PBS plus 0.1 % sodium azide is added per 100mg of faeces. The faecal pellets are dispersed by microtip sonication for 10 to 20 second bursts at 50% duty. The faecal suspensions are subsequently pelleted in a microfuge for 5 min, and the supernatants transferred to a fresh tube and held frozen until assays are performed.
  • Bound antibody is detected by the addition of an anti-mouse Ig Horseradish peroxidase conjugate diluted 1/1000 in PBST containing 0.4% skim milk powder and incubated for 2 h at 37 0 C. Reactions are developed using immunopure o- phenylenediamine with H 2 O 2 as the substrate. Absorbance is read at 492 nm in a plate reader. The serum antibody titre is designated as the reciprocal of the dilution of specific antibody that gives an OD 492 value of five times the background value.
  • BALB/c mice are pre-infected for 30 days with the potential vaccine strain(s) and the S. Typhimurium AaroA strain and LB control at doses of 1-2 x 10 9 bacteria according to experiment 1.
  • the pre-infected mice are then challenged at day 30 via oral infection according to Experiment 1 with 1x10 8 cfu of a S. Typhimurium wild type strain (4/74) to assess the protection conferred by the vaccine compared to control BALB/c mice.
  • Mice successfully immunised following infection with the metabolism mutant strain(s) and aroA strains show no symptoms of typhoid fever according to the accepted scoring system.
  • cfu's in above organs are enumerated and ELISA assays are performed to detect the presence of antibodies against Salmonella LPS (see Experiment 3 for methodology).
  • Troutt H. F., Galland, J. C, Osburn, B.I., Brewer, R. L., Braun, R.K., Schmitz, J.A., Sears, P., Childers, A.B., Richey, E., Mather, E. et al. (2001) Prevalence of Salmonella spp in cull (market) dairy cows at slaughter. Journal of the American Veterinary Medical Association, 219, 1212-1215.
  • primer sequence crrredr ⁇ 400> 6 aaatggcgcc caaaggcgcc attcttcact gcggcaagaa catatgaata tcctccttag 60 ⁇ 210> 7 ⁇ 211 > 60 ⁇ 212> DNA ⁇ 213> Artificial ⁇ 220>

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Mycology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to a mutant pathogenic bacterium, preferably a mutant intracellular pathogenic bacterium, that is incapable of expressing one or more metabolic genes; preferably the mutant pathogenic bacterium is attenuated. A mutant pathogenic bacterium of the invention may be incapable of expressing two or more metabolic genes, which encode proteins involved in the same or in different metabolic pathways, such as glycolysis and sugar transport. The invention also relates to vaccines comprising a live attenuated mutant pathogenic bacterium of the invention, methods for their production and their use for the protection of humans or animals against infection with a pathogenic bacterium or against the pathogenic effects of infection.

Description

Mutant Pathogenic Bacteria and Live Attenuated Vaccine Compositions
Field of the Invention
The present invention relates to mutant pathogenic bacteria, particularly mutant intracellular pathogenic bacteria, useful in the preparation of attenuated pathogenic bacteria, to attenuated pathogenic bacteria, to such bacteria carrying a heterologous gene, to vaccines comprising live attenuated pathogenic bacteria, optionally carrying a heterologous gene, useful for the prevention of microbial pathogenesis, and to methods for the preparation of such vaccines and bacteria.
Background to the Invention
The mammalian cell provides a uniquely stable environment for the replication and survival of intracellular bacteria. The host cell provides constant temperature and pH regulation, a ready supply of nutrients for growth, and protection against antimicrobial agents. Intracellular bacteria can be divided into strict and facultative bacteria (1). Strict intracellular bacteria cannot be cultivated in broth medium and can replicate only in vivo or in tissue-cultured cells in vitro. Strict intracellular bacterial pathogens include Chlamydia (responsible for genital and ophthalmic infections in humans), Coxiella (the etiological agent of Q fever), Ehrlichia (headache, fever and chills in humans) and the Rikettsias. In contrast, facultative intracellular bacteria are able to replicate in broth medium as well as within mammalian cells. Certain facultative intracellular pathogens are able to enter and multiply in phagocytic cells such as neutrophils or macrophages. The intracellular environment of a phagocyte may protect the bacteria during the early stages of infection or until they develop a full complement of virulence factors. The intracellular environment also guards the bacteria against the activities of extracellular bactericides, antibodies, drugs, etc. Facultative intracellular parasites that are able to replicate in phagocytic cells include Yersinia, Legionella pneumophila, Salmonella, Mycobacterium tuberculosis and Mycobacerium leprae, the agents of tuberculosis and leprosy respectively. Other facultative intracellular pathogens are able to replicate in nonphagocytic cells and include Bartonella, associated with several human diseases, Brucella, Franciscella tularensis, Listeria, Salmonella and Tropheryma whippelii (associated with Whipple's disease).
Examples of bacterial intracellular pathogens (1) and the diseases that they cause are:
Organism Disease
Mycobacterium tuberculosis Tuberculosis
Mycobacterium leprae Leprosy
Listeria monocytogenes Listeriosis
Salmonella typhi Typhoid Fever
Shigella dysenteriae Bacillary dysentery
Yersinia pestis Plague
Brucella species Brucellosis
Legionella pneumophila Pneumonia
Rickettsiae Typhus; Rocky Mountain Spotted Fever
Chlamydia Chlamydia; Trachoma
Burkholderia mallei Glanders
Burkholderia pseudomallei Melioidosis
Although there is a long history of success in developing vaccines against extracellular bacterial pathogens, the development of vaccines against intracellular bacterial pathogens has proven to be more challenging (2). Intracellular bacteria have developed an array of weaponry that enables them to evade host cell immune mechanisms. These include slow growth phenotypes in phagocytic cells, and the ability to shield themselves from host cell defence mechanisms. The invasion and growth of intracellular pathogens in host cells also involves many virulence factors acting in concert. Therefore, unlike extracellular pathogens, it is unusual to identify a single gene product such as a toxin, which provides an overriding role in the pathogenesis of the disease and may provide a target for antimicrobials. Furthermore, antigens that play roles in the pathogenesis of disease caused by intracellular pathogens are likely to have limited visibility to the immune system and are generally not good targets for antibody mediated protection. Although antibodies have been developed that provide protection against some intracellular pathogens, the level of protection that they offer is relatively low. For example, antibodies can protect against low but not high virulence strains of F. tularensis (3), or can provide protection against low challenge doses of B. pseudomallei administered by the intraperitoneal route, but not administered by the aerosol route of infection (4,5).
The need to devise effective vaccines against intracellular bacteria has been driven by the emergence of multi-antibiotic resistant strains over the past 20 - 30 years, which pose a serious future threat for human and animal welfare. Furthermore, certain intracellular bacterial pathogens may act as candidates for biowarfare agents. For example, Brucella suis, Brucella melitensis, Brucella abortus, Francisella tularensis, Burkholderia mallei and Burkholdeήa pseudomallei are strains that may form a basis for the development of putative biowarfare agents.
Salmonella is a facultative intracellular pathogen that causes gastroenteritis and fatal systemic disease in mammals including humans, cattle and pigs. Typhoidal Salmonella serovars, such as Salmonella enterica serovars Typhi and Paratyphi cause an estimated 20 million cases of salmonellosis and 200,000 human deaths worldwide per annum (6). Salmonella infections occur as a result of ingestion of contaminated food and water. An important source of Salmonella infections in humans arises from contaminated poultry, pig, and cattle products. A randomised UK National Survey carried out in 2004 for faecal carriage of Salmonella in slaughter pigs, cattle and sheep identified S. Typhimurium in 11.1 % of caecal samples and 2.1 % of carcass swabs of 2509 pigs (7). Other studies have shown a high prevalence of S. Typhimurium in cull cattle, especially after transport. In one survey a pre-transport prevalence of 1 % increased to 21 % at the abattoir (8). A further study found 21.3% of 5087 caecal and colonic samples from cull dairy cows in the USA contained Salmonella (9).
Because salmonellosis has such global effects on human health, development of reliable vaccines is critical (for review see (10)). Many vaccines using Salmonella mutants have failed due to either over- or underattenuation of the vaccine strain (11 ,12). In systemic infections, Salmonella penetrates the small intestinal barrier by invading gut epithelial cells. From there Salmonella can gain access to the mesenteric lymph nodes where the bacteria are engulfed by phagocytic cells such as macrophages. Once inside macrophages, Salmonella is compartmentalised into an intracellular phagosome, which is modified to become the "Salmonella containing vacuole" (SCV). The SCV acts as a shield that prevents lysosomal fusion and exposure to host cell antimicrobial agents (13,14). Salmonella also resides within an SCV during invasion of epithelial cells (15). The Salmonella must acquire nutrients for replication in epithelial cells and macrophages.
It has previously been suggested that due to metabolic redundancy, the scope for antimicrobials based on the exploitation of metabolism-related genes was limited (16).
Statement of Invention
The invention provides a mutant pathogenic bacterium, preferably a mutant intracellular pathogenic bacterium, that is incapable of expressing one or more metabolic genes.
Such a mutation can be a deletion, an insertion, a substitution or a combination thereof, provided that the mutation leads to the failure to express a functional protein (functional silencing). Preferably the mutation is a deletion. Preferably the metabolic gene or genes are deleted, however other mutations, for example partial deletion of the gene or mutation in sequences that regulate expression of the metabolic gene or genes may be used to achieve functional silencing.
Preferably the mutant pathogenic bacterium is attenuated as a result of functional silencing of one or more metabolic genes. However, the invention also relates to mutant pathogenic bacteria that are incapable of expressing one or more metabolic genes, but are not attenuated, such mutants can be subjected to further mutation in metabolic or other virulence genes to provide a multiple mutant attenuated form of the pathogenic bacteria. Surprisingly, it has been found that mutation in one or multiple genes that involve proteins implicated in metabolism enables the development of novel antimicrobial live attenuated vaccine strains.
A mutant pathogenic bacterium of the invention, which may or may not be attenuated, can be incapable of expressing two or more metabolic genes. The two or more metabolic genes can encode proteins that are involved in the same metabolic pathway, or in different metabolic pathways. The metabolic gene or genes may be involved in glycolysis and/or sugar transport. A metabolic gene or genes involved in glycolysis can encode a phosphofructokinase, examples of such genes are pfkA and pfkB. Metabolic genes involved in sugar transport include those of sugar phosphotransferase systems, such as one or more metabolic genes that encode components of a glucose, mannose and/or galactose transport system, for example ptstil, err, glk, ptsG, manXYZ, mglABC and galP.
Until the present invention, metabolic pathways such as glycolysis and sugar transport were not identified as necessary for virulence. The present inventors are the first to recognise that metabolic genes involved in glycolysis and/or sugar transport are required for infection and thus that mutation in genes involved in these processes may provide attenuated strains of intracellular pathogenic bacteria useful as vaccines. Multiple signature tagged mutagenesis (STM) screens would not have identified glycolysis and sugar transport as being required for infection (17,18).
The inventors have investigated substrates and metabolic pathways required by S. Typhimurium for the infection of cultured murine macrophages and human epithelial cells and for the systemic infection of mice. S. Typhimurium causes a self-limited gastroenteritis in humans and results in a systemic typhoid-like disease in mice. Infected mice are frequently used as an experimental model for human typhoid diseases (19). A problem in the development of Salmonella vaccines is the variety of typhoidal and non-typhoidal salmonelloses caused by various serovars and strains. Since the central metabolic pathways of sugar transport and glycolysis are likely to be found in all Salmonella serovars, introduction of mutations into metabolic gene or genes of any disease causing strain of Salmonella may provide potential candidate strains for development as live attenuated vaccines.
In particular embodiments, the invention relates to mutant pathogenic bacteria, preferably mutant intracellular pathogenic bacteria, incapable of expressing:
(a) pfkA and/or pfkB;
(b) ptsHI, err, and/or glk;
(c) pfkA, pfkB, ptsHI, err, and glk;
(d) pfkA, pfkB, ptsHI, and err;
(e) pfkA, pfkB and glk,
(f) pfkA, pfkB and ptsHI
(g) pfkA, pfkB, ptsHI and glk (h) ptsHI;
(i) ptsHI, err
0) ptsHI, glk
(k) glk, ptsG and manXYZ.
(I) pfkA, pfkB, glk, ptsG and/or manXYZ
(m) mglABC and/or galP
(n) mglABC, galP, ptsHI
(o) mglABC, galP, ptsHI and err
(p) mglABC, galP, ptsHI
(q) mglABC, galP, ptsG and manXYZ
(r) pfkA, pfkB, ptsHI, err, mglABC and galP
(s) pfkA, pfkB, ptsHI, mglABC and galP; or,
(t) pfkA, pfkB, mglABC and gal.
Preferred attenuated strains include mutant pathogenic bacteria, preferably mutant intracellular pathogenic bacteria, incapable of expressing: (a) pfkA and pfkB; (b) ptsHI, err, and/or glk;
(c) pfkA, pfkB, ptsHI, err, and glk;
(d) p/ikA, pfkB, ptsHI, and err; (Θ) pfkA, pfkB and glk,
(f) pfkA, pfkB and ptsHI
(g) ptsHI;
(h) ptsHI, err;
(i) ptsHI, glk; or,
(f) glk, ptsG and manXYZ.
(k) ptsHI, mglABC and galP
(I) ptsHI, err, mglABC and galP
(m) pfkA, pfkB, mglABC and gal P
(n) pfkA, pfkB, ptsHI, err, mglABC and galP
(o) pfkA, pfkB, ptsHI, mglABC and gal.
Particularly preferred attenuated strains are glycolysis mutant strains in which phosphofructokinase is functionally silenced, such as ApfkAB strains in which phosphofructokinase genes pfkA and pfkB are functionally silenced; sugar transport mutant strains in which sugar transport genes are functionally silenced, such as glucose transport mutant strains AptsHlcrrAglk in which ptsHI, err, and glk are functionally silenced; or glucose transport mutant strains ΔptsHI in which ptsHI is functionally silenced. Also preferred are strains in which both glycolysis and sugar transport genes are functionally silenced, for example mutant strains in which ApfkAB is combined with the AptsHlcrr and AgIk mutations, or in which ApfkAB is combined with AptsHI; to minimise the risk of reversion of an attenuated strain to virulence it may be advantageous for two independent metabolic pathways to be rendered inoperable, e.g. both glycolysis (e.g., ApfkAB) and sugar transport (e.g., AptsHI, or AptsHlcrrΔglk). Thus, in a preferred embodiment, the attenuated strain is selected from AptsHI, AptsHlcrr, AptsGAmanXYZAglk, AptsHlcrrAglk or ApfkAB.
In S. Typhimurium, phosphofructokinase is encoded by two genes pfkA and pfkB; both the pfkA and pfkB genes must be mutated, preferably deleted, in order to functionally silence phosphofructokinase and thereby effect attenuation in virulence of S. Typhimurium in the infection models described herein. The S. Typhimurium ΔpfkAB mutant was observed to be avirulent in infected mice. The fur of infected mice was found to be slightly ruffled, suggesting that the strain provokes an immunogenic response protective against future infections by S. Typhimurium. This indicates that S. Typhimurium ApfkAB mutant is a promising candidate for a successful live attenuated vaccine for animals and humans.
The inability of the S. Typhimurium AptsHlcrrAglk mutant to replicate in RAW 264.7 macrophages and HeLa epithelial cells and impairment of survival in RAW macrophages indicates potential use of this strain as a live attenuated vaccine.
In a further aspect, a mutant pathogenic bacterium of the invention, which is attenuated is also incapable of expressing one or more other virulence genes, such as aroA, aroC, aroD, crp, cya, htrA, nirB, rpoS, ssaV, surA, Salmonella pathogenicity island 2 (SPI2) or combinations thereof.
The attenuated metabolic mutant bacteria of the invention provide a highly immunogenic live oral vaccine, suitable as a carrier of a gene or genes that express protective antigens cloned from other pathogens (20-22). A pathogenic bacterium of the invention may carry a heterologous gene or genes. Given the number of different immunisations that can be administered to humans, pets and farm animals, combined administration of several immunogens is desirable. A live attenuated pathogenic bacterium of the invention can be used as a recombinant carrier for a heterologous gene or genes, encoding antigens selected from other pathogenic microorganisms or viruses. Administration of such a recombinant carrier has the advantage that protective immunity can be induced against two or more diseases at the same time. The live attenuated mutant pathogenic bacteria according to the present invention provide suitable carriers for heterologous genes.
The construction of recombinant carriers can be performed using standard molecular biology techniques. Therefore, the invention also relates to mutant pathogenic bacteria of the invention, into which a heterologous gene has been inserted (herein termed "recombinant bacteria" or "recombinant carriers"). A heterologous gene can be obtained from pathogenic bacteria; suitable bacterial heterologous genes include a gene encoding a bacterial toxin, such as an Actinobacillus pleuropneumoniae toxin, Clostridium toxin, outer membrane protein and the like. A heterologous gene can, be derived from pathogenic herpesviruses (e.g., the genes encoding the structural proteins of herpesviruses), retroviruses (e.g., the gp160 envelope protein), adenoviruses and the like. Another possibility is insertion of a heterologous a gene encoding a protein involved in triggering the immune system, such as an interleukin or an interferon, or another gene involved in immune regulation.
A pathogenic bacterium of the invention can be a bacterial intracellular pathogen, such as a virulent strain of Salmonella, Bartonella, Burkholderia, Brucella, Chlamydia, Coxiella, Ehrlichia, Francisella, Legionella, Listeria, Mycobacterium, Nocardia, Rickettsia, Shigella, Tropheryma or Yersinia. Preferably the pathogenic bacterium is a typhoidal Salmonella serovar, such as a Salmonella enterica Typhi or Paratyphi. The Salmonella enterica Typhi can be a Salmonella Typhimurium.
The invention further provides a live attenuated vaccine for protection against infection with a pathogenic bacterium or the pathogenic effects thereof, comprising a live attenuated mutant pathogenic bacterium of the invention and a pharmaceutically acceptable carrier.
In addition to an immunogenically effective amount of the live attenuated mutant pathogenic bacteria of the invention, a vaccine according to the present invention also contains a pharmaceutically acceptable carrier, such as a vaccine-compatible pharmaceutically acceptable (i.e., sterile and non-toxic) liquid, semisolid, or solid diluent. Liquid diluents include water, but may, for example also comprise culture fluid in which the bacteria were cultured. Another suitable liquid carrier is a solution of physiological salt concentration. Any carrier known in the art may be used. Exemplary carriers include, but are not limited to, polyoxyethylene sorbitan monolaurate, magnesium stearate, methyl- and propylhydroxybenzoate, talc, alginates, starches, lactose, sucrose, dextrose, sorbitol, mannitol, gum acacia, calcium phosphate, mineral oil, cocoa butter, and oil of theobroma. Pharmaceutically acceptable carriers useful in the present invention may comprise stabilisers such as SPGA, carbohydrates (e.g. sorbitol, mannitol, starch, sucrose, glucose, dextrin), proteins for example albumin or casein, protein-containing agents such as bovine serum or skimmed milk and buffers (e.g., phosphate buffer). When such stabilisers are present in the vaccine, the vaccine can be freeze-dried. Thus, a live attenuated vaccine of the invention can be provided in freeze-dried form.
The vaccine compositions can be packaged in forms convenient for delivery. The compositions can be enclosed within a capsule, caplet, sachet, cachet, gelatin, paper, or other container. These delivery forms are preferred when compatible with entry of the immunogenic composition into the recipient organism and, particularly, when the immunogenic composition is being delivered in unit dose form. The dosage units can be packaged, e.g., in tablets, capsules, suppositories or cachets. The vaccine compositions may be introduced into the subject to be immunized by any conventional method including, e.g., by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, or subcutaneous injection; by oral, transdermal, sublingual, intranasal, anal, or vaginal, delivery. In a preferred embodiment, the vaccine is delivered intraperitoneal.
The useful dosage to be administered will vary depending on the age, weight and animal vaccinated, the mode of administration and the type of pathogen against which immunisation is sought. The treatment may consist of a single dose or a plurality of doses over a period of time.
The vaccine may comprise any dose of bacteria, sufficient to evoke an immune response. Suitably the dose can be 109 bacteria or less.
A live attenuated vaccine of the invention may comprise an adjuvant. Optionally, one or more compounds having adjuvant activity may be included in the vaccine. Adjuvants are non-specific stimulators of the immune system. They enhance the immune response of the host to the vaccine. Examples of adjuvants known in the art are Freund's Complete and Incomplete adjuvant, vitamin E, non-ionic block polymers, muramyldipeptides, ISCOMs (immune stimulating complexes), saponins, mineral oil, vegetable oil, and Carbopol. Adjuvants, especially suitable for mucosal application are, for example, E. coli heat-labile toxin (LT) or Cholera toxin (CT). Other suitable adjuvants are for example aluminium hydroxide, aluminium phosphate or aluminium oxide, oil-emulsions (e.g., of Bayol F® or Marcol 52®, saponins or vitamin-E solubilisate).
For administration to animals or humans, the vaccine according to the present invention can be given orally, intranasally, intradermal^, subcutaneously, by aerosol or intramuscularly. For application to poultry, wing web and eye-drop administration are appropriate routes. Preferably the live attenuated vaccine is formulated for oral administration.
The invention yet further provides for the use of a live attenuated pathogenic bacterium of the invention for the manufacture of a vaccine for the protection of humans or animals against infection with a pathogenic bacterium or against the pathogenic effects of infection.
Additionally, there is provided a method for the preparation of a vaccine, comprising admixing a live attenuated pathogenic bacterium according to the invention with a pharmaceutically acceptable carrier.
Also, the invention provides a method for immunizing a human or animal against infection with a pathogenic bacterium, comprising administering to the human or animal a vaccine of the invention.
Furthermore, the invention relates to a method of confirming that a bacterial strain is a mutant, potentially attenuated, pathogenic bacterial strain by virtue of not expressing one or more metabolic genes, which comprises confirming that one or more metabolic genes are functionally silenced. Additionally, the invention relates to a method of confirming that a bacterial strain is a mutant attenuated pathogenic bacterial strain by virtue of not expressing one, two or more metabolic genes. Such methods may comprise identifying a mutation in the metabolic gene or genes, and/or a mutation in sequences that regulate expression of the metabolic gene or genes. The mutation identified can be a deletion, an insertion, a substitution or a combination thereof, provided that the mutation leads to the failure to express a functional protein (functional silencing). Preferably the mutation identified is a deletion. Preferably the mutation identified is deletion of the entire coding sequence of the metabolic gene or genes; however, the mutation identified can be, for example, a partial deletion of the gene or a mutation in sequences that regulate expression of the metabolic gene or genes provided that the mutation results in functional silencing. Methods for identifying such mutations include sequencing, restriction enzyme digestion, PCR or other hybridisation- based methods such as in situ hybridisation, northern or southern blotting, and the like. Analysis of proteins expressed may be performed to confirm functional silencing of the metabolic gene or genes; protein analysis methods may involve detection of absence of expression of protein from the metabolic gene or genes, or detection of expression of a non-functional mutant form of the protein. Analysis of proteins expressed may comprise use of a ligand or ligands specific for the protein encoded by the metabolic gene(s) to identify absence of functional protein, or to identify presence of a mutated, non-functional form of protein encoded by the metabolic gene(s). Methods that employ specific ligands include immunological methods based on use of specific antibodies, such as ELISA, immunostaining and western blotting methods. Analysis of protein may involve testing to determine if protein encoded by the metabolic gene(s) is active.
List of Figures
Fig. 1. Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and ΔpfkAB strains in RAW 264.7 macrophages. The data are the statistical mean from three biological replicate experiments.
Fig. 2. Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and ApfkAB strains in HeLa epithelial cells. The data are the statistical mean from three biological replicate experiments. Fig. 3. Salmonella cfu's from BALB/c mouse livers and spleens (n=5) after 72 hours infection.
Fig. 4. Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and AptsHlcrrΔglk strains in RAW macrophages. The data are the statistical mean from three biological replicate experiments.
Fig. β. Intracellular replication rate assays of S. Typhimurium 4/74 wild type (WT) and AptsHlcrrΔglk strains in cultured human HeLa epithelial cells. The data are the statistical mean from three biological replicate experiments.
Examples
Example 1 : Mutant construction and testing
Mutants were constructed according to published procedures (23). Briefly, 60 base pair (bp) oligonucleotides were designed that contain flanking DNA sequences of the target genes to be deleted (Table 1). The oligonucleotides were used to amplify, by polymerase chain reaction (PCR), a cassette containing the kanamycin or the chloramphenicol resistance gene carried by plasmids pKD4 and pKD3 respectively (Table 2; (23)). The resulting PCR product was then transformed by electroporation into Salmonella Typhimurium 4/74 (24) carrying plasmid pKD46 (Table 2). Plasmid pKD46 encodes a recombinase enzyme that facilitates homologous recombination of the target gene with the kanamycin or the chloramphenicol resistance cassette resulting in complete replacement of the deleted gene.
The kanamycin/chloramphenicol resistance region of the deleted gene was transferred by P22 transduction into S. Typhimurium wild type strain 4/74 (25) to avoid unwanted genetic recombination events that may have occurred during the original transformation. The transductants were screened on green agar plates to obtain lysogen free colonies (26). The kanamycin or chloramphenicol resistance gene was then removed via a further recombination event by transforming the cells with the FLP-recombinase expression plasmid, pCP20 (27). The FLP- recombinase removed the kanamycin or chloramphenicol resistance gene and the strains were cured of the pCP20 plasmid by growing the strain at 420C. The resultant strains, JH3478, JH3479 and JH3521 (Table 2) carry complete deletions of the specified genes and are free of genes conferring antibiotic resistance (23), For construction of multiple gene deletions, the above process was repeated on the original mutant strain (Table 2).
Confirmation of gene deletion.
The complete absence of the deleted gene(s) was confirmed by DNA sequencing across the deleted regions of the chromosome (data not shown).
Primer Name Primer DNA sequence pfkaredf CAATAGATTTCATTTTGCATTCCAAAGTTCAG
AGGTAGTCGTGTAGGCTGGAGCTGCTTC pfkaredr AGGCCTGATAAGCGTAGCGCCATCAGGCGCGC
AAAAACAACATATGAATATCCTCCTTAG pfkbredf ATTAAGTGCCAGACTGAAATCAGCCTAA
CAGGAGGTAACGGTGTAGGCTGGAGCTGCTTC pfkbredr AACCGATTTTCCGTTATCCCCCTCGGCGA
GGGGGAAACGACATATGAATATCCTCCTTAG ptshredf TTAGTTCCACAACACTAAACCTATAAGTT
GGGGAAATACAGTGTAGGCTGGAGCTGCTTC crrredr AAATGGCGCCCAAAGGCGCCATTCTTCAC
TGCGGCAAGAACATATGAATATCCTCCTTAG glkredf TGACAAAGACTTATTTTGACTTTAGCGGA
GCAGTAGAAGAGTGTAGGCTGGAGCTGCTTC glkredr CTTTTGTAGGCCGGATAAGGCGTTTATGCCA
CCATCTGGCCATATGAATATCCTCCTTAG ptslrevlnti GCAGTTCCTGTTTGTAGATTTCAATCTC
TTTGCGCAGCGCCATATGAATATCCTCCTTAG crrredf TCCACGAGATGCGGCCCAATTTACTGCTTAGGA
GAAGATCGTGTAGGCTGGAGCTGCTTC ptsgredf GAACGTAGAAAAGCACAAATACTCAGGA
GCACTCTCAATTGTGTAGGCTGGAGCTGCTTC ptsgredr GCCGAATGGCTGCCTTAATTCTCCCCAACATCA
TTACTGCCATATGAATATCCTCCTTAG manxredf TGTCAAGTTGATGTGTTGACAATAATAAAG GAGGTAGCAAGTGTAGGCTGGAGCTGCTTC manzredr AAAAAACGGGGCCGTTTGGCCCCGGTAGT GTACAACAGCCCATATGAATATCCTCCTTAG
Table 1. Primers used to construct S. Typhimurium gene deletion mutants (23)
S. Typhimurium strains Relevant genotype Reference
4/74 Wild type strain (24)
SL1344 rpsL' hisG' (25)
JH3386 4/74 ApfkA::Kn This study
JH3460 4/74 ApfkBv. Cm This study
JH3486 4/74 ApfkAv.Kn, ApfkBv.Cm This study
JH3536 4/74 AptsHlcrr:. Cm This study
JH3494 4/74 Δg Ik:: Kn This study
JH3540 4/74 AptsHlcrr.Cm, Δg//c::Kn This study
JH3502 4/74 Acrr. Kn This study
JH3537 4/74 ΔpteH/::Cm This study
JH3504 4/74 AptsG: :Cm This study
JH3501 4/74 AmanXYZ:: Cm This study
JH3541 4/74 AmanXYZ This study
AT1011 4/74 ΔpfeG::Cm, AmanXYZ This study
AT1012 4/74 AptsG/.Cm, AglkrKn This study
AT1013 4/74 AmanXYZ, Aglk::Kn This study
AT1014 4/74 ΔpfsG::Cm, AmanXYZ, Δg//c::Kn This study
Table 2, Strains and plasmids used.
Macrophage infection assays.
Infection assays in murine macrophages were performed according to (28). Briefly, murine macrophages (RAW264.7; obtained from American Type Culture Collection, Rockville, MD) were grown in MEM medium (Sigma) supplemented with 10% fetal bovine serum (FBS; Sigma), L-glutamine (2 mM final concentration; Sigma) and 1x non-essential amino acids (Sigma) as described (28). For infections, 1x105 macrophage cells were seeded into each well of a 12-well cell culture plate and infected with complement-opsonized S. Typhimurium 4/74 and mutant strains at a multiplicity of infection (MOI) of 1 :1 (bacteria:cells) (28). To minimize SPM expression, bacteria were grown overnight on Luria broth plates at 370C and suspended in phosphate buffered saline (PBS) before opsonization.
To increase the uptake of Salmonella, plates were centrifuged at 100Og for 5 min, and this was defined as time 0 hours (h). After 1 h of phagocytosis, extracellular bacteria were killed using 100 μg ml"1 gentamicin (Sigma). The media was replaced after 1 h with medium containing 10 μg ml"1 gentamicin. Incubations were continued for 2 h and 18 h. To estimate the amount of intracellular bacteria at each time point, cells were lysed using 1 % Triton X-100, and samples were taken for viable counts (28).
HeLa cell infection assays.
Infection assays in human HeLa epithelial cells (obtained from American Type Culture Collection, Rockville, MD) were performed according to (29). Briefly, HeLa cells were grown in DMEM medium (Sigma) supplemented with 10% fetal bovine serum (Sigma), 2mM L-glutamine (Sigma) and 2OmM HEPES buffer (Sigma). Approximately, 1x105 HeLa cells were seeded into each well of a 6-well cell culture plates and infected with S. Typhimurium 4/74 and mutant strains at an MOI of 10:1. Prior to infection the S. Typhimurium strains had been grown to an OD600 of 1.2 to allow expression of the SPI-1 Type 3 secretion system. To increase the uptake of Salmonella, plates were centrifuged at 1000 g for 5 min, and this was defined as time 0 h. After 1 h of infection, extracellular bacteria were killed with 30 μg ml"1 gentamicin. The media was replaced after 1 h with medium containing 5 μg ml"1 gentamicin. Incubations were continued for 2 h and 6 h. To estimate the amount of intracellular bacteria at each time point, cells were lysed using 0.1 % SDS, and samples were taken for viable counts (28). Mouse infection assays.
Mouse infection experiments were performed as described (30) with some modifications. Liquid cultures of 4/74 and the S. Typhimurium ΔpfkAB strain, JH3486, were grown statically in 50ml of Luria Broth (LB; (31) at 370C overnight in 50ml Falcon tubes (Corning Inc.) (32). The following day bacteria were resuspended at a final cell density of 1x104 colony forming units (cfu) ml"1 in sterile Phosphate Buffered Saline (PBS). Five female BALB/c mice (Charles River U.K. Ltd.) were infected with 200μl of bacterial suspension via the intraperitoneal (IP) cavity at a final dose of 2x103 Salmonella cfu (30). The infection was permitted to proceed for 72 hours at which time point the mice were sacrificed by cervical dislocation and the spleens and livers surgically removed (30). Following homogenization of the organs in a stomacher (Seward Tekmar), serial dilutions of the suspensions in PBS were spread onto LB agar plates and bacterial colonies enumerated after overnight incubation at 37°C (32).
Phosphofructokinase is required for intracellular replication and survival of S. Typhimurium in cultured RAW macrophages.
Glycolysis is the sequence of catabolic reactions that converts sugars into pyruvate with the concomitant synthesis of ATP and NADH. It is the foundation of both aerobic and anaerobic respiration and is found in nearly all organisms (33). The enzyme phosphofructokinase irreversibly converts β-D-fructose 6-phosphate into β-D-fructose1 , 6-bisphosphate and is encoded by two genes in most bacteria (pfkA and pfkB) (34). An S. Typhimurium mutant strain in which the pfkAB genes were completely deleted was constructed according to procedures described above. The marker antibiotic resistance genes (kanamycin and chloramphenicol) were completely removed from the chromosome of the S. Typhimurium ApfkAB strain. The growth of the marker-free S. Typhimurium ApfkAB strain was inhibited by kanamycin and chloramphenicol.
The 322-fold decrease in the number of cfu's from recovered intracellular S. Typhimurium wild type and S. Typhimurium ApfkAB bacteria after 18h infection showed that the latter strain was unable to replicate within RAW 264.7 macrophages (Fig 1A, Table 3A). Furthermore, the 34-fold decrease in the cfu's of recovered intracellular S. Typhimurium ApfkAB bacteria after 18h compared to 2h infection showed that the S. Typhimurium ApfkAB strain was unable to survive within RAW 264.7 macrophages (Fig 1 , Table 3A). Infection assays have also shown that intracellular replication of the S. Typhimurium ApfkA and ApfkB single mutants is not attenuated compared to the wild type strain in RAW 264.7 macrophages (data not shown). This demonstrates that deletion of both pfkA and pfkB genes are required for functional silencing of phosphofructokinase for attenuation.
2 hours S. D. 18 hours S.D.
4/74 24.3 3.4 200.4 16.4
ApfkAB 21.2 3.0 0.6 0.2
4/74 24.8 1.6 57.8 1.6
AptsHlcrrΔglk 25.6 3.6 19.0 1.8
B
2 hours S.D. 6 hours S.D.
4/74 120.0 13.7 641.6 129.6
ApfkAB 86.4 6.5 143.6 27.7
4/74 120.0 13.7 641.6 129.6
AptsHlcrrΔglk 38.5 8.6 51.0 11.2
Table 3. Intracellular replication rate and survival assays for S. Typhimurium 4/74 (wild type), ApfkAB and AptsHlcrrglk strains in (A) cultured RAW 264.7 macrophages and (B) HeLa epithelial cells expressed as percentages of original inoculum. S.D. is standard deviation from three biological replicate experiments.
Phosphofructokinase is required for intracellular replication of S. Typhimurium in cultured HeLa epithelial cells.
The intracellular replication rates of the S. Typhimurium 4/74 wild type and ApfkAB mutant strains were determined in cultured human HeLa epithelial cells. A comparison of the intracellular replication rate of S. Typhimurium ApfkAB bacteria between 2 and 6 hours showed that the mutant strain replicated less than 2-fold in HeLa cells (Fig. 2, Table 3B).
Phosphofructokinase is avirulent in the mouse typhoid infection model.
BALB/c mice were infected (IP) with the S. Typhimurium ΔpfkAB mutant and the wild type strain. Salmonella were recovered and enumerated from the spleen and liver of mice after 72 hours infection. The results demonstrated that the S. Typhimurium ApfkAB mutant was severely attenuated by approximately 102 fold compared to the wild type strain in the spleen and liver of infected mice. (Fig. 3, Table 4). Mice infected with the S. Typhimurium wild type strain showed severe symptoms of systemic disease after 72 hours. The fur of mice infected with the S. Typhimurium ApfkAB strain was slightly ruffled after 72 hours.
Strain cfu's per spleen (SD) cfu's per liver (SD)
4/74 1853333 (1205047) 1051333 (685875)
ApfkAB 32828 (21395) 14912 (6923)
Table 4. Mean S. typhimurium wild type and S. typhimurium ApfkAB cfu's recovered from BALB/c mice liver and spleens 72 h post- infection (n=5).
Sugar transport is required for intracellular replication of S. Typhimurium in RAW 264.7 macrophages.
An S. Typhimurium mutant strain containing complete deletions of the ptsHI, err and glk genes was constructed according to the procedures described above. The ptsHI and err genes encode components of a phosphotransferase (PTS) system, which detects, transports, and phosphorylates several sugars and sugar derivatives into the bacterial cell (35). The glk gene encodes glucose kinase, which specifically phosphorylates intracellular glucose (36). In vitro growth experiments on M9 minimal salts media showed that, as expected, the S. Typhimurium AptsHlcrrΔglk mutant was unable to utilize glucose for growth (37). Infection assays of the S. Typhimurium wild type and AptsHlcrrΔglk strains were performed in RAW 264.7 macrophage. The data are the statistical mean from three biological replicate experiments (Fig. 4). A comparison of the intracellular replication rate of S. Typhimurium AptsHlcrrΔglk bacteria at 2 and 18 hours showed that the strain was unable to replicate in RAW 264.7 macrophages and may be impaired for survival (Fig. 4, Table 3A).
Sugar transport is required for intracellular replication of S. Typhimurium in HeLa epithelial cells.
The intracellular replication rates of the S. Typhimurium 4/74 wild type and
AptsHlcrrΔglk strains were compared in cultured HeLa cells. The S. Typhimurium
AptsHlcrrΔglk strain was unable to replicate in HeLa epithelial cells (Fig. 5, Table
3B).
The intracellular phenotype of the S. Typhimurium ApfkAB mutant demonstrated that phosphofructokinase and glycolysis are required for intracellular replication and survival of S. Typhimurium in RAW macrophages.
The S. Typhimurium ApfkAB mutant was unable to replicate within human HeLa epithelial cells suggesting phosphofructokinase and glycolysis are necessary for growth within this cell type.
The severe attenuation of the AptsHlcrrΔglk mutant compared to the wild type strain suggests glucose is the major sugar required for intracellular replication and survival of S. Typhimurium in RAW 264.7 macrophages and HeLa epithelial cells.
The S. Typhimurium pfkAB mutant is severely attenuated during intraperitoneal infection of BALB/c mice demonstrating that phosphofructokinase and glycolysis are required for Salmonella survival and replication within mice.
Example 2: Oral live vaccine strain verification Experiment 1 : Attenuation of potential vaccine strains
Female BALB/c mice (6 to 8 weeks old) and housed under specific-pathogen-free conditions are used for all experiments. For each animal experiment, a single bacterial dose of 1 x 109 to 2 x 109 bacteria in LB broth is administered by gavage to mice (n=5; day 0). Control mice are given an equal volume of sterile LB broth. The mice are infected with 0.2 ml of stationary phase bacteria grown aerobically overnight at 370C in Luria Bertani (LB) broth and then concentrated in fresh LB broth to approximately 5 x 109 to 10 x 109 per ml. The Salmonella strains used to infect the mice are: wild type strain (S. Typhimurium 4/74) and 5 potential vaccine strains: S. Typhimurium ApfkAB, AptsHlcrrAglk, AptsHlcrr, AptsHI, AptsGAmanXYZAglk. Attenuation of the metabolic mutant strains were determined by evaluation of the mice according to humane procedures based on Home Office Licensing for Animal Experimentation. Between 4 and 7 days after inoculation the majority of animals inoculated with a virulent Salmonella strain will develop mild to moderate symptoms of systemic infection consisting of reduced activity, anorexia, hunched posture and ruffled fur. Each of these symptoms should appear with various degrees of development from the 4th day onward and therefore will be scored from 1 to 3 for "slightly developed" to "highly developed" at each checking point. Animals with three or more of these symptoms scored as highly developed will constitute the end point of the experiment and animals will be humanely killed. Any dyspnoeic animals will be killed at first sight. The results from this experiment are shown in Table 5. According to the above scoring procedure, after 4 days, mice infected with the wild-type strain displayed all the symptoms of a severe systemic typhoid infection and were humanely killed. The S. Typhimurium AptsGAmanXYZAglk strain displayed attenuated upon infection and mice were killed after 9 days (Table 5). The S. Typhimurium ApfkAB, AptsHlcrrAglk, AptsHlcrr, AptsHI strains were avirulent upon infection and continued to survive past 3 weeks post infection (Table 5).
Conclusion: The S. Typhimurium ApfkAB, AptsHlcrrAglk, AptsHlcrr, AptsHI, AptsGAmanXYZAglk strains are all attenuated or avirulent for infection of BALB/c mice and therefore verified as candidates for potential vaccine strains.
Figure imgf000022_0001
Figure imgf000023_0001
Table 5. Percentage survival of BALB/c mice infected with S. Typhimurium metabolic mutant strains.
Experiment 2: Clearance of potential vaccine strains
To determine whether complete sterility has been achieved, Experiment 1 is repeated and bacterial cfu's enumerated from spleen, liver, mesenteric lymph nodes and Peyer's patches every 14 days for a total of 50 days as described in Experiment 1. A positive outcome with respect to strain validation as potential vaccines would be no cfu's are recovered from the organs of mice infected with attenuated metabolism mutant strain(s), a positive control aroA strain, or LB control (sterility).
Experiment 3: Elicitation of an immune response by potential vaccine strains
Experiment 2 is repeated after 30 days infection using potential vaccine strain(s), S. Typhimurium ΔaroA strain and an LB control. Faecal and blood samples are collected after day 30. Faecal samples are weighed, and 1.0 ml of PBS plus 0.1 % sodium azide is added per 100mg of faeces. The faecal pellets are dispersed by microtip sonication for 10 to 20 second bursts at 50% duty. The faecal suspensions are subsequently pelleted in a microfuge for 5 min, and the supernatants transferred to a fresh tube and held frozen until assays are performed. Blood samples are collected from the tail vein and incubated overnight at 40C followed by microcentrifugation for 1 min before the serum collection. The titre of anti-Salmonella lipopolysaccharide (LPS) in faecal supernatant and blood serum samples is determined by end-point ELISA. Briefly, 96-well commercially available immunoplates are coated overnight at 40C with 0.01 mg/ml Salmonella LPS. Following blocking with 4% skim milk powder in PBS for 1 h at 370C, serial dilutions of mouse sera in PBST (0.05% Tween20 in PBS) containing 0.4% skim milk powder are added to the wells and the plates incubated for 2h at 370C. Bound antibody is detected by the addition of an anti-mouse Ig Horseradish peroxidase conjugate diluted 1/1000 in PBST containing 0.4% skim milk powder and incubated for 2 h at 370C. Reactions are developed using immunopure o- phenylenediamine with H2O2 as the substrate. Absorbance is read at 492 nm in a plate reader. The serum antibody titre is designated as the reciprocal of the dilution of specific antibody that gives an OD492 value of five times the background value.
Experiment 4: Virulent challenge
BALB/c mice are pre-infected for 30 days with the potential vaccine strain(s) and the S. Typhimurium AaroA strain and LB control at doses of 1-2 x 109 bacteria according to experiment 1. The pre-infected mice are then challenged at day 30 via oral infection according to Experiment 1 with 1x108 cfu of a S. Typhimurium wild type strain (4/74) to assess the protection conferred by the vaccine compared to control BALB/c mice. Mice successfully immunised following infection with the metabolism mutant strain(s) and aroA strains show no symptoms of typhoid fever according to the accepted scoring system. Optionally cfu's in above organs are enumerated and ELISA assays are performed to detect the presence of antibodies against Salmonella LPS (see Experiment 3 for methodology).
References
1. Cossart, P., Pizarro-Cerda, J., Lecut, M. (2005) Cellular Microbiology, ASM Press. 2. Titball, R.W. (2008) Vaccines against intracellular bacterial pathogens. Drug discovery today, 13, 596-600.
3. Fulop, M., Mastroeni, P., Green, M. and Titball, R.W. (2001) Role of antibody to lipopolysaccharide in protection against low- and high-virulence strains of Francisella tularensis. Vaccine, 19, 4465-4472.
4. Jones, S. M., Ellis, J. F., Russell, P., Griffin, K.F. and Oyston, P. C. (2002) Passive protection against Burkholderia pseudomallei infection in mice by monoclonal antibodies against capsular polysaccharide, lipopolysaccharide or proteins. Journal of medical microbiology, 51 , 1055-1062.
5. Nelson, M., Prior, J. L., Lever, M.S., Jones, H. E., Atkins, T.P. and Titball, R.W. (2004) Evaluation of lipopolysaccharide and capsular polysaccharide as subunit vaccines against experimental melioidosis. Journal of medical microbiology, 53, 1177-1182.
6. Crump, JA, Luby, SP. and Mintz, E. D. (2004) The global burden of typhoid fever. Bulletin of the World Health Organization, 82, 346-353.
7. Davies, R.H., Dalziel, R., Gibbens, J. C1 Wilesmith, J.W., Ryan, J. M., Evans, S.J., Byrne, C1 Paiba, GA, Pascoe, S.J. and Teale, CJ. (2004) National survey for Salmonella in pigs, cattle and sheep at slaughter in Great Britain (1999-2000). Journal of applied microbiology, 96, 750-760.
8. Beach, J. C1 Murano, E.A. and Acuff, G. R. (2002) Prevalence of Salmonella and Campylobacter in beef cattle from transport to slaughter. Journal of food protection, 65, 1687-1693.
9. Troutt, H. F., Galland, J. C, Osburn, B.I., Brewer, R. L., Braun, R.K., Schmitz, J.A., Sears, P., Childers, A.B., Richey, E., Mather, E. et al. (2001) Prevalence of Salmonella spp in cull (market) dairy cows at slaughter. Journal of the American Veterinary Medical Association, 219, 1212-1215.
10. Guzman, C.A., Borsutzky, S., Griot-Wenk, M., Metcalfe, I. C1 Pearman, J., Collioud, A., Favre, D. and Dietrich, G. (2006) Vaccines against typhoid fever. Vaccine, 24, 3804-3811.
11. Levine, M. M., Galen, J., Barry, E., Noriega, F., Chatfield, S., Sztein, M., Dougan, G. and Tacket, C. (1996) Attenuated Salmonella as live oral vaccines against typhoid fever and as live vectors. Journal of biotechnology, 44, 193-196. 12. Ward, S.J., Douce, G., Figueiredo, D., Dougan, G. and Wren, B.W. (1999) lmmunogenicity of a Salmonella typhimurium aroA aroD vaccine expressing a nontoxic domain of Clostridium difficile toxin A. Infection and immunity, 67, 2145-2152.
13. Abrahams, G. L. and Hensel, M. (2006) Manipulating cellular transport and immune responses: dynamic interactions between intracellular Salmonella enterica and its host cells. Cellular microbiology, 8, 728-737.
14. Haraga, A., Ohlson, M. B. and Miller, S.I. (2008) Salmonellae interplay with host cells. Nature reviews, 6, 53-66.
15. Brumell, J. H., Steele-Mortimer, O. and Finlay, B. B. (1999) Bacterial invasion: Force feeding by Salmonella. CurrBiol, 9, R277-280.
16. Becker, D., Selbach, M., Rollenhagen, C1 Ballmaier, M., Meyer, T. F., Mann, M. and Bumann, D. (2006) Robust Salmonella metabolism limits possibilities for new antimicrobials. Nature, 440, 303-307.
17. Lehoux, D. E., Sanschagrin, F., Kukavica-lbrulj, I., Potvin, E. and Levesque, R. C. (2004) Identification of novel pathogenicity genes by PCR signature- tagged mutagenesis and related technologies. Methods in molecular biology (Clifton, N.J, 266, 289-304.
18. Morgan, E., Campbell, J. D., Rowe, S. C1 Bispham, J., Stevens, M. P., Bowen, A.J., Barrow, P.A., Maskell, D.J. and Wallis, T.S. (2004) Identification of host-specific colonization factors of Salmonella enterica serovar Typhimurium. Molecular microbiology, 54, 994-1010.
19. Tsolis, R.M., Kingsley, R.A., Townsend, S. M., Ficht, T.A., Adams, L.G. and Baumler, AJ. (1999) Of mice, calves, and men. Comparison of the mouse typhoid model with other Salmonella infections. Advances in experimental medicine and biology, 473, 261-274.
20. Aggarwal, A., Kumar, S., Jaffe, R., Hone, D., Gross, M. and Sadoff, J. (1990) Oral Salmonella: malaria circumsporozoite recombinants induce specific CD8+ cytotoxic T cells. The Journal of experimental medicine, 172, 1083-1090.
21. Formal, S. B., Baron, L.S., Kopecko, DJ. , Washington, O., Powell, C. and Life, CA. (1981) Construction of a potential bivalent vaccine strain: introduction of Shigella sonnei form I antigen genes into the galE Salmonella typhi Ty21a typhoid vaccine strain. Infection and immunity, 34, 746-750.
22. Wu, J.Y., Newton, S., Judd, A., Stacker, B. and Robinson, W.S. (1989) Expression of immunogenic epitopes of hepatitis B surface antigen with hybrid flagellin proteins by a vaccine strain of Salmonella. Proceedings of the National Academy of Sciences of the United States of America, 8β, 4726-4730.
23. Datsenko, K.A. and Wanner, B. L. (2000) One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proceedings of the National Academy of Sciences of the United States of America, 97, 6640-6645.
24. Wray, C. and Sojka, WJ. (1978) Experimental Salmonella typhimurium infection in calves. Research in veterinary science, 25, 139-143.
25. Hoiseth, S. K. and Stacker, B.A. (1981) Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines. Nature, 291 , 238-239.
26. Smith, H. O. and Levine, M. (1967) A phage P22 gene controlling integration of prophage. Virology, 31 , 207-216.
27. Cherepanov, P.P. and Wackernagel, W. (1995) Gene disruption in Escherichia coli: TcR and KmR cassettes with the option of Flp-catalyzed excision of the antibiotic-resistance determinant. Gene, 158, 9-14.
28. Eriksson, S., Bjorkman, J., Borg, S., Syk, A., Pettersson, S., Andersson, D.I. and Rhen, M. (2000) Salmonella typhimurium mutants that downregulate phagocyte nitric oxide production. Cellular microbiology, 2, 239-250.
29. Hautefort, I., Thompson, A., Eriksson-Ygberg, S., Parker, M. L., Lucchini, S., Danino, V., Bongaerts, R.J., Ahmad, N., Rhen, M. and Hinton, J. C. (2008) During infection of epithelial cells Salmonella enterica serovar Typhimurium undergoes a time-dependent transcriptional adaptation that results in simultaneous expression of three type 3 secretion systems. Cellular microbiology, 10, 958-984.
30. Oswald, I. P., Lantier, F., Moutier, R., Bertrand, M. F. and Skamene, E. (1992) Intraperitoneal infection with Salmonella abortusovis is partially controlled by a gene closely linked with the lty gene. Clinical and experimental immunology, 87, 373-378.
31. Bertani, G. (1951) Studies on lysogenesis. I. The mode of phage liberation by Iysogenic Escherichia coli. Journal of bacteriology, 62, 293-300.
32. Baumler, A.J., Tsolis, R.M., Valentine, P.J., Ficht, TA and Heffron, F. (1997) Synergistic effect of mutations in invA and IpfC on the ability of Salmonella typhimurium to cause murine typhoid. Infection and immunity, 65, 2254-2259.
33. Fraenkel, D. G. (1996) Glycolysis. 2 ed. ASM Press, Washington DC.
34. Riley, M. (1993) Functions of the gene products of Escherichia coli. Microbiological reviews, 57, 862-952.
35. Postma, P.W., Lengeler, J.W. and Jacobson, G. R. (1993) Phosphoenolpyruvate:carbohydrate phosphotransferase systems of bacteria. Microbiological reviews, 57, 543-594.
36. Meyer, D., Schneider-Fresenius, C, Horlacher, R., Peist, R. and Boos, W. (1997) Molecular characterization of glucokinase from Escherichia coli K- 12. Journal of bacteriology, 179, 1298-1306.
37. Mackey, B. M. and Derrick, CM. (1982) A comparison of solid and liquid media for measuring the sensitivity of heat-injured Salmonella typhimurium to selenite and tetrathionate media, and the time needed to recover resistance. The Journal of applied bacteriology, 53, 233-242.
Sequence listing
<110> Plant Bioscience Limited
<120> Mutant Pathogenic Bacteria and Live Attenuated Vaccine
Compositions <130> pc780117WO <150> GB0900974.7 <151 > 2009-01-21 <160> 14
<170> Patentln version 3.3 <210> 1 <211> 60 <212> DNA <213> Artificial <220>
<223> Primer sequence pfkaredf <400> 1 caatagattt cattttgcat tccaaagttc agaggtagtc gtgtaggctg gagctgcttc 60 <210> 2 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence pfkaredr <400> 2 aggcctgata agcgtagcgc catcaggcgc gcaaaaacaa catatgaata tcctccttag 60 <210> 3 <211> 60 <212> DNA <213> Artificial <220>
<223> primer sequence pfkbredf <400> 3 attaagtgcc agactgaaat cagcctaaca ggaggtaacg gtgtaggctg gagctgcttc 60 <210> 4 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence pfkbredr <400> 4 aaccgatttt ccgttatccc cctcggcgag ggggaaacga catatgaata tcctccttag 60 <210> 5 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence ptshredf <400> 5 ttagttccac aacactaaac ctataagttg gggaaataca gtgtaggctg gagctgcttc 60 <210> 6 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence crrredr <400> 6 aaatggcgcc caaaggcgcc attcttcact gcggcaagaa catatgaata tcctccttag 60 <210> 7 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence glkredf <400> 7 tgacaaagac ttattttgac tttagcggag cagtagaaga gtgtaggctg gagctgcttc 60 <210> 8 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence glkredr <400> 8 cttttgtagg ccggataagg cgtttatgcc accatctggc catatgaata tcctccttag 60 <210> 9 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence ptslrevlnti <400> 9 gcagttcctg tttgtagatt tcaatctctt tgcgcagcgc catatgaata tcctccttag 60 <210> 10 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence crrredf <400> 10 tccacgagat gcggcccaat ttactgctta ggagaagatc gtgtaggctg gagctgcttc 60 <210> 11 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence ptsgredf <400> 11 gaacgtagaa aagcacaaat actcaggagc actctcaatt gtgtaggctg gagctgcttc 60 <210> 12 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence ptsgredr <400> 12 gccgaatggc tgccttaatt ctccccaaca tcattactgc catatgaata tcctccttag 60 <210> 13 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence manxredf <400> 13 tgtcaagttg atgtgttgac aataataaag gaggtagcaa gtgtaggctg gagctgcttc 60 <210> 14 <211 > 60 <212> DNA <213> Artificial <220>
<223> primer sequence manzredr <400> 14 aaaaaacggg gccgtttggc cccggtagtg tacaacagcc catatgaata tcctccttag 60

Claims

CLAIMS:
1. A mutant pathogenic bacterium that is incapable of expressing one or more metabolic genes.
2. A mutant pathogenic bacterium according to claim 1 , characterised in that the pathogenic bacterium is an intracellular pathogenic bacterium.
3. A mutant pathogenic bacterium according to claim 1 or 2, characterised in that it is attenuated.
4. A mutant pathogenic bacterium according to any of claims 1 to 3, characterised in that the bacterium is incapable of expressing two or more metabolic genes.
5. A mutant pathogenic bacterium according to claim 4, characterised in that the two or more genes encode proteins involved in the same or in different metabolic pathways.
6. A mutant pathogenic bacterium according to any of claims 1 to 5, characterised in that the metabolic gene or genes are involved in glycolysis and/or sugar transport.
7. A mutant pathogenic bacterium according to any of claims 1 to 6, characterised in that the metabolic gene or genes encode a phosphofructokinase.
8. A mutant pathogenic bacterium according to any of claims 1 to 7, characterised in that the metabolic genes are pfkA and pfkB.
9. A mutant pathogenic bacterium according to any of claims 1 to 8, characterised in that the bacterium is incapable of expressing one or more metabolic genes that encode components of a sugar phosphotransferase system.
10. A mutant pathogenic bacterium according to claim 9, characterised in that the one or more metabolic genes encode components of a glucose, mannose and/or galactose transport system.
11. A mutant pathogenic bacterium according to claim 10, characterised in that the one or more metabolic genes that encode components of a glucose, mannose and/or galactose transport system are selected from ptsHI, err, glk, ptsG, manXYZ, mglABC and galP.
12. A mutant pathogenic bacterium according to any of claims 1 to 11 , characterised in that it is incapable of expressing:
(a) pfkk and/or pfkB;
(b) ptsHI, err, and/or glk;
(c) pfkk, pfkB, ptsHI, err, and glk;
(d) pfkk, pfkB, ptsHI, and err;
(e) pfkk, pfkB and glk,
(f) pfkk, pfkB and ptsHI
(g) pfkA, pfkB, ptsHI and glk (h) ptsHI;
(I) ptsHI, err φ ptsHI, glk
(k) glk, ptsG and manXYZ.
(I) pfkA, pfkB, glk, ptsG and/or manXYZ
(m) mglABC and/or galP
(n) mglABC, galP, ptsHI
(o) mglABC, galP, ptsHI and err
(p) mglABC, galP, ptsHI
(q) mglABC, galP, ptsG and manXYZ
(r) pfkA, pfkB, ptsHI, err, mglABC and galP
(s) pfkA, pfkB, ptsHI, mglABC and galP; or,
(t) pfkA, pfkB, mglABC and galP.
13. A mutant pathogenic bacterium according to any of claims 1 to 12 wherein the bacterial strain is selected from AptsHI, AptsHlcrr, AptsGAmanXYZAglk, AptsHlcrrAglk or ApfkAB.
14. A mutant pathogenic bacterium according to any of claims 1 to 13, which is attenuated and which is incapable of expressing one or more other virulence gene or genes, such as aroA and/or rpoS.
15. A mutant pathogenic bacterium according to any of claims 1 to 14, characterised in that the bacterium carries a heterologous gene.
16. A mutant pathogenic bacterium according to any of claims 1 to 15, characterised in that the pathogenic bacterium is a Salmonella, Bartonella, Brucella, Burkholderia, Chlamydia, Coxiella, Ehrlichia, Francisella, Legionella, Listeria, Mycobacter, Nocardia, Rickettsia, Shigella or Tropheryma
17. A mutant pathogenic bacterium according to any of claims 1 to 16, characterised in that the pathogenic bacterium is a typhoidal Salmonella serovar.
18. A mutant pathogenic bacterium according to claim 16, characterised in that the typhoidal Salmonella serovar is a Salmonella enterica Typhi or Paratyphi.
19. A mutant pathogenic bacterium according to claim 18, characterised in that the Salmonella enterica Typhi is a Samonella Typhimurium.
20. A live attenuated vaccine for protection against infection with a pathogenic bacterium or the pathogenic effects thereof, comprising a live attenuated mutant pathogenic bacterium according to any one of claims 3 to 19 and a pharmaceutically acceptable carrier.
21. A live attenuated vaccine according to claim 20, characterised in that it is formulated for oral administration.
22. A live attenuated vaccine according to claim 20 or 21 , characterised in that it is in freeze-dried form.
23. A live attenuated vaccine according to any of claims 20 to 22, characterised in that it comprises an adjuvant.
24. Use of a live attenuated mutant pathogenic bacterium according to any one of claims 3 to 19 for the manufacture of a vaccine for the protection of humans or animals against infection with a pathogenic bacterium or against the pathogenic effects of infection.
25. A method for the preparation of a vaccine according to any one of claims 20 to 22, comprising admixing a live attenuated mutant pathogenic bacterium according to any one of claims 3 to 18 with a pharmaceutically acceptable carrier.
26. A method for immunizing a human or animal against infection with a pathogenic bacterium, comprising administering to the human or animal a vaccine according to any of claims 20 to 22.
PCT/GB2010/050086 2009-01-21 2010-01-21 Mutant pathogenic bacterial and live attenuated vaccine compositions WO2010084350A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0900974.7 2009-01-21
GBGB0900974.7A GB0900974D0 (en) 2009-01-21 2009-01-21 Mutant pathogenic bacteria and live attenuated vaccine compositions

Publications (1)

Publication Number Publication Date
WO2010084350A1 true WO2010084350A1 (en) 2010-07-29

Family

ID=40446122

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2010/050086 WO2010084350A1 (en) 2009-01-21 2010-01-21 Mutant pathogenic bacterial and live attenuated vaccine compositions

Country Status (2)

Country Link
GB (1) GB0900974D0 (en)
WO (1) WO2010084350A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015103104A1 (en) * 2014-01-06 2015-07-09 The United States Of America, As Represented By The Secretary Of Agriculture Attenuated salmonella enterica
CN113088454A (en) * 2021-04-08 2021-07-09 武汉轻工大学 Method for separating intact bacteria from infected cells

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB900974A (en) 1957-08-28 1962-07-11 British Oxygen Co Ltd Electric arc working apparatus
US4550081A (en) * 1980-05-19 1985-10-29 The Board Of Trustees Of The Leland Stanford Jr. University Non-reverting salmonella

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB900974A (en) 1957-08-28 1962-07-11 British Oxygen Co Ltd Electric arc working apparatus
US4550081A (en) * 1980-05-19 1985-10-29 The Board Of Trustees Of The Leland Stanford Jr. University Non-reverting salmonella

Non-Patent Citations (50)

* Cited by examiner, † Cited by third party
Title
ABRAHAMS, G.L.; HENSEL, M.: "Manipulating cellular transport and immune responses: dynamic interactions between intracellular Salmonella enterica and its host cells", CELLULAR MICROBIOLOGY, vol. 8, 2006, pages 728 - 737
AGGARWAL, A.; KUMAR, S.; JAFFE, R.; HONE, D.; GROSS, M.; SADOFF, J.: "Oral Salmonella: malaria circumsporozoite recombinants induce specific CD8+ cytotoxic T cells", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 172, 1990, pages 1083 - 1090
BAUMLER, A.J.; TSOLIS, R.M.; VALENTINE, P.J.; FICHT, T.A.; HEFFRON, F.: "Synergistic effect of mutations in invA and IpfC on the ability of Salmonella typhimurium to cause murine typhoid", INFECTION AND IMMUNITY, vol. 65, 1997, pages 2254 - 2259, XP055027538
BEACH, J.C.; MURANO, E.A.; ACUFF, G.R.: "Prevalence of Salmonella and Campylobacter in beef cattle from transport to slaughter", JOURNAL OF FOOD PROTECTION, vol. 65, 2002, pages 1687 - 1693
BECKER ET AL.: "ROBUST SALMONELLA METABOLISM LIMITS POSSIBILITIES FOR NEW ANTIMICROBIALS", NATURE 16 MAR 2006, vol. 440, no. 7082, 16 March 2006 (2006-03-16), pages 303 - 307, XP002575493, ISSN: 1476-4687 *
BECKER, D.; SELBACH, M.; ROLLENHAGEN, C.; BALLMAIER, M.; MEYER, T.F.; MANN, M.; BUMANN, D.: "Robust Salmonella metabolism limits possibilities for new antimicrobials", NATURE, vol. 440, 2006, pages 303 - 307, XP002575493, DOI: doi:10.1038/nature04616
BERTANI, G.: "Studies on lysogenesis. . The mode of phage liberation by lysogenic Escherichia coli", JOURNAL OF BACTERIOLOGY, vol. 62, 1951, pages 293 - 300
BOWDEN STEVEN D ET AL: "Glucose and Glycolysis Are Required for the Successful Infection of Macrophages and Mice by Salmonella enterica Serovar Typhimurium", INFECTION AND IMMUNITY, vol. 77, no. 7, July 2009 (2009-07-01), pages 3117 - 3126, XP002575494, ISSN: 0019-9567 *
BRUMELL, J.H.; STEELE-MORTIMER, O.; FINLAY, B.B.: "Bacterial invasion: Force feeding by Salmonella", CURR BIOL, vol. 9, 1999, pages 277 - 280
CHEREPANOV, P.P.; WACKERNAGEL, W.: "Gene disruption in Escherichia coli: TcR and KmR cassettes with the option of Flp-catalyzed excision of the antibiotic-resistance determinant", GENE, vol. 158, 1995, pages 9 - 14
CHIN A M ET AL: "GENETIC EXPRESSION OF ENZYME I ACTIVITY OF THE PHOSPHOENOLPYRUVATE SUGAR PHOSPHOTRANSFERASE SYSTEM IN PTS-HI DELETION STRAINS OF SALMONELLA-TYPHIMURIUM", JOURNAL OF BACTERIOLOGY, vol. 169, no. 2, 1987, pages 894 - 896, XP002575492, ISSN: 0021-9193 *
COSSART, P.; PIZARRO-CERDA, J.; LECUT, M.: "Cellular Microbiology", 2005, ASM PRESS
CRUMP, J.A.; LUBY, S.P.; MINTZ, E.D.: "The global burden of typhoid fever", BULLETIN OF THE WORLD HEALTH ORGANIZATION, vol. 82, 2004, pages 346 - 353
DATSENKO, K.A.; WANNER, B.L.: "One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 97, 2000, pages 6640 - 6645, XP002210218, DOI: doi:10.1073/pnas.120163297
DAVIES, R.H.; DALZIEL, R.; GIBBENS, J.C.; WILESMITH, J.W.; RYAN, J.M.; EVANS, S.J.; BYRNE, C.; PAIBA, G.A.; PASCOE, S.J.; TEALE, C: "National survey for Salmonella in pigs, cattle and sheep at slaughter in Great Britain", JOURNAL OF APPLIED MICROBIOLOGY, vol. 96, 1999, pages 750 - 760
ERIKSSON, S.; BJORKMAN, J.; BORG, S.; SYK, A.; PETTERSSON, S.; ANDERSSON, D.I.; RHEN, M.: "Salmonella typhimurium mutants that downregulate phagocyte nitric oxide production", CELLULAR MICROBIOLOGY, vol. 2, 2000, pages 239 - 250
FELDHEIM D A ET AL: "Physiological consequences of the complete loss of phosphoryl-transfer proteins HPr and FPr of the phosphoenolpyruvate:sugar phosphotransferase system and analysis of fructose (fru) operon expression in Salmonella typhimurium", JOURNAL OF BACTERIOLOGY 1990 US, vol. 172, no. 9, 1990, pages 5459 - 5469, XP002575491, ISSN: 0021-9193 *
FORMAL, S.B.; BARON, L.S.; KOPECKO, D.J.; WASHINGTON, O.; POWELL, C.; LIFE, C.A.: "Construction of a potential bivalent vaccine strain: introduction of Shigella sonnei form I antigen genes into the galE Salmonella typhi Ty21a typhoid vaccine strain", INFECTION AND IMMUNITY, vol. 34, 1981, pages 746 - 750, XP002248097
FRAENKEL, D.G.: "Glycolysis", 1996, ASM PRESS
FULOP, M.; MASTROENI, P.; GREEN, M.; TITBALL, R.W.: "Role of antibody to lipopolysaccharide in protection against low- and high-virulence strains of Francisella tularensis", VACCINE, vol. 19, 2001, pages 4465 - 4472, XP004296108, DOI: doi:10.1016/S0264-410X(01)00189-X
GERMANIER R ET AL: "ISOLATION AND CHARACTERIZATION OF GAL E MUTANT TY 21A OF SALMONELLA TYPHI: A CANDIDATE STRAIN FOR A LIVE, ORAL TYPHOID VACCINE", JOURNAL OF INFECTIOUS DISEASES, UNIVERSITY OF CHICAGO PRESS, CHICAGO, IL, vol. 131, no. 5, 1 May 1975 (1975-05-01), pages 553 - 558, XP009014131, ISSN: 0022-1899 *
GUZMAN, C.A.; BORSUTZKY, S.; GRIOT-WENK, M.; METCALFE, I.C.; PEARMAN, J.; COLLIOUD, A.; FAVRE, D.; DIETRICH, G.: "Vaccines against typhoid fever", VACCINE, vol. 24, 2006, pages 3804 - 3811, XP028010843, DOI: doi:10.1016/j.vaccine.2005.07.111
HARAGA, A.; OHLSON, M.B.; MILLER, S.I.: "Salmonellae interplay with host cells", NATURE REVIEWS, vol. 6, 2008, pages 53 - 66
HAUTEFORT, I.; THOMPSON, A.; ERIKSSON-YGBERG, S.; PARKER, M.L.; LUCCHINI, S.; DANINO, V.; BONGAERTS, R.J.; AHMAD, N.; RHEN, M.; HI: "During infection of epithelial cells Salmonella enterica serovar Typhimurium undergoes a time-dependent transcriptional adaptation that results in simultaneous expression of three type 3 secretion systems", CELLULAR MICROBIOLOGY, vol. 10, 2008, pages 958 - 984
HOISETH S K ET AL: "AROMATIC-DEPENDENT SALMONELLA TYPHIMURIUM ARE NON-VIRULENT AND EFFECTIVE AS LIVE VACCINES", NATURE, NATURE PUBLISHING GROUP, LONDON, GB, vol. 291, 21 May 1981 (1981-05-21), pages 238/239, XP000882816, ISSN: 0028-0836 *
HOISETH, S.K.; STOCKER, B.A.: "Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines", NATURE, vol. 291, 1981, pages 238 - 239, XP000882816, DOI: doi:10.1038/291238a0
JONES, S.M.; ELLIS, J.F.; RUSSELL, P.; GRIFFIN, K.F.; OYSTON, P.C.: "Passive protection against Burkholderia pseudomallei infection in mice by monoclonal antibodies against capsular polysaccharide, lipopolysaccharide or proteins", JOURNAL OF MEDICAL MICROBIOLOGY, vol. 51, 2002, pages 1055 - 1062, XP002421923
KOPECKO D J ET AL: "Genetic stability of vaccine strain Salmonella Typhi Ty21a over 25 years", INTERNATIONAL JOURNAL OF MEDICAL MICROBIOLOGY, URBAN UND FISCHER, DE, vol. 299, no. 4, 1 April 2009 (2009-04-01), pages 233 - 246, XP026068183, ISSN: 1438-4221, [retrieved on 20090101] *
LEHOUX, D.E.; SANSCHAGRIN, F.; KUKAVICA-IBRULJ, I.; POTVIN, E.; LEVESQUE, R.C.: "Identification of novel pathogenicity genes by PCR signature- tagged mutagenesis and related technologies", METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J, vol. 266, 2004, pages 289 - 304
LEVINE, M.M.; GALEN, J.; BARRY, E.; NORIEGA, F.; CHATFIELD, S.; SZTEIN, M.; DOUGAN, G.; TACKET, C.: "Attenuated Salmonella as live oral vaccines against typhoid fever and as live vectors", JOURNAL OF BIOTECHNOLOGY, vol. 44, 1996, pages 193 - 196, XP004036865, DOI: doi:10.1016/0168-1656(95)00094-1
LOVINGSHIMER M R ET AL: "Construction of an inducible, pfkA and pfkB deficient strain of Escherichia coli for the expression and purification of phosphofructokinase from bacterial sources", PROTEIN EXPRESSION AND PURIFICATION, ACADEMIC PRESS, SAN DIEGO, CA, vol. 46, no. 2, 1 April 2006 (2006-04-01), pages 475 - 482, XP024908700, ISSN: 1046-5928, [retrieved on 20060401] *
MACKEY, B.M.; DERRICK, C.M.: "A comparison of solid and liquid media for measuring the sensitivity of heat-injured Salmonella typhimurium to selenite and tetrathionate media, and the time needed to recover resistance", THE JOURNAL OF APPLIED BACTERIOLOGY, vol. 53, 1982, pages 233 - 242
MEYER, D.; SCHNEIDER-FRESENIUS, C.; HORLACHER, R.; PEIST, R.; BOOS, W.: "Molecular characterization of glucokinase from Escherichia coli K-12", JOURNAL OF BACTERIOLOGY, vol. 179, 1997, pages 1298 - 1306
MORGAN, E.; CAMPBELL, J.D.; ROWE, S.C.; BISPHAM, J.; STEVENS, M.P.; BOWEN, A.J.; BARROW, P.A.; MASKELL, D.J.; WALLIS, T.S.: "Identification of host-specific colonization factors of Salmonella enterica serovar Typhimurium", MOLECULAR MICROBIOLOGY, vol. 54, 2004, pages 994 - 1010
MORITA T ET AL: "Accumulation of glucose 6-phosphate or fructose 6-phosphate is responsible for destabilization of glucose transporter mRNA in Escherichia coli", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, INC, US, vol. 278, no. 18, 2 May 2003 (2003-05-02), pages 15608 - 15614, XP002274626, ISSN: 0021-9258 *
MUNOZ-ELIAS ERNESTO J ET AL: "Carbon metabolism of intracellular bacteria", CELLULAR MICROBIOLOGY, vol. 8, no. 1, January 2006 (2006-01-01), pages 10 - 22, XP002575495, ISSN: 1462-5814 *
NELSON, M.; PRIOR, J.L.; LEVER, M.S.; JONES, H.E.; ATKINS, T.P.; TITBALL, R.W.: "Evaluation of lipopolysaccharide and capsular polysaccharide as subunit vaccines against experimental melioidosis", JOURNAL OF MEDICAL MICROBIOLOGY, vol. 53, 2004, pages 1177 - 1182, XP002407041, DOI: doi:10.1099/jmm.0.45766-0
NNALUE N A ET AL: "Vaccination route, infectivity and thioglycollate broth administration: effects on live vaccine efficacy of auxotrophic derivatives of Salmonella choleraesuis", MICROBIAL PATHOGENESIS, ACADEMIC PRESS LIMITED, NEW YORK, NY, US, vol. 7, no. 4, 1 October 1989 (1989-10-01), pages 299 - 310, XP023312797, ISSN: 0882-4010, [retrieved on 19891001] *
OSWALD, I.P.; LANTIER, F.; MOUTIER, R.; BERTRAND, M.F.; SKAMENE, E.: "Intraperitoneal infection with Salmonella abortusovis is partially controlled by a gene closely linked with the Ity gene", CLINICAL AND EXPERIMENTAL IMMUNOLOGY, vol. 87, 1992, pages 373 - 378
PATERSON G K ET AL: "Redundancy in the requirement for the glycolytic enzymes phosphofructokinase (Pfk) 1 and 2 in the in vivo fitness of Salmonella enterica serovar Typhimurium", MICROBIAL PATHOGENESIS, ACADEMIC PRESS LIMITED, NEW YORK, NY, US, vol. 46, no. 5, 1 May 2009 (2009-05-01), pages 261 - 265, XP026057451, ISSN: 0882-4010, [retrieved on 20090205] *
POSTMA P W: "DEFECTIVE ENZYME II-B-GLC OF THE PHOSPHOENOL PYRUVATE SUGAR PHOSPHO TRANSFERASE SYSTEM LEADING TO UNCOUPLING OF TRANSPORT AND PHOSPHORYLATION IN SALMONELLA-TYPHIMURIUM", JOURNAL OF BACTERIOLOGY, vol. 147, no. 2, 1981, pages 382 - 389, XP002575490, ISSN: 0021-9193 *
POSTMA, P.W.; LENGELER, J.W.; JACOBSON, G.R.: "Phosphoenolpyruvate:carbohydrate phosphotransferase systems of bacteria", MICROBIOLOGICAL REVIEWS, vol. 57, 1993, pages 543 - 594, XP001051826
RILEY, M.: "Functions of the gene products of Escherichia coli", MICROBIOLOGICAL REVIEWS, vol. 57, 1993, pages 862 - 952
SMITH, H.O.; LEVINE, M.: "A phage P22 gene controlling integration of prophage", VIROLOGY, vol. 31, 1967, pages 207 - 216, XP023058088, DOI: doi:10.1016/0042-6822(67)90164-X
TITBALL, R.W.: "Vaccines against intracellular bacterial pathogens", DRUG DISCOVERY TODAY, vol. 13, 2008, pages 596 - 600, XP022797176, DOI: doi:10.1016/j.drudis.2008.04.010
TROUTT, H.F.; GALLAND, J.C.; OSBURN, B.I.; BREWER, R.L.; BRAUN, R.K.; SCHMITZ, J.A.; SEARS, P.; CHILDERS, A.B.; RICHEY, E.; MATHER: "Prevalence of Salmonella spp in cull (market) dairy cows at slaughter", JOURNAL OF THE AMERICAN VETERINARY MEDICAL ASSOCIATION, vol. 219, 2001, pages 1212 - 1215
TSOLIS, R.M.; KINGSLEY, R.A.; TOWNSEND, S.M.; FICHT, T.A.; ADAMS, L.G.; BAUMLER, A.J.: "Of mice, calves, and men. Comparison of the mouse typhoid model with other Salmonella infections", ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY, vol. 473, 1999, pages 261 - 274
WARD, S.J.; DOUCE, G.; FIGUEIREDO, D.; DOUGAN, G.; WREN, B.W.: "Immunogenicity of a Salmonella typhimurium aroA aroD vaccine expressing a nontoxic domain of Clostridium difficile toxin A", INFECTION AND IMMUNITY, vol. 67, 1999, pages 2145 - 2152
WRAY, C.; SOJKA, W.J.: "Experimental Salmonella typhimurium infection in calves", RESEARCH IN VETERINARY SCIENCE, vol. 25, 1978, pages 139 - 143
WU, J.Y.; NEWTON, S.; JUDD, A.; STOCKER, B.; ROBINSON, W.S.: "Expression of immunogenic epitopes of hepatitis B surface antigen with hybrid flagellin proteins by a vaccine strain of Salmonella", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 86, 1989, pages 4726 - 4730, XP002348920, DOI: doi:10.1073/pnas.86.12.4726

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015103104A1 (en) * 2014-01-06 2015-07-09 The United States Of America, As Represented By The Secretary Of Agriculture Attenuated salmonella enterica
US9868769B2 (en) 2014-01-06 2018-01-16 The United States Of America, As Represented By The Secretary Of Agriculture Mutated Salmonella enteriaca
US10351606B2 (en) 2014-01-06 2019-07-16 The United States Of America, As Represented By The Secretary Of Agriculture Mutated Salmonella enterica
CN113088454A (en) * 2021-04-08 2021-07-09 武汉轻工大学 Method for separating intact bacteria from infected cells

Also Published As

Publication number Publication date
GB0900974D0 (en) 2009-03-04

Similar Documents

Publication Publication Date Title
US8889121B2 (en) Bacterium comprising a regulated rfaH nucleic acid
US6399074B1 (en) Live attenuated salmonella vaccines to control avian pathogens
US10517939B2 (en) Live attenuated vaccines
La Ragione et al. Efficacy of a live attenuated Escherichia coli O78∶ K80 vaccine in chickens and turkeys
US20080274139A1 (en) Attenuated microorganisms for the treatment of infection
HU205262B (en) Process for producing new, nonreverting living vaccine and stock against shigella
DE60112413T2 (en) SSA INACTIVATED SALMONELLA VACCINES
WO2010045620A1 (en) Recombinant bacterium capable of eliciting an immune response against streptococcus pneumoniae
JP2009531029A (en) Attenuated Salmonella live vaccine
CN104797268B (en) A kind of novel shigella attenuated live vaccine
WO2012092226A1 (en) Veterinary vaccine composition against infections caused by salmonella
JP2020505036A (en) Induction of protective immunity against antigen
JP2002507414A (en) Attenuated bacteria used in vaccines
ES2824402T3 (en) Attenuated vaccines against Pasteurella multocida and procedures for their manufacture and use
US20230090746A1 (en) Attenuated salmonella synthesizing antigens for vaccinating against helicobacter pylori
US20050019335A1 (en) Salmonella vaccine
WO2010084350A1 (en) Mutant pathogenic bacterial and live attenuated vaccine compositions
JP2009529895A (en) Attenuated Salmonella live vaccine
US20220218810A1 (en) New immunogenic compositions
Zhao et al. Regulated delayed attenuation enhances the immunogenicity and protection provided by recombinant Salmonella enterica serovar Typhimurium vaccines expressing serovar Choleraesuis O-polysaccharides
WO2001046428A1 (en) Vaccine strains against infection with pathogenic bacteria
EP1037664A1 (en) Vaccines containing attenuated bacteria
WO2000073462A9 (en) Attenuated mutant enteropathogenic e. coli (epec) strains, process for their production and their use
WO2017209917A1 (en) Live attenuated catfish vaccine and method of making
KR20170010540A (en) Salmonella enteritidis mutant strains hid2092 and hid2114 and pharmaceutical composition using the same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10702162

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10702162

Country of ref document: EP

Kind code of ref document: A1