WO2001049867A1 - M cell directed vaccines - Google Patents

M cell directed vaccines Download PDF

Info

Publication number
WO2001049867A1
WO2001049867A1 PCT/US2001/000426 US0100426W WO0149867A1 WO 2001049867 A1 WO2001049867 A1 WO 2001049867A1 US 0100426 W US0100426 W US 0100426W WO 0149867 A1 WO0149867 A1 WO 0149867A1
Authority
WO
WIPO (PCT)
Prior art keywords
vaccine
cell
protein
cells
nucleic acid
Prior art date
Application number
PCT/US2001/000426
Other languages
French (fr)
Inventor
David W. Pascual
Original Assignee
Research And Development Institute, Inc.
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 Research And Development Institute, Inc. filed Critical Research And Development Institute, Inc.
Priority to US10/169,492 priority Critical patent/US20040033486A1/en
Priority to EP01901811A priority patent/EP1257654A1/en
Priority to AU27672/01A priority patent/AU2767201A/en
Publication of WO2001049867A1 publication Critical patent/WO2001049867A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/255Salmonella (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/12011Reoviridae
    • C12N2720/12022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32611Poliovirus
    • C12N2770/32622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention is in the general field of vaccine development.
  • the present invention is in the general field of vaccine development.
  • infectious agents or cancer for human, livestock, and wildlife More specifically, the
  • present invention provides DNA vaccines directed to follicle-associated epithelium. Even
  • the invention is directed to polycation conjugated M cell ligand (e.g.,
  • enteric adheins enteric adheins
  • DNA complex vaccine compositions and diagnostic and therapeutic uses
  • HSV simplex virus
  • HIV-l Boyer
  • rotavirus Herrmann et al, Jlnfec Dis (1996) 174(Suppl.l):S93-S97 and Chen et al, J Virol
  • DNA immunization has a number of attractive features including ease of
  • adjuvant e.g., cytokines
  • peripheral sites e.g., intradermal or intramuscular sites. While these methods can elicit
  • mucosal immunity i.e., both antibody, particularly IgA, and cellular (cytotoxic T
  • lymphocyte (CTL) immunity induction TTL lymphocyte (CTL) immunity induction
  • Transepithelial transport of antigens and pathogens is the first step in the induction
  • Mucosal inductive tissues are sites in the small intestine or
  • mucosal lymphocytes for the development of mucosal immunity
  • M cells a specialized lymphoid tissue barrier to the underlying lymphoid tissue.
  • epithelium would be advantageous from both investigational and therapeutic standpoints.
  • drugs can be a powerful aid to route compounds to a certain target population.
  • insulin Sobolev et al, JBiol Chem (1998) 273 -.7928-7933) and would include receptor
  • carrier complex can be designed for cell-specific targeting by selecting the appropriate receptor ligand. For example, efficient transfer of DNA to the intestinal epithelial cells by
  • transferrin-polylysine conjugates and M cell lectins have been used to successfully transfect
  • M cells or follicle associated epithelium are not restricted to M cells or follicle associated epithelium and as M cell lectins
  • enteropathic Escherichia coli induces EEC.
  • EEC enteropathic Escherichia coli
  • Reovirus is an enteric pathogen and infects the host following attachment to
  • the protein ⁇ l is a 45
  • This invention exploits receptor mediated endocytosis as a means of introducing
  • M cell ligands chemically coupling M cell ligands to a polymeric chain of basic amino acids (e.g.,
  • DNA can be delivered to appropriate tissue types to obtain enhanced in vivo
  • HIV human immunodeficiency virus
  • Brucella in vivo.
  • the mucosa shows the ability of the protein ⁇ l to mediate efficient gene transfer to the nasal-associated lymphoid tissue (NALT) in vivo.
  • NALT nasal-associated lymphoid tissue
  • the present invention is based, in part, on the observation that a DNA vaccine
  • constructs show improved mucosal IgA antibody responses when compared to DNA applied
  • the present invention is further based on the induced anti-
  • DNA vaccines to specific cells of the follicle associated epithelium, preferably M-cells, for
  • DNA vaccine compositions comprising a polypeptide (or other complexing agent) linked
  • polypeptide-DNA complexes in which the polypeptide is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-N-DNA complexe
  • the DNA structural sequence preferably encodes an immunogenic antigen from an
  • infectious agent but also may encode other immunogens, such as a tumor specific antigen,
  • the present invention provides the ability to
  • infectious agent be it bacterial, fungal, viral, protozoan, parasitic or protective molecule
  • an M cell specific ligand includes an M cell specific ligand, a nucleic acid sequence encoding an immunogen, and a
  • nucleic acid binding moiety Preferably, the nucleic acid will be DNA although RNA
  • the binding moiety preferably is a polypeptide, however, other
  • binding and complexing agents may be utilized so long as they stabilize or protect the
  • nucleic acid and protein components of the vaccine from degradtaion and facilitate their
  • a polypeptide binding moiety preferably comprises a polymeric chain of basic
  • amino acid residues and a contemplated polymeric chain would comprise polylysine.
  • the M cell specific ligand is selected from the group consisting of the
  • protein ⁇ l of a reovirus or is (or is derived from) an adhesin of Salmonella or a polio virus.
  • a polypeptide binding moiety would further comprise an M cell specific
  • ligand and may be expressed as a fusion protein.
  • nucleotide vaccines in which the immunogen to be delivered
  • an immunogen expressed by an infectious agent such as a
  • immunogens are derived from or,
  • tuberculobacillus tuberculobacillus, leprosy bacillus, malaria parasite, diphtheria bacillus, tetanus
  • immunogen and are operably linked to transcription regulatory elements are a preferred
  • the vaccines of the present invention are preferably formulated with a
  • contemplated immunomodulators include cytokines, lymphokines,
  • these vaccines induce a protective
  • contemplated vaccines will tolerize a host vaccinated against appropriate
  • administration through a route selected from the group consisting of oral, nasal, vaginal,
  • nucleotide vaccines as
  • assaying for mucosal immunity comprising the steps of administering the vaccine to an
  • antigen expressing cells In this assay, lysing of antigen expressing cells is indicative of
  • mucosal B cells example mucosal B cells, T cells, lamina intestinal isolates, mtraepithelial isolates, Peyer's
  • a fusion protein comprising a nucleic acid binding moiety and an M cell specific
  • the binding moiety encodes a polymeric chain of basic amino
  • nucleic acids such as polylysine.
  • Associated vectors comprising these nucleic acids, such as
  • Contemplated nucleic acids would be in an operable linkage, and would include both sense and antisense
  • polypeptides comprising a
  • the immunogen also serves as a nucleic acid binding moiety and an M cell specific ligand.
  • the immunogen also serves as a promoter for cleavage of the immunogen.
  • antibodies may be encoded by such fusion proteins. It is also contemplated that antibodies may be
  • test purposes that include an M cell specific ligand and a nucleic acid binding moiety
  • Figure 1 shows that our recombinant reovirus protein ⁇ l can bind murine nasal M
  • FIG. 1 shows sustained mucosal IgA responses against the reporter gene product
  • Figure 3 shows induced cytolytic T cell responses against the reporter gene product
  • FIG. 4 shows the mucosal intestinal IgA response of mice immunized with one of
  • HIN D ⁇ A vaccine constructs presenting gpl60, gpl40(c) or gp 140(s).
  • Figure 5A and 5B show enhanced cytolytic activity (cell-mediated immunity) against
  • adjuvant refers to a substance added to a vaccine to
  • antibody refers to an immunoglobulin molecule that has a
  • Antibodies are classified according to their mode of action as
  • the tenn "antigen" refers to a substance recognized as foreign by the
  • immune system and can be an immunogen.
  • DNA vaccine specifically refers to a therapeutic or
  • prophylactic pharmaceutical formulation that contains a nucleic acid that encodes a protein
  • such a DNA vaccine contains a
  • plasmid nucleic acids may be encoded in plasmid nucleic acids.
  • enteric adhesin refers to a peptide, protein, carbohydrate
  • expression refers to the expression of peptides or proteins
  • the DNA vaccine or associated delivery vector that are encoded by, for example, the DNA vaccine or associated delivery vector.
  • immunogen refers to a process that increases or
  • immunogen refers to a antigen that is capable of eliciting
  • an immunogen usually has a fairly high
  • infectious agent refers to a microorganism (or associated with
  • ligand refers to any molecule that binds to another
  • a soluble molecule such as a hormone or neurofransmitter, that binds to a
  • M cell(s) and “follicle associated epithelium” refer to
  • MALT mucosal associated lymphoreticular tissue
  • GALT associated lymphoid tissue
  • BALT bronchus associated lymphoid tissue
  • NALT lymphoid tissue
  • M cell specific ligand refers to a molecule that selectively
  • enteric adhesin protein ⁇ l of reovirus
  • transferrin and certain other M cell lectins are not
  • the transferrin receptor is not limited to M
  • carbohydrates with said linkages which are not follicle associated epithelium cells (e.g.,
  • the M cell specific ligands are preferred.
  • membrane refers to any membrane surface in a host
  • organism preferably a mammal such as a human being or agriculturally important animal
  • nucleic acid includes DNA and RNA molecules and is
  • nucleic acid sequence and “polynucleotide.”
  • nucleic acid binding moiety refers to compositions and
  • Polybasic chains of amino acids are particularly contemplated for this purpose, as are, for example, synthetic
  • polymeric chain refers to compounds formed by the joining of
  • polymeric chain of basic amino acids i.e., polybasic
  • DNA binding sequence that is rich in basic amino acids, such as lysine, arginine,
  • amino acids are suitable so long as the length of the stretch of basic amino acids is within
  • the polymeric chain of basic amino acids can be a homopolymer of a
  • basic amino acid or it can comprise more than one kind of basic amino acid residue.
  • polypeptide refers to an amino acid sequence including, but not
  • proteins and protein fragments are proteins and protein fragments, naturally derived or synthetically produced.
  • reovirus refers to a genus of the family Reoviridae
  • reovirus 1 is the type species.
  • transcriptional factors refer to a class of proteins that bind
  • tumor specific immunogens refer to immunogens that are
  • tumor cells preferentially expressed by tumor cells, more preferably immunogens that are selectively
  • vaccination refers to the introduction of vaccine into the
  • vaccine generally refers to a therapeutic or prophylactic
  • an immune response preferably a protective immune response.
  • such a component would be a protein encoded by nucleic acids that is expressed
  • This invention provides DNA vaccines, preferably polybasic-M cell ligand
  • conjugate-polynucleotide complexes which, when directly introduced into a vertebrate in
  • the present invention is based, in part, on the ability of
  • Mucosal inductive tissues are sites within the mucosa that support the development of B and
  • T lymphocytes to become stimulated against a specific pathogen or vaccine component
  • compositions and methods To specifically induce such a mucosal immune response, the compositions and
  • methods of the present invention employ ligands formulated to preferentially or specifically
  • M target the specialized epithelium that surrounds mucosal inductive tissues referred to as M
  • a ligand binds M cells to mediate internalization of the dislcosed DNA vaccine.
  • the M cell ligand is an adhesin of a pathogen, preferably an enteric
  • adhesin of a pathogen such as a ⁇ l protein of a reovirus. Additionally, adhesins from
  • Salmonella and poliovirus as well as other infectious agents having the same tissue tropism
  • nucleotide sequences encoding said proteins would be appropriate.
  • nucleotide sequences encoding said proteins would be appropriate.
  • the immunogen may be an enteric adhesin of a pathogen
  • nucleotide 1 such as an intimin of an enteropathic Escherichia coli.
  • nucleotide 1 such as an intimin of an enteropathic Escherichia coli.
  • sequences encoding said intimin protein include but are not limited to polynucleotides
  • immunogen is an enteric adhesin receptor of a pathogen such as an Tir of an enteropathic Escherichia coli.
  • pathogen such as an Tir of an enteropathic Escherichia coli.
  • protem include but are not limited to polynucleotides comprising nucleotide sequences as
  • the immunogen is an
  • enteric adhesin of a pathogen such as an invasin of Salmonella typhimurium, Yersinia pestis
  • nucleotide sequence for example, the nucleotide
  • sequences encoding said invasin proteins include but are not limited to polynucleotides
  • nucleotide sequences as set forth in accession numbers: AF140550; Z48169;
  • a contemplated polynucleotide is a nucleic acid which
  • the polynucleotide is a polydeoxyribonucleic acid comprising immunogen (or
  • polynucleotide comprises
  • ribosomes amenable to translation by the eukaryotic cellular machinery (ribosomes, tRNAs, and other
  • HIN human immunodeficiency virus
  • the animals' immune system is activated to launch a protective immune response.
  • MHC major histocompatibility system
  • polynucleotide generating immune responses to an encoded protein are referred to herein as polynucleotide
  • the described vaccine works by inducing the vaccinated animal to
  • the present formulations encoding various selected antigens may be administered to
  • tuberculosis e.g., BCG
  • malaria e.g., surface antigen
  • MSA-2 Pye et al, Vaccine (1997) 15(9):1017-1023), diptheria (e.g., diptheria toxoid: U.S. Patent No. 4,691,006 ), tetanus (e.g., tetanus toxin: Fairweather et al, Infect Immun (1987)
  • leishmania e.g., Leishmania major promastigotes: Lasri et al, Vet Res.
  • salmonella e.g., covalently bound capsular polysaccharide (Ni) with
  • schistomiasis e.g., major antigen of Schistosoma mansoni (Sm28 GST): Auriault et al,
  • measles e.g., the surface glycoprotein and fusion protein of
  • mumps e.g., mumps
  • H ⁇ hemagglutinin-neuraminidase
  • herpes e.g., HSN-2 surface glycoproteins (gB2 and gD2): Corey et al,
  • influenza e.g., immunodommant peptide from hemagglutinin: Novak
  • the present invention further provides recombinant DNA molecules (rDNAs) that
  • the vaccines are produced using conventional eukaryotic cells.
  • a rDNA molecule is a
  • a vector contemplated by the present invention is at least capable of
  • Expression control elements that are used for regulating the expression of an
  • operably linked protein encoding sequence include, but are not
  • inducible promoters are limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements.
  • the inducible promoter is readily controlled, such as being
  • the vector containing a coding nucleic acid molecule will be any suitable nucleic acid molecule.
  • prokaryotic replicon i.e., a DNA sequence having the ability to direct autonomous
  • prokaryotic host cell such as a bacterial host cell, transformed therewith.
  • prokaryotic host cell such as a bacterial host cell
  • vectors that include a prokaryotic replicon may also be used.
  • a gene whose expression confers a detectable marker such as a drug resistance include a gene whose expression confers a detectable marker such as a drug resistance.
  • Typical bacterial drug resistance genes are those that confer resistance to ampicillin or tetracycline.
  • Vectors that include a prokaryotic replicon can further include a prokaryotic or
  • a bacteriophage promoter capable of directing the expression (transcription and translation) of the coding gene sequences in a bacterial host cell, such as E. coli.
  • a promoter is an expression
  • control element formed by a DNA sequence that permits binding of RNA polymerase and
  • Promoter sequences compatible with bacterial hosts are typically
  • vector plasmids Typical of such vector plasmids are pUC8, pUC9, pBR322
  • Expression vectors compatible with eukaryotic cells preferably those compatible with
  • vertebrate cells can also be used to form a rDNA molecules that contains a coding sequence.
  • Eukaryotic cell expression vectors are well known in the art and are available from several sources.
  • Typical of such vectors are pSVL and pKSV-10
  • Eukaryotic cell expression vectors used to construct the DNA vaccine molecules of the
  • present invention may further include a selectable marker that is effective in an eukaryotic cell,
  • a preferred drug resistance marker is the gene
  • neomycin phosphotransferase neomycin phosphotransferase
  • the marker can be present on a separate plasmid, and the two vectors are introduced by co- transfection of the host cell, and selected by culturing in the appropriate drag for the selectable
  • the M cell ligand-polybasic conjugates according to the invention may be produced
  • coupling may be carried out by means of various techniques Icnown to persons skilled in
  • nucleic acid molecule that encodes an M cell ligand protein of the
  • nucleic acid molecule is then preferably placed in operable linkage
  • the expression unit is used to transform a suitable host and the
  • transformed host is cultured under conditions that allow the production of the recombinant
  • the recombinant protein is isolated from the medium or from the cells;
  • coding sequences may be obtained from genomic fragments and used directly in appropriate
  • sequences, expression vectors, and transformation methods are dependent on the type of host
  • Suitable restriction sites can, if
  • the polybasic components may vary in terms of their size and amino acid sequence.
  • the conjugate to be modified by increasing the ability to bind to the receptor, by suitable
  • express plasmids can be obtained, of which the plasmid containing the desired sequence can be
  • the nucleic acids which are to be transported into the cell may be DNAs or RNAs, with
  • nucleic acids may be modified, provided that
  • ligand-polybasic conjugates can be efficiently absorbed into living cells and internalized.
  • conjugates or complexes according to the invention are not apparently harmful to cell
  • the ratio of nucleic acid to conjugate can vary within a wide range, and it is not
  • nucleic acid which is to be transported the size of the polybasic component and the number and
  • nucleic acid which is favorable to the particular appUcation. This ratio can first of all be adjusted
  • negative charges of the nucleic acid are not an obstacle to transportation into the cell.
  • precipitation is to mix the two components together first of all at a high (about 1 molar)
  • complex according to the invention is an immunogen structural gene encoded by the nucleic acid
  • the invention further relates to a process for introducing nucleic acid or acids into human
  • Antibodies against M cell ligand-polybasic moiety protein conjugate or complex may be
  • linking reagents such as those supplied by Pierce Chemical Co., Rockford, IL,
  • the hapten peptides can be extended at
  • hnmortaUzed cell lines which secrete the desired monoclonal antibodies may be prepared using
  • lymphocytes or spleen cells as is generally known.
  • desired antibodies are screened by immunoassay in which the antigen is the peptide hapten,
  • the cells can be cultured either in vitro or by production in ascites fluid.
  • the desired monoclonal antibodies are then recovered from the culture supernatant or
  • fragments is often preferable, especially in a therapeutic context, as these fragments are generally
  • the antibodies or fragments may also be produced, using current technology, by
  • antibodies specific for the M cell ligand polybasic moiety conjugate can also be produced in the context of chimeras with multiple species origin.
  • antibodies specific for the M cell ligand polybasic moiety conjugate can also be produced in the context of chimeras with multiple species origin.
  • antibodies specific for the M cell ligand polybasic moiety conjugate can also be produced in the context of chimeras with multiple species origin.
  • antibodies specific for the M cell ligand polybasic moiety conjugate can be produced in the context of chimeras with multiple species origin.
  • carrier vectors or specific gene sequences may be used successfully.
  • Various methods are
  • encoding a protein can be produced which alter the amino acid sequence of the encoded protein.
  • amino acids are tolerated without affecting protein function. Similar amino acids can be those
  • isoleucine and valine are both pairs of similar amino acids. Similarity between amino acid pairs
  • the altered expressed protein may have an altered amino acid sequence, yet still eUcits immune responses which react with the antigen protein, and
  • fragments should encode a protein or peptide which eUcits antibodies that crossreact with the
  • immunogenic protein and are considered to be functional equivalents.
  • the amount of expressible DNA or transcribed RNA to be introduced into a vaccine is the amount of expressible DNA or transcribed RNA to be introduced into a vaccine
  • transcriptional and translational promoters used as well as subject size, e.g., human versus bison
  • immune response may depend on the level of protein expression and on the immunogenicity of
  • an effective dose ranges of about 1 ng to 5 mg, 100 ng to
  • mterleukin-12 protein or other
  • cytokines e.g. GM-CSF
  • the polynucleotide may be associated with adjuvants or other agents which affect the
  • the formulation it is desirable for the formulation to be in a
  • physiologically acceptable solution such as, but not limited to, sterile saline or sterile buffered saUne.
  • active immunogenic ingredients can be mixed with excipients or carriers which are
  • the DNA vaccine complexes may contain minor amounts of
  • auxiUary substances such as wetting or emulsifying agents, pH buffering agents, and/or
  • adjuvants which enhance the effectiveness of the vaccine.
  • adjuvants wliich may be any adjuvant.
  • CGP 11637 N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP);
  • CGP 19835A referred to as MTP-PE
  • RIBI which contains three components extracted from bacteria, monophosphoryl Upid A,
  • the effectiveness of an adjuvant may be determined by measuring the amount of
  • vaccines wliich are also comprised of the various adjuvants. Such additional formulations and
  • the DNA vaccines of the present invention may be formulated into compositions as
  • compositions include but are not limited to the acid
  • addition salts formed with free amino groups of the peptide which are formed with inorganic acids, e.g., hydrochloric acid or phosphoric acids; and organic acids, e.g., acetic, oxalic, tartaric,
  • Salts formed with the free carboxyl groups may also be derived from inorganic
  • bases e.g., sodium, potassium, ammonium, calcium, or ferric hydroxides, and organic bases,
  • isopropylarnine trimethylamine
  • 2-ethylamino-ethanol histidine
  • procaine e.g., isopropylarnine, trimethylamine, 2-ethylamino-ethanol, histidine, andprocaine.
  • the M cell ligand-polybasic moiety (or conjugate)-polynucleotide compositions are provided.
  • Precise amounts of the active ingredient required to be administered may depend on the
  • the DNA vaccines of the present invention may be given in a single dose or multiple
  • a multiple dose schedule is one in which a primary course of vaccination may
  • intervals as required to maintain and or reinforce the immune response e.g., at 1 to 4 months for
  • the vaccines of the present invention are useful for administration to domesticated or
  • Vaccines of the present invention may be used to treat agricultural animals, as well as humans.
  • Vaccines of the present invention may be used to treat livestock, as well as humans.
  • MBP maltose-binding protein
  • fusion protein was purified by affinity chromatography using amylose resin according to
  • recombinant protein ⁇ l referred to as recombinant protein ⁇ l .
  • Example 2 Preparation of recombinant fusion protein ⁇ l-polylysine-DNA complex
  • the recombinant protein ⁇ l was covalently linked to poly-L-lysine (PL) according
  • SPDP succinimidyl 3-(2-pyridyldithio)propionate
  • modified protein ⁇ 1 was then mixed with the 20 mg of mercaptopropionate-modified PL under
  • MBP-PL conjugates were similarly generated.
  • conjugate- DNA complex the protein ⁇ l-PL conjugate in 125 ⁇ l of HS was added dropwise into an equal
  • mice L cells were incubated with mouse L cells (CCL-1, American Type Culture Collection, Manassas, VA),
  • RFL-6 fibroblast cells (CCL-192, ATCC), and Caco-2 cells (HTB-37, ATCC) and binding was
  • biotinylated monoclonal anti-reo virus protein ⁇ l antibody (HB-167,
  • the mouse L cells, RFL-6 cells, and Caco-2 cells were used for targeting gene
  • the mouse L cells have been used as the in vitro model for
  • DMEM lninimiim essential medium
  • conjugate-DNA complexes were removed, and cells were incubated with complete media for
  • DNA complexes containing 8 ⁇ g ⁇ l-PL and pCMV ⁇ -gal (Life Technologies), with or without
  • chloroquine were added and incubated for 24 hours. The cells were then incubated with fresh
  • the Luc gene was used as a reporter gene to assay protein ⁇ l-PL conjugate-
  • pCMVLuc was
  • FDG fluorescein-mono- ⁇ -D-galactopyranoside
  • NALT were incubated with excess unmodified protein ⁇ l in order to inhibit biotinylated protein
  • Tissue Tek @ Cryomold (Miles Inc., Elkhard, IN) with their ventral surfaces oriented toward the bottom of the mold. The palates were then frozen in Tissue
  • Example 7 In vivo analysis of intranasal immunization with ⁇ l conjugates
  • mice induced mucosal IgA responses in mice.
  • Data depicts the mean endpoint titers ( ⁇ SE) for mice
  • mice/group Significant differences between protein ⁇ l-PL-pCMVLuc and pCMVLuc only
  • mice received three i.n.
  • mice The mean endpoint liters ( ⁇ SE) for mice
  • mice were immunized with one of three designated HJV D ⁇ A vaccine
  • mice/group received three intranasal immunizations either of naked D ⁇ A or of the identified M
  • the D ⁇ Avaccine formulation improved mucosal IgA responses when compared to
  • mice received a formulated vaccine, naked D ⁇ A version, protein
  • mice i.n. -immunized with the formulated vaccine as opposed to mice immunized with the naked D ⁇ A alone.
  • Pulmonary lymph nodes (LRL ⁇ ) and splenocytes from immunized mice were restimulated in vitro with cells expressing gpl20 or beta-galactosidase (neg. control), and were subsequently examined for cytolytic activity. The observed killing was specific since negative

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This invention provides a vaccine that can direct gene transfer to follicle associated epithelium or M cells to induce mucosal immunity using M cell ligands for receptor-mediated endocytosis. Also provided are polynucleotides sequences encoding M cell ligand-polybasic component fusion proteins, host cells, and methods of producing such proteins recombinantly and chemically. Further, methods are described for immunizing animal and human subjects against bacterial, viral, parasitic, fungal infectious agents or cancer and methods for assaying mucosal immunity using this vaccine.

Description

M CELL DIRECTED VACCINES
Technical Field
The present invention is in the general field of vaccine development. The present
invention provides methods and compositions useful for, among other purposes, the
identification, diagnosis, prevention and treatment of bacterial, viral, parasitic, fungal
infectious agents or cancer for human, livestock, and wildlife. More specifically, the
present invention provides DNA vaccines directed to follicle-associated epithelium. Even
more specifically, the invention is directed to polycation conjugated M cell ligand (e.g.,
enteric adheins)-DNA complex vaccine compositions and diagnostic and therapeutic uses
thereof.
Background of the Invention
Aspects of this invention are discussed in Wu et al, Gene Therapy (2000)
7(l):61-69, herein incorporated by reference in its entirety. All publications and patent
applications mentioned or identified in this specification are incorporated by reference to the
same extent as if each individual publication or patent application was specifically and
individually indicated to be incorporated by reference.
Recent studies have shown the utility of DNA vaccination for inducing protective
immunity in experimental animals exposed to influenza (Fynan et al, Proc Natl Acad Sci
USA (1993) 90: 11478-11482 and Robinson et al, Vaccine (1993) 11 :957-960), herpes
simplex virus (HSV) (Gillichan et al, JInfDis (1998) 177:1155-1161), HIV-l (Boyer), rotavirus (Herrmann et al, Jlnfec Dis (1996) 174(Suppl.l):S93-S97 and Chen et al, J Virol
(1998) 72:5757-5761), and Borrelia burgdorgeri infections (Simon et al, J Immunol (1996)
26:2831-2840). DNA immunization has a number of attractive features including ease of
preparation for encoding desired protective immunogens, co-expression of immunogens, co-
expression of adjuvant (e.g., cytokines), no requirement for large-scale protein purifications,
and ease of delivery. However, conventional DNA vaccine technology immunizes the host
at peripheral sites, e.g., intradermal or intramuscular sites. While these methods can elicit
systemic cell-mediated and antibody-dependent responses, most infectious agents infect via
a mucosal surface, and such DNA immunizations at peripheral sites do not result in optimal
mucosal immunity (i.e., both antibody, particularly IgA, and cellular (cytotoxic T
lymphocyte (CTL) immunity induction).
This lack of mucosal immunity induction has prompted attempts to deliver DNA
vaccines to mucosal surfaces. For example, successful induction of mucosal immunity has
been accomplished using DNA vaccines by intraoral jet delivery (Chen et al, Vaccine
(1999) 17(23-24):3171-3176); co-administration of a DNA vaccine with a polymer by
intranasal injection (Hamajima et al, Clinical Immunol Immunopathol (1998) 88(2):205-
210); co-administration of a DNA vaccine with IL-12 (Okada et al, J Immunol (1997)
159(7):3638-3647); intravaginal administration of DNA vaccines (Wang et al, Vaccine
(1997) 15(8):821-825); oral delivery of micro-encapsulated DNA vaccines (Jones et al, Dev
Biol Stand (1998) 92:149-155); and parenteral and mucosal injection of DNA vaccines
(Shroff et al, Vaccine (1999) 18(3-4):222-230). However, these methods generally lack mucosal surface selectivity. Nevertheless, they illustrate the desire to observe mucosal
immunity as the end-point in determining the efficacy of these vaccines.
Transepithelial transport of antigens and pathogens is the first step in the induction
of a mucosal immune response. Mucosal inductive tissues are sites in the small intestine or
in the nasal passages where vaccine antigens are taken to be processed and presented to
mucosal lymphocytes for the development of mucosal immunity (Frey et al, Behring Inst
Mitt (1997) 98:376-389). In the intestine, the delivery of antigen across the epithelial
barrier to the underlying lymphoid tissue is accomplished by M cells, a specialized
epithelial cell type that occurs only in the lymphoid follicle-associated epithelium (Frey et
al, 1997). Further, such follicle-associated epithelium is found in the nasal lymphoid
tissues (believed to be sites of induction of mucosal immune responses to airborne antigens;
Giannasca et al, Infect Immun (1997) 65(10):4288-4289). Selective and efficient transport
of antigen by M cells is considered an essential requirement for effective mucosal vaccines.
Thus, targeting of M cells by taking advantage of their capacity to endocytose particles,
including those particles comprising gene transfer vehicles and DNA vaccines, has
generated great interest as selective transfer of genes across the follicle-associated
epithelium would be advantageous from both investigational and therapeutic standpoints.
Although viruses can be efficient gene transfer vehicles, progress has been made
toward developing non- viral delivery systems. Coupling of a specific ligand to vaccines or
drugs can be a powerful aid to route compounds to a certain target population. One of the
most promising means is by exploiting receptor-mediated endocytosis pathways using selective ligands. In this method, DNA-ligand complexes are internalized by targeted cells
when the ligand binds to its respective cell-surface receptor. Such receptor-mediated gene
transfer has been accomplished using a variety of receptors by conjugating DNA to their
cognate ligands such as asialo-orosomucoid Wu et al, JBiol Chem (1989) 264:16985-
16987 and Wu et al, JBiol Chem (1994) 269:11542-11546), transferrin (Lozier et al,
Human Gene Ther (1994) 5:313-322; Wagner et al, Proc Natl Acad Sci USA (1990)
87:3410-3414; and Wagner et al, Proc Natl Acad Sci USA (1992) 89:6099-6103), lectins
(Batra et al, Gene Ther (1994) 1:255-260), folate (Leamon et al, Biochem J
(1993)291:855-860), lung surfactant protein (Ross et al, Human Gene Ther (1995) 6:31-
40), insulin (Sobolev et al, JBiol Chem (1998) 273 -.7928-7933) and would include receptor
specific monoclonal antibodies (Chen et al, FEBSLett (1994) 338:167-169 and Kang et al,
J Pharmacol Exp Therapeut (1994) 269:344-350).
Receptor-mediated gene transfer has some advantages over the other methods of in
vivo gene transfer. Compared to attenuated viral vectors, it shares tissue-specificity, but
receptor-mediated gene transfer minimizes the use of viral gene elements, obviating the
concerns regarding genomic integration. Further, it lessens concerns with the
proinflammatory properties often associated with viral vectors (Simon et al, Human Gene
Ther (1993) 4:771-780; Yang et al, J Virol (1996) 70:7209-7212; and van Ginkel et al, J
Immunol (1997) 159:685-693). The DNA-ligand complex is believed to be internalized by
receptor-dependent endocytosis rendering transfection to be minimally toxic. The conjugate
carrier complex can be designed for cell-specific targeting by selecting the appropriate receptor ligand. For example, efficient transfer of DNA to the intestinal epithelial cells by
transferrin-polylysine conjugates and M cell lectins have been used to successfully transfect
gastrointestinal cells in vitro (Batra et al, Cancer Gene Ther (1994) 1(3): 185-192 and
Curiel et al, Am JRespir Cell Mol Biol (1992) 6(3):247-252). However, as transferrin
receptors are not restricted to M cells or follicle associated epithelium and as M cell lectins
can potentially bind to any α-linked galactose (Giannasca et al, 1997), the use of these
systems in vivo is limited.
The surface properties of many enteric pathogens are important in the establishment
of the pathogen in the host. For example, enteropathic Escherichia coli (EPEC) induce
characteristic attaching and effacing (A E) lesions on epithelial cells of Peyer's patches
(Hartland et al, Mol Microbiol (1999) 32(1):151-158). This event is mediated, in part, by
binding of the bacterial outer membrane protein, intimin, to a second EPEC protein, Tir
(translocated intimin receptor), which is exported by the bacteria and integrated into the host
cell plasma membrane. Both of these protein have been shown to bind to host cells in vitro
(Hartland et al, 1999 and DeVinney et al, Cell Mol Life Sci (1999) 55(6-7):961-976).
Reovirus is an enteric pathogen and infects the host following attachment to
intestinal Peyer's patch M cells (Lee et al, Virology (1981) 108:156-63 and Mah et al, J
Virol (1990) 179:95-103). Thus, as with other enteric pathogens, reovirus exploits M cells
as a means to gain entry into the host. Mediating reovirus attachment is the adhesin, σl,
which is expressed as a viral coat protem (Lee et al, 1981). The protein σl is a 45
kilodalton protein that polymerizes via its N-terminus (Mah et al., 1990) to form a tetramer when isolated from reovirus-infected cells or purified as a recombinant protein from E. coli
(Bassel-Assel-Duby et al, J Virol (1987) 61:1834-1841). In vitro analysis has
demonstrated that neutral liposomes comprising σl protein can be taken up by rat Peyer's
patches (Rubas et al., J Microencapsul (1990) 7(3):385-395). Thus, enteric pathogen
adhesins make more effective targeting ligands than either transferrin or M cell lectins
(Batra et al, 1994, Curiel et al, 1992 and Giannasca et al, 1997).
This invention exploits receptor mediated endocytosis as a means of introducing
DNA into cells using M cell ligands for specific targeting of DNA vaccines to follicle
associated epithelium of nasal or gastrointestinal origin. We have discovered that, by
chemically coupling M cell ligands to a polymeric chain of basic amino acids (e.g.,
polylysine), DNA can be delivered to appropriate tissue types to obtain enhanced in vivo
mucosal IgA antibody and T cell responses against an encoded antigen.
Further, to demonstrate the efficacy of the present vaccine design, we have applied
this concept to reporter gene products, β-galactosidase and luciferase, as well as vaccine
antigens derived from human immunodeficiency virus (HIV) and Brucella in vivo. Using
these systems, enhanced mucosal IgA antibody responses can be demonstrated between
animals vaccinated with DNA only (that is, DNA not included in our formulation) and those
vaccinated with conjugated DNA complexes.
Our presently formulated DNA vaccine induces improved mucosal IgA antibody
responses and promotes sustained CTL responses, demonstrating efficacious vaccination via
the mucosa. Further, as the present invention shows the ability of the protein σl to mediate efficient gene transfer to the nasal-associated lymphoid tissue (NALT) in vivo, we have
demonstrated that systemic and mucosal immunity to intranasally delivered DNA as part of
a M cell ligand complex is achievable.
Summary of the Invention
The present invention is based, in part, on the observation that a DNA vaccine
protected from the mucosal environment can be effectively used to vaccinate a host by
targeting the mucosa. Data described herein shows that appropriately formulated DNA
constructs show improved mucosal IgA antibody responses when compared to DNA applied
directly to a mucosal surface. The present invention is further based on the induced anti-
vaccine antibody and cellular immune responses produced by vaccinated mice, cattle, and
bison. Based on these observations, the present invention provides compositions and
methods for use in a variety of animals, particularly humans, livestock, and wildlife.
It is therefore an object of this invention to provide a method for inducing mucosal
immunity using receptor mediated endocytosis pathways to deliver nucleotide, particularly
DNA, vaccines to specific cells of the follicle associated epithelium, preferably M-cells, for
example, of nasal and gastrointestinal origin. It is also an object of this invention to provide
DNA vaccine compositions comprising a polypeptide (or other complexing agent) linked
electrostatically to (or otherwise associated or complexed with) a DNA structural sequence.
Particularly contemplated are polypeptide-DNA complexes, in which the polypeptide is
comprised of a polymeric chain of basic amino acid residues and an M cell specific ligand. The DNA structural sequence preferably encodes an immunogenic antigen from an
infectious agent, but also may encode other immunogens, such as a tumor specific antigen,
against which the induction of an immune response is desired, but also including antigens
against which a host might be tolerized. The present invention provides the ability to
produce a previously unknown protein ~ and to elicit an immune response against such
proteins ~ using the cloned nucleic acid molecules derived, for example, from any given
infectious agent be it bacterial, fungal, viral, protozoan, parasitic or protective molecule
against cancer.
Consistent with the foregoing, a preferred embodiment of the present invention
includes an M cell specific ligand, a nucleic acid sequence encoding an immunogen, and a
nucleic acid binding moiety. Preferably, the nucleic acid will be DNA although RNA
vaccines are contemplated.
In such vaccines, the binding moiety preferably is a polypeptide, however, other
binding and complexing agents may be utilized so long as they stabilize or protect the
nucleic acid and protein components of the vaccine from degradtaion and facilitate their
delivery, by various routes of administration, to the target mucosal tissues. Thus, for
example, a polypeptide binding moiety preferably comprises a polymeric chain of basic
amino acid residues and a contemplated polymeric chain would comprise polylysine.
In general, the M cell specific ligand is selected from the group consisting of the
protein σl of a reovirus, or is (or is derived from) an adhesin of Salmonella or a polio virus.
M cell tropic fragments of the foregoing also are contemplated.In a preferred embodiment of the invention, a polypeptide binding moiety would further comprise an M cell specific
ligand and may be expressed as a fusion protein.
Also contemplated are nucleotide vaccines in which the immunogen to be delivered
to the target mucosal tissue is an immunogen expressed by an infectious agent such as a
microorganism or is a tumor specific antigen. Preferred immunogens are derived from or,
like an expressed toxin, are associated with a bacterium, protozoan, parasite, virus, fungus,
prion, tuberculobacillus, leprosy bacillus, malaria parasite, diphtheria bacillus, tetanus
bacillus, Leishmania, Salmonella, Schistoma, measles virus, mumps virus, herpes virus,
HIN, cancer and influenza virus. Plasmid vectors in which DΝA sequences encode such an
immunogen and are operably linked to transcription regulatory elements are a preferred
embodiment of the present invention.
The vaccines of the present invention are preferably formulated with a
pharmaceutically acceptable excipient or an adjuvant such as an immunomodulator.
Examples of contemplated immunomodulators include cytokines, lymphokines,
interleukins, interferons and growth factors. Preferably, these vaccines induce a protective
iinmune response in a host vaccinated against the immunogen. In other embodiments of the
invention, contemplated vaccines will tolerize a host vaccinated against appropriate
immunogens.
Vaccines formulated in unit dosage form, and vaccines packaged with instructions
for the use of the vaccine to induce an immune response against said immunogen or disease
with which said immunogen is associated are preferred. Therapeutic as well as prophylactic vaccines also are contemplated. Moreover, preferred vaccines are formulated for
administration through a route selected from the group consisting of oral, nasal, vaginal,
rectal and urethral routes of administration.
Another preferred embodiment of the present invention provides a method for
immunizing a host against an immunogen by administering the nucleotide vaccines as
described above. In addition, other embodiments of the invention provide a method for
assaying for mucosal immunity comprising the steps of administering the vaccine to an
animal which is free of infection of the infectious agent whose antigen is to be tested;
isolating cells from the animal; and co-incubating said isolated cells with heterologous
antigen expressing cells. In this assay, lysing of antigen expressing cells is indicative of
mucosal immunity in the vaccinated animal.
Use of the foregoing assay method is contemplated with isolated cells including, for
example mucosal B cells, T cells, lamina propria isolates, mtraepithelial isolates, Peyer's
patches cells, lymph nodes, nasal passages, NALT, adenoids and vaginal epithelium. The
the additional step of evaluating the animal's cytokine profile also is contemplated.
A related embodiment of the present invention provides an isolated nucleic acid
encoding a fusion protein comprising a nucleic acid binding moiety and an M cell specific
ligand. In such nucleic acids, the binding moiety encodes a polymeric chain of basic amino
acid residues such as polylysine. Associated vectors comprising these nucleic acids, such as
expression vectors, are expressly contemplated. Moreover, the polypeptide expression
products of such vectors also may be used as immunogens in vaccines. Contemplated nucleic acids would be in an operable linkage, and would include both sense and antisense
orientations relative to transcriptional elements comprising the vector. Host cells
comprising or transformed with such vectors are also contemplated.
Another embodiment of the invention includes methods of expressing fusion
proteins from such cells. Particularly contemplated are isolated polypeptides comprising a
nucleic acid binding moiety and an M cell specific ligand. Optionally, the immunogen also
may be encoded by such fusion proteins. It is also contemplated that antibodies may be
generated that bind selectively or preferentially to such polypeptides, as opposed to the
immunogen or to the M cell specific ligand or nucleic acid binding moiety themselves.
Yet another embodiment of the present invention relates to various kits for assay and
other test purposes that include an M cell specific ligand and a nucleic acid binding moiety
as well as the other constructs and components described above.
Other objects, features and advantages of the present invention will become apparent
from the following detailed description. It should be understood, however, that the detailed
description and specific examples, while indicating preferred embodiments of the invention
are given by way of illustration only, since various changes and modifications within the
spirit and scope of the invention will become apparent to those skilled in the art from this
detailed description.
Brief Description of the Drawings Figure 1 shows that our recombinant reovirus protein σl can bind murine nasal M
cells.
Figure 2 shows sustained mucosal IgA responses against the reporter gene product,
luciferase.
Figure 3 shows induced cytolytic T cell responses against the reporter gene product,
β-galactosidase.
Figure 4 shows the mucosal intestinal IgA response of mice immunized with one of
three designated HIN DΝA vaccine constructs presenting gpl60, gpl40(c) or gp 140(s).
Figure 5A and 5B show enhanced cytolytic activity (cell-mediated immunity) against
target cells expressing HIN gpl20 from biopsies from mice immunized mtranasally with an
M cell-formulated HIN DΝA vaccine.
Detailed Description of the Invention
Definitions
As used herein, the term "adjuvant" refers to a substance added to a vaccine to
improve the immune response.
As used herein, the term "antibody" refers to an immunoglobulin molecule that has a
specific amino acid sequence by virtue of which it interacts only with the antigen that
induced its synthesis in cells of the lymphoid series (especially plasma cells) or with antigen
closely related to it. Antibodies are classified according to their mode of action as
agglutinins, bacteriolysins, haemolysins, opsonins, precipitins, etc. As used herein, the tenn "antigen" refers to a substance recognized as foreign by the
immune system and can be an immunogen.
As used herein, the term "DNA vaccine" specifically refers to a therapeutic or
prophylactic pharmaceutical formulation that contains a nucleic acid that encodes a protein
or peptide against which a vaccinated host is induced to mount an immune response,
preferably a protective immune response. Preferably, such a DNA vaccine contains a
complete eukaryotic expression system encoding the molecular machinery for the
expression of such a protein or peptide subunit vaccine. For example, such a DNA vaccine
may be encoded in plasmid nucleic acids.
As used herein, the term "enteric adhesin" refers to a peptide, protein, carbohydrate
or other class of compound that allows for or facilitates pathogen attachment to M cells as a
means to gain entry to the host. For example, a reovirus σl protein having a molecular
weight of 47 kDa is an enteric adhesin (Nagata et al, Nucleic Acids Res (1984)
12(22):8699-710).
As used herein, the term "expression" refers to the expression of peptides or proteins
that are encoded by, for example, the DNA vaccine or associated delivery vector. After
expression of such a peptide or protein by, for example, an M cell to which a DNA vaccine
has been targeted, such expression by the M cell would lead to the induction of an immune
response by a vaccinated host against that encoded immunogen. As used herein, the term "immunization" refers to a process that increases or
enhances an organism's reaction to antigen and therefore improves its ability to resist or
overcome infection.
As used herein, the term "immunogen" refers to a antigen that is capable of eliciting
(inducing) an immune response. For example, an immunogen usually has a fairly high
molecular weight (usually greater than 10,000 daltons). Thus, for example, a variety of
macromolecules such as peptides, proteins, lipoproteins, polysaccharides, some nucleic
acids, and certain of the teichoic acids, can act as immunogens.
As used herein, the term "infectious agent" refers to a microorganism (or associated
substance such as a toxin) that affects or communicates disease through invasion and
multiplication of said substance in body tissues, which may be clinically unapparent or
result in local cellular injury due to competitive metabolism, toxins, intracellular replication
or antigen antibody response.
As used herein, the term "ligand" refers to any molecule that binds to another; in
normal usage a soluble molecule such as a hormone or neurofransmitter, that binds to a
receptor.
As used herein, the term "M cell(s)" and "follicle associated epithelium" refer to
specialized mucosal cells overlying mucosal associated lymphoreticular tissue (MALT), gut
associated lymphoid tissue (GALT), bronchus associated lymphoid tissue (BALT) and nasal
associated lymphoid tissue (NALT) and any other corresponding mucosal cells that are
known to or become known to persons skilled in the art. As used herein, the term "M cell specific ligand" refers to a molecule that selectively
binds to a receptor available on the surface of follicle associated epithelial cell
subpopulations, and an M cell specific physiologic effect accompanies that binding (e.g.,
uptake of pathogen). For example, the enteric adhesin, protein σl of reovirus, is an M cell
specific ligand. By way of distinction, transferrin and certain other M cell lectins are not
considered M cell specific ligands because: 1) the transferrin receptor is not limited to M
cells (e.g., neurons express these receptors: Taylor et al, JComp Physiol (1991)
1 1 (5):521 -524) and would not select for follicle associated epithelium subpopulations; and
2) because certain M cell lectins select for α-linked galactose, and many cells possess
carbohydrates with said linkages which are not follicle associated epithelium cells (e.g.,
hepatocytes: Oda et al, JBiol Chem (1988) 263(25): 12576-12583). While M cell ligands
(rather than M cell specfic ligands) are contemplated for the compositions and methods of
certain embodiments of the present invention, the M cell specific ligands are preferred.
As used herein, the term "mucosal" refers to any membrane surface in a host
organism, preferably a mammal such as a human being or agriculturally important animal,
that is covered by mucous.
As used herein, the term "nucleic acid" includes DNA and RNA molecules and is
used synonymously with the terms "nucleic acid sequence" and "polynucleotide."
As used herein, the term "nucleic acid binding moiety" refers to compositions and
substances that are capable of binding to or complexing with DNA and serving as a vehicle
to deliver the compositions of the present invention to their target M cells. Polybasic chains of amino acids are particularly contemplated for this purpose, as are, for example, synthetic
compounds known to persons skilled in the art that have appropriate ionic charges to form
complexes with DNA.
As used herein, "polymeric chain" refers to compounds formed by the joining of
smaller, usually repeating, units linked by covalent bonds.
As used herein, the term "polymeric chain of basic amino acids" (i.e., polybasic)
refers to a DNA binding sequence that is rich in basic amino acids, such as lysine, arginine,
and ornithine, that is typically about ten to 300 residues long. D-isomers of these basic
amino acids are suitable so long as the length of the stretch of basic amino acids is within
the prescribed length. The polymeric chain of basic amino acids can be a homopolymer of a
basic amino acid or it can comprise more than one kind of basic amino acid residue.
As used herein, "polypeptide" refers to an amino acid sequence including, but not
limited to, proteins and protein fragments, naturally derived or synthetically produced.
As used herein, the term "reovirus" refers to a genus of the family Reoviridae
infecting vertebrates only. Transmission is horizontal and infected species include humans,
birds, cattle, monlceys, sheep, swine, and bats. Reovirus 1, reovirus 2, and reovirus 3 infect
mammals, and reovirus 1 is the type species.
As used herein, the term "transcriptional factors" refer to a class of proteins that bind
to a promoter or to a nearby sequence of DNA to facilitate or prevent transcription
initiation. As used herein, "tumor specific immunogens" refer to immunogens that are
preferentially expressed by tumor cells, more preferably immunogens that are selectively
expressed by tumor cells.
As used herein, the term "vaccination" refers to the introduction of vaccine into the
body of an animal (or host) for the purpose of inducing immunity.
As used herein, the term "vaccine" generally refers to a therapeutic or prophylactic
pharmaceutical formulation that contains a component against which a vaccinated host is
induced to mount an immune response, preferably a protective immune response. For
example, such a component would be a protein encoded by nucleic acids that is expressed
by a vaccinated host to form an expressed protein or peptide subunit vaccine.
General
This invention provides DNA vaccines, preferably polybasic-M cell ligand
conjugate-polynucleotide complexes which, when directly introduced into a vertebrate in
vivo, including mammals such as humans, induces the expression of encoded proteins
within the animal. Prior to the present invention, the art had taught that DNA vaccines
represent an efficient method of inducing immunity against a given pathogen if the
responsible gene for eliciting protection is identified. As described below, the present
inventors have found that the described DNA vaccine formulations improve the targeting of
DNA to mucosal inductive tissues. The present invention is based, in part, on the ability of
such vaccine formulations to selectively and preferentially target mucosal inductive tissues. Mucosal inductive tissues are sites within the mucosa that support the development of B and
T lymphocytes to become stimulated against a specific pathogen or vaccine component or
subunit. If the antigen or vaccine can reach this site, there is a strong likelihood that a
mucosal immune response will be induced.
To specifically induce such a mucosal immune response, the compositions and
methods of the present invention employ ligands formulated to preferentially or specifically
target the specialized epithelium that surrounds mucosal inductive tissues referred to as M
cells. Thus, a ligand binds M cells to mediate internalization of the dislcosed DNA vaccine.
In one embodiment, the M cell ligand is an adhesin of a pathogen, preferably an enteric
adhesin of a pathogen, such as a σl protein of a reovirus. Additionally, adhesins from
Salmonella and poliovirus, as well as other infectious agents having the same tissue tropism
would be appropriate. For example, the nucleotide sequences encoding said proteins
include but are not limited to polynucleotides comprising nucleotide sequences as set forth
in GenBank accession numbers: J02325; M10491; AF059719; AF059718; AF059717;
AF059716; U74293; and U74292.
In another embodiment, the immunogen may be an enteric adhesin of a pathogen
such as an intimin of an enteropathic Escherichia coli. For example, the nucleotide
sequences encoding said intimin protein include but are not limited to polynucleotides
comprising nucleotide sequences as set forth in GenBank accession numbers: U38618;
AJ223063; Y13111; Y13112; AF043226; and U62657. In another embodiment, the
immunogen is an enteric adhesin receptor of a pathogen such as an Tir of an enteropathic Escherichia coli. For example, the nucleotide sequences encoding the intimin receptor
protem include but are not limited to polynucleotides comprising nucleotide sequences as
set forth in accession number: AF113597. In another embodiment, the immunogen is an
enteric adhesin of a pathogen such as an invasin of Salmonella typhimurium, Yersinia pestis
mdpseudotuberculosis and enteropathic Escherichia coli. For example, the nucleotide
sequences encoding said invasin proteins include but are not limited to polynucleotides
comprising nucleotide sequences as set forth in accession numbers: AF140550; Z48169;
X53368; U25631; andM17448.
In general, it is the formulation of an appropriate DNA conjugate or complex (or
other delivery vector) to deliver the DNA to a target M cell that improves host immune
responses against a specific pathogen or other immunogen. For example, such a vaccine
may be comprised of a polybasic conjugate/DNA complex by incorporating an M cell
ligand. Thus, for any given immunogen encoded by nucleic acids that can be used for
eliciting a host response, such a response can be enhanced through effective targeting
mediated by M cell ligands.
In a preferred embodiment, a contemplated polynucleotide is a nucleic acid which
contains essential regulatory elements such that upon introduction into a living vertebrate
cell, it is able to direct the cellular machinery to produce translation products encoded by the
structural gene sequence component of the polynucleotide. In one embodiment of the
invention, the polynucleotide is a polydeoxyribonucleic acid comprising immunogen (or
antigen) structural genes or fragments thereof operatively linked to a transcriptional promoter(s). In another embodiment of the invention the polynucleotide comprises
polyribonucleic acid encoding antigen structural genes or fragments thereof which are
amenable to translation by the eukaryotic cellular machinery (ribosomes, tRNAs, and other
translation factors). Where the protein encoded by the polynucleotide is one which does not
normally occur in that animal except in pathological conditions, (i.e. an heterologous
protein) such as proteins associated with human immunodeficiency virus (HIN) and
Brucella, the animals' immune system is activated to launch a protective immune response.
Because these exogenous proteins are produced by the animals' own tissues, the expressed
proteins are processed by the major histocompatibility system (MHC) in a fashion
analogous to when an actual infection occurs. The result, as shown in this disclosure, is
induction of immune responses against an antigen. Polynucleotides for the purpose of
generating immune responses to an encoded protein are referred to herein as polynucleotide
or DΝA vaccines. The described vaccine works by inducing the vaccinated animal to
produce antibodies or cell-mediated immune responses specific for the vaccine. The
production of these antibodies or cell-mediated immune responses will protect the host upon
subsequent exposure to the infectious agent.
The present formulations encoding various selected antigens may be administered to
immunize individuals against, but not limited to, diseases such as tuberculosis (e.g., BCG
antigen: Kumar et al, Immunology (1999) 97(3):515-521), leprosy (e.g., antigen 85
complex: Νaito et al, Vaccine (1999) 18(9-10):795-798), malaria (e.g., surface antigen
MSA-2: Pye et al, Vaccine (1997) 15(9):1017-1023), diptheria (e.g., diptheria toxoid: U.S. Patent No. 4,691,006 ), tetanus (e.g., tetanus toxin: Fairweather et al, Infect Immun (1987)
55(11):2541-2545), leishmania (e.g., Leishmania major promastigotes: Lasri et al, Vet Res
(1999) 30(5):441-449), salmonella (e.g., covalently bound capsular polysaccharide (Ni) with
porin, both isolated from S. typhi.: Singh et al, Microbiol Immunol (1999) 43(6):535-542),
schistomiasis (e.g., major antigen of Schistosoma mansoni (Sm28 GST): Auriault et al,
Pept Res (1991) 4(1):6-11), measles (e.g., the surface glycoprotein and fusion protein of
measles virus: Machamer et al, Infect Immun (1980) 27(3):817-825), mumps (e.g.,
hemagglutinin-neuraminidase (HΝ) viral gene product: Brown et al, J Infect Dis (1996)
174(3):619-622), herpes (e.g., HSN-2 surface glycoproteins (gB2 and gD2): Corey et al,
JAMA (1999) 282(4):331-340), AIDS (e.g., gpl60: Pontesilli et al, Lancet (1999)
354(9182):948-949), influenza (e.g., immunodommant peptide from hemagglutinin: Novak
et al, J Clin Invest (1999) 104(12):R63-67) and cancer (see Wang RF., JMol Med (1999)
77(9):640-655). Administration of the formulation to a host results in stimulation of the
host's immune system to produce a protective immune response.
The present invention further provides recombinant DNA molecules (rDNAs) that
contain a coding sequence. The vaccines are produced using conventional eukaryotic
plasmid expression systems for the encoded gene. As used herein, a rDNA molecule is a
DNA molecule that has been subjected to molecular manipulation in situ. Methods for
generating rDNA molecules are well known in the art, for example, see Sambrook et al,
Molecular Cloning (1989). In the preferred rDNA molecules, a coding DNA sequence is
operably linked to expression control sequences and/or vector sequences. The choice of vector and/or expression control sequences to which one of the protem
encoding sequences of the present invention is operably linked depends directly, as is well
known in the art, on the functional properties desired, e.g., protein expression, and the host
cell to be transformed. A vector contemplated by the present invention is at least capable of
directing the replication or insertion into the host chromosome, and preferably also
expression, of the structural gene included in the rDNA molecule.
Expression control elements that are used for regulating the expression of an
operably linked protein encoding sequence are known in the art and include, but are not
limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements. Preferably, the inducible promoter is readily controlled, such as being
responsive to a host cell's environment.
In one embodiment, the vector containing a coding nucleic acid molecule will
include a prokaryotic replicon, i.e., a DNA sequence having the ability to direct autonomous
replication and maintenance of the recombinant DNA molecule extrachromosomally in a
prokaryotic host cell, such as a bacterial host cell, transformed therewith. Such replicons are well known in the art. In addition, vectors that include a prokaryotic replicon may also
include a gene whose expression confers a detectable marker such as a drug resistance.
Typical bacterial drug resistance genes are those that confer resistance to ampicillin or tetracycline.
Vectors that include a prokaryotic replicon can further include a prokaryotic or
bacteriophage promoter capable of directing the expression (transcription and translation) of the coding gene sequences in a bacterial host cell, such as E. coli. A promoter is an expression
control element formed by a DNA sequence that permits binding of RNA polymerase and
transcription to occur. Promoter sequences compatible with bacterial hosts are typically
provided in plasmid vectors containing convenient restriction sites for insertion of a DNA
segment of the present invention. Typical of such vector plasmids are pUC8, pUC9, pBR322
and pBR329 available from Biorad Laboratories, (Richmond, CA), pPL and pKK223 available
from Pharmacia, Piscataway, N. J.
Expression vectors compatible with eukaryotic cells, preferably those compatible with
vertebrate cells, can also be used to form a rDNA molecules that contains a coding sequence.
Eukaryotic cell expression vectors are well known in the art and are available from several
commercial sources. Typically, such vectors are provided containing convenient restriction sites
for insertion of the desired DNA segment. Typical of such vectors are pSVL and pKSV-10
(Pharmacia), pBPV-l/pML2d (International Biotechnologies, Inc.), pTDTl (ATCC, #31255),
the vector pCDM8 described herein, and the like eukaryotic expression vectors.
Eukaryotic cell expression vectors used to construct the DNA vaccine molecules of the
present invention may further include a selectable marker that is effective in an eukaryotic cell,
preferably a drug resistance selection marker. A preferred drug resistance marker is the gene
whose expression results in neomycin resistance, i.e., the neomycin phosphotransferase (neo)
gene. (Southern et al, J. Mol. Anal. Genet. 1:327-341, 1982.) Alternatively, the selectable
marker can be present on a separate plasmid, and the two vectors are introduced by co- transfection of the host cell, and selected by culturing in the appropriate drag for the selectable
marker.
The M cell ligand-polybasic conjugates according to the invention may be produced
chemically or by the recombinant method. Coupling by the chemical method can be carried out
in a manner known per se for the coupling of peptides and if necessary the individual
components may be provided with linker substances before the coupling reaction (this
procedure is necessary when there is no functional group suitable for coupling available at the
outset, such as a mercapto or alcohol group).
Depending on the desired properties of the conjugates, particularly the desired stability
thereof, coupling may be carried out by means of various techniques Icnown to persons skilled in
the art, including but not limited to the following techniques. For example, the use of disulphide
bridges, which can be cleaved again under reductive conditions (e.g., using succiriimidyl pyridyl
dithiopropionate, are contemplated. See Jung et al, Biochem Biophys Res Comm 101:599-606
(Jul. 30, 1981). Also contemplated is the use of compounds which are largely stable under
biological conditions (e.g., thioethers, by reacting maleimido linkers with sulfhydryl groups of
the linker bound to the second component). Further comtemplated is the use of bridges that are
unstable under biological conditions, e.g., ester bonds, or using acetal or ketal bonds which are
unstable under weakly acidic conditions.
The production of the conjugates according to the invention by the recombinant method
offers the advantage of producing precisely defined, uniform compounds, whereas chemical
coupling produces conjugate mixtures which then have to be separated. The recombinant preparation of the conjugates according to the invention can be carried
out using methods known for the production of chimeric polypeptides. The present invention
further provides methods for producing a protein of the invention using nucleic acid molecules
herein described. In general terms, the production of a recombinant form of a protein typically
involves the following steps:
First, a nucleic acid molecule is obtained that encodes an M cell ligand protein of the
invention. If the encoding sequence is uninterrupted by introns, it is directly suitable for
expression in any host. The nucleic acid molecule is then preferably placed in operable linkage
with suitable control sequences, as described above, to form an expression unit containing the
protein open reading frame. The expression unit is used to transform a suitable host and the
transformed host is cultured under conditions that allow the production of the recombinant
protein. Optionally the recombinant protein is isolated from the medium or from the cells;
recovery and purification of the protein may not be necessary in some instances where some
impurities may be tolerated.
Each of the foregoing steps can be done in a variety of ways. For example, the desired
coding sequences may be obtained from genomic fragments and used directly in appropriate
hosts. The construction of expression vectors that are operable in a variety of hosts is
accomplished using appropriate replicons and control sequences, as set forth above. The control
sequences, expression vectors, and transformation methods are dependent on the type of host
cell used to express the gene and were discussed in detail earlier. Suitable restriction sites can, if
not normally available, be added to the ends of the coding sequence so as to provide an excisable gene to insert into these vectors. A skilled artisan can readily adapt any host/expression system
known in the art for use with the nucleic acid molecules of the invention to produce recombinant
protein. The polybasic components may vary in terms of their size and amino acid sequence.
Production by genetic engineering has the advantage of allowing the M cell Hgand component of
the conjugate to be modified, by increasing the ability to bind to the receptor, by suitable
mutations, for example, or by shortening the M cell ligand component to the part of the
molecule which is responsible for the binding to the receptor. It is particularly expedient for the
recombinant preparation of the conjugates according to the invention to use a vector which
contains the sequence coding for the M cell ligand component as well as a polylinker into which
the required sequence coding for the polybasic component is inserted, hi this way, a set of
express plasmids can be obtained, of which the plasmid containing the desired sequence can be
used as necessary in order to express the conjugate according to the invention.
The nucleic acids which are to be transported into the cell may be DNAs or RNAs, with
no restrictions as to the nucleotide sequence. The nucleic acids may be modified, provided that
this modification does not affect the polyanionic nature of the nucleic acids; these modifications
include, for example, the substitution of the phosphodiester group by phosphorothioates or the
use of nucleoside analogues.
With regard to the size of the nucleic acids the invention again permits a wide range of
uses. There is no lower limit brought about by the transporting system according to the
invention; thus, any lower limit which might arise would be for reasons specific to the particular intended use use or target specificity. It is also possible to convey different nucleic acids into the
cell at the same time using the conjugates according to the invention.
Within the scope of the present invention it has been possible to demonstrate that M cell
ligand-polybasic conjugates can be efficiently absorbed into living cells and internalized. The
disclosed conjugates or complexes according to the invention are not apparently harmful to cell
growth. This means that they can be administered repeatedly and thus ensure a constantly high,
expression level of the genes and nucleotide sequences inserted into the cell.
The ratio of nucleic acid to conjugate can vary within a wide range, and it is not
absolutely necessary to neutralize all the charges of the nucleic acid. This ratio will have to be
adjusted for each individual case depending on criteria such as the size and structure of the
nucleic acid which is to be transported, the size of the polybasic component and the number and
distribution of its charges, so as to achieve a ratio of transportability and biological activity of the
nucleic acid which is favorable to the particular appUcation. This ratio can first of all be adjusted
coarsely, for example by using the delay in the speed of migration of the DNA in a gel (e.g.,
using the mobility shift on an agarose gel) or by density gradient centrifugation. Once this
provisional ratio has been obtained, it may be expedient to carry out transporting tests with the
radioactively labeled complex with respect to the maximum available activity of the nucleic acid
in the cell and then reduce the proportion of conjugate if necessary so that the remaining
negative charges of the nucleic acid are not an obstacle to transportation into the cell.
The preparation of the M cell ligand-polybasic conjugate/nucleic acid complexes, which
are also a subject of the invention, can be carried out using methods known per se for the complexing of polyionic compounds. One possible way of avoiding uncontrolled aggregation or
precipitation is to mix the two components together first of all at a high (about 1 molar)
concentration of common salt and subsequently to adjust to physiological saline concentration
by dialysis or dilution. Preferably, the concentrations of DNA and conjugate used in the
complex forming reaction are not too high (more than 100 μg/ml), to ensure that the complexes
are not precipitated, as would be known to persons skilled in the art.
A preferred nucleic acid component of the M cell ligand-polybasic moiety-nucleic acid
complex according to the invention is an immunogen structural gene encoded by the nucleic
acids. The invention further relates to a process for introducing nucleic acid or acids into human
or animal cells, preferably forming a complex which is soluble under physiological conditions.
Antibodies against M cell ligand-polybasic moiety protein conjugate or complex may be
prepared by immunizing suitable mammalian hosts using the peptides, polypeptides or proteins
alone or conjugated to suitable carriers. Methods for preparing immunogenic conjugates with
carriers such as BSA, KLH, or other carrier proteins are well Icnown in the art. In some
circumstances, direct conjugation using, for example, carbodiimide reagents may be effective; in
other instances linking reagents such as those supplied by Pierce Chemical Co., Rockford, IL,
may be desirable to provide accessibility to the hapten. The hapten peptides can be extended at
either the amino or carboxy terminus with a Cys residue or interspersed with cysteine residues,
for example, to facilitate linking to a carrier. Administration of the immunogens is conducted
generally by injection over a suitable time period and with use of suitable adjuvants, as is generally understood in the art. During the immunization schedule, titers of antibodies are taken
to determine adequacy of antibody formation.
While the polyclonal antisera produced in this way may be satisfactory for some
applications, for pharmaceutical compositions, use of monoclonal preparations is preferred.
hnmortaUzed cell lines which secrete the desired monoclonal antibodies may be prepared using
the standard method of Kohler and Milstein or modifications which effect immortalization of
lymphocytes or spleen cells, as is generally known. The iinmortalized cell lines secreting the
desired antibodies are screened by immunoassay in which the antigen is the peptide hapten,
polypeptide or protein. When the appropriate immortalized cell culture secreting the desired
antibody is identified, the cells can be cultured either in vitro or by production in ascites fluid.
The desired monoclonal antibodies are then recovered from the culture supernatant or
from the ascites supernatant. Fragments of the monoclonals or the polyclonal antisera which
contain the immunologically significant portion can be used as antagonists, as well as the intact
antibodies. Use of immuiiologically reactive fragments, such as the Fab, Fab', of F(ab')2
fragments is often preferable, especially in a therapeutic context, as these fragments are generally
less immunogenic than the whole immunoglobulin.
The antibodies or fragments may also be produced, using current technology, by
recombinant means. Regions that bind specifically to the desired regions of the gene products
can also be produced in the context of chimeras with multiple species origin. Alternatively, antibodies specific for the M cell ligand polybasic moiety conjugate can
be humanized antibodies or human antibodies, as described in U. S. Patent No. 5,585,089 by
Queen et al. See also Riechmann et al, Nature (1988) 332: 323-27.
There are many embodiments of the instant invention which persons skilled in the art
can appreciate from the specification. Thus, different transcriptional promoters, terminators,
carrier vectors or specific gene sequences may be used successfully. Various methods are
known for such constructs which, upon introduction into mammalian cells, induces the
expression, in vivo, of the polynucleotide thereby producing the encoded protein. It is readily
apparent to those skilled in the art that variations or derivatives of the nucleotide sequence
encoding a protein can be produced which alter the amino acid sequence of the encoded protein.
It is well known in the biological arts that certain amino acid substitutions can be made
in protein sequences without affecting the function of the protein. Generally, conservative
amino acids are tolerated without affecting protein function. Similar amino acids can be those
that are similar in size and/or charge properties, for example, aspartate and glutamate, and
isoleucine and valine are both pairs of similar amino acids. Similarity between amino acid pairs
has been assessed in the art in a number of ways. For example, Dayhoff et al. (1978) in Atlas of
Protein Sequence and Structure, Volume 5, Supplement 3, Chapter 22, pages 345-352, which is
incorporated by reference herein, provides frequency tables for amino acid substitutions which
can be employed as a measure of amino acid similarity. Dayhoff et al. 's frequency tables are
based on comparisons of amino acid sequences for proteins having the same function from a
variety of different evolutionary sources. The altered expressed protein may have an altered amino acid sequence, yet still eUcits immune responses which react with the antigen protein, and
are considered functional equivalents. In addition, fragments of the full length genes which
encode portions of the full length immunogenic protein may also be constructed. These
fragments should encode a protein or peptide which eUcits antibodies that crossreact with the
immunogenic protein, and are considered to be functional equivalents.
The amount of expressible DNA or transcribed RNA to be introduced into a vaccine
recipient will have a very broad dosage range and may depend on the strength of the
transcriptional and translational promoters used as well as subject size, e.g., human versus bison
(i.e., in bison, 5 mg of DNA can be an effective dose). In addition, the magnitude of the
immune response may depend on the level of protein expression and on the immunogenicity of
the expressed gene product. In general, an effective dose ranges of about 1 ng to 5 mg, 100 ng to
2.5 mg, 1 μg to 750 μg, and preferably about 10 μg to 300 μg of DNA is administered
intranasally. It is also contemplated that booster vaccinations may be provided. Following
vaccination with M cell ligand-polybasic conjugate-polynucleotide complexes, boosting with the
encoded antigen products is also contemplated. Parenteral administration, such as intravenous,
intramuscular, subcutaneous or other means of administration of mterleukin-12 protein (or other
cytokines, e.g. GM-CSF), concurrently with or subsequent to intranasal introduction of the M
cell Ugand-polybasic conjugate-polynucleotide complex of this invention may be advantageous.
The polynucleotide may be associated with adjuvants or other agents which affect the
recipient's immune system. In this case, it is desirable for the formulation to be in a
physiologically acceptable solution, such as, but not limited to, sterile saline or sterile buffered saUne. The active immunogenic ingredients can be mixed with excipients or carriers which are
pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients
include but are not limited to water, saline, dextrose, glycerol, ethanol, or the like and
combinations thereof.
hi addition, if desired, the DNA vaccine complexes may contain minor amounts of
auxiUary substances such as wetting or emulsifying agents, pH buffering agents, and/or
adjuvants which enhance the effectiveness of the vaccine. Examples of adjuvants wliich may be
effective include but are not limited to: aluminum hydroxide;
N-acetyl-muramyl-L-threonyl-D-isoglutamme (thr-MDP);
N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP);
N-acetylmuramyl-L-alanyl-D-isoglutam^^
-glycero-3-hydroxyphosphoryloxy)-ethylamine (CGP 19835A, referred to as MTP-PE); and
RIBI, which contains three components extracted from bacteria, monophosphoryl Upid A,
trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80
emulsion. The effectiveness of an adjuvant may be determined by measuring the amount of
antibodies directed against the immunogen resulting from administration of the immunogen in
vaccines wliich are also comprised of the various adjuvants. Such additional formulations and
modes of administration are known in the art and can also be used.
The DNA vaccines of the present invention may be formulated into compositions as
neutral or salt forms. Pharmaceutically acceptable salts include but are not limited to the acid
addition salts (formed with free amino groups of the peptide) which are formed with inorganic acids, e.g., hydrochloric acid or phosphoric acids; and organic acids, e.g., acetic, oxalic, tartaric,
or maleic acid. Salts formed with the free carboxyl groups may also be derived from inorganic
bases, e.g., sodium, potassium, ammonium, calcium, or ferric hydroxides, and organic bases,
e.g., isopropylarnine, trimethylamine, 2-ethylamino-ethanol, histidine, andprocaine.
The M cell ligand-polybasic moiety (or conjugate)-polynucleotide compositions are
administered in a manner compatible with the dosage formulation, and in such amount as will be
prophylactically and/or therapeutically effective. The quantity to be administered, which is
generally in the range of about 100 to 1,000 μg of protein per dose, more generally in the range
of about 5 to 500 μg of protein per dose, depends on the subject to be treated, the capacity of the
individual's immune system to synthesize antibodies, and the degree of protection desired.
Precise amounts of the active ingredient required to be administered may depend on the
judgment of the physician and may be peculiar to each individual, but such a determination is
within the skill of such a practitioner.
The DNA vaccines of the present invention may be given in a single dose or multiple
dose schedule. A multiple dose schedule is one in which a primary course of vaccination may
include 1 to 10 or more separate doses, followed by other doses administered at subsequent time
intervals as required to maintain and or reinforce the immune response, e.g., at 1 to 4 months for
a second dose, and if needed, a subsequent dose(s) after several months.
Immunization by DNA injection allows the ready assembly of multicomponent subunit
vaccines. Simultaneous immunization with multiple influenza genes has recently been reported.
(Donnelly et al, Vaccines (1994) pp 55-59). The inclusion in a DNA vaccine of genes whose products activate different arms of the immune system may also provide thorough protection
from subsequent challenge.
The vaccines of the present invention are useful for administration to domesticated or
agricultural animals, as well as humans. Vaccines of the present invention may be used to
prevent and/or combat infection of any agricultural animals. The techniques for adrriinistering
these vaccines to animals and humans are known to those skilled in the veterinary and human
health fields, respectively.
Except as may be noted hereafter, contemplated techniques for cloning, DNA
isolation, ampUfication and purification, for enzymatic reactions involving DNA ligase, DNA
polymerase, restriction endonucleases and the like, and various separation techniques are those
well known and commonly employed by those skilled in the art. A number of standard
techniques are described in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual,
Second Edition, Cold Spring Harbor Laboratory, Plainview, N.Y.; Maniatis et al. (1982)
Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Plainview, N.Y.;
Wu (ed.) (1993) Meth Enzymol. 218, Part I; Wu (ed.) (1979) Meth Enzymol 68; Wu et al. (eds.)
(1983) Meth Enzymol 100 and 101; Grossman et al. (eds.) Meth Enzymol 65; Miller (ed.) (1972)
Experiments in Molecular Genetics, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.;
Old et al. (1981) Principles of Gene Manipulation, University of California Press, Berkeley;
Schleif etal. (1982) Practical Methods in Molecular Biology; Glover (ed.) (1985) DNA Cloning
Vol. I and It, IRL Press, Oxford, UK; Hames et al. (eds.) (1985) Nucleic Acid Hybridization,
URL Press, Oxford, UK; Setlow et al (1979) Genetic Engineering: Principles and Methods, Vols. 1-4, Plenum Press, New York. Abbreviations and nomenclature, where employed, are
deemed standard in the field and commonly used in professional journals such as those cited
herein.
The following examples are provided to illustrate the present invention without,
however, limiting the same thereto.
Examples
Example 1. Production of recombinant reovirus σl protein
The cloned protein σl cDNA from reovirus serotype 3 strain in the pST3-S 1
Banjerjea et al, Virology (1988) 167:601-612) was kindly provided by Dr. Wolfgang K. Jolik
from Duke University Medical Center. For its expression in E. coli, using PCR, a 1.4 kb cDNA
fragment containing the restriction endonuclease sites, EcoR 1 and Pst 1, was inserted into the
polylinker site of an E. coli expression plasmid, pMAL-C2 (New England Biolabs, Beverly,
MA). The resultant, pMAL-C2-S 1 , was used to transform E. coli, strain BL21 (DE3 ; Novagen,
Madison, WI). Upon induction with IPTG, the maltose-binding protein (MBP) -protein σl
fusion protein was induced in the cytoplasm of E. coli. The clear lysate of E. coli containing the
fusion protein was purified by affinity chromatography using amylose resin according to
manufacturer's directions (New England Biolabs). This MBP::protein σl fusion protein is
referred to as recombinant protein σl . Example 2. Preparation of recombinant fusion protein σl-polylysine-DNA complex
The recombinant protein σl was covalently linked to poly-L-lysine (PL) according
to the methods of Wagner et α/(1990). Protein σl was purified and resuspended in phosphate-
buffered saline (PBS), pH 7.3. To generate the ditMopyridine linker, both protein σl and PL
were each modified with succinimidyl 3-(2-pyridyldithio)propionate (SPDP; Sigma Chemical
Co., St. Louis, MO). Briefly, in separate vessels, ten milligrams of protein σl in 5 ml PBS, pH
7.3, and twenty milHgrams of PL (Sigma), with an average chain length of 270 lysine
monomers, in 1 ml of 75 mM sodium acetate were each vigorously mixed to react with SPDP in
15 mM ethanolic solution for one hour. The resulting SPDP modified protein σl was then
dialyzed against PBS, pH 7.3, and the modified PL was then dialyzed against 20 mM sodium
acetate to remove unbound SPDP. To generate the mercaptopropionate linker, the resultant PL
with άithiopyridine linker was further mixed with 23 mg dithiothreitol (DTT) in sodium
bicarbonate solution, pH 7.5, for one hour under argon. The mercaptopropionate PL was
dialyzed against 20 mM sodium acetate to remove free DTT. The 10 mg of dithiopyridine-
modified protein σ 1 was then mixed with the 20 mg of mercaptopropionate-modified PL under
argon at room temperature for 18 hours. The resultant reaction generated what is referred to as
protein σl-PL conjugate. This conjugate was dialyzed to remove unreacted
mercaptopropionate-PL using a membrane with an exclusion of 100 kilodaltons, against HEPES
buffered saline (20 mM HEPES, 100 mM sodium chloride, pH 7.4; HS). Protein σl-PL
concentration was determined using a Bradford assay (Pierce, Rockford, IL). For control
transfections, MBP-PL conjugates were similarly generated. For the formation of conjugate- DNA complex, the protein σl-PL conjugate in 125 μl of HS was added dropwise into an equal
volume of HS containing the plasmid DNA and incubated at room temperature for 30 minutes to
form conjugate-DNA complex.
Example 3. Cell Ugand binding assay
To assess the cell-binding capacity of the protein σl and protein σl-PL conjugates,
an immunofluorescent binding assay was performed. The protein σl and σl-PL conjugates
were incubated with mouse L cells (CCL-1, American Type Culture Collection, Manassas, VA),
RFL-6 fibroblast cells (CCL-192, ATCC), and Caco-2 cells (HTB-37, ATCC) and binding was
assessed using 20 μg/ml of biotinylated monoclonal anti-reo virus protein σl antibody (HB-167,
ATCC) and SA-PE (Southern Biotech. Assoc, Birmingham, AL), and specific binding was then
assessed using flow cytometry. No staining was obtained with normal rabbit serum or in the
presence of SA-PE only.
Example 4. Cell culture and transfection with plasmid DNA
The mouse L cells, RFL-6 cells, and Caco-2 cells were used for targeting gene
transfer by protein σl-PL conjugate. The mouse L cells have been used as the in vitro model for
reovirus protein σl binding studies. Cells were maintained in complete media: Dulbecco's
lninimiim essential medium (DMEM; BioWhittaker, Walkersville, MD), supplemented with
10% fetal bovine serum (Life Technologies, Grand Island, NY) at 37°C under 5% CO2. For Luc
assay, 2.5 X 105 cells were added to each well of the 12-well plate and allowed to adhere overnight. The conjugate-DNA complexes were added and incubated for another 24 hours in
complete media. For chloroquine treatment, the cells were incubated with protein σl-PL-DNA
complexes and 100 μM chloroquine for 4 hours at 37°C. Four hours after incubation, the
conjugate-DNA complexes were removed, and cells were incubated with complete media for
another 24 hours. The cells were lysed to assay reporter gene activity. For β-Gal assay, 5X 105
cells were added to each well of 6- ell plate and allowed to adhere overnight. The conjugate-
DNA complexes containing 8 μg σl-PL and pCMVβ-gal (Life Technologies), with or without
chloroquine, were added and incubated for 24 hours. The cells were then incubated with fresh
media for another 24 hours prior to flow cytometry analysis.
Example 5. Assays for reporter gene detection
The Luc gene was used as a reporter gene to assay protein σl-PL conjugate-
mediated transfection. A 1.4 kb Luc gene fragment flanked with Hind DI and EcoR V was
extracted from pSPKuci(+) (Promega, Madison, WI). The pCMVLuciferase (pCMVLuc) was
constructed by ligating the 1.4 kb luciferase gene into the polylinker site in pcDNA3.1 (+)
(hivitrogen, Carlsbad, CA). The cells were lysed with lx luciferase lysis buffer (Promega,
Madison, WI). Twenty μl of supernatant of cell lysates were mixed with 100 μl of Luc assay
buffer and assayed with a luminometer (LUMAT LB 9507, EG&G Berthold, Germany). The
relative light units from the total lysates were used to express the Luc activities produced from
each transfection. Expression of β-Gal was visualized by incubating the transfected cells with PBS
solution containing 1 mg/ml of 5-boromo-4-chloro-3-indolyl-β-galactopyranoside (X-Gal,
Boeringer Mannheim, Indianapolis, IN) at 37°C for 16 hr. To quantify the transfection
efficiency, cells having been transfected with the constructs pCMV-β-Gal (Life Technologies)
were harvested, loaded with 200 μM fluorescein-mono-β-D-galactopyranoside (FDG; Molecular
Probe, Eugene, OR) for 30 minutes at 37°C and diluted with cold PBS to a final concentration of
2.5X105 cells/ml. Flow cytometry analysis was performed using a Becton Dickinson
FACSCalibur.
Example 6. Histochemical determination of fusion protein σl binding to NALT
NALT tissues were collected as previously described (Asanuma et al, J Immunol
Methods (1997) 202:123-131 and Heritage et al, Am JRespir Crit Care Med (1997) 156(4 Pt
1): 1256-1262). Palates with visible NALT were washed in DMEM, and prior to binding with
biotinylated protein σl (following standard procedures), NALT were first incubated in DMEM
alone or in the presence of 500 μg/ml of protein σl in DMEM with gentle rotation on a
GeneMate orbital Shaker (hitermountain Scientific Co., Bountiful, UT) for 45 minutes at 4°C.
NALT were incubated with excess unmodified protein σl in order to inhibit biotinylated protein
σl binding, and thus, show specificity of binding to the NALT. NALT were then washed gently
in DMEM and incubated in 50 μg/ml biotinylated protein σl in DMEM, and were again rotated
gently for 45 min at 4°C. Following incubation, NALT were removed, rinsed gently in PBS, and
then arranged in 15 mm by 15 mm Tissue Tek@Cryomold (Miles Inc., Elkhard, IN) with their ventral surfaces oriented toward the bottom of the mold. The palates were then frozen in Tissue
Tek® O.C.T. compound embedding media and stored at -80°C until use. For immunoperoxidase
staining, frozen NALT sections, previously treated with biotinylated protein σl, were cut at 5
mm, air dried, fixed in acetone at 4°C, and air dried before rehydration.
Frozen sections were rehydrated in Dulbecco's PBS (DPBS) containing 0.2% normal
goat serum (NGS). A 1:250 dilution of SA-HRP conjugate (BioSource International,
Camarillo, CA) was added for 45 min at room temperature. The location of the HRP was
visuaUzed upon reaction with the precipitable substrate, 3-amino-ethylcarbazole (AEC: Sigma).
Example 7. In vivo analysis of intranasal immunization with σl conjugates
Luciferase
Intranasal (i.n.) immunization with protein σl-polylysine (PL) conjugate enhances
induced mucosal IgA responses in mice. Data depicts the mean endpoint titers (± SE) for mice
immunized i.n. with protein σl-PL-pCMVLuciferase (Luc) or uncomplexed pCMVLuc (5
mice/group). Significant differences between protein σl-PL-pCMVLuc and pCMVLuc only
were determined by student t-test. *p<0.05. **p<0.005. (See Figure 2).
Example 8. β-galactosidase
Intranasal (i.n.) immunization with protein σl-PL-pCMVβ-galactosidase (βgal)
stimulates βgal-specific CTL responses in mice. BALB/c mice received three i.n.
immunizations with either protein σl-PL-pCMVβgal or pCMVβgal. Immune splenocytes were
able to lyse 5ICr loaded βgal-expressing fibroblasts (BC-βgal), but not irrelevant BC-envelope (BC-env) targets. The mucosally formulated DNA was as efficient in stimulating βgal-specific
CTLs as those mice receiving naked DNA. (See Figure 3).
Example 10. HJV
Intranasal (i.n.) immunization with protein σl -polylysine (PL) conjugate was conducted
to enhance induced mucosal IgA responses in mice. The mean endpoint liters (± SE) for mice
immunized i.n. with protein σl-PL-pCMVgpl60 and σl-PL-pCMNgpl40 or uncomplexed
pCMNgpl60 and pCMVgpl40 (5 mice/group) was compared. Significant differences between
protein σl-PL-pCMVgρl60 and σl-PL-pCMVgpl40 versus pCMVgpl60 and ρCMNgpl40
only were deteπnined by student t-test. Using the mucosal DΝA formulation, the same
magnitude of IgG antibody response is observed as was observed for the anti-reporter gene
responses.
Experimentally, mice were immunized with one of three designated HJV DΝA vaccine
constructs, that is gpl60, gp 140(c) and gp 140(s), as indicated in Fig 4. Each group (5
mice/group) received three intranasal immunizations either of naked DΝA or of the identified M
cell DΝA vaccine formulation. As indicated, the mucosal intestinal IgA response was elevated
10 weeks after the initial immunization when compared to intranasal naked DΝA immunization.
Thus, the DΝAvaccine formulation improved mucosal IgA responses when compared to
conventional naked DΝA immunization. Example ll. Brucella
Intranasal (i.n.) immunization with protein σl -polylysine (PL) conjugate enhances
induced mucosal IgA and IgG responses in bison. The mean endpoint liters (± SE) for bison
immunized i.n. with protein σl-PL-pCMVL7/L12 ribosomal protein or uncomplexed
pCMVL7/L12 ribosomal protein (5 bison/group) was compared. Significant differences
between protein σl-PL-pCMVL7/L12 ribosomal protein versus pCMVL7/L12 ribosomal
protein only were determined by student t-test. Using our mucosal DNA formulation, we
observed increases in serum IgG and vaginal IgA and IgG anti-L7/L12 antibody titer in bison.
Example 12. HIN gp!20
Intranasal immunization with an M cell-formulated HIN DΝA vaccine promotes
enhanced cytolytic activity (cell-mediated immunity) against target cells expressing HIN gpl20
as shown in Figs 5 A and 5B. Mice received a formulated vaccine, naked DΝA version, protein
sigmal by the intranasal route three times at one week intervals or were left imimmunized.
Mice were sacrificed six weeks subsequent to this initial immunization to procure specified
tissues, hi a dose-dependent fashion, the lungs from only mice receiving only the formulated
vaccine showed effector function. These results show that the vaccine as formulated is superior to naked DΝA in stimulating gpl20-specific immunity.
Data also indicated that antigen restimulation specifically enhances CTL responses from
mice i.n. -immunized with the formulated vaccine as opposed to mice immunized with the naked DΝA alone. Pulmonary lymph nodes (LRLΝ) and splenocytes from immunized mice were restimulated in vitro with cells expressing gpl20 or beta-galactosidase (neg. control), and were subsequently examined for cytolytic activity. The observed killing was specific since negative
targets were not lysed, and other mechanisms of vaccination failed to stimulate cytolytic activity.
The foregoing detailed description has been given for clearness of understanding only
and no unnecessary limitations should be understood therefrom as modifications will be obvious
to those skilled in the art. While the invention has been described in connection with specific
embodiments thereof, it will be understood that it is capable of further modifications and this
application is intended to cover any variations, uses, or adaptations of the invention following, in
general, the principles of the invention and including such departures from the present disclosure
as come within known or customary practice within the art to which the invention pertains and
as may be applied to the essential features hereinbefore set forth and as follows in the scope of
the appended claims.

Claims

ClaimsWe claim:
1. A composition comprising:
an M cell specific Hgand;
a nucleic acid sequence encoding an immunogen; and
a nucleic acid binding moiety.
2. The composition of claim 1, wherein said nucleic acid sequence is a DNA
sequence.
3. The composition of claim 1, wherein said binding moiety is a polypeptide.
4. The composition of claim 3, wherein said polypeptide comprises a polymeric
chain of basic amino acid residues.
5. The composition of claim 4, wherein said polymeric chain comprises polylysine.
6. The composition of claim 3, wherein said polypeptide further comprises said M
cell specific ligand.
7. The composition of claim 1 , wherein said immunogen is selected from the group
consisting of immunogens expressed by infectious agents and tumor specific antigens.
8. The composition of claim 7, wherein said infectious agent is selected from the
group consisting of bacterium, parasite, virus, fungus, prion, tuberculobacillus, leprosy bacillus,
malaria parasite, diphtheria bacillus, tetanus bacillus, Leishmania, SalmoneUa, Schistoma,
measles virus, mumps virus, herpes virus, FflV, cancer and influenza virus.
9. The composition of claim 1, wherein said M cell specific Hgand is selected from
the group consisting of the protein σl of a reovirus, adhesin derived from Salmonella and
adhesin derived from poUo virus and M cell tropic fragments thereof.
10. The composition of claim 9, wherein said M cell specific ligand is the protein
σl of a reovirus and M cell tropic fragments thereof.
11. The composition of claim 2, wherein said DNA sequence further comprises a
plasmid vector in wliich said DNA sequence encoding an immunogen is operably linked to
transcription regulatory elements.
12. A vaccine comprising the composition of any of claims 1 to 11 and a
pharmaceutically acceptable excipient.
13. The vaccine of claim! 2, which induces a protective immune response in a
vaccinated host against said immunogen.
14. The vaccine of claim 12, further comprising an adjuvant.
15. The vaccine of claim 14, wherein said adjuvant comprises an
immunomodulator.
16. The vaccine of claim 15, wherein said immunomodulator is selected from the
group consisting of cytokines, lymphokines, mterleukins, interferons and growth factors.
17. The vaccine of claim 12, wherein the vaccine is formulated in unit dosage form.
18. The vaccine of claim 12, further packaged with instructions for the use of the
vaccine to mduce an immune response against said immunogen or against the disease with
which said immunogen is associated.
19. The vaccine of claim 12, wherein the vaccine is a therapeutic vaccine.
20. The vaccine of claim 12, wherein the vaccine is formulated for adrninistration
through a route selected from the group consisting of oral, nasal, vaginal, rectal and urethral
routes of administration.
21. A method for immunizing a host against an immunogen, comprising the step of
administeriiig the vaccine of claim 12 to the host.
22. A method for assaying for mucosal immunity comprising the steps of
administering the vaccine of claim 12 to an animal which is free of infection of the
infectious agent whose antigen is to be tested;
isolating cells from the animal; and
co-incubating said isolated cells with heterologous antigen expressing cells, wherein
lysing of antigen expressing cells is indicative of mucosal immunity in the animal.
23. The method of claim 22, wherein said isolated cells are selected from the group
consisting of mucosal B cells, T cells, lamina propria isolates, intraepithelial isolates, Peyer's
patches cells, lymph nodes, nasal passages, NALT, adenoids and vaginal epithelium.
24. The method of claim 22, comprising the additional step of evaluating the
animal's cytokine profile.
25. An isolated nucleic acid encoding a fusion protein comprising a nucleic acid
binding moiety and an M cell specific Hgand.
26. The nucleic acid of claim 25, wherein said binding moiety comprises a
polymeric chain of basic amino acid residues.
27. The nucleic acid of claim 26, wherein said polymeric chain comprises
polylysine.
28. The nucleic acid of claim 25, wherein said M cell Hgand is selected from the
group consisting of: protem σl of a reovirus, adhesin derived from Salmonella and adhesin
derived from polio virus and M cell tropic fragments thereof.
29. A vector comprising the nucleic acid of any of claims 25 to 28.
30. The vector of claim 29, wherein said vector is an expression vector.
31. A polypeptide comprising the expression product of the vector of claim 30.
32. The vector of claim 29, wherein said nucleic acid is in operable linkage and
wherem the operable linkage is selected from the group consisting of sense and antisense
orientations relative to transcriptional elements comprising the vector.
33. A host cell comprising the vector of claim 29.
34. A method of expressing a fusion protein comprising the step of expressing the
vector of claim 30.
35. An isolated polypeptide comprising a nucleic acid binding moiety and an M cell
specific ligand.
36. The polypeptide of claim 35, wherein said binding moiety comprises a
polymeric chain of basic amino acid residues.
37. The polypeptide of claim 36, wherein said polymeric chain comprises polylysine.
38. The polypeptide of claim 35, wherein said M cell Hgand is selected from the
group consisting of: protein σl of a reovirus, adhesin derived from Salmonella and adhesin
derived from poHo virus and M cell tropic fragments thereof.
39. An isolated antibody that binds to the polypeptide of claim 35.
40. A kit comprising:
an M cell specific ligand; and
a nucleic acid bmding moiety.
41. The kit of claim 40, wherein said binding moiety is a polypeptide.
42. The kit of claim 41, wherein said polypeptide comprises a polymeric cham of
basic amino acid residues.
43. The kit of claim 41, wherein said polymeric chain comprises polylysine.
44. The kit of claim 41, wherein said polypeptide further comprises said M cell
specific Hgand.
45. The kit of claim 40, wherem said M cell specific ligand is selected from the
group consisting of: protein σ 1 of a reovirus, adhesin derived from Salmonella and adhesin
derived from polio virus and M cell tropic fragments thereof.
46. The kit of claim 45, wherein said M cell specific Hgand is the protein σl of a
reovirus and M cell tropic fragments thereof.
PCT/US2001/000426 2000-01-06 2001-01-08 M cell directed vaccines WO2001049867A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/169,492 US20040033486A1 (en) 2001-01-08 2001-01-08 M cell directed vaccines
EP01901811A EP1257654A1 (en) 2000-01-06 2001-01-08 M cell directed vaccines
AU27672/01A AU2767201A (en) 2000-01-06 2001-01-08 M cell directed vaccines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17478600P 2000-01-06 2000-01-06
US60/174,786 2000-01-06

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/169,492 A-371-Of-International US20040033486A1 (en) 2000-01-06 2001-01-08 M cell directed vaccines
US10/660,787 Continuation-In-Part US20040109871A1 (en) 2000-01-06 2003-09-12 M cell directed vaccines

Publications (1)

Publication Number Publication Date
WO2001049867A1 true WO2001049867A1 (en) 2001-07-12

Family

ID=22637521

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/000426 WO2001049867A1 (en) 2000-01-06 2001-01-08 M cell directed vaccines

Country Status (3)

Country Link
EP (1) EP1257654A1 (en)
AU (1) AU2767201A (en)
WO (1) WO2001049867A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2460969A (en) * 2004-06-23 2009-12-23 Ian A Ferguson Nasally-administered vaccines
AU2012201102B2 (en) * 2004-06-23 2014-05-08 Ferguson, Ian Andrew Agents and methods for early diagnosis and monitoring of Alzheimer's disease and other neurological disorders

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
FREY A ET AL: "Targeting of mucosal vaccines to Peyer's patch M cells.", BEHRING INSTITUTE MITTEILUNGEN, (1997 FEB) (98) 376-89. REF: 69, XP000995438 *
GIANNASCA P.J. ET AL: "Targeted delivery of antigen to hamster nasal lymphoid tissue with M - cell -directed lectins.", INFECTION AND IMMUNITY, (1997) 65/10 (4288-4298)., XP000995425 *
HADDAD, A. (1) ET AL: "Targeted M cell immunization for HIV-1 ENV DNA vaccines.", FASEB JOURNAL, (APRIL 20, 2000) VOL. 14, NO. 6, PP. A1204. PRINT. MEETING INFO.: JOINT ANNUAL MEETING OF THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS AND THE CLINICAL IMMUNOLOGY SOCIETY SEATTLE, WASHINGTON, USA MAY 12-16, 2000, XP000995418 *
NEUTRA, MARIAN R. ET AL: "Antigen uptake by M cells for effective mucosal vaccines", MUCOSAL VACCINES (1996), 41-55. EDITOR(S): KIYONO, HIROSHI;OGRA, PEARAY L.; MCGHEE, JERRY R. PUBLISHER: ACADEMIC, SAN DIEGO, CALIF., XP000996131 *
WAGNER E: "TRANSFERRIN-POLYCATION CONJUGATES AS CARRIERS FOR DNA UPTAKE INTO CELLS", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA,US,NATIONAL ACADEMY OF SCIENCE. WASHINGTON, vol. 87, no. 9, 1 May 1990 (1990-05-01), pages 3410 - 3414, XP000368690, ISSN: 0027-8424 *
WANG, X. (1) ET AL: "In vivo induction of cytotoxic T lymphocytes by M cell vaccination.", FASEB JOURNAL, (APRIL 20, 2000) VOL. 14, NO. 6, PP. A1204. PRINT. MEETING INFO.: JOINT ANNUAL MEETING OF THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS AND THE CLINICAL IMMUNOLOGY SOCIETY SEATTLE, WASHINGTON, USA MAY 12-16, 2000, XP000995419 *
WU Y ET AL: "Gene transfer facilitated by a cellular targeting molecule, reovirus protein sigma1.", GENE THERAPY, (2000 JAN) 7 (1) 61-9., XP000995427 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2460969A (en) * 2004-06-23 2009-12-23 Ian A Ferguson Nasally-administered vaccines
GB2460969B (en) * 2004-06-23 2010-02-17 Ian A Ferguson Vaccines for intranasal administration
AU2012201102B2 (en) * 2004-06-23 2014-05-08 Ferguson, Ian Andrew Agents and methods for early diagnosis and monitoring of Alzheimer's disease and other neurological disorders

Also Published As

Publication number Publication date
EP1257654A1 (en) 2002-11-20
AU2767201A (en) 2001-07-16

Similar Documents

Publication Publication Date Title
US11104916B2 (en) Compositions and methods for alphavirus vaccination
KR101650364B1 (en) Fusion proteins for use as immunogenic enhancers for inducing antigen-specific t cell responses
Ellis New technologies for making vaccines
CN100489105C (en) Immune responses against HPV antigens elicited by compositions comprising an HPV antigen and a stress protein or an expression vector
Fennelly et al. Mucosal DNA vaccine immunization against measles with a highly attenuated Shigella flexneri vector
TWI674269B (en) A vaccine composition comprising an immunogenic protein and combination adjuvants for use in eliciting antigen-specific t-cell responses
US20100303847A1 (en) Compositions of toll-like receptor agonists and papillomavirus antigens and methods of use thereof
Partidos et al. The adjuvant effect of a non‐toxic mutant of heat‐labile enterotoxin of Escherichia coli for the induction of measles virus‐specific CTL responses after intranasal co‐immunization with a synthetic peptide
EP2478915A1 (en) CyaA-carried polypeptide(s) and use to induce both therapeutic and prophylactic immune responses
US6737521B1 (en) Delivery and expression of a hybrid surface protein on the surface of gram positive bacteria
JP2013520487A (en) Immunogenic proteins and compositions
JP4653934B2 (en) Bacteriophage mediated immunization methods
WO2023023940A1 (en) Immunogen for inducing broad-spectrum anti-coronavirus t cell vaccine and use thereof
AU674311B2 (en) Delivery and expression of a hybrid surface protein on the surface of gram positive bacteria
US20040109871A1 (en) M cell directed vaccines
TW202206598A (en) A vaccine against sars-cov-2 and preparation thereof
Shi et al. The expression of membrane protein augments the specific responses induced by SARS-CoV nucleocapsid DNA immunization
Toussaint et al. Genetic immunisation of cattle against bovine herpesvirus 1: glycoprotein gD confers higher protection than glycoprotein gC or tegument protein VP8
WO2002072015A2 (en) M cell directed vaccines
Chin et al. Manipulating systemic and mucosal immune responses with skin-deliverable adjuvants
US20040033486A1 (en) M cell directed vaccines
WO2001049867A1 (en) M cell directed vaccines
Wu et al. Gene transfer facilitated by a cellular targeting molecule, reovirus protein σ1
JP2002514061A (en) Compositions and methods for administering pneumococcal DNA
Burnette Recombinant subunit vaccines

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2001901811

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10169492

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 2001901811

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2001901811

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP