US20170065700A1 - Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide - Google Patents

Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide Download PDF

Info

Publication number
US20170065700A1
US20170065700A1 US15/308,899 US201515308899A US2017065700A1 US 20170065700 A1 US20170065700 A1 US 20170065700A1 US 201515308899 A US201515308899 A US 201515308899A US 2017065700 A1 US2017065700 A1 US 2017065700A1
Authority
US
United States
Prior art keywords
seq
lps
baumannii
vaccine composition
lipopolysaccharide
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
US15/308,899
Other languages
English (en)
Inventor
Michael James McConnell
Meritxell García Quintanilla
Marina Pulido Toledano
María Pilar Pérez Romero
Jerónimo PACHÓN DÍAZ
Juan José INFANTE VIÑOLO
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.)
Fundacion Publica Andaluza Para La Gestion de la Investigacion En Salud De Sevilla
Vaxdyn SL
Servicio Andaluz de Salud
Original Assignee
Fundacion Publica Andaluza Para La Gestion de la Investigacion En Salud De Sevilla
Vaxdyn SL
Servicio Andaluz de Salud
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 Fundacion Publica Andaluza Para La Gestion de la Investigacion En Salud De Sevilla, Vaxdyn SL, Servicio Andaluz de Salud filed Critical Fundacion Publica Andaluza Para La Gestion de la Investigacion En Salud De Sevilla
Publication of US20170065700A1 publication Critical patent/US20170065700A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/104Pseudomonadales, e.g. Pseudomonas
    • 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
    • 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
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • C07K14/212Moraxellaceae, e.g. Acinetobacter, Moraxella, Oligella, Psychrobacter
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • C07K16/1217Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Neisseriaceae (F)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • A61K2039/521Bacterial cells; Fungal cells; Protozoal cells inactivated (killed)
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention refers, in general, to the field of pharmacology and immunology, and, in particular, to a vaccine composition suitable for the prophylactic treatment of an infection caused by an Acinetobacter baumannii strain in a mammal.
  • Acinetobacter baumannii is a Gram-negative coccabacillus with clinical importance in the hospital environment. This organism is highly distributed in soil and environmental water sources (Baumann, P. 1968 . J. Bacteriol. 96, 39-42), and can cause different types of infections as a nosocomial pathogen such as pneumonia, bacteremia, meningitis and skin and soft tissue infection, among others (Garcia-Quintanilla et al., 2013 . Curr. Pharm. Biotechnol . In press). This pathogen typically infects patients receiving mechanical ventilation and patients sustaining burn injury (Mu ⁇ oz-Price y Weinstein. 2008 . N. Engl. J. Med. 358, 1271-1281).
  • the experimental vaccines that have been described for A. baumannii can be classified into two broad groups, vaccines consisting of a single purified antigen, and multicomponent vaccines.
  • the outer membrane protein OmpA Lio et al., 2012 . PLoS One 7, e29446
  • the biofilm associated protein Bap (Fattahian et al., 2011 . Microb. Pathog. 51, 402-406)
  • the membrane transporter Ata (Bentancor et al., 2012 . Infect. Immun. 80, 3381-3388)
  • the surface polysaccharide poly-N-acetyl- ⁇ -(1-6)-glucosamine Boentancor et al., 2012 .
  • Infect Immun 80, 651-656 have been described as good candidates due to their capacity to elicit a specific immune response. However, the experiments testing survival after active immunization have only demonstrated that OmpA provides partial protection, and that the expression of Bap has not been clearly demonstrated in strains that do no form biofilm.
  • the approaches that employ multicomponent vaccines include outer membrane complexes (McConnell et al., 2011 . Infect. Immun. 79, 518-526), outer membrane vesicles (outer membrane vesicles; McConnell et al., 2011 . Vaccine 29, 5705-5710) and inactivated whole cells (McConnell y Pachón, 2010 . Vaccine 29: 1-5).
  • LPS lipopolysaccharide
  • Outer membrane vesicles are vesicles derived from the bacterial outer membrane that are secreted from numerous Gram-negative bacteria (Kulp et al., 2010 . Annu. Rev. Microbiol. 64, 163-184). OMVs are spherical vesicles of approximately 20-200 nm that are composed of outer membrane proteins, periplasmic proteins and LPS (Kuehn et al., 2005 . Genes Dev. 19, 2645-2655; Mashburn et al., 2005 . Nature. 437, 422-425).
  • OMVs have been shown to participate in the detection of quorum sensing, the transport of virulence factors and the transfer of genes, indicating that they play a role in bacterial patogenesis. It has also been demonstrated that OMVs can deliver proteins to the interior of host cells through fusion with lipid rafts, suggesting that OMVs can be used to transport bacterial products over large distances (Kesty et al., 2004 . Embo J. 23, 4538-4549). A recent study by Kwon et al demonstrated that a clinical isolate of A. baumannii secreted OMVs during growth in vitro (Kwon et al., 2009 . FEMS Microbiol. Lett. 297, 150-156). A proteomic analysis of the OMVs demonstrated that they contain multiple virulence factors and immunomodulating proteins, suggesting that OMVs play an important role in the pathogenesis of A. baumannii.
  • Vaccines based on OMVs have been developed for various Gram-negative bacteria including Neisseria meningitidis, Helicobacter pylori , and Vibrio cholerae (Bjune et al., 1991 . NIPH Ann. 14, 125-130; Keenan et al., 2003 . FEMS Immunol. Med. Microbiol. 36, 199-205; Bishop et al., 2012 . J. Infect. Dis. 205, 412-21). Immunization with OMVs has been shown to induce antibodies against multiple bacterial antigens, and the capacity to provide protective immunity in animal models of infection. In addition, the OMVs isolated form N.
  • meningitids serogroup B have been shown to be safe and immunogenic in humans, and have been used to control an outbreak of meningococcus meningitis in New Zealand (Nokleby et al., 2007 . Vaccine. 25, 3080-84).
  • This aspect of the invention refers to a pharmaceutical composition, preferably a vaccine composition, suitable for the prophylactic treatment (before infection) of an infection caused by an Acinetobacter baumannii strain in a mammal, which comprises:
  • the Acinetobacter baumannii strain deficient in lipopolysaccharide is characterized by the partial or complete inactivation of the genes selected from the list consisting of IpxA, IpxB and/or IpxC.
  • the pharmaceutical composition preferably the vaccine composition, further comprises a, preferably recombinant, polypeptide selected from the list consisting of:
  • the pharmaceutical composition preferably the vaccine composition, further comprises a purified outer membrane protein sequence of A. baumannii selected from the list consisting of: SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46; SEQ ID NO: 47; SEQ ID NO: 48; SEQ ID NO: 49; SEQ ID NO: 50; SEQ ID NO: 51; SEQ ID NO: 52; SEQ ID NO: 53; SEQ ID NO: 54; SEQ ID NO: 55; SEQ ID NO: 56; SEQ ID NO: 57;
  • the pharmaceutical composition preferably the vaccine composition, further comprises fusion recombinant polypeptides sequences SEQ ID NO 24 and/or 25 and/or the amino acid sequence coded by nucleotide sequence SEQ ID NO 26.
  • the Acinetobacter baumannii strain deficient in lipopolysaccharide comprises or is transformed, transduced or transfected with a nucleotide sequence capable of coding for any of the amino acid sequences as defined in any of the precedent paragraphs so that such strain is capable of producing the exogenous expression any of these amino acid sequences.
  • said pharmaceutical composition preferably the vaccine composition, further comprises a vector, such as viral vector, a plasmid or an expression cassette comprising a nucleotide sequence capable of coding for any of the amino acid sequences as defined in any of the precedent paragraphs and expressing said amino acid sequences.
  • a vector such as viral vector, a plasmid or an expression cassette comprising a nucleotide sequence capable of coding for any of the amino acid sequences as defined in any of the precedent paragraphs and expressing said amino acid sequences.
  • the Acinetobacter baumannii strain deficient in lipopolysaccharide (LPS) is inactivated.
  • said strain or cell is derived from ATCC strain 19606.
  • This aspect of the invention refers to the pharmaceutical composition, preferably the vaccine composition, of the aspect A of the invention or of any of its preferred embodiments, for use in the prophylactic treatment or for the active immunization of an infection caused by A. baumannii in a mammal, preferably in a human.
  • This aspect of the invention refers to a vaccine composition
  • a vaccine composition comprising an antibody (monoclonal or polyclonal) or a fragment thereof, preferably selected from the list consisting of Fab, Fab′, Fab′-SH, Fv, scFv, F(ab′) 2 , Vhh, nanobody and diabody, having affinity or binding affinity against the Acinetobacter baumannii strain deficient in lipopolysaccharide (LPS) and/or against an outer membrane vesicle (OMV) derived therefrom.
  • said antibody or fragment thereof specifically binds the Acinetobacter baumannii strain deficient in lipopolysaccharide (LPS) and/or to the outer membrane vesicle (OMV) derived therefrom.
  • the term “specifically binding” means that the antibody binds to the LPS deficient strain or to the OMV derived therefrom with an affinity KD of lower than or equal to 10 ⁇ 6 M (monovalent affinity).
  • the antibody may have substantially greater affinity for the target antigen compared to other unrelated molecules.
  • the antibody may also have substantially greater affinity for the target antigen compared to homologs, e.g. at least 1.5-fold, 2-fold, 5-fold 10-fold, 100-fold, 10 ⁇ 3 -fold, 10 ⁇ 4 -fold, 10 ⁇ 5 -fold, 10 ⁇ 6 -fold or greater relative affinity for the target antigen.
  • affinities may be readily determined using conventional techniques, such as by equilibrium dialysis; by radioimmunoassay using radiolabeled target antigen; or by another method known to the skilled artisan.
  • the affinity data may be analyzed, for example, by the method described in [Kaufman R J, Sharp P A. (1982) Amplification and expression of sequences cotransfected with a modular dihydrofolate reductase complementary dna gene. J Mol Biol. 159:601-621].
  • antibody is used in the broadest sense and includes fully assembled antibodies, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), antibody fragments that can bind the antigen (e.g., Fab′, F′(ab)2, Fv, single chain antibodies, diabodies), camelbodies and recombinant peptides comprising the forgoing as long as they exhibit the desired biological activity.
  • a “functional fragment” or “antigen-binding antibody fragment” of an antibody/immunoglobulin hereby is defined as a fragment of an antibody/immunoglobulin (e.g., a variable region of an IgG) that retains the antigen-binding region.
  • An “antigen-binding region” of an antibody typically is found in one or more hypervariable region(s) of an antibody, i.e., the CDR-1, -2, and/or -3 regions; however, the variable “framework” regions can also play an important role in antigen binding, such as by providing a scaffold for the CDRs.
  • variable region refers to the amino acid residues of the variable domains VH and VL of an antibody or functional fragment which are responsible for antigen-binding.
  • Nonlimiting examples of antibody fragments include Fab, Fab′, F(ab′)2, Fv, domain antibody (dAb), complementarity determining region (CDR) fragments, single-chain antibodies (scFv), single chain antibody fragments, diabodies, triabodies, tetrabodies, minibodies, linear antibodies [Johnson G, Wu T T. (2000) Kabat database and its applications: 30 years after the first variability plot. Nucleic Acids Res.
  • chelating recombinant antibodies tribodies or bibodies, intrabodies, nanobodies, small modular immunopharmaceuticals (SMI Ps), an antigen-binding-domain immunoglobulin fusion protein, a camelized antibody, a VHH containing antibody, or muteins or derivatives thereof, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide, such as a CDR sequence, as long as the antibody retains the desired biological activity; and multispecific antibodies formed from antibody fragments [Chothia C, Lesk A M. (1987) Canonical structures for the hypervariable regions of immunoglobulins. J Mol Biol.
  • the pharmaceutical or vaccine composition is obtained or obtainable after immunizing a mammal with the vaccine composition as defined in the first aspect of the invention.
  • This aspect of the invention refers to the vaccine composition as defined in aspect C of the invention, for use in the therapeutic treatment (after the infection), or in the passive immunization, of an infection caused by A. baumannii in a mammal, preferably in a human.
  • This aspect of the invention refers to an Acinetobacter baumannii strain deficient in lipopolysaccharide (LPS), transformed, transduced or transfected with a nucleotide sequence capable of coding for any of the amino acid sequences as defined in aspect A of the invention so that such strain is capable of producing the exogenous expression any of said amino acid sequences.
  • said A. baumannii deficient strain is use as a medicament.
  • This aspect of the invention refers to a method for the production of antibodies or fragments thereof, preferably selected from the list consisting of Fab, Fab′, Fab′-SH, Fv, scFv, F(ab′) 2 , Vhh, nanobody and diabody, which comprises:
  • Serotype specific variability in A. baumannii relies on variations in epitopes of the LPS component.
  • LPS component By eliminating the LPS component according to the method as detailed in aspect F above, we focus the antibody raising to antibodies against less variable epitopes of the bacterial surface, not related to the LPS component. These epitopes are conserved among A. baumannii strains or individual isolates belonging to all A. baumannii international clones used in A. baumannii classification. This method thus provides the tools for a universal treatment against A. baumannii infections independently of the origin of the strain(s) responsible for the host infection.
  • This aspect of the invention refers to an antibody or fragment thereof produced, obtained or obtainable by the method of aspect F of the invention.
  • This aspect of the invention refers to the use of an Acinetobacter baumannii strain deficient in lipopolysaccharide (LPS) and/or an outer membrane vesicle (OMV) derived therefrom, for the production of antibodies or fragments thereof having affinity or binding affinity or capable of specifically binding such strain and/or OMV.
  • LPS lipopolysaccharide
  • OMV outer membrane vesicle
  • the present invention refers to compositions and vaccines that consist of whole cells deficient in LPS and/or outer membrane vesicles of Acientobacter baumannii ( A. baumannii ) capable of conferring protection against infection caused by infectious pathogens.
  • the authors of the present invention demonstrate that inactivated cells of A. baumannii deficient in LPS and/or outer membrane vesicles from A. baumannii , upon being inoculated produce immunization, which provides protection against posterior infection by said bacteria, which demonstrates the utility of these cells or strains as prophylactic vaccines against infections caused by A. baumannii.
  • a first aspect of the present invention refers to an Acinetobacter cell or strain that is deficient in LPS, hereinafter cell or strain of the invention.
  • Acinetobacter are strictly aerobic non-fermenting and non-motile bacilli that are oxidase negative and appear in pairs by microscopy. They are distributed widely in nature, and are important in soil and contribute to its mineralization.
  • inactivated cell in the present invention is a cell that does not have the ability to replicate but that conserves its immunogenic capacity.
  • the cells of the present invention are inactivated prior to their inoculation to prevent their replication in the host, and therefore prevent invention produced by their administration.
  • the inactivation of the cells of the invention can be performed using diverse methods known in the state of the art for example, although not limited to, adsorption, heat, ultraviolet light, ionizing radiation, ultrasound, phenol, formol, formaldehyde, crystal violet, glyceraldehyde, ethylene oxide, propiolactone, ethylenamina, bromoethyleneamina or formalin.
  • the cells of the invention are inactivating with formalin.
  • the cells of the invention are from the species Acinetobacter baumannii and they are inactivated with formalin.
  • the deficiency in LPS can be achieved by partial or compete inactivation of one or various cellular molecules of nucleic acids that encode the endogenous genes for the LPS subunits, particularly IpxA, IpxB and/or IpxC of LPS.
  • the cell or strain of Acinetobacter deficient in LPS is obtained by deletions and/or insertions of one or various nucleotides in nucleic acid sequences encoding the gene involved in the biosynthesis of LPS and/or the sequences that control their expression.
  • the deletions and/or insertions can be generation by homologous recombination, insertion of transposons, or other adequate methods known in the state of the art.
  • the sequence is inactivated e.g. by construction of a suicide vector that contains the gene IpxA, IpxB, IpxC, IpxD, IpxK, IpxL and/or IpxM or any of their combination, or interrupting with a marker gene for selection, transforming the target cells with the vector and screening for positive cells that are negative for LPS expression.
  • the cell or strain of the invention is preferably an A. baumannii cell, particularly an attenuated A. baumannii cell or other Acinetobacters; Acinetobacter baylyi, A. beijerinckii, A. bereziniae, A. boissieri, A. bouvetii, A. brisouii, A. calcoaceticus, A. outfitri, A. guillouiae, A. grimontii, A. gyllenbergii, A. haemolyticus, A. indicus, A. johnsonii, A. junii, A. lwoffii, A. nectaris, A.
  • nosocomialis A. parvus, A. pittii, A. puyangensis, A. radioresistens, A. rudis, A. schindleri, A. soli, A. tandoii, A. tjernbergiae, A. towneri, A. ursingii or A. venetianus.
  • Acinetobacter refers to the kingdom Bacteria, phylum Proteobacteria, class Gammaproteobacteria, order Pseudomonadales, family Moraxellaceae.
  • cells or strains of the Acinetobacter baumannil in the present invention are those cells pertaining to the domain Bacteria, phylum Proteobacteria, class Gammaproteobacteria, order Pseudomonadales, family Moraxellaceae, genus Acinetobacer , species Acinetobacter baumannii.
  • LPS lipopolysaccharide
  • lipooligosaccharide is a component that is found on the external membrane of various Gram-negative bacteria.
  • LPS is used often and interchangeably with “endotoxin”, due to its history of discovery.
  • LPS consists of a polysaccharide chain and the rest is lipid, known as lipid A, which is responsible for the endotoxin activity.
  • the polysaccharide chain is variable between different bacterias and determines the serotype.
  • Endotoxin is of approximately 10 kDa in size, but can form large aggregates of up to 1000 kDa. Humans are able to produce antibodies against LPS, but in general these antibodies can only protect against bacteria of a specific serotype.
  • Endotoxin is responsible for many of the clinical manifestations of infections caused by Gram-negative bacteria such as Neisseria meningitidis and Acinetobacter baumannii.
  • composition of the invention comprising:
  • the nucleic acid and/or the amino acid sequence or polypeptide is recombinant.
  • polypeptide is selected from
  • composition of the invention comprises the amino acid sequence SEQ ID NO: 28 and the amino acid sequence SEQ ID NO: 27.
  • the composition of the invention comprises a fusion protein that consists of at least 2, preferably 3, more preferably 4, amino acid sequences from the following list consisting of: SEQ ID NO: 1 to SEQ ID NO: 23 or a variant of these sequences having at least 85% identity with the sequences SEQ ID NO: 1 to SEQ ID NO: 23.
  • the fusion protein comprises the amino acid sequence SEQ ID NO: 24 or the amino acid sequence SEQ ID NO: 25.
  • the composition of the invention comprises a nucleotide sequence, hereinafter nucleotide sequence of the invention, capable of transcribing an amino acid sequence described in the invention. More preferably, the nucleotide sequence is SEQ ID NO: 26.
  • the fragments described previously differ in amino acid sequence by at least one amino acid.
  • the most preferred variations are those having at least 85%, or more, including 90%, 93% or more, and preferably 95% or more, 96% or more, 97% or more, 98% or more, 99% or more of sequence identity with any of the polypeptides shown in SEQ ID NO:1 to SEQ ID NO:25.
  • the invention refers to a sequence variant characterize by at least one (at least two, at least three, at least four) mutation(s) in relation to any of the polypeptides SEQ ID NO:1 to SEC ID NO:25.
  • mutation can refer to any mutation selected by insertion(s), deletion(s), and substitution(s).
  • substitution(s) can refer to any mutation selected by insertion(s), deletion(s), and substitution(s).
  • substitution(s) can refer to any mutation selected by insertion(s), deletion(s), and substitution(s).
  • substitution(s) can refer to any mutation selected by insertion(s), deletion(s),
  • composition of the invention comprises an expression vector (hereinafter expression vector of the invention), comprising a nucleotide sequence of the invention.
  • composition of the invention also comprises outer membrane vesicles, hereinafter outer membrane vesicles of the invention, deficient in LPS.
  • the composition of the invention also comprises at least one or the proteins purified from the membrane of A. baumanni with amino acid sequence SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46; SEQ ID NO: 47; SEQ ID NO: 48; SEQ ID NO: 49; SEQ ID NO: 50; SEQ ID NO: 51; SEQ ID NO: 52; SEQ ID NO: 53; SEQ ID NO: 54; SEQ ID NO: 55; SEQ ID NO: 56; SEQ ID NO: 57; SEQ ID NO: 58; SEQ ID NO: 59; SEQ ID NO:
  • At least one of the proteins of the outer membrane of A. baumannii is obtained by a process comprising:
  • the composition of the invention is a pharmaceutical composition, more preferably also comprises an acceptable pharmaceutical vehicle, and still more preferably, also comprises another active ingredient.
  • composition of the invention also comprises an adjuvant.
  • composition of the invention is a vaccine.
  • the term “vaccine” refers to an antigenic preparation employed for inducing an immune response to a disease. They are prepared from antigens that, once inside the host, provoke an immune response through the production of antibodies, and generate immunologic memory producing transient or permanent immunity. It is noted that as used herein the term “vaccine” can also be understood as a preparation from antibodies or fragments thereof suitable for the therapeutic treatment or for passive immunization of an infection caused by an Acinetobacter strains, in particular from an A. baumannii strain.
  • a third aspect of the invention refers to a nucleotide sequence, hereinafter second nucleotide sequence of the invention, which encodes any of SEQ ID NO 1 to 25, 27 or 28 or any combination thereof.
  • Said second nucleotide sequence of the invention also includes nucleic acid sequence SEQ ID NO: 26.
  • a fourth aspect of the invention refers to an expression vector, hereinafter expression vector of the invention, comprising the second nucleotide sequence of the invention.
  • the cell of the invention comprises the expression vector of the invention.
  • the nucleic acid can be localized in a recombinant vector.
  • the recombinant vector is a prokaryotic vector, that is a vector that contains elements for replication and/or integration into the genome of prokaryotic cells.
  • the recombinant vector contains the nucleic acid molecule of the present invention operatively linked to an expression control sequence.
  • the control sequence is preferably a sequence controlling the active expression in Acinetobacter , particularly in A. baumannii .
  • the vector can be an extrachromosomic vector adequate for integration in the chromosome. Examples of such as vectors are known by experts in the field, for example in Sambrook et al. supra.
  • a fifth aspect of the invention refers to an outer membrane vesicle that is deficient in LPS.
  • the outer membrane vesicle is obtained from the cell or strain of the invention.
  • a sixth aspect refers to the composition of the invention for use as a medicament, or alternatively, to the use of the composition of the invention for the manufacture of a medicament.
  • the term “medicament” or “pharmaceutical composition” as used in this report, makes reference to any substance used for the prevention, alleviation, treatment or cure of a disease in man or animals.
  • the context of the present invention refers to a composition comprising the composition of the invention.
  • This composition of the invention comprises inactivated cells of the genus Acinetobacter and/or outer membrane vesicles of the same in a quantity therapeutically effective, that is able to inducer an immune response in the organism in which they are administered against an organism of the genus Acinetobacter .
  • the term “medicament” or “pharmaceutical composition” therefore is known as a vaccine.
  • the dosing for obtaining the effective therapeutic quantity depends on a variety of factors such as for example, age, weight, sex, tolerante, . . . of the mammal.
  • the “effective therapeutic quantity” refers to the quantity of inactive cells of the genus Acinetobacter and/or outer membrane vesicles that produce the desired effect, and in general are determined by the therapeutic effect that is desired.
  • a seventh aspect of the invention refers to the composition of the invention for the prevention, the improvement or the treatment of an infection caused by A. baumannii , en a mammal, or alternatively, to the use of the composition of the invention for the elaboration of a medicament for the prevention, improvement or treatment of an infection caused by A. baumannii in a mammal.
  • diseases produced by organisms of the genus Acientobacter those diseases in which the causal agent of the pathology is from the genus Acinetobacter , or any of its metabolic products.
  • the genus Acinetobacter produces diverse pathologies for example but not limited to, bacteremia, meningitis, urinary tract infections, skin and soft tissue infections, surgical site infections and pneumonia.
  • diseases produced by organisms of the genus Acinetobacter are selected from a list that consists of bacteremia, meningitis, urinary tract infections, skin and soft tissue infections, surgical site infections and pneumonia.
  • the medicaments and compositions of the invention can be used alone or in combination with other medicaments or compositions for the treatment of diseases produced by organisms from the genus Acinetobacter.
  • Both the medicaments and the compositions of the invention can also include pharmaceutically acceptable vehicles or excipients
  • the medicaments and compositions of the invention may be used either alone or in combination with other medicaments or compositions for the treatment or prevention of diseases caused by organisms of the genus Acinetobacter.
  • excipient makes reference to a substance that helps in the absorption of the elements of the composition of the medicaments of the invention, stabilizing said elements, activating or helping the preparation of the medicament such that it provides consistency or flavours that make it more palatable.
  • the excipients can maintain the ingredients together, like for example is the case with starches, sugars, cellulose, sweetners, coloring agents, the function of protecting the medicament, for example isolating it form air and/or humidity, the function of filling the pill, capsule or any other form of presentation, for example, the case of dibasic calcium phosphate, the function for facilitating dissolution of the components and their absorption in the intestine, without excluding other types of excipients described in this paragraph.
  • the vehicle in the same way as the excipient, is a substance that is used in the medicament to dilute any of the components of the present invention to a desired volume or weight.
  • the pharmaceutically acceptable vehicle is an inert substance or of similar action to any of the elements of the present invention.
  • the function of the vehicle is to facilitate the incorporation of other elements, permit better dosing and administration and give consistency and form to the medicament.
  • the pharmaceutically acceptable vehicle is the diluent.
  • the adjuvants and pharmaceutically acceptable vehicle that can be used in the composition of the invention are those vehicles known by experts in the field.
  • the term “adjuvant” refers to any agent that does not poses antigenic activity in and of itself, that can be used to stimulate the immune system to increase the response to a vaccine.
  • adjuvants for example but not limited to, aluminium phosphate, aluminium hydroxide, toll-like receptor agonists, cytokines, squaline, Freunds incomplete and complete adjuvants.
  • the adjuvant is selected for a list that consists of aluminium phosphate, aluminium hydroxide, toll-like receptor agonists, cytokines, squaline, Freunds incomplete and complete adjuvants.
  • the adjuvant is aluminium phosphate.
  • active ingredient refers to any component that potentially provides pharmacological activity or other different effect in the diagnosis, cure, alleviation, treatment or prevention of a disease or that affects the structure or function of the human or animal body.
  • the term includes those components that promote a chemical change in the elaboration of the drug and are present in the same and a modified form that provides specific activity or the effect.
  • An eighth aspect of the invention refers to the composition of the invention for conferring protection against an infection caused by A. baumannii en a mammal, or alternatively, the use of the composition of the invention in the elaboration of a medicament for conferring protection against an infection caused by A. baumannii in a mammal.
  • Another aspect of the invention refers to the fusion protein or peptide of the invention or the composition of the first or second aspect of the invention or the pharmaceutical composition of the invention that can be administered once or various, such as two, three, four, five, six, seven, eight, nine or ten or more times. There are no particular limitations relative to the quantity of the active ingredient per dose.
  • An additional aspect of the present invention refers to a composition that consists o fan antibody or fragment thereof that is capable of binding to SEQ ID NO: 27 or SEQ ID NO: 28 or a fusion protein as defined in the second aspect or the fusion protein of the invention, wherein preferably said composition is a pharmaceutical composition, preferably a vaccine, and wherein said pharmaceutical composition is used in the treatment or prevention of an infection caused by A. baumannii.
  • a ninth aspect of the invention refers to an antibody or an active fragment thereof obtainable by immunization of a mammal with the composition of the first or second aspect of the invention or with the fusion protein of the invention, preferably said antibody or active fragment consists of a composition in which preferably said composition is a pharmaceutical composition and said pharmaceutical composition is used as a therapy, particularly for the treatment of infections caused by A. baumannii.
  • a tenth aspect of the invention refers to a method for preparing a cell of A. baumannii as described above.
  • this method consists of the steps to (i) provide a bacterial cell deficient in LPS, particularly a cell of Acinetobacter , (ii) insert a recombinant nucleic acid molecule in said bacterial cell, encoding said nucleic acid molecule a fusion peptide that consists of (a) at least on domain of the polypeptide wherein said domain is capable of producing an immune response in mammal and (b) a domain of escape for the phagolysosome, and (iii) culture the obtained cell in agreement with step (ii) in adequate conditions.
  • a cell capable of expressing said nucleic acid is obtained. More preferably, the cell is a cell of A. baumannii.
  • this method consists of step of (i) providing a bacterial cell deficient in LPS, particularly an Acinetobacter cell, (ii) inserting a recombinant nucleic acid molecule in said bacteria, encoding said nucleic acid molecule a peptide or polypeptide for escape from the phagolysosome, and (iii) culture the obtained cell in agreement with step (ii) in adequate conditions.
  • the method of the present invention consists of inserting at least one recombinant nucleic acid molecule in the bacterial cell, said molecule encoding a peptide or polypeptide capable of producing an immune response in mammals.
  • infection in the present invention is that pathology generated by the invasion or colonization of any host tissue by any organisms of the genus Acinetobacter , preferably Acinetobacter baumannii.
  • soft tissue in the present invention, is all non-bony tissue of an organism.
  • prevention as understood in the present invention consists of avoiding the appearance of damage whose cause is cells of the genus Acinetobacter , or any derivative or metabolic product of the same.
  • antigen in the report refers to a molecule (generally a protein or polysaccharide) that can induce the formation of antibodies.
  • a molecule generally a protein or polysaccharide
  • antigens There many different types of molecules that can act as antigens, such as proteins, peptides, polysaccharides, and more rarely other molecules such as nucleic acids.
  • resistance refers to any mechanism of defense developed by bacteria against a drug.
  • the mechanisms of resistance acquired and transmitted by bacteria are the most important and consist primarily of: the production of enzymes that inactivate antibiotics, appearance of modifications that impede the arrival of the drug to its target and/or alteration of the target itself.
  • One bacterial strain can develop various resistance mechanisms against one or many antibiotics and in the same way an antibiotic can be inactivated by distinct mechanisms from diverse bacterial species.
  • polynucleotide and “nucleic acid” are used interchangeably, referring to polymeric forms of nucleotides of any length, both of DNA and RNA
  • amino acid sequence refers to a polymeric form of amino acids of any length that can be chemically or biochemically modified.
  • FIG. 1 Stability and endotoxin content of IB010.
  • A Genomic DNA from three independent cultures of ATCC 19606 and IB010 was extracted and amplified using primers specific for the IpxD gene. The band corresponding to approximately 1000 Kb corresponds to the intact IpxD gene, whereas the faster migrating band corresponds to the IpxD gene with a deletion of 462 nucleotides.
  • B Endotoxin levels of ATCC 19606 and IB010 determined by the Limulus Amebocyte Assay. Bars represent the median values of three independent cultures, and error bars represent the standard error of the mean. EU; endotoxin units.
  • FIG. 2 Antibody response to immunization with IB010.
  • IgG1 (C) and IgG2c (D) levels were measured in 21-day serum were measured by ELISA in ATCC 19606 vaccinated, IB010 vaccinated and control mice.
  • box and whisker plots represent the interquartile ranges and ranges, respectively, and horizontal lines represent median values.
  • * p ⁇ 0.05 compared to levels in control mice at the same time point # p ⁇ 0.05 compared to 7-day samples from the same experimental group, ⁇ p ⁇ 0.05 compared to 21-day samples in ATCC 19606 vaccinated mice.
  • FIG. 4 Effect of vaccination on post-infection pro-inflammatory cytokine levels.
  • Data points represent cytokine levels from individual mice, and horizontal lines represent median values from groups of mice. * p ⁇ 0.05 compared to control mice, # p ⁇ 0.05 compared to ATCC 19606 vaccinated mice.
  • FIG. 5 Effect of vaccination on survival in a mouse model of disseminated A. baumannii infection.
  • FIG. 6 Protein profile of the A. baumannii OMVs without LPS.
  • FIG. 7 Effect of 2,2 Bipyridyl, iron chelator, in the protein profile of OMVs.
  • the strains ATCC 19606 and IB010 were used to analyze the effect of 2,2 bypiridyl (BIP) on the protein profile of the OMVs after culturing for 24 hours.
  • BIP 2,2 bypiridyl
  • a concentration of 200 mcM was used while in the case of IB010 100 and 150 mcM was used.
  • After the purification of OMVs the amount of protein was quantified using Bradford method and 10 mcg of the protein was visualized in a 10% polyacrylamide gel with Coomassie stain.
  • FIG. 8 Effect of 2,2 Bipyridyl, iron chelator, on the production of OMVs.
  • the strains ATCC 19606 and IB010 were used to analyze the effect of 2,2 bypiridyl (BIP) on the production of the OMVs after culturing for 24 hours.
  • BIP 2,2 bypiridyl
  • FIG. 9 Visualization of OMVs.
  • OMVs purified form ATCC 19606 were fixed using glutaraldehyde at 1.6% and stained with osmium tetroxide and lead and uranium and visualized by electron microscopy.
  • FIG. 10 Visualization of purified OMVs.
  • OMVs purified form IB010 were fixed using glutaraldehyde at 1.6% and stained with osmium tetroxide and lead and uranium and visualized by electron microscopy.
  • FIG. 11 Protein profile of OMVs without LPS.
  • the strains IB010 and 167R were used to produce OMVs and the amount of protein was quantified from each sample. Ten mcg of the protein was visualized in a 10% polyacrylamide gel with Coomassie stain.
  • A. baumannii ATCC 19606 is an antibiotic susceptible reference strain.
  • An LPS-deficient derivative of ATCC 19606 was obtained by plating an overnight culture of ATCC 19606 on Mueller Hinton agar containing 10 mg/I of colistin, as described previously (Clinical Laboratory Standards Institute 2013) Strains with mutations in the genes involved in LPS biosynthesis were identified by sequencing the IpxA, IpxC and IpxD genes of the colistin resistant mutants that were present after overnight growth at 37° C.
  • a strain with a large deletion in the IpxD gene was identified and designated IB010. Resistance to colistin was confirmed by broth microdilution according to Clinical Laboratory Standard Institute guidelines [23].
  • LPS Limulus Amebocyte Assay
  • the IWC vaccines (both LPS-containing and LPS-deficient) were prepared as described based on a previously described method (Moffatt et al., 2010 . Antimicrob. Agents Chemother. 54, 4971-4977). Briefly, the ATCC 19606 and IB010 strains were grown in Mueller-Hinton broth to OD 600 of 0.8. In the case of IB010, 10 ⁇ g/ml of colistin were added to the culture. In order to confirm that no reversion to wild type occurred during growth of IB010, three independent cultures of ATCC 19606 and IB010 were grown, and genomic DNA was isolated from each culture using the QIAmp DNA Mini Kit (Qiagen).
  • the IpxD specific primers 5′ GCTAATTGGTGAAGGTAGTC 3′ and 5′ GACGAATCGTTTGAATCTGC 3′ were used to amplify genomic DNA from the cultures in order to confirm that the deletion in IpxD of IB010 was present after growth.
  • bacteria were washed extensively in phosphate buffer saline before inactivation in 0.5 M formalin for 18 h with shaking at room temperature. Complete inactivation of the bacteria was confirmed by plating on blood agar. The concentration of inactivated cells was adjusted to 1 ⁇ 10 10 cells/ml and combined 1:1 (v/v) with the aluminium-based adjuvant, Alhydrogel 2% (w/v) (InvivoGen). Vaccination was carried out in 6 to 8-week-old, female C57BL/6 mice by intramuscular injection of 100 ⁇ l of the vaccine into each quadriceps muscle on days 0 and 14. Control mice were injected similarly with a mixture of phosphate buffer saline and adjuvant.
  • This model produces a disseminated infection after intraperitoneal instillation of the inoculum, typically resulting in death within 24 to 48 hours.
  • Post-infection bacterial loads were determined in vaccinated and control mice 12 h after infection. Mice were euthanized with an overdose of thiopental and after collection of blood samples from the retro-orbital sinus, spleens were aseptically removed, weighed and homogenized in 2 ml of physiological saline. Serial log dilutions were plated on blood agar plates for bacterial quantification. Serum levels of interleukin-1 ⁇ (IL-1 ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), and interleukin-6 (IL-6) were determined in mice at 12 h post-infection using BD OptEIA mouse kits (BD Biosciences).
  • IL-1 ⁇ interleukin-1 ⁇
  • TNF- ⁇ tumor necrosis factor alpha
  • IL-6 interleukin-6
  • ELISAs indirect enzyme-linked immunosorbent assays
  • 96-well plates were coated with 5 ⁇ 10 7 bacterial cells/well in phosphate buffer saline by incubating at 4° C. overnight.
  • ELISAs were performed using sera collected on days 0, 7 and 21 as described previously [28].
  • Antibody titers were measured against the strain which was used to immunize the mouse, and were defined as the dilution in which spectrophotometric readings were at least 0.1 units above background wells (wells containing no serum).
  • Antibody titers, bacterial loads, and cytokine levels were compared using the Kruskal-Wallis H test and the Mann-Whitney U test for independent samples, and the Friedmann and Wilcoxon tests for dependent samples. The Bonferroni correction was applied when appropriate. Survival data were compared using the log-rank test. All statistics were performed using SPSS version 15.0 software (SPSS Inc.), and a p value of 0.05 was considered significant.
  • Vaccinated and control mice were infected with 2.0 ⁇ 10 6 cfu (300 ⁇ LD 50 ) of the ATCC 19606 strain, and 12 hours after infection spleen bacterial loads were determined ( FIG. 3 ).
  • IB010 vaccination reduced the number of bacteria in spleens approximately 1000-fold compared to control mice (p ⁇ 0.05; Mann-Whitney U test). Spleen bacterial loads in IB010 vaccinated mice were not significantly different than in mice immunized with inactivated ATCC 19606 cells.
  • Vaccine efficacy was tested by infecting immunized and control mice with 2.25 ⁇ 10 6 cfu (340.9 ⁇ LD 50 ) of the ATCC 19606 strain seven days after the second immunization, and survival was monitored over seven days ( FIG. 5 ). All mice vaccinated with the IB010 vaccine were protected from challenge, whereas all control mice died within 48 hours (P ⁇ 0.001; log-rank test). As expected, all mice immunized with the ATCC 19606 strain survived challenge, similar to results that were previously reported (McConnell et al., 2011 . Infect. Immun. 79, 518-526; McConnell et al., 2011 .
  • Vaccine 29 5705-5710; McConnell y Pachón, 2010 . Vaccine 29, 1-5).
  • immunized and control mice were infected with 1.05 ⁇ 10 6 cfu (2.18 ⁇ LD 50 ) of the previously characterized A. baumannii clinical isolate Ab-154 [29].
  • control mice succumbed to infection within 48 hours (p ⁇ 0.001; log-rank test), indicating that immunization with IB010 can provide cross protection against challenge with a heterologous strain.
  • This example relates to the development of a vaccine against A. baumannii based on OMVS purified from said cultures of mutants without LPS.
  • the strains ATCC 19606T and its mutant without LPS IB010 which was generated in our laboratory from the ATCC 19606T strain and contains a deletion of 462 nucleotides between positions 103 and 565 of the gene IpxD.
  • the pellet is resuspended in PBS and the absence of viable bacteria is confirmed by plating.
  • the OMVs are stored at ⁇ 80° C.
  • OMVs were purified form the LPS mutant of a clinical isolate of A. baumannii Ab-167 which contains an ISAba1 insertion in the IpxC gene. Proteins were quantified and visualized on acrylamide gels by Coomassie staining. And proteins were quantified by Bradford and 2D Quant kit.
  • A. baumannii ATCC 19606 was grown in 1 liter of Mueller-Hinton broth to an optical density at 600 nm (OD600) of 0.6, and pelleted bacteria were resuspended in 10 ml of 10 mM phosphate buffer, pH 7.2, and lysed by sonication. Unlysed cells were removed by centrifugation at 4,000 ⁇ g for 5 min, and the supernatant was centrifuged at 20,000_g for 1 h to pellet cell envelopes. Inner membranes were selectively solubilized with 5 ml of 2% N-laurylsarcosinateby incubation at 37° C. for 30 min. The insoluble fraction was pelleted by centrifugation at 20,000_g for 1 h and then washed with 2 ml of 62.5 mM Tris-Cl, pH 6.8.
  • Endotoxin was extracted from the preparation by use of a cold detergent wash step in which proteins were resuspended in 5% SDS and incubated at 4° C. for 10 min. SDS and endotoxin were subsequently removed by precipitating in methanol chloroform and resuspended in PBS.
  • the purified proteins at a concentration of 500 mcg/ml were mixed with aluminum phosphate adjuvant at a 1:1 ration.
  • Acinetobacter cell according to the preceding claim obtained through partial or complete inactivation of one or various of the nucleic acids encoding the endogenous LPS biosynthesis genes.
  • Acinetobacter cell according to any one of the preceding claims, wherein the cell is obtained by deletions, and/or insertions of one or various nucleotides in the coding sequences of the genes.
  • Acinetobacter cell according to any one of the preceding claims, wherein the cell is an attenuated Acinetobacter cell.
  • composition comprising:
  • composition according to the preceding claim wherein the nucleic acid molecule is recombinant and the polypeptide is recombinant.
  • composition according to any one of claims 6 - 7 wherein the polypeptide is selected from:
  • composition according to any one of claims 6 - 8 further comprising the amino acid sequence SEQ ID NO: 28 and the amino acid sequence SEQ ID NO: 27.
  • composition according to any one of claims 6 - 9 further comprising a fusion protein comprising at least 2, preferably 3, more preferably 4 amino acid sequences form the list SEQ ID NO: 1 to SEQ ID NO: 23 or a variant of these sequences having at least 85% sequence identity with SEQ ID NO. 1 to SEQ ID NO: 23.
  • composition according to claim 10 wherein the fusion protein further comprises the amino acid sequence SEQ ID NO: 24 or the amino acid sequence SEQ ID NO: 25.
  • composition according to any one of claims 6 - 11 wherein the composition comprises a nucleotide sequence capable of transcribing an amino acid sequence as described in any one of claims 8 - 11 .
  • composition according to claim 12 wherein the nucleotide sequence is the SEQ ID NO: 26.
  • composition according to any one of claims 6 - 13 further comprising an expression vector comprising the nucleotide sequence according to any one of claims 12 - 13
  • composition according to any one of claims 6 - 14 further comprising outer membrane vesicles deficient in LPS, or cells according to any one of claims 1 to 5 .
  • composition according to any one of claims 6 - 14 further comprising at least one of the purified outer membrane proteins of A. baumannii with amino acid sequence SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46; SEQ ID NO: 47; SEQ ID NO: 48; SEQ ID NO: 49; SEQ ID NO: 50; SEQ ID NO: 51; SEQ ID NO: 52; SEQ ID NO: 53; SEQ ID NO: 54; SEQ ID NO: 55; SEQ ID NO: 56; SEQ ID NO: 57; SEQ ID NO: 58; SEQ ID NO: 59
  • composition according to any one of claims 6 - 15 wherein the cells or the outer membrane vesicles are designed to produce the amino acid sequences according to any one of claim 8 - 11 or 16 and/or comprising the nucleotides or nucleic acids according to any one of claims 12 - 13 .
  • composition according to any one of claims 7 to 16 and 19 that is a pharmaceutical composition is a pharmaceutical composition.
  • composition according to claim 22 further comprising a pharmaceutically acceptable vehicle.
  • composition according to any one of claim 22 or 23 further comprising another active ingredient.
  • composition according to any one of claims 22 - 24 further comprising an adjuvant.
  • composition according to any one of claims 7 to 16 , 19 to 22 and 25 , wherein the composition is a vaccine.
  • composition according to any one of claims 7 to 16 , 19 to 22 , and 25 for use as a medicament for use as a medicament.
  • composition according to any one of claims 7 to 16 , 19 to 22 , and 25 for the prevention, improvement or the treatment of an infection caused by A. baumannii in un mammal.
  • composition is a pharmaceutical composition and wherein said composition is used in therapy, particularly for the treatment and prevention of infection caused by A. baumannii.
  • composition comprising:
  • composition according to claim 3 wherein the polypeptide is selected from:
  • composition according to any one of claims 3 - 4 , wherein the composition comprises a nucleotide sequence capable of transcribing an amino acid sequence described in claim 4 , preferably SEQ ID NO: 26.
  • composition according to any one of claims 3 - 5 further comprising an outer membrane vesicle deficient in LPS, or cells according to any one of claims 1 and 2 .
  • composition according to any one of claims 3 - 6 further comprising at least one of the proteins purified from the outer membrane of A. baumannii SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46; SEQ ID NO: 47; SEQ ID NO: 48; SEQ ID NO: 49; SEQ ID NO: 50; SEQ ID NO: 51; SEQ ID NO: 52; SEQ ID NO: 53; SEQ ID NO: 54; SEQ ID NO: 55; SEQ ID NO: 56; SEQ ID NO: 57; SEQ ID NO: 58; SEQ ID NO: 59;
  • composition according to any one of claims 3 - 7 wherein the cells or the outer membrane vesicles are designed to produce the polypeptide sequences according to any one of claims 4 and 7 , and/or comprising the nucleic acid sequences of claim 5 .
  • composition according to any one of claims 3 - 7 and 10 wherein the composition is a pharmaceutical composition.
  • composition according to any one of claims 3 - 7 , 10 and 11 for use as a medicament.
  • composition according to any one of claims 3 - 7 , 10 and 11 for use in the prevention, improvement or treatment of an infection caused by A. baumannii in a mammal.
  • An antibody or active fragment thereof obtained by immunization of a mammal with the composition according to any one of claims 3 to 7 , 10 and 11 .
  • composition is a pharmaceutical composition, and where said composition is used in therapy, particularly for the treatment or prevention of an infection caused by A. baumannii.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
US15/308,899 2014-05-05 2015-05-05 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide Abandoned US20170065700A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP14382164.3 2014-05-05
EP14382164.3A EP2942389A1 (en) 2014-05-05 2014-05-05 Vaccine against Acinetobacter baumannii based on lipopolysaccharide-deficient cellular components
PCT/EP2015/059870 WO2015169809A1 (en) 2014-05-05 2015-05-05 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/059870 A-371-Of-International WO2015169809A1 (en) 2014-05-05 2015-05-05 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/878,491 Division US11160856B2 (en) 2014-05-05 2018-01-24 Vaccine against Acinetobacter baumannii based on cellular components deficient in lipopolysaccharide

Publications (1)

Publication Number Publication Date
US20170065700A1 true US20170065700A1 (en) 2017-03-09

Family

ID=51059396

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/308,899 Abandoned US20170065700A1 (en) 2014-05-05 2015-05-05 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
US15/878,491 Active 2035-08-19 US11160856B2 (en) 2014-05-05 2018-01-24 Vaccine against Acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
US17/488,545 Pending US20220152184A1 (en) 2014-05-05 2021-09-29 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/878,491 Active 2035-08-19 US11160856B2 (en) 2014-05-05 2018-01-24 Vaccine against Acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
US17/488,545 Pending US20220152184A1 (en) 2014-05-05 2021-09-29 Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide

Country Status (9)

Country Link
US (3) US20170065700A1 (da)
EP (2) EP2942389A1 (da)
JP (1) JP6775424B2 (da)
CN (1) CN107073095B (da)
DK (1) DK3140391T3 (da)
ES (1) ES2848066T3 (da)
MX (1) MX2016014564A (da)
PT (1) PT3140391T (da)
WO (1) WO2015169809A1 (da)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019060428A1 (en) * 2017-09-20 2019-03-28 The United States Government As Represented By The Department Of Veterans Affairs BACTERIAL PROTEIN COMPOSITIONS AND USES THEREOF
CN110746496A (zh) * 2019-09-30 2020-02-04 四川大学华西医院 一种鲍曼不动杆菌的pal重组蛋白及其编码基因和它们的应用
CN112203683A (zh) * 2018-04-10 2021-01-08 杜雷安教育基金会行政处 作为佐剂的假鼻疽伯克霍尔德氏菌复合外膜囊泡
US11160856B2 (en) 2014-05-05 2021-11-02 Vaxdyn S.l. Vaccine against Acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
WO2022140382A1 (en) * 2020-12-22 2022-06-30 Biological Mimetics, Inc. Irradiated whole-cell immunogens of acinetobacter baumannii

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11819532B2 (en) * 2018-04-23 2023-11-21 Hoffmann-La Roche Inc. Peptide macrocycles against Acinetobacter baumannii
CN110283745B (zh) * 2019-06-27 2021-05-11 浙江工业大学 医院不动杆菌fk2及其在降解有机污染物中的应用
CN113234625B (zh) * 2021-05-08 2022-05-24 哈尔滨师范大学 一株皮特不动杆菌及其应用
EP4169529A1 (en) 2021-10-20 2023-04-26 Vaxdyn S.L. Lipopolysaccharide (lps) deficient acinetobacter baumannii multivalent vaccine
CN116462743B (zh) * 2023-06-14 2023-09-19 四川大学华西医院 鲍曼不动杆菌翻译延伸因子重组蛋白、制备方法及应用
CN116478953B (zh) * 2023-06-14 2023-09-12 四川大学华西医院 鲍曼不动杆菌DlaT重组蛋白、制备方法及应用

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101575587B (zh) * 2009-06-03 2011-01-05 中国农业大学 一种流产布鲁氏菌重组菌及其在制备疫苗中的应用
ES2366735B1 (es) * 2009-10-08 2012-09-13 Fundación Pública Andaluza Para La Gestión De La Investigación En Salud De Sevilla Vacuna frente a acinetobacter baumannii.
GEP20197052B (en) * 2011-09-14 2019-12-25 Angeles Biomedical Research Center Los Hyr1-derived compositions and methods of treatment using same
WO2014048976A1 (en) * 2012-09-25 2014-04-03 Vaxdyn Sl Anti-acinetobacter baumannii immunogens
CN102977208B (zh) * 2012-12-12 2015-04-08 大连医科大学 鲍曼不动杆菌特异性卵黄免疫球蛋白的制备方法、应用及药物组合物、制剂和试剂盒
EP2942389A1 (en) 2014-05-05 2015-11-11 Servicio Andaluz De Salud Vaccine against Acinetobacter baumannii based on lipopolysaccharide-deficient cellular components

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11160856B2 (en) 2014-05-05 2021-11-02 Vaxdyn S.l. Vaccine against Acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
WO2019060428A1 (en) * 2017-09-20 2019-03-28 The United States Government As Represented By The Department Of Veterans Affairs BACTERIAL PROTEIN COMPOSITIONS AND USES THEREOF
US11291712B2 (en) 2017-09-20 2022-04-05 The United States Government As Represented By The Department Of Veterans Affairs Bacterial protein compositions and uses thereof
CN112203683A (zh) * 2018-04-10 2021-01-08 杜雷安教育基金会行政处 作为佐剂的假鼻疽伯克霍尔德氏菌复合外膜囊泡
CN110746496A (zh) * 2019-09-30 2020-02-04 四川大学华西医院 一种鲍曼不动杆菌的pal重组蛋白及其编码基因和它们的应用
WO2022140382A1 (en) * 2020-12-22 2022-06-30 Biological Mimetics, Inc. Irradiated whole-cell immunogens of acinetobacter baumannii

Also Published As

Publication number Publication date
DK3140391T3 (da) 2021-01-11
PT3140391T (pt) 2021-01-06
EP3140391B1 (en) 2020-10-07
US20180256700A1 (en) 2018-09-13
JP2017514873A (ja) 2017-06-08
US20220152184A1 (en) 2022-05-19
CN107073095B (zh) 2022-04-19
CN107073095A (zh) 2017-08-18
EP2942389A1 (en) 2015-11-11
EP3140391A1 (en) 2017-03-15
MX2016014564A (es) 2017-05-30
US11160856B2 (en) 2021-11-02
WO2015169809A1 (en) 2015-11-12
JP6775424B2 (ja) 2020-10-28
ES2848066T3 (es) 2021-08-05

Similar Documents

Publication Publication Date Title
US20220152184A1 (en) Vaccine against acinetobacter baumannii based on cellular components deficient in lipopolysaccharide
Garcia-Quintanilla et al. First steps towards a vaccine against Acinetobacter baumannii
Badmasti et al. Immunological evaluation of OMV (PagL)+ Bap (1-487aa) and AbOmpA (8-346aa)+ Bap (1-487aa) as vaccine candidates against Acinetobacter baumannii sepsis infection
JP5931724B2 (ja) StreptococcusPneumoniaeに対するワクチンおよび組成物
Lei et al. DNA vaccine encoding OmpA and Pal from Acinetobacter baumannii efficiently protects mice against pulmonary infection
KuoLee et al. Intranasal immunization protects against Acinetobacter baumannii-associated pneumonia in mice
Butler et al. Immunomodulation therapy offers new molecular strategies to treat UTI
Singh et al. Vaccine development to control the rising scourge of antibiotic-resistant Acinetobacter baumannii: A systematic review
Wang et al. Immunization with outer membrane vesicles of avian pathogenic Escherichia coli O78 induces protective immunity in chickens
US9597386B2 (en) Outer membrane proteins of Histophilus somni and methods thereof
Gregory et al. Soluble antigens derived from Coxiella burnetii elicit protective immunity in three animal models without inducing hypersensitivity
Ganie et al. Structure, regulation, and host interaction of outer membrane protein U (OmpU) of Vibrio species
CN102648003B (zh) 选择性裂解的全细胞疫苗
Yang et al. Subunit vaccines for Acinetobacter baumannii
Liu et al. Pseudomonas fluorescens: identification of Fur-regulated proteins and evaluation of their contribution to pathogenesis
KR102040665B1 (ko) 약독화된 폐렴구균 균주를 포함하는 약제학적 조성물 및 이의 용도
Guo et al. Evaluation of the immunogenicity and protective ability of a pili subunit, SBP2’, of Streptococcus suis serotype 2
Ma et al. Where are we and how far is there to go in the development of an Acinetobacter vaccine?
KR101795524B1 (ko) 마이코플라즈마 감염증용 백신
Yousief et al. Immunization with the basic membrane protein (BMP) family ABC transporter elicits protection against Enterococcus faecium in a murine infection model
CN115151559A (zh) 葡萄球菌肽和使用方法
CA2547425A1 (en) Orally-administered live bacterial vaccines for plague
Orne Vaccinogenic and Therapeutic Applications of Burkholderia Polysaccharides
JP2003507433A (ja) 敗血症治療において治療に用いる対象としての外膜タンパク質a、ペプチドグリカン関連リポタンパク質およびムレインリポタンパク質
Seco Towards glycoconjugate vaccines against carbapenem-resistance Klebsiella pneumoniae

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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