US20210290667A1 - Mutant Bordetella Strains and Methods of Use - Google Patents

Mutant Bordetella Strains and Methods of Use Download PDF

Info

Publication number
US20210290667A1
US20210290667A1 US17/337,367 US202117337367A US2021290667A1 US 20210290667 A1 US20210290667 A1 US 20210290667A1 US 202117337367 A US202117337367 A US 202117337367A US 2021290667 A1 US2021290667 A1 US 2021290667A1
Authority
US
United States
Prior art keywords
pertactin
deficient
mice
bordetella
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/337,367
Inventor
Luis Solans
Camille Locht
Anne Tsicopoulos
Saliha Ait-Yahia Sendid
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institut Pasteur de Lille
Institut National de la Sante et de la Recherche Medicale INSERM
Original Assignee
Institut Pasteur de Lille
Institut National de la Sante et de la Recherche Medicale INSERM
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 Institut Pasteur de Lille, Institut National de la Sante et de la Recherche Medicale INSERM filed Critical Institut Pasteur de Lille
Priority to US17/337,367 priority Critical patent/US20210290667A1/en
Publication of US20210290667A1 publication Critical patent/US20210290667A1/en
Priority to US18/338,314 priority patent/US20230381224A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/42Respiratory system, e.g. lungs, bronchi or lung 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
    • A61K39/099Bordetella
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • C07K14/235Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Bordetella (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
    • 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
    • A61K2039/10Brucella; Bordetella, e.g. Bordetella pertussis; Not used, see subgroups
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation

Definitions

  • the invention relates generally to the fields of microbiology, immunology, allergy, and medicine. More particularly, the invention relates to live attenuated Bordetella pertussis strains deficient in pertactin, and their use as prophylactic and therapeutic agents in various disease settings.
  • Microbial organisms and their components have long been known to affect the immune systems of mammals. Infection with virulent bacteria and viruses can cause severe illness or death. Contributing toward this, purified components of bacteria and viruses can also cause pathology by inducing inflammatory responses or otherwise causing the immune system to behave in an undesirable manner. Despite this, vaccines including whole bacteria, viruses, or parts thereof have not only proven to be one of the most powerful tools that medicine has developed to prevent serious infections, but also can cause other beneficial effects.
  • BPZE1 live attenuated pertussis vaccine candidate named BPZE1 (see WO2007104451 A1) was found to not only protect against virulent Bordetella pertussis , but also to exert potent anti-allergic and anti-asthma effects by dampening hyperimmune responses to allergens (see WO2013066272A1).
  • BPZE1P pertactin-deficient mutant of BPZE1P (deposited in accordance with the requirements of the Budapest Treaty with the Collection Nationale de Cultures de Microorganismes (“CNCM”), 25. Rue du Dondel Roux. Paris Cedex 15, 75724. France on Dec.
  • BPZE1P colonized lungs as efficiently as BPZE1 and induced protective immunity against B. pertussis challenge as efficiently as BPZE1.
  • BPZE1P colonized the mouse lungs significantly better than BPZE1. Therefore, pertactin-deficient B. pertussis strains such as BPZE1P may be advantageous in protecting against respiratory tract inflammation in subjects with high pre-existing titers of pertactin antibodies, including those previously vaccinated with pertactin-containing acellular vaccines.
  • the airway inflammation can be associated with one or more of airway resistance, eosinophil infiltration in the lungs of the subject, and/or increased amounts of inflammatory cytokines in the lungs of the subject. Colonization of the respiratory tract of the subject can result in reduction or prevention of such airway resistance, eosinophil infiltration, and/or increased amounts of inflammatory cytokines.
  • compositions including a pharmaceutically acceptable carrier and live pertactin-deficient Bordetella bacteria capable of colonizing the respiratory tract of a subject and reducing or preventing the development of airway inflammation in the subject.
  • the live pertactin-deficient Bordetella bacteria can lack a functional gene encoding pertactin, also be deficient in tracheal cytotoxin (TCT), pertussis toxin (PTX), and/or dermonecrotic toxin (DNT).
  • TCT tracheal cytotoxin
  • PTX pertussis toxin
  • DNT dermonecrotic toxin
  • the live pertactin-deficient Bordetella bacteria can be BPZE1P.
  • the airway inflammation can be caused by exposure to an allergen, and the subject can be one diagnosed with asthma, interstitial lung disease, or allergic rhinitis; one having greater than 10 ng per ml of anti-pertactin antibodies in its serum; or one that has previously been immunized with a vaccine containing pertactin or a pertactin-like antigen.
  • pertactin is the outer surface membrane protein produced by Bordetella pertussis and its close relatives, such as Bordetella parapertussis that is involved in the binding of Bordetella bacteria to host cells as described in Leininger et al., Proc. Nat'l. Acad. Sci. USA, 1991, 88:345-9. conserveed regions in this protein, such as its passenger and autotransporter domains, contribute directly to the overall virulence and pathogenicity of these organisms.
  • PTX pertussis toxin, a major virulence factor of B. pertussis , which induces metabolic changes and alters immune responses in the host as described in Saukkonen et al., Proc. Natl. Acad. Sci. USA., 1992, 89:118-122.
  • DNT pertussis dermonecrotic toxin
  • lethal toxin a toxin found in B. pertussis which induces inflammation, vasoconstriction and dermonecrotic lesions in sites where B. pertussis colonize the respiratory tract.
  • TCT refers to tracheal cytotoxin, a disaccharide tetrapeptide derivative of peptidoglycan synthesized by bordetellae, which induces the production of interleukin-1 and nitric oxide synthase, and causes stasis of cilia and lethal effects on respiratory epithelial cells.
  • a “pertactin-deficient” Bordetella strain is one that exhibits at least less than 50% (e.g., less than 50, 40, 30, 20, 10, 5, 4, 3, 2, or 1%) of the pertactin activity found in BPZE1 under the conditions described in the Examples section below, one that exhibits no detectable pertactin activity, or one that exhibits not detectable expression of pertactin as determined by Western blotting.
  • the term “functional” when referring to a toxin or virulence factor in a bacterial strain means that (i) the toxin/virulence factor expressed by the strain has not been mutated to eliminate or at least reduce by greater than 50% its enzymatic activity compared to the non-mutated version of the toxin/factor, and/or (ii) that a bacterial strain expressing the toxin/factor has not been engineered or selected to eliminate or at least reduce by greater than 50% the number of molecules of that toxin/factor compared to the starting strain from which the engineered or selected strain was derived.
  • mammalian subject or “subject” encompasses any of various warm-blooded vertebrate animals of the class Mammalia, including human beings.
  • FIG. 1A is a diagram showing the structure of a plasmid used in the construction of BPZE1P.
  • FIG. 1B is another diagram showing the construction of BPZE1P. and photographs of gels showing the results of PCR amplification of the prn UPg and prn Log fragments.
  • FIG. 1C is photographs of immunoblots showing the presence of pertactin in BPZE1, but not in BPZE1P lysates and supernatants.
  • FIG. 2 is a graph showing the results of lung colonization in Balb/c mice using either BPZE1 or BPZE1P.
  • FIG. 3 is a graph showing total IgG titers after BPZE1P or BPZE1 administration to mice.
  • FIG. 4A is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 10 6 viable B. pertussis bacteria of the BPSM strain.
  • FIG. 4B is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 10 6 viable B. pertussis bacteria of the BPSMP strain.
  • FIG. 4C is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 10 6 viable B. pertussis bacteria of the B1917 strain.
  • FIG. 5 is a graph showing the fitness of BPZE1 and BPZE1P in mice preimmunized with an acellular B. pertussis vaccine (aPv).
  • FIG. 6 is a diagram of the experimental protocol of an assay for airway responsiveness in allergic mice vaccinated with BPZE1, BPZE1P or left unvaccinated, and a graph showing the results of the assay.
  • FIG. 7A is a graph showing the total airway cell population infiltration in the bronchoalveolar (BAL) fluid of the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 7B is a graph showing the percentage of eosinophils in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 7C is a graph showing the percentage of neutrophils in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 7D is a graph showing the percentage of lymphocytes in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 7E is a graph showing the percentage of macrophages in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8A is a graph showing the amount of IL-1 ⁇ normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8B is a graph showing the amount of IL-1 ⁇ normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8C is a graph showing the amount of IL-6 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8D is a graph showing the amount of IL-13 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8E is a graph showing the amount of CXCL1 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8F is a graph showing the amount of CXCL9 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8G is a graph showing the amount of CXCL10 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 8H is a graph showing the amount of GM-CSF normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6 .
  • FIG. 9 is a diagram of the experimental protocol of an assay for airway responsiveness in a therapeutic model of allergic mice vaccinated with BPZE1, BPZE1P or left unvaccinated, and a graph showing the results of the assay.
  • Bordetella strains deficient in pertactin and their use in stimulating anti- Bordetella immune responses as well as in preventing and treating respiratory tract inflammation.
  • the below described embodiments illustrate representative examples of these methods. Nonetheless, from the description of these embodiments, other aspects of the invention can be made and/or practiced based on the description provided below.
  • Microbiological methods are described in Methods for General and Molecular Microbiology (3d Ed). Reddy et al., ed., ASM Press. Immunological methods are generally known in the at and described in methodology treatises such as Current Protocols in Immunology. Coligan et al., ed., John Wiley & Sons, New York. Techniques of molecular biology are described in detail in treatises such as Molecular Cloning: A Laboratory Manual, 2nd ed., vol.
  • Bordetella species such as Bordetella pertussis, Bordetella parapertussis , and Bordetella bronchiseptica that are deficient in pertactin expression (e.g., those that express at least 50, 60, 70, 80, 90, 95, 96, 97, 98, or 99% less pertactin than do corresponding strains) can be used to generate immune responses against Bordetella species, as well as to treat and/or prevent respiratory tract inflammation such as that which occurs in allergic asthma.
  • Bordetella species such as Bordetella pertussis, Bordetella parapertussis , and Bordetella bronchiseptica that are deficient in pertactin expression (e.g., those that express at least 50, 60, 70, 80, 90, 95, 96, 97, 98, or 99% less pertactin than do corresponding strains) can be used to generate immune responses against Bordetella species, as well as to treat and/or prevent respiratory tract inflammation such
  • Live, attenuated, pertactin-deficient Bordetella pertussis and live, attenuated, pertactin-deficient Bordetella parapertussis are preferred for use for treating or preventing allergic respiratory tract inflammation in human subjects.
  • the live attenuated pertactin-deficient Bordetella strains described herein can be made by adapting methods known in the art such as those described in the Examples section below.
  • the starting strain can be any suitable Bordetella species. Examples of Bordetella species include B. pertussis, B. parapertussis , and B. bronchiseptica. B. pertussis is preferred for use as the starting strain for vaccines and methods for preventing pertussis infection.
  • Bordetella strains for use as starting strains are available from established culture collections (e.g., the American Type Culture Collection in Manassas, Va.) or can be isolated from natural reservoirs (e.g., a patient having pertussis) by known techniques (e.g., as described in Aoyama et al., Dev. Biol. Stand, 73:185-92, 1991).
  • Bordetella strains which express functional pertactin can be made deficient in this molecule or its activity by selection or, preferably for stability purposes, mutagenesis (e.g., deletion of the native prn gene as described below).
  • Bordetella species deficient in pertactin can also be isolated from natural sources (e.g., human subjects or other mammals infected or colonized with such strains). Because insufficient attenuation of a pathogenic strain of Bordetella might cause a pathological infection in a subject, it is preferred that the pertactin-deficient Bordetella strain used also have lower levels of other functional virulence factors.
  • pertactin-deficient Bordetella strain retains the ability to colonize a subject and exert a protective effect on respiratory tract inflammation, it must not be overly attenuated. Attenuation might be achieved by mutating the strain to reduce its expression of pertactin and one or more (e.g., 1, 2, 3, 4, 5 or more) of the following: pertussis toxin (PTX), dermonecrotic toxin (DNT), tracheal cytotoxin (TCT), adenylate cyclase (AC), lipopolysaccharide (LPS), filamentous hemagglutinin (FHA), or any of the bvg-regulated components.
  • PTX pertussis toxin
  • DNT dermonecrotic toxin
  • TCT tracheal cytotoxin
  • AC adenylate cyclase
  • LPS lipopolysaccharide
  • FHA filamentous hemagglutinin
  • Attenuation might also be achieved by mutating the strain to reduce the biological activity of pertactin and one or more (e.g., 1, 2, 3, 4, 5 or more) of the following: pertussis toxin (PTX), dermonecrotic toxin (DNT), tracheal cytotoxin (TCT), adenylate cyclase (AC), lipopolysaccharide (LPS), filamentous hemagglutinin (FHA), or any of the bvg-regulated components.
  • pertussis toxin PTX
  • DNT dermonecrotic toxin
  • TCT tracheal cytotoxin
  • AC adenylate cyclase
  • LPS lipopolysaccharide
  • FHA filamentous hemagglutinin
  • Bordetella strain deficient in functional pertactin, functional PTX, functional DNT, and functional TCT was able to colonize the respiratory tract of subjects, induce immune responses against Bordetella , and reduce or prevent the development of allergic and inflammatory responses.
  • Bordetella strains such as BPZE1P, which are deficient in these four virulence factors and can colonize a subject and induce immune responses targeting Bordetella strains and/or reduce or prevent the development of allergic and inflammatory responses are preferred.
  • a variety of methods are known in the art for attenuating an infectious bacterial strain. These include passaging the strain in vitro until virulence is lost, non-specific chemical mutagenesis followed by screening and selection based on phenotype, and using targeted molecular biology techniques such as those described in the Examples section below (including allelic exchange) and in Methods for General and Molecular Microbiology (3d Ed), Reddy et al., ed., ASM Press. Using these methods, the genes encoding pertactin. PTX, and/or DNT can be deleted or mutated to an enzymatically inactive form (which is preferred where it is desired to retain the toxin's antigenicity).
  • TCT production can be significantly (e.g., >than 99.99, 99.90, 99.8, 99.7, 99.6, 99.5, 99.0, 98, 97, 96, 95, or 90%) reduced by replacing the native ampG gene (unlike other species, B. pertussis ampG does not actively recycle TCT-containing peptidoglycan) with a heterologous (e.g., from E. coli or another gram-negative species) ampG gene, or by mutating the native ampG gene such that it is active at recycling peptidoglycan.
  • native ampG gene unlike other species, B. pertussis ampG does not actively recycle TCT-containing peptidoglycan
  • a heterologous ampG gene e.g., from E. coli or another gram-negative species
  • Modification of a starting strain to reduce or remove toxin/virulence factor activity can be confirmed by sequencing the genomic DNA or genes encoding the toxins of the modified strains. Southern. Northern, and/or Western blotting might also be used to confirm that the target genes have been deleted or that expression of the target factors has been reduced or removed. Biological activity can also be evaluated to confirm reduction or removal of toxin/virulence factor activity. Once the modifications have been confirmed, the modified strains can be tested for the ability to colonize a subject and to induce protective immunity against Bordetella infection or to reduce or prevent the development of allergic and inflammatory responses by known methods such as those described in the Examples section below.
  • the live attenuated Bordetella strains described herein can be used in compositions that protect a mammalian subject from developing a Bordetella infection (e.g., pertussis), or to reduce the symptoms of such an infection. They can also be used to reduce or prevent the development of allergic and inflammatory responses in a subject such as asthma, allergic rhinitis, interstitial lung disease, food allergies, peanut allergy, venom allergies, atopic dermatitis, contact hypersensitivity, and anaphylaxis.
  • the live attenuated Bordetella strains are typically formulated with a pharmaceutically acceptable excipient.
  • pharmaceutically acceptable excipients include, e.g., buffered saline solutions, distilled water, emulsions such as an oil/water emulsion, various types of wetting agents, sterile solutions, and the like.
  • the vaccines can be packaged in unit dosage form for convenient administration to a subject.
  • a single dose of between 1 ⁇ 10 4 to 1 ⁇ 10 9 e.g., 1 ⁇ 10 4 . 5 ⁇ 10 4 , 1 ⁇ 10 5 , 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 +/ ⁇ 10, 20, 30, 40, 50, 60, 70, 80, or 90%
  • live bacteria of the selected attenuated Bordetella strain and any excipient can be separately contained in packaging or in an administration device.
  • the vaccine can be contained within an administration device such as a syringe, spraying device, or insufflator.
  • compositions described herein can be administered to a mammalian subject (e.g., a human being, a human child or neonate, a human adult, a human being at high risk from developing complications from pertussis, a human being with lung disease, a human being that is or will become immunosuppressed, and a human being having or at high risk for developing respiratory tract inflammation such as asthma, allergic rhinitis, or interstitial lung disease) by any suitable method that deposits the bacteria within the composition in the respiratory tract or other mucosal compartment.
  • a mammalian subject e.g., a human being, a human child or neonate, a human adult, a human being at high risk from developing complications from pertussis, a human being with lung disease, a human being that is or will become immunosuppressed, and a human being having or at high risk for developing respiratory tract inflammation such as asthma, allergic rhinitis, or interstitial lung disease
  • the compositions may be administered by inhalation or intranas
  • the pertactin-deficient Bordetella strains described herein can be formulated as compositions for administration to a subject.
  • a suitable number of live bacteria are mixed with a pharmaceutically suitable excipient or carrier such as phosphate buffered saline solutions, distilled water, emulsions such as an oil/water emulsions, various types of wetting agents, sterile solutions and the like.
  • the vaccine can be lyophilized and then reconstituted prior to administration.
  • pharmaceutically suitable excipients or carriers which are compatible with mucosal (particularly nasal, bronchial, or lung) administration are preferred for colonizing the respiratory tract. See Remington's Pharmaceutical Sciences, a standard text in this field, and USP/NF.
  • each dose of a composition can include a sufficient number of live pertactin-deficient Bordetella bacteria to result in colonization of the mucosal site, e.g., approximately (i.e., +/ ⁇ 50%) 5 ⁇ 10 3 to 5 ⁇ 10 9 bacteria.
  • the dose can include approximately 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , or 1 ⁇ 10 10 live pertactin-deficient Bordetella bacteria.
  • the dose may be given once or on multiple (2, 3, 4, 5, 6, 7, 8 or more) occasions at intervals of 1, 2, 3, 4, 5, or 6 days or 1, 2, 3, 4, 5, or 6 weeks, or 1, 2, 3, 4, 5, 6, or 12 months.
  • sufficient amounts of a composition are administered to result in colonization and the protective and/or anti-inflammatory response.
  • Additional amounts are administered after the induced protective and/or anti-inflammatory response wanes (e.g., after the subject resumes suffering from the symptoms of respiratory tract inflammation).
  • Subjects which can be administered a composition containing live pertactin-deficient Bordetella bacteria can include any capable of being colonized with a selected live pertactin-deficient Bordetella bacterial strain.
  • the subject can be a mammal such as a human being.
  • Human subjects having, or at high risk of developing, respiratory tract inflammation such as those having or prone to developing allergic asthma or allergic rhinitis are preferred recipients of the composition.
  • compositions can be used in subjects regardless of their titers of anti-pertactin antibodies, the composition may be used in those having measurable titers (e.g., greater than 10, 20, 50, 100, 200, or 500 ng per ml of serum) of anti-pertactin antibodies and those having previously been immunized with a vaccine containing pertactin or a pertactin-like antigen because pertactin-deficient Bordetella bacterial strains are not subject to pertactin-targeting immune responses.
  • measurable titers e.g., greater than 10, 20, 50, 100, 200, or 500 ng per ml of serum
  • compositions in dampening respiratory tract inflammation can be assessed by known methods, e.g., measuring the number of inflammatory cells, IgE titers, levels of pro-inflammatory cytokines/chemokines (such as eotaxin, GM-CSF. IFN ⁇ , IL-4, IL-5, IL-8, IL-10, IL-12, IL-13, IL-17A, IL-17F, IL-18, and TNF ⁇ ) in fluid taken from the respiratory tract (e.g., bronchoalveolar lavage fluid), or clinical parameters such as spirometry or the level of dyspnea, coughing, wheezing, or respiratory capacity.
  • cytokines/chemokines such as eotaxin, GM-CSF. IFN ⁇ , IL-4, IL-5, IL-8, IL-10, IL-12, IL-13, IL-17A, IL-17F, IL-18, and TNF ⁇
  • fluid taken from the respiratory tract e.g., bron
  • Improvement in any of one or more of these parameters indicates that the composition is effective.
  • Animal models of allergic respiratory tract inflammation can also be used to assess the effectiveness of a composition, see e.g., U.S. Pat. No. 8,986,709.
  • Escherichia coli DH5 ⁇ , SM10 and B. pertussis BPZE1, BPSM (Menozzi et al., Infect Immun 1994; 62:769-778) and B1917 (Bart et al. Genome Announc 2014; 2(6)) were used in this study.
  • the Bordetella strains were cultured at 37° C. on Bordet-Gengou agar (BG), supplemented with 1% glycerol and 10% defibrinated sheep blood. After growth, the bacteria were harvested by scraping the plates and resuspended in phosphate-buffered saline (PBS) at the desired density. For liquid culture the Bordetella strains were grown at 37° C.
  • E. coli strains used for cloning procedures were growth in LB broth or LB agar plates.
  • streptomycin (Sm) was used at 100 ⁇ g/ml
  • gentamycin (Gm) was used at 10 ⁇ g/ml
  • ampicillin (Amp) was used at 100 ⁇ g/ml.
  • the gene coding for pertactin, in BPZE1, a 739-bp fragment downstream (prn LO) of the prn gene and a 759-bp fragment upstream (prn UP) of the prn gene were cloned into pSS4940 to introduce the prn deletion in the BPZE1 and BPSM genomes by homologous recombination. Referring to FIGS.
  • the upstream and downstream prn flanking region were PCR amplified using prn_KO_fw (ATCCTCAAGCAAGACTGCGAGCTG) (SEQ ID NO:1)) and OL_prn_KO_rv (GGGGATAGACCCTCCTCGCTTGGATGCCAGGTGGAGAGCA) (SEQ ID NO:2)), and OL_prn_KO_fw (TGCTCTCCACCTGGCATCCAAGCGAGGAGGGTCTATCCCC) (SEQ ID NO:3)) and prn_KO_rv (CCATCATCCTGTACGACCGCCT) (SEQ ID NO:4)), respectively, as primers.
  • prn_KO_fw ATCCTCAAGCAAGACTGCGAGCTG
  • OL_prn_KO_rv GGGGATAGACCCTCCTCGCTTGGATGCCAGGTGGAGAGCA
  • OL_prn_KO_fw TGCTCTCCACCTGGCATCCAAGCGAGGAGGGTCTATCCCC
  • the resulting fragment (containing the prn deletion) was inserted into the TOPO Blunt® vector (ThermoFisher Scientific) and then excised as a KpnI-NotI fragment.
  • the excised KpnI-NotI fragment was inserted into KpnI- and NotI-digested pSS4940, a pSS4245 (Inatsuka et al., infect immun 2010; 78 2901-2909) derivative.
  • the resulting plasmid was transformed into E. coli SM10, which was then conjugated with BPZE1.
  • PCR was used to confirm deletion of the entire prin gene by amplifying the flanking regions covering the construction using prnKO_UP (TTCTGCGCGAACAGATCAAAC) (SEQ ID NO:5))—prnKOin_UPrv (CTGCTGGTCATCGGCGAAGT) (SEQ ID NO:6)) for the 5′ region and prnKOin_LOfw (CGCCCATTCTCCCTGTTCC) (SEQ ID NO:7))—prnKO_LO (GAACAGGAACTGGAACAGGCG) (SEQ ID NO:8)) for the 3′ region.
  • a strain carrying the prn deletion was selected and named BPZE1P.
  • the same strategy was used to construct a pertactin-deficient BPSM mutant, named BPSMP.
  • BPZE1 and BPZE1P lysates and supernatants were tested by immunoblotting of BPZE1 and BPZE1P lysates and supernatants, using purified Prn (List Biological laboratories) as control for correct size of the band.
  • Prn List Biological laboratories
  • BPZE1 and BPZE1P strains were plated onto BG blood agar and incubated for 48 h at 37° C. After growth, the bacteria were scraped off the plates, resuspended in 10 ml of Stainer-Scholte medium and grown for 4 days at 37° C. The bacteria were then harvested by centrifugation.
  • the supernatants were recovered and treated with trichloroacetic acid (TCA) as described previously (Solans et al., PLoS Pathog 2014; 10:e1004183) for protein concentration.
  • TCA trichloroacetic acid
  • the bacterial pellets were resuspended in PBS complemented with an EDTA-free protease inhibitor cocktail (Roche) and lysed using a French pressure cell. Bacterial debris were removed by centrifugation for 30 minutes at 15.000 ⁇ g, and the supernatants were recovered for immunoblotting. Proteins were separated by 12% SDS-PAGE and then transferred onto a Nitrocellulose membrane using the Criterion rM cell system (Bio-Rad).
  • mice Groups of 18 six-week old mice were inoculated intranasally with 20 ⁇ l PBS containing 10 6 viable bacteria as described previously (Mielcarek et al., supra). At the indicated time points (3 hours, 3 days, 7 days, 14 days, 21 days and 28 days), 3 mice per group were sacrificed, and lungs were harvested and homogenized for measuring total number of colony formation units (CFU). Statistical analysis was done by a 2-way ANOVA test, using post hoc comparison Bonferroni test with confidence intervals of 95%. Referring to FIG. 2 , both BPZE1 and BPZE1P colonized the animals equally well. Both strains exhibited a peak of multiplication 3 days post vaccination and colonization persisted for 4 weeks. No statistically significant difference was observed between these strains in their ability to colonize the mouse lungs.
  • BPZE1P is as Immunogenic and Protective Against Challenge with Virulent B. pertussis as BPZE1
  • Immunity induced by BPZE1P in comparison with BPZE1 was measured by antibody titration of mouse immune serum after nasal vaccination.
  • Groups of 8 mice were vaccinated intranasally with 10 5 viable BPZE1 or BPZE1P.
  • the mice were bled, and total IgG titers were measured against total BPSM lysate.
  • Blood was centrifuged for 5 min. at 5,000 ⁇ g to separate the serum from the cells.
  • Antibody titers against B. pertussis were estimated using enzyme-linked immunosorbent assays (ELISA) as described previously (Mielcarek et al., supra), using total B. pertussis BPSM lysate at 1 ⁇ g of total protein per well.
  • ELISA enzyme-linked immunosorbent assays
  • mice The protective efficacy of BPZE1P compared with BPZE1 was tested in a suboptimal protection protocol by measuring the CFU counts in the lungs 7 days post challenge, and comparing a na ⁇ ve group with the vaccinated groups.
  • Groups of 8 six-week old mice were vaccinated intranasally with 20 ⁇ l PBS containing 105 viable BPZE1 or BPZE1P. or were left unvaccinated.
  • All mice were challenged intranasally with 20 ⁇ l PBS containing 10 6 viable BPSM. BPSMP or B1917.
  • Three hours after the challenge 3 mice per group were sacrificed, and lungs were harvested and homogenized for CFU counting.
  • mice per group were sacrificed 7 days after challenge for CFU counting. Three hours post infection, 3 mice were euthanized, and their lungs were harvested to determine the CFU counts shortly after challenge. Seven days post-infection, the remaining 5 mice were euthanized, their lungs were harvested, and the CFU counts were measured.
  • Statistical analysis was done applying a parametric 2-way ANOVA test, using post hoc Bonferroni comparison test with a confidence interval of 95%, *, p ⁇ 0.005; **, p ⁇ 0.001; ***, p ⁇ 0.0001.
  • vaccination with either strain protected against challenge with BPSM, BPSMP and B1917 equally well.
  • mice having pre-existing antibodies against pertactin were vaccinated subcutaneously with 1 ⁇ 5 of the human dose of the acellular pertussis vaccine (aPv; INFANRIX®, GSK; containing inactivated pertussis toxin, filamentous hemagglutinin and pertactin).
  • aPv acellular pertussis vaccine
  • INFANRIX® GSK
  • aPv acellular pertussis vaccine
  • the mice were boosted with the same dose of aPv.
  • the mice were infected intranasally with 10 6 BPZE1 or BPZE1P.
  • mice Seven days post-infection, the remaining 5 mice were euthanized, the lungs harvested, and the CFU counts were measured. Statistical analysis was done applying a parametric 2-way ANOVA test, using post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p ⁇ 0.0001.
  • BPZE1P colonized the lungs significantly better than BPZE1.
  • Mice infected with BPZE1P had almost 10 4 CFU in the lungs 7 days after administration, while the CFU counts in the lungs of the mice given BPZE1 reached 102 CFU.
  • mice The effect of vaccination with BPZE1P on airway responsiveness was investigated in allergic mice as described in the protocol shown in FIG. 6 .
  • Groups of 4 weeks-old mice were vaccinated intranasally with 20 ⁇ l PBS containing 10 6 viable BPZE1 or BPZE1P, or were left unvaccinated.
  • the mice were sensitized intranasally with 20 ⁇ l of house dust mite (HDM; Stallergenes S. A.) of 5 index of reactivity (IR) of Dermnaophagoidesfarinae extract (Derf 5IR) or 20 ⁇ l of PBS as control.
  • HDM house dust mite
  • IR index of reactivity
  • Derf 5IR Dermnaophagoidesfarinae extract
  • mice Ten days later, the mice where challenged intranasally with 20 ⁇ l of Derf 5IR or PBS daily for 5 days, and two days later, the mice were anesthetized and intubated intratracheally for mechanical ventilation using the FLEXIVENT® (SCIREQ®) device. The mice were then exposed to increasing concentrations of nebulized methacholine (0-50 mg/mL in PBS) (Sigma-Aldrich) to determine the resistance in their respiratory airways using plethysmography. Statistical analysis was done by applying a parametric 2-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p ⁇ 0.0001; **, p ⁇ 0.001; *, p ⁇ 0.005. In FIG.
  • BAL Bronchoalveolar lavage
  • FIG. 7C lymphocytes
  • FIG. 7D lymphocytes
  • FIG. 7E macrophages
  • FIG. 7A Statistical analysis was done by applying a parametric one-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p ⁇ 0.0001; *,p ⁇ 0.005.
  • BPZE1 or BPZE1P vaccination significantly reduced the recruitment of total cells in the airways after allergen exposure and challenge, compared to the non-vaccinated mice ( FIG. 7A ). This reduction reflected essentially the reduction in eosinophil recruitment in the vaccinated mice ( FIG. 7B ), whereas there was no significant change in the percentages of neutrophils or lymphocytes ( FIGS. 7C and D) between the vaccinated and non-vaccinated mice.
  • FIGS. 7C and D A small, but significant increase in the percentage of macrophages was observed in the mice that were vaccinated with BPZE1P ( FIG. 7E ).
  • the right lung lobes were harvested and directly frozen in liquid Nitrogen for protein extraction.
  • the lung lobes were resuspended in 1 mL of lysis buffer, PBS with 0.5% nonidet P40 and protease inhibitor cocktail (Roche @), and homogenized at 4° C. using T-18 ULTRA-TURRAX@ (IKA®).
  • the samples were centrifuged, and the supernatants were collected for total protein quantification using the PIERCETM BCA protein assay (Thermo Fisher Scientific), and for cytokine and chemokine measurements using the Cytokine 20-Plex Mouse Panel (INVITROGENTM, Thermo Fisher Scientific) per the manufacturer's specifications. Referring to FIG.
  • cytokine levels are represented as the normalization of the cytokine/chemokine quantification against total proteins measured in the lung lobe.
  • Statistical analysis was done by applying a parametric one-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, **, p ⁇ 0.001, *; p ⁇ 0.005.
  • HDM Dense 5IR-treated mice produced significantly increased levels of IL1 ⁇ and IL1 ⁇ in the lungs, compared to non-treated mice.
  • KC induced CXCL1
  • MIG CXCL9
  • IP-10 CXCL10
  • GM-CSF GM-CSF
  • mice were either sensitized intranasally with Derf 5IR or administrated PBS, and then either vaccinated with 10 6 BPZE1 or BPZE1P. or left unvaccinated.
  • the mice were challenged intranasally with Derf 5IR or PBS during 5 days.
  • the mice were anesthetized, and their resistance in the respiratory airway was measured by plethysmography as described above.
  • mice vaccinated with either BPZE1 or BPZE1P present significantly lower levels of airway resistance compared to those that were not vaccinated. Again, the airway resistance of the vaccinated mice was indistinguishable from that of the control group, which were not sensitized nor challenged with Derf 5IR.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Pain & Pain Management (AREA)
  • Wood Science & Technology (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Physiology (AREA)
  • Developmental Biology & Embryology (AREA)

Abstract

A method of reducing or preventing the development of airway inflammation in a subject includes the step of infecting the respiratory tract of a subject an amount of a composition including a pharmaceutically acceptable carrier and live attenuated pertactin-deficient Bordetella bacteria sufficient to colonize the respiratory tract of the subject. The step of infecting the subject with the live attenuated pertactin-deficient Bordetella bacteria results in reduction or prevention of the development of airway inflammation in the subject.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation application of U.S. patent application Ser. No. 16/863,972 filed on Apr. 30, 2020, which is a continuation application of U.S. patent application Ser. No. 15/472,436 filed on Mar. 29, 2017 (now U.S. Pat. No. 10,682,377), which claims the priority of U.S. provisional patent application Ser. No. 62/314,843 filed on Mar. 29, 2016.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 27, 2017, is named 7056-0074_SL.txt and is 2,134 bytes in size
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • Not applicable.
  • FIELD OF THE INVENTION
  • The invention relates generally to the fields of microbiology, immunology, allergy, and medicine. More particularly, the invention relates to live attenuated Bordetella pertussis strains deficient in pertactin, and their use as prophylactic and therapeutic agents in various disease settings.
  • BACKGROUND
  • Microbial organisms and their components have long been known to affect the immune systems of mammals. Infection with virulent bacteria and viruses can cause severe illness or death. Contributing toward this, purified components of bacteria and viruses can also cause pathology by inducing inflammatory responses or otherwise causing the immune system to behave in an undesirable manner. Despite this, vaccines including whole bacteria, viruses, or parts thereof have not only proven to be one of the most powerful tools that medicine has developed to prevent serious infections, but also can cause other beneficial effects. For example, in experimental models, a live attenuated pertussis vaccine candidate named BPZE1 (see WO2007104451 A1) was found to not only protect against virulent Bordetella pertussis, but also to exert potent anti-allergic and anti-asthma effects by dampening hyperimmune responses to allergens (see WO2013066272A1).
  • Developing safe and effective vaccines nonetheless remains challenging for several reasons. Among these, despite modern molecular biology techniques and significant advances in our understanding of microbiology and immunology, it remains quite difficult to produce a vaccine product that is sufficiently attenuated to not cause significant pathology, while at the same time sufficiently immunogenic to induce an effective and long-lasting immune response against the target pathogen. In the case of live attenuated whole-cell bacterial vaccines, arriving at an optimal level of attenuation is particularly troublesome because overattenuation by reducing the amount or activity of virulence factors can result in a vaccine that is poorly immunogenic and/or unable to survive or replicate in a subject for a sufficient time after administration to induce an immune response.
  • SUMMARY
  • Described herein is the development of pertactin-deficient Bordetella strains, and their use in inducing protective immune responses against pathologic Bordetella infection as well as in treating or preventing respiratory tract inflammation such as that observed in allergic asthma. Pertactin, an outer membrane protein of Bordetella bacteria, serves as a virulence factor by promoting adhesion to a variety of cells. In the experiments described below, it was discovered that a pertactin-deficient mutant of BPZE1, termed BPZE1P (deposited in accordance with the requirements of the Budapest Treaty with the Collection Nationale de Cultures de Microorganismes (“CNCM”), 25. Rue du Docteur Roux. Paris Cedex 15, 75724. France on Dec. 12, 2016 under accession number CNCM-I-5150), was able to colonize the respiratory tract, induce antibody responses against Bordetella, and protect against and treat allergic lung disease. The discovery was surprising because as others have shown that pertactin was required for Bordetella to resist neutrophil-mediated clearance, B. pertussis deficient in this virulence factor would have been expected to be cleared too rapidly to allow the induction of a protective immune responses. See, Inatsuka et al. Infect. Immun. 2010; 78: 2901-2909.
  • In the absence of anti-pertactin antibodies. BPZE1P colonized lungs as efficiently as BPZE1 and induced protective immunity against B. pertussis challenge as efficiently as BPZE1. In the presence of anti-pertactin antibodies, BPZE1P colonized the mouse lungs significantly better than BPZE1. Therefore, pertactin-deficient B. pertussis strains such as BPZE1P may be advantageous in protecting against respiratory tract inflammation in subjects with high pre-existing titers of pertactin antibodies, including those previously vaccinated with pertactin-containing acellular vaccines.
  • Accordingly described herein are methods of reducing or preventing the development of airway inflammation in a subject by administering to the respiratory tract of the subject an amount of a composition including a pharmaceutically acceptable carrier and live pertactin-deficient Bordetella bacteria sufficient to colonize the respiratory tract of the subject and thereby reduce or prevent the development of airway inflammation in the subject. In these methods, the airway inflammation can be associated with one or more of airway resistance, eosinophil infiltration in the lungs of the subject, and/or increased amounts of inflammatory cytokines in the lungs of the subject. Colonization of the respiratory tract of the subject can result in reduction or prevention of such airway resistance, eosinophil infiltration, and/or increased amounts of inflammatory cytokines.
  • Also described herein are compositions including a pharmaceutically acceptable carrier and live pertactin-deficient Bordetella bacteria capable of colonizing the respiratory tract of a subject and reducing or preventing the development of airway inflammation in the subject.
  • In the methods and compositions described herein, the live pertactin-deficient Bordetella bacteria can lack a functional gene encoding pertactin, also be deficient in tracheal cytotoxin (TCT), pertussis toxin (PTX), and/or dermonecrotic toxin (DNT). The live pertactin-deficient Bordetella bacteria can be BPZE1P. The airway inflammation can be caused by exposure to an allergen, and the subject can be one diagnosed with asthma, interstitial lung disease, or allergic rhinitis; one having greater than 10 ng per ml of anti-pertactin antibodies in its serum; or one that has previously been immunized with a vaccine containing pertactin or a pertactin-like antigen.
  • As used herein, “pertactin” is the outer surface membrane protein produced by Bordetella pertussis and its close relatives, such as Bordetella parapertussis that is involved in the binding of Bordetella bacteria to host cells as described in Leininger et al., Proc. Nat'l. Acad. Sci. USA, 1991, 88:345-9. Conserved regions in this protein, such as its passenger and autotransporter domains, contribute directly to the overall virulence and pathogenicity of these organisms.
  • As used herein, the abbreviation “PTX” refers to pertussis toxin, a major virulence factor of B. pertussis, which induces metabolic changes and alters immune responses in the host as described in Saukkonen et al., Proc. Natl. Acad. Sci. USA., 1992, 89:118-122.
  • As used herein the abbreviation “DNT” refers to pertussis dermonecrotic toxin (also called lethal toxin), a toxin found in B. pertussis which induces inflammation, vasoconstriction and dermonecrotic lesions in sites where B. pertussis colonize the respiratory tract. See Fukui-Miyazaki et al., BMC Microbiol. 2010, 10:247.
  • As used herein the abbreviation “TCT” refers to tracheal cytotoxin, a disaccharide tetrapeptide derivative of peptidoglycan synthesized by bordetellae, which induces the production of interleukin-1 and nitric oxide synthase, and causes stasis of cilia and lethal effects on respiratory epithelial cells. See Luker et al., Proc. Natl. Acad. Sci. USA., 1993, 90, 2365-2369
  • As used herein, a “pertactin-deficient” Bordetella strain is one that exhibits at least less than 50% (e.g., less than 50, 40, 30, 20, 10, 5, 4, 3, 2, or 1%) of the pertactin activity found in BPZE1 under the conditions described in the Examples section below, one that exhibits no detectable pertactin activity, or one that exhibits not detectable expression of pertactin as determined by Western blotting.
  • The term “functional” when referring to a toxin or virulence factor in a bacterial strain means that (i) the toxin/virulence factor expressed by the strain has not been mutated to eliminate or at least reduce by greater than 50% its enzymatic activity compared to the non-mutated version of the toxin/factor, and/or (ii) that a bacterial strain expressing the toxin/factor has not been engineered or selected to eliminate or at least reduce by greater than 50% the number of molecules of that toxin/factor compared to the starting strain from which the engineered or selected strain was derived.
  • The term “mammal”. “mammalian subject” or “subject” encompasses any of various warm-blooded vertebrate animals of the class Mammalia, including human beings.
  • Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention. All publications, patents, and patent applications mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions will control. In addition, the particular embodiments discussed below are illustrative only and not intended to be limiting.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a diagram showing the structure of a plasmid used in the construction of BPZE1P.
  • FIG. 1B is another diagram showing the construction of BPZE1P. and photographs of gels showing the results of PCR amplification of the prn UPg and prn Log fragments.
  • FIG. 1C is photographs of immunoblots showing the presence of pertactin in BPZE1, but not in BPZE1P lysates and supernatants.
  • FIG. 2 is a graph showing the results of lung colonization in Balb/c mice using either BPZE1 or BPZE1P.
  • FIG. 3 is a graph showing total IgG titers after BPZE1P or BPZE1 administration to mice.
  • FIG. 4A is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 106 viable B. pertussis bacteria of the BPSM strain.
  • FIG. 4B is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 106 viable B. pertussis bacteria of the BPSMP strain.
  • FIG. 4C is a graph showing BPZE1- and BPZE1P-mediated protection in Balb/c mice challenged intranasally with 106 viable B. pertussis bacteria of the B1917 strain.
  • FIG. 5 is a graph showing the fitness of BPZE1 and BPZE1P in mice preimmunized with an acellular B. pertussis vaccine (aPv).
  • FIG. 6 is a diagram of the experimental protocol of an assay for airway responsiveness in allergic mice vaccinated with BPZE1, BPZE1P or left unvaccinated, and a graph showing the results of the assay.
  • FIG. 7A is a graph showing the total airway cell population infiltration in the bronchoalveolar (BAL) fluid of the allergic mice of the experiments shown in FIG. 6.
  • FIG. 7B is a graph showing the percentage of eosinophils in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6.
  • FIG. 7C is a graph showing the percentage of neutrophils in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6.
  • FIG. 7D is a graph showing the percentage of lymphocytes in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6.
  • FIG. 7E is a graph showing the percentage of macrophages in the cells in the BAL fluid of the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8A is a graph showing the amount of IL-1α normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8B is a graph showing the amount of IL-1β normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8C is a graph showing the amount of IL-6 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8D is a graph showing the amount of IL-13 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8E is a graph showing the amount of CXCL1 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8F is a graph showing the amount of CXCL9 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8G is a graph showing the amount of CXCL10 normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 8H is a graph showing the amount of GM-CSF normalized against total proteins measured in the lung lobe in the allergic mice of the experiments shown in FIG. 6.
  • FIG. 9 is a diagram of the experimental protocol of an assay for airway responsiveness in a therapeutic model of allergic mice vaccinated with BPZE1, BPZE1P or left unvaccinated, and a graph showing the results of the assay.
  • DETAILED DESCRIPTION
  • Described herein are Bordetella strains deficient in pertactin and their use in stimulating anti-Bordetella immune responses as well as in preventing and treating respiratory tract inflammation. The below described embodiments illustrate representative examples of these methods. Nonetheless, from the description of these embodiments, other aspects of the invention can be made and/or practiced based on the description provided below.
  • General Methodology
  • Methods involving conventional microbiological, immunological, molecular biological, and medical techniques are described herein. Microbiological methods are described in Methods for General and Molecular Microbiology (3d Ed). Reddy et al., ed., ASM Press. Immunological methods are generally known in the at and described in methodology treatises such as Current Protocols in Immunology. Coligan et al., ed., John Wiley & Sons, New York. Techniques of molecular biology are described in detail in treatises such as Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3, Sambrook et al., ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2001; and Current Protocols in Molecular Biology, Ausubel et al., ed., Greene Publishing and Wiley-Interscience, New York. General methods of medical treatment are described in McPhee and Papadakis, Current Medical Diagnosis and Treatment 2010, 49th Edition, McGraw-Hill Medical, 2010; and Fauci et al., Harrison's Principles of Internal Medicine, 17th Edition, McGraw-Hill Professional, 2008.
  • Pertactin-Deficient Bordetella Strains
  • Bordetella species such as Bordetella pertussis, Bordetella parapertussis, and Bordetella bronchiseptica that are deficient in pertactin expression (e.g., those that express at least 50, 60, 70, 80, 90, 95, 96, 97, 98, or 99% less pertactin than do corresponding strains) can be used to generate immune responses against Bordetella species, as well as to treat and/or prevent respiratory tract inflammation such as that which occurs in allergic asthma. Live, attenuated, pertactin-deficient Bordetella pertussis and live, attenuated, pertactin-deficient Bordetella parapertussis are preferred for use for treating or preventing allergic respiratory tract inflammation in human subjects. The live attenuated pertactin-deficient Bordetella strains described herein can be made by adapting methods known in the art such as those described in the Examples section below. The starting strain can be any suitable Bordetella species. Examples of Bordetella species include B. pertussis, B. parapertussis, and B. bronchiseptica. B. pertussis is preferred for use as the starting strain for vaccines and methods for preventing pertussis infection. Several suitable Bordetella strains for use as starting strains are available from established culture collections (e.g., the American Type Culture Collection in Manassas, Va.) or can be isolated from natural reservoirs (e.g., a patient having pertussis) by known techniques (e.g., as described in Aoyama et al., Dev. Biol. Stand, 73:185-92, 1991).
  • Bordetella strains which express functional pertactin can be made deficient in this molecule or its activity by selection or, preferably for stability purposes, mutagenesis (e.g., deletion of the native prn gene as described below). Alternatively. Bordetella species deficient in pertactin can also be isolated from natural sources (e.g., human subjects or other mammals infected or colonized with such strains). Because insufficient attenuation of a pathogenic strain of Bordetella might cause a pathological infection in a subject, it is preferred that the pertactin-deficient Bordetella strain used also have lower levels of other functional virulence factors. On the other hand, to ensure that the pertactin-deficient Bordetella strain retains the ability to colonize a subject and exert a protective effect on respiratory tract inflammation, it must not be overly attenuated. Attenuation might be achieved by mutating the strain to reduce its expression of pertactin and one or more (e.g., 1, 2, 3, 4, 5 or more) of the following: pertussis toxin (PTX), dermonecrotic toxin (DNT), tracheal cytotoxin (TCT), adenylate cyclase (AC), lipopolysaccharide (LPS), filamentous hemagglutinin (FHA), or any of the bvg-regulated components. Attenuation might also be achieved by mutating the strain to reduce the biological activity of pertactin and one or more (e.g., 1, 2, 3, 4, 5 or more) of the following: pertussis toxin (PTX), dermonecrotic toxin (DNT), tracheal cytotoxin (TCT), adenylate cyclase (AC), lipopolysaccharide (LPS), filamentous hemagglutinin (FHA), or any of the bvg-regulated components. Examples of methods for making such mutants are described herein and in U.S. Pat. No. 9,119,804. In the experiments presented below, a Bordetella strain deficient in functional pertactin, functional PTX, functional DNT, and functional TCT was able to colonize the respiratory tract of subjects, induce immune responses against Bordetella, and reduce or prevent the development of allergic and inflammatory responses. Accordingly, Bordetella strains, such as BPZE1P, which are deficient in these four virulence factors and can colonize a subject and induce immune responses targeting Bordetella strains and/or reduce or prevent the development of allergic and inflammatory responses are preferred.
  • A variety of methods are known in the art for attenuating an infectious bacterial strain. These include passaging the strain in vitro until virulence is lost, non-specific chemical mutagenesis followed by screening and selection based on phenotype, and using targeted molecular biology techniques such as those described in the Examples section below (including allelic exchange) and in Methods for General and Molecular Microbiology (3d Ed), Reddy et al., ed., ASM Press. Using these methods, the genes encoding pertactin. PTX, and/or DNT can be deleted or mutated to an enzymatically inactive form (which is preferred where it is desired to retain the toxin's antigenicity). TCT production can be significantly (e.g., >than 99.99, 99.90, 99.8, 99.7, 99.6, 99.5, 99.0, 98, 97, 96, 95, or 90%) reduced by replacing the native ampG gene (unlike other species, B. pertussis ampG does not actively recycle TCT-containing peptidoglycan) with a heterologous (e.g., from E. coli or another gram-negative species) ampG gene, or by mutating the native ampG gene such that it is active at recycling peptidoglycan.
  • Modification of a starting strain to reduce or remove toxin/virulence factor activity can be confirmed by sequencing the genomic DNA or genes encoding the toxins of the modified strains. Southern. Northern, and/or Western blotting might also be used to confirm that the target genes have been deleted or that expression of the target factors has been reduced or removed. Biological activity can also be evaluated to confirm reduction or removal of toxin/virulence factor activity. Once the modifications have been confirmed, the modified strains can be tested for the ability to colonize a subject and to induce protective immunity against Bordetella infection or to reduce or prevent the development of allergic and inflammatory responses by known methods such as those described in the Examples section below.
  • Compositions for Modulating Immune Reponses
  • The live attenuated Bordetella strains described herein can be used in compositions that protect a mammalian subject from developing a Bordetella infection (e.g., pertussis), or to reduce the symptoms of such an infection. They can also be used to reduce or prevent the development of allergic and inflammatory responses in a subject such as asthma, allergic rhinitis, interstitial lung disease, food allergies, peanut allergy, venom allergies, atopic dermatitis, contact hypersensitivity, and anaphylaxis. For use in therapeutic or prophylactic compositions, the live attenuated Bordetella strains are typically formulated with a pharmaceutically acceptable excipient. Examples of pharmaceutically acceptable excipients include, e.g., buffered saline solutions, distilled water, emulsions such as an oil/water emulsion, various types of wetting agents, sterile solutions, and the like.
  • The vaccines can be packaged in unit dosage form for convenient administration to a subject. For example, a single dose of between 1×104 to 1×109 (e.g., 1×104. 5×104, 1×105, 5×105, 1×106, 5×106, 1×107, 5×107, 1×108, 5×108, or 1×109+/−10, 20, 30, 40, 50, 60, 70, 80, or 90%) live bacteria of the selected attenuated Bordetella strain and any excipient can be separately contained in packaging or in an administration device. The vaccine can be contained within an administration device such as a syringe, spraying device, or insufflator.
  • Formulations/Dosage/Administration
  • The compositions described herein can be administered to a mammalian subject (e.g., a human being, a human child or neonate, a human adult, a human being at high risk from developing complications from pertussis, a human being with lung disease, a human being that is or will become immunosuppressed, and a human being having or at high risk for developing respiratory tract inflammation such as asthma, allergic rhinitis, or interstitial lung disease) by any suitable method that deposits the bacteria within the composition in the respiratory tract or other mucosal compartment. For example, the compositions may be administered by inhalation or intranasal introduction, e.g., using an inhaler, a syringe, an insufflator, a spraying device, etc.
  • The pertactin-deficient Bordetella strains described herein can be formulated as compositions for administration to a subject. A suitable number of live bacteria are mixed with a pharmaceutically suitable excipient or carrier such as phosphate buffered saline solutions, distilled water, emulsions such as an oil/water emulsions, various types of wetting agents, sterile solutions and the like. In some cases, the vaccine can be lyophilized and then reconstituted prior to administration. The use of pharmaceutically suitable excipients or carriers which are compatible with mucosal (particularly nasal, bronchial, or lung) administration are preferred for colonizing the respiratory tract. See Remington's Pharmaceutical Sciences, a standard text in this field, and USP/NF.
  • When formulated for mucosal administration, each dose of a composition can include a sufficient number of live pertactin-deficient Bordetella bacteria to result in colonization of the mucosal site, e.g., approximately (i.e., +/−50%) 5×103 to 5×109 bacteria. For administration to human subjects, the dose can include approximately 1×106, 5×106, 1×107, 5×107, 1×108, 5×108, 1×109, 5×109, or 1×1010 live pertactin-deficient Bordetella bacteria. The dose may be given once or on multiple (2, 3, 4, 5, 6, 7, 8 or more) occasions at intervals of 1, 2, 3, 4, 5, or 6 days or 1, 2, 3, 4, 5, or 6 weeks, or 1, 2, 3, 4, 5, 6, or 12 months. Generally, sufficient amounts of a composition are administered to result in colonization and the protective and/or anti-inflammatory response. Additional amounts are administered after the induced protective and/or anti-inflammatory response wanes (e.g., after the subject resumes suffering from the symptoms of respiratory tract inflammation).
  • Subjects which can be administered a composition containing live pertactin-deficient Bordetella bacteria can include any capable of being colonized with a selected live pertactin-deficient Bordetella bacterial strain. For example, the subject can be a mammal such as a human being. Human subjects having, or at high risk of developing, respiratory tract inflammation such as those having or prone to developing allergic asthma or allergic rhinitis are preferred recipients of the composition. While the composition can be used in subjects regardless of their titers of anti-pertactin antibodies, the composition may be used in those having measurable titers (e.g., greater than 10, 20, 50, 100, 200, or 500 ng per ml of serum) of anti-pertactin antibodies and those having previously been immunized with a vaccine containing pertactin or a pertactin-like antigen because pertactin-deficient Bordetella bacterial strains are not subject to pertactin-targeting immune responses.
  • The effectiveness of the compositions in dampening respiratory tract inflammation can be assessed by known methods, e.g., measuring the number of inflammatory cells, IgE titers, levels of pro-inflammatory cytokines/chemokines (such as eotaxin, GM-CSF. IFNγ, IL-4, IL-5, IL-8, IL-10, IL-12, IL-13, IL-17A, IL-17F, IL-18, and TNFα) in fluid taken from the respiratory tract (e.g., bronchoalveolar lavage fluid), or clinical parameters such as spirometry or the level of dyspnea, coughing, wheezing, or respiratory capacity. Improvement in any of one or more of these parameters (at least 10, 20, 30, 40, 50, 60, 70, 80, 90% or more improved compared to a subject not receiving the composition) indicates that the composition is effective. Animal models of allergic respiratory tract inflammation can also be used to assess the effectiveness of a composition, see e.g., U.S. Pat. No. 8,986,709.
  • EXAMPLES Example 1—Construction and Characterization of a Pertactin-Deficient Strain of B. pertussis
  • Escherichia coli DH5α, SM10 and B. pertussis BPZE1, BPSM (Menozzi et al., Infect Immun 1994; 62:769-778) and B1917 (Bart et al. Genome Announc 2014; 2(6)) were used in this study. The Bordetella strains were cultured at 37° C. on Bordet-Gengou agar (BG), supplemented with 1% glycerol and 10% defibrinated sheep blood. After growth, the bacteria were harvested by scraping the plates and resuspended in phosphate-buffered saline (PBS) at the desired density. For liquid culture the Bordetella strains were grown at 37° C. in modified Stainer-Scholte medium (Imaizumi et al. Infect Immun 1983; 41:1138-1143) containing 1 g/l heptakis (2,6-di-o-methyl) β-cyclodextrin (Sigma). E. coli strains used for cloning procedures were growth in LB broth or LB agar plates. When required, streptomycin (Sm) was used at 100 μg/ml, gentamycin (Gm) at 10 μg/ml and ampicillin (Amp) at 100 μg/ml.
  • To delete prn, the gene coding for pertactin, in BPZE1, a 739-bp fragment downstream (prn LO) of the prn gene and a 759-bp fragment upstream (prn UP) of the prn gene were cloned into pSS4940 to introduce the prn deletion in the BPZE1 and BPSM genomes by homologous recombination. Referring to FIGS. 1A and 1B, the upstream and downstream prn flanking region were PCR amplified using prn_KO_fw (ATCCTCAAGCAAGACTGCGAGCTG) (SEQ ID NO:1)) and OL_prn_KO_rv (GGGGATAGACCCTCCTCGCTTGGATGCCAGGTGGAGAGCA) (SEQ ID NO:2)), and OL_prn_KO_fw (TGCTCTCCACCTGGCATCCAAGCGAGGAGGGTCTATCCCC) (SEQ ID NO:3)) and prn_KO_rv (CCATCATCCTGTACGACCGCCT) (SEQ ID NO:4)), respectively, as primers. These fragments then served as template for a PCR elongation using prn_KO_fw and prn_KO_rv as primers. The resulting fragment (containing the prn deletion) was inserted into the TOPO Blunt® vector (ThermoFisher Scientific) and then excised as a KpnI-NotI fragment. The excised KpnI-NotI fragment was inserted into KpnI- and NotI-digested pSS4940, a pSS4245 (Inatsuka et al., infect immun 2010; 78 2901-2909) derivative. The resulting plasmid was transformed into E. coli SM10, which was then conjugated with BPZE1. Following two successive homologous recombination events, as described elsewhere (Mielcarek et al., PLoS Pathog 2006; 2:e65), referring to FIG. 1B. PCR was used to confirm deletion of the entire prin gene by amplifying the flanking regions covering the construction using prnKO_UP (TTCTGCGCGAACAGATCAAAC) (SEQ ID NO:5))—prnKOin_UPrv (CTGCTGGTCATCGGCGAAGT) (SEQ ID NO:6)) for the 5′ region and prnKOin_LOfw (CGCCCATTCTCCCTGTTCC) (SEQ ID NO:7))—prnKO_LO (GAACAGGAACTGGAACAGGCG) (SEQ ID NO:8)) for the 3′ region. A strain carrying the prn deletion was selected and named BPZE1P. The same strategy was used to construct a pertactin-deficient BPSM mutant, named BPSMP.
  • The presence of pertactin was tested by immunoblotting of BPZE1 and BPZE1P lysates and supernatants, using purified Prn (List Biological laboratories) as control for correct size of the band. For protein extraction, BPZE1 and BPZE1P strains were plated onto BG blood agar and incubated for 48 h at 37° C. After growth, the bacteria were scraped off the plates, resuspended in 10 ml of Stainer-Scholte medium and grown for 4 days at 37° C. The bacteria were then harvested by centrifugation. The supernatants were recovered and treated with trichloroacetic acid (TCA) as described previously (Solans et al., PLoS Pathog 2014; 10:e1004183) for protein concentration. The bacterial pellets were resuspended in PBS complemented with an EDTA-free protease inhibitor cocktail (Roche) and lysed using a French pressure cell. Bacterial debris were removed by centrifugation for 30 minutes at 15.000×g, and the supernatants were recovered for immunoblotting. Proteins were separated by 12% SDS-PAGE and then transferred onto a Nitrocellulose membrane using the Criterion rM cell system (Bio-Rad). After blocking with 5% w/v skim milk powder in PBS 0.01% TWEEN® 20 for 30 min, the membrane was incubated with an anti-pertactin monoclonal antibody pertactin at 1:1,000 dilution. Goat-anti mouse-HRP (Abcam) was then added at a 1:10,000 dilution, and the blot was developed using chemiluminescent substrates (GE Healthcare). As shown in FIG. 1C, an anti-pertactin antibody reactive protein co-migrating with purified pertactin was detected in the supernatant of BPZE1, but not in the supernatant of BPZE1P. This protein was not detected in the bacterial cell lysate of either BPZE1 or BPZE1P.
  • Example 2—BPZE1P Colonizes Mice as Well as BPZE1
  • Groups of 18 six-week old mice were inoculated intranasally with 20 μl PBS containing 106 viable bacteria as described previously (Mielcarek et al., supra). At the indicated time points (3 hours, 3 days, 7 days, 14 days, 21 days and 28 days), 3 mice per group were sacrificed, and lungs were harvested and homogenized for measuring total number of colony formation units (CFU). Statistical analysis was done by a 2-way ANOVA test, using post hoc comparison Bonferroni test with confidence intervals of 95%. Referring to FIG. 2, both BPZE1 and BPZE1P colonized the animals equally well. Both strains exhibited a peak of multiplication 3 days post vaccination and colonization persisted for 4 weeks. No statistically significant difference was observed between these strains in their ability to colonize the mouse lungs.
  • Example 3—BPZE1P is as Immunogenic and Protective Against Challenge with Virulent B. pertussis as BPZE1
  • Immunity induced by BPZE1P in comparison with BPZE1 was measured by antibody titration of mouse immune serum after nasal vaccination. Groups of 8 mice were vaccinated intranasally with 105 viable BPZE1 or BPZE1P. Four weeks later, the mice were bled, and total IgG titers were measured against total BPSM lysate. Blood was centrifuged for 5 min. at 5,000×g to separate the serum from the cells. Antibody titers against B. pertussis were estimated using enzyme-linked immunosorbent assays (ELISA) as described previously (Mielcarek et al., supra), using total B. pertussis BPSM lysate at 1 μg of total protein per well. Statistical analysis was performed using GRAPHPAD PRISM® Prism software. As shown in FIG. 3, BPZE1- and BPZE1P-vaccinated mice exhibited much higher antibody titers than did naïve control mice. No significant difference in antibody titers between BPZE1- and BPZE1P-vaccinated mice was detected.
  • The protective efficacy of BPZE1P compared with BPZE1 was tested in a suboptimal protection protocol by measuring the CFU counts in the lungs 7 days post challenge, and comparing a naïve group with the vaccinated groups. Groups of 8 six-week old mice were vaccinated intranasally with 20 μl PBS containing 105 viable BPZE1 or BPZE1P. or were left unvaccinated. Four weeks later, all mice were challenged intranasally with 20 μl PBS containing 106 viable BPSM. BPSMP or B1917. Three hours after the challenge 3 mice per group were sacrificed, and lungs were harvested and homogenized for CFU counting. The remaining 5 mice per group were sacrificed 7 days after challenge for CFU counting. Three hours post infection, 3 mice were euthanized, and their lungs were harvested to determine the CFU counts shortly after challenge. Seven days post-infection, the remaining 5 mice were euthanized, their lungs were harvested, and the CFU counts were measured. Statistical analysis was done applying a parametric 2-way ANOVA test, using post hoc Bonferroni comparison test with a confidence interval of 95%, *, p<0.005; **, p<0.001; ***, p<0.0001. As shown in FIGS. 4A-C, vaccination with either strain protected against challenge with BPSM, BPSMP and B1917 equally well. These results show that the deletion of prn does not impact on the protective capacity of the live attenuated vaccine—either against the laboratory strain BPSM, its pertactin-deficient derivative BPSMP, or clinical isolate B1917.
  • Example 4—BPZE1P Increases Pulmonary Vaccine Uptake in aPv-Vaccinated Mice
  • The ability of BPZE1P to colonize the lungs of mice having pre-existing antibodies against pertactin was investigated. Groups of 8 six-week old mice were vaccinated subcutaneously with ⅕ of the human dose of the acellular pertussis vaccine (aPv; INFANRIX®, GSK; containing inactivated pertussis toxin, filamentous hemagglutinin and pertactin). Four weeks later, the mice were boosted with the same dose of aPv. Four weeks after boosting, the mice were infected intranasally with 106 BPZE1 or BPZE1P. Three hours post infection, 3 mice were euthanized, and the lungs were harvested to determine the CFU counts. Seven days post-infection, the remaining 5 mice were euthanized, the lungs harvested, and the CFU counts were measured. Statistical analysis was done applying a parametric 2-way ANOVA test, using post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p<0.0001. Referring to FIG. 5, at 3 hours post-administration, no significant difference in colonization was seen between the two strains. In contrast, seven days after inoculation. BPZE1P colonized the lungs significantly better than BPZE1. Mice infected with BPZE1P had almost 104 CFU in the lungs 7 days after administration, while the CFU counts in the lungs of the mice given BPZE1 reached 102 CFU. These data show that the deletion of the prn gene improves BPZE1 pulmonary take in mice pre-immunized with pertactin-containing aP vaccine.
  • Example 5—BPZE1P and BPZE1 Protect Equally Well Against Allergic Airway Inflammation
  • The effect of vaccination with BPZE1P on airway responsiveness was investigated in allergic mice as described in the protocol shown in FIG. 6. Groups of 4 weeks-old mice were vaccinated intranasally with 20 μl PBS containing 106 viable BPZE1 or BPZE1P, or were left unvaccinated. Four weeks later, the mice were sensitized intranasally with 20 μl of house dust mite (HDM; Stallergenes S. A.) of 5 index of reactivity (IR) of Dermnaophagoidesfarinae extract (Derf 5IR) or 20 μl of PBS as control. Ten days later, the mice where challenged intranasally with 20 μl of Derf 5IR or PBS daily for 5 days, and two days later, the mice were anesthetized and intubated intratracheally for mechanical ventilation using the FLEXIVENT® (SCIREQ®) device. The mice were then exposed to increasing concentrations of nebulized methacholine (0-50 mg/mL in PBS) (Sigma-Aldrich) to determine the resistance in their respiratory airways using plethysmography. Statistical analysis was done by applying a parametric 2-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p<0.0001; **, p<0.001; *, p<0.005. In FIG. 6, comparisons between Derf 51R and BPZE1+Derf 5IR are represented the solid line, and comparisons between Derf 5IR and BPZE1P+Derf 5IR are represented the dashed line. Both the BPZE1- and the BPZE1P-vaccinated mice presented significantly less resistance in their airways after treatment with methacholine at 6, 12 and 25 mg/ml compared to the non-vaccinated mice. The resistance of the vaccinated mice was comparable to that of the PBS control group, which was not sensitized, nor challenged throughout the entire experiment.
  • Example 6—Measurement of Lung Cell Infiltration and Cytokine Profiles
  • Airway cell population infiltration in the allergic mice of the experiment shown in FIG. 6 and discussed immediately above was assessed. After plethysmography measurements, Bronchoalveolar lavage (BAL) fluids were collected to measure the cell infiltration in the airway. Cells from the BAL fluids were harvested by centrifugation at 1,200 rpm for 5 min at 4° C., resuspended in PBS for cell counting using the Shandon cytospin 4 (Thermo Fisher Scientific) and stained with May Griinwald Giemsa (DiffQuik@) for cell type counting. Total cell numbers were measured in the BAL fluid of mice (FIG. 7A), and percentages of eosinophils (FIG. 7B), neutrophils (FIG. 7C), lymphocytes (FIG. 7D) and macrophages (FIG. 7E) were calculated. Statistical analysis was done by applying a parametric one-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p<0.0001; *,p<0.005. BPZE1 or BPZE1P vaccination significantly reduced the recruitment of total cells in the airways after allergen exposure and challenge, compared to the non-vaccinated mice (FIG. 7A). This reduction reflected essentially the reduction in eosinophil recruitment in the vaccinated mice (FIG. 7B), whereas there was no significant change in the percentages of neutrophils or lymphocytes (FIGS. 7C and D) between the vaccinated and non-vaccinated mice. A small, but significant increase in the percentage of macrophages was observed in the mice that were vaccinated with BPZE1P (FIG. 7E).
  • Following BAL, the right lung lobes were harvested and directly frozen in liquid Nitrogen for protein extraction. The lung lobes were resuspended in 1 mL of lysis buffer, PBS with 0.5% nonidet P40 and protease inhibitor cocktail (Roche @), and homogenized at 4° C. using T-18 ULTRA-TURRAX@ (IKA®). The samples were centrifuged, and the supernatants were collected for total protein quantification using the PIERCE™ BCA protein assay (Thermo Fisher Scientific), and for cytokine and chemokine measurements using the Cytokine 20-Plex Mouse Panel (INVITROGEN™, Thermo Fisher Scientific) per the manufacturer's specifications. Referring to FIG. 8, cytokine levels are represented as the normalization of the cytokine/chemokine quantification against total proteins measured in the lung lobe. Statistical analysis was done by applying a parametric one-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, **, p<0.001, *; p<0.005.
  • As shown in FIG. 8, HDM (Derf 5IR)-treated mice produced significantly increased levels of IL1α and IL1β in the lungs, compared to non-treated mice. Vaccination with BPZE1 or BPZE1P prior to HDM sensitization significantly decreased these levels (FIGS. 8A and B). A similar trend was observed for IL6 and IL13 although the differences between the vaccinated and the non-vaccinated mice did not reach statistical significance (FIGS. 8C and D). Significantly lower levels of induced CXCL1 (KC), CXCL9 (MIG), CXCL10 (IP-10) and GM-CSF were observed in the vaccinated mice compared to the mice which were only treated with HDM (FIG. 8F-H). Generally, there was no statistical difference between the BPZE1-vaccinated and the BPZE1P-vaccinated mice.
  • Example 7
  • Vaccination of pre-sensitized subjects with either BPZE1 or BPZE1P present significantly lower levels of airway resistance compared to those that were not vaccinated. As shown in the diagram of FIG. 9, groups of 5-week-old mice were either sensitized intranasally with Derf 5IR or administrated PBS, and then either vaccinated with 106 BPZE1 or BPZE1P. or left unvaccinated. Nine days later, the mice were challenged intranasally with Derf 5IR or PBS during 5 days. Two days after last challenge, the mice were anesthetized, and their resistance in the respiratory airway was measured by plethysmography as described above. Statistical analysis was done by applying a parametric 2-way ANOVA test, using the post hoc Bonferroni comparison test with a confidence interval of 95%, ***, p<0.0001, **; p<0.001. Comparisons between Derf 5IR and BPZE1+Derf 5IR are represented by the solid line, and comparisons between Derf 5IR and BPZE1P+Derf 5IR are represented by the dashed line. The mice vaccinated with either BPZE1 or BPZE1P present significantly lower levels of airway resistance compared to those that were not vaccinated. Again, the airway resistance of the vaccinated mice was indistinguishable from that of the control group, which were not sensitized nor challenged with Derf 5IR.
  • OTHER EMBODIMENTS
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (5)

What is claimed is:
1. A composition comprising a pharmaceutically acceptable carrier and live attenuated pertactin-deficient Bordetella bacteria able to colonize the respiratory tract of a subject, wherein the live attenuated pertactin-deficient Bordetella bacteria are deficient in at least one virulence factor selected from the group consisting of tracheal cytotoxin (TCT), pertussis toxin (PTX), and dermonecrotic toxin (DNT).
2. The composition of claim 1, wherein the live attenuated pertactin-deficient Bordetella bacteria lack a functional gene encoding pertactin.
3. The composition of claim 1, wherein the live attenuated pertactin-deficient Bordetella bacteria are deficient in at least two virulence factors selected from the group consisting of tracheal cytotoxin (TCT), pertussis toxin (PTX), and dermonecrotic toxin (DNT).
4. The composition of claim 1, wherein the live attenuated pertactin-deficient Bordetella bacteria are deficient in tracheal cytotoxin (TCT), pertussis toxin (PTX), and dermonecrotic toxin (DNT).
5. A method of reducing or preventing the development of allergic airway inflammation in a subject having greater than 10 nanogram per ml of serum antibodies to Bordetella pertussis pertactin, the method comprising the step of administering to the respiratory tract of the subject an amount of a composition comprising a pharmaceutically acceptable carrier and live attenuated pertactin-deficient Bordetella bacteria able to colonize the respiratory tract of a subject, wherein the live attenuated pertactin-deficient Bordetella bacteria are deficient in at least one virulence factor selected from the group consisting of tracheal cytotoxin (TCT), pertussis toxin (PTX), and dermonecrotic toxin (DNT).
US17/337,367 2016-03-29 2021-06-02 Mutant Bordetella Strains and Methods of Use Abandoned US20210290667A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/337,367 US20210290667A1 (en) 2016-03-29 2021-06-02 Mutant Bordetella Strains and Methods of Use
US18/338,314 US20230381224A1 (en) 2016-03-29 2023-06-20 Mutant Bordetella Strains and Methods of Use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662314843P 2016-03-29 2016-03-29
US15/472,436 US10682377B2 (en) 2016-03-29 2017-03-29 Mutant Bordetella strains and methods of use
US16/863,972 US11065276B2 (en) 2016-03-29 2020-04-30 Mutant Bordetella strains and methods of use
US17/337,367 US20210290667A1 (en) 2016-03-29 2021-06-02 Mutant Bordetella Strains and Methods of Use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/863,972 Continuation US11065276B2 (en) 2016-03-29 2020-04-30 Mutant Bordetella strains and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/338,314 Division US20230381224A1 (en) 2016-03-29 2023-06-20 Mutant Bordetella Strains and Methods of Use

Publications (1)

Publication Number Publication Date
US20210290667A1 true US20210290667A1 (en) 2021-09-23

Family

ID=58488973

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/472,436 Active 2037-07-15 US10682377B2 (en) 2016-03-29 2017-03-29 Mutant Bordetella strains and methods of use
US16/863,972 Active US11065276B2 (en) 2016-03-29 2020-04-30 Mutant Bordetella strains and methods of use
US17/337,367 Abandoned US20210290667A1 (en) 2016-03-29 2021-06-02 Mutant Bordetella Strains and Methods of Use
US18/338,314 Pending US20230381224A1 (en) 2016-03-29 2023-06-20 Mutant Bordetella Strains and Methods of Use

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/472,436 Active 2037-07-15 US10682377B2 (en) 2016-03-29 2017-03-29 Mutant Bordetella strains and methods of use
US16/863,972 Active US11065276B2 (en) 2016-03-29 2020-04-30 Mutant Bordetella strains and methods of use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/338,314 Pending US20230381224A1 (en) 2016-03-29 2023-06-20 Mutant Bordetella Strains and Methods of Use

Country Status (8)

Country Link
US (4) US10682377B2 (en)
EP (1) EP3436034A1 (en)
JP (1) JP6993984B2 (en)
CN (2) CN109069535B (en)
AU (2) AU2017242541B2 (en)
CA (1) CA3018126A1 (en)
SG (1) SG11201807783UA (en)
WO (1) WO2017167834A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11285201B2 (en) 2006-03-10 2022-03-29 Institut Pasteur De Lille Attenuated Bordetella strains
US11446372B2 (en) * 2017-10-18 2022-09-20 Institut Pasteur De Lille Bordetella strains expressing serotype 3 fimbriae
US11819545B2 (en) 2009-04-28 2023-11-21 Institut National De La Santé Et De La Recherche Médicale (Inserm) Vaccine for prophylaxis or treatment of an allergen-driven airway pathology

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6993984B2 (en) * 2016-03-29 2022-02-03 アンスティチュ パスツール ドゥ リール Mutant Bordetella strain and usage
CA3195783A1 (en) 2020-11-17 2022-05-27 Keith Rubin Treatment and prevention of neuropathology associated with neurodegenerative diseases
WO2024018061A1 (en) 2022-07-22 2024-01-25 Institut National de la Santé et de la Recherche Médicale Use of bordetella strains for the treatment of chronic obstructive pulmonary disease

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1184459A2 (en) * 1991-07-16 2002-03-06 Aventis Pasteur Limited Manipulation of gene copy number in Bordetella
US11065276B2 (en) * 2016-03-29 2021-07-20 Institut Pasteur De Lille Mutant Bordetella strains and methods of use

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6713072B1 (en) 1987-11-02 2004-03-30 Chiron S.R.L. Immunologically active polypeptides with altered toxicity useful for the preparation of an antipertussis vaccine
FR2718750B1 (en) 1994-04-19 1996-06-14 Pasteur Institut Recombinant proteins of the filamentous hemagglutinin of Bordetella, in particular, B. Pertussis, production and application to the production of foreign proteins or vaccinating active principles.
ES2136290T3 (en) * 1994-04-28 1999-11-16 Takeda Chemical Industries Ltd METHOD FOR SEPARATING PROTECTIVE COMPONENTS OF BORDETELLA PERTUSSIS.
FR2754543B1 (en) * 1996-10-11 1998-12-31 Pasteur Institut BORDETELLA STRAIN DEFICIENT IN TOXIN PRODUCTION AND EXPRESSING HYDRID PROTEIN, LIPOSOMES COMPRISING FHA AND THEIR USES AS VACCINES, AND THE USE OF FHA TO STIMULATE IMMUNE RESPONSES
FR2840319B1 (en) 2002-05-30 2004-08-20 Pasteur Institut BORDETELLA STRAINS MADE DEFICIENT BY GENETIC ATTENUATION
JP2007526208A (en) 2003-04-11 2007-09-13 メディミューン,インコーポレーテッド Methods for treating or preventing respiratory symptoms
US9119804B2 (en) 2006-03-10 2015-09-01 Institut Pasteur De Lille Live attenuated bordetella strains as a single dose vaccine against whooping cough
DK2069503T3 (en) 2006-11-15 2016-02-15 Folia Biotech Inc PAPAYAMOSAIKVIRUS-BASED VACCINES AGAINST INFLUENZA
EP2134359A4 (en) 2007-02-23 2010-06-16 Penn State Res Found Use of an avirulent bordetella mutant as a live vaccine vector
JP2010530423A (en) 2007-06-18 2010-09-09 へミスフェリックス・バイオファーマ,インコーポレーテッド Early intervention of viral infection using immune activators
AU2010243708C1 (en) * 2009-04-28 2014-10-02 Inserm (Institut National De La Sante Et De La Recherche Medicale) Vaccine for prophylaxis or treatment of an allergen-driven airway pathology
EP2944320A1 (en) 2009-06-15 2015-11-18 National University of Singapore Influenza vaccine, composition, and methods of use
SG11201401904YA (en) * 2011-11-02 2014-05-29 Univ Singapore Effect of an attenuated bordetella strain against allergic disease
CN102793915B (en) * 2012-07-25 2013-10-23 天津康希诺生物技术有限公司 Production method of acellular pertussis vaccine
EP2722338A1 (en) 2012-10-17 2014-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Novel recombinant Bordetella strains

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1184459A2 (en) * 1991-07-16 2002-03-06 Aventis Pasteur Limited Manipulation of gene copy number in Bordetella
US11065276B2 (en) * 2016-03-29 2021-07-20 Institut Pasteur De Lille Mutant Bordetella strains and methods of use

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Locht C International Microbiology: the official journal of the Spanish Society for Microbiology 2: 137-144, 1999 *
Srigley et al. Clin. Microbiol, Newsletter 37: 61-65, 2015 *
Williams et al. Emerg. Infect. Dis. 22: 319-322, February 2016 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11285201B2 (en) 2006-03-10 2022-03-29 Institut Pasteur De Lille Attenuated Bordetella strains
US11819545B2 (en) 2009-04-28 2023-11-21 Institut National De La Santé Et De La Recherche Médicale (Inserm) Vaccine for prophylaxis or treatment of an allergen-driven airway pathology
US11446372B2 (en) * 2017-10-18 2022-09-20 Institut Pasteur De Lille Bordetella strains expressing serotype 3 fimbriae

Also Published As

Publication number Publication date
US10682377B2 (en) 2020-06-16
CA3018126A1 (en) 2017-10-05
AU2017242541A1 (en) 2018-10-11
AU2022221438A1 (en) 2022-10-06
CN109069535B (en) 2022-03-18
WO2017167834A1 (en) 2017-10-05
EP3436034A1 (en) 2019-02-06
US20170283890A1 (en) 2017-10-05
US11065276B2 (en) 2021-07-20
SG11201807783UA (en) 2018-10-30
US20200323901A1 (en) 2020-10-15
JP2019513722A (en) 2019-05-30
US20230381224A1 (en) 2023-11-30
AU2017242541B2 (en) 2022-06-30
CN114767718A (en) 2022-07-22
JP6993984B2 (en) 2022-02-03
CN109069535A (en) 2018-12-21

Similar Documents

Publication Publication Date Title
US11065276B2 (en) Mutant Bordetella strains and methods of use
Solans et al. IL-17-dependent SIgA-mediated protection against nasal Bordetella pertussis infection by live attenuated BPZE1 vaccine
Fennelly et al. Bordetella pertussis expresses a functional type III secretion system that subverts protective innate and adaptive immune responses
KR101225304B1 (en) Mycoplasma hyopneumoniae avirulent-adjuvanted live vaccine
JP2019513722A5 (en)
US11446372B2 (en) Bordetella strains expressing serotype 3 fimbriae
US20080254062A1 (en) Use of an avirulent bordetella mutant as a live vaccine vector
WO2010132932A1 (en) A temperature sensitive vaccine strain of mycoplasma hyopneumoniae and uses thereof
Sukumar et al. Active and passive immunizations with Bordetella colonization factor A protect mice against respiratory challenge with Bordetella bronchiseptica
WO2020049133A1 (en) Adenylate cyclase catalytic domain deficient bordetella strains
Mann et al. Use of a genetically defined double mutant strain of Bordetella bronchiseptica lacking adenylate cyclase and type III secretion as a live vaccine
Hur et al. Immunologic study and optimization of Salmonella delivery strains expressing adhesin and toxin antigens for protection against progressive atrophic rhinitis in a murine model
Zhang et al. Two Bordetella bronchiseptica attenuated vaccine candidates confer protection against lethal challenge with B. Bronchiseptica and Pasteurella multocida toxin in mouse models
Rajeev et al. Expression of a truncated Pasteurella multocida toxin antigen in Bordetella bronchiseptica
Oyant et al. Role of Bordetella bronchiseptica adenylate cyclase in nasal colonization and in development of local and systemic immune responses in piglets
Kawai et al. A causal relationship between Bordetella pertussis and Bordetella parapertussis infections
Byeon et al. Generation of an attenuated Salmonella-delivery strains expressing adhesin and toxin antigens for progressive atrophic rhinitis, and evaluation of its immune responses in a murine model
CN116355827A (en) Recombinant bacillus calmette-guerin medicine for treating tuberculosis and application thereof
Verma et al. Comparative Study of the Adjuvanticity of FIA and Phosphatidylcholine Liposomes for Purified Mycobacterial RNA
Goel Interleukin-6 Contributes To Immunity Against Bordetella Pertussis.
Pilione et al. Use of a Genetically Defined Double Mutant

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION