US20090081256A1 - Transgenic plant-based vaccines - Google Patents

Transgenic plant-based vaccines Download PDF

Info

Publication number
US20090081256A1
US20090081256A1 US12/190,383 US19038308A US2009081256A1 US 20090081256 A1 US20090081256 A1 US 20090081256A1 US 19038308 A US19038308 A US 19038308A US 2009081256 A1 US2009081256 A1 US 2009081256A1
Authority
US
United States
Prior art keywords
plant
mammal
antigen
subunit
cholera toxin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/190,383
Inventor
William H.R. Langridge
Jie Yu
Takeshi Arakawa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Loma Linda University
Original Assignee
Loma Linda University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/296,981 external-priority patent/US6777546B2/en
Priority claimed from US09/771,536 external-priority patent/US20030165543A1/en
Priority claimed from US11/001,153 external-priority patent/US20050186219A1/en
Application filed by Loma Linda University filed Critical Loma Linda University
Priority to US12/190,383 priority Critical patent/US20090081256A1/en
Publication of US20090081256A1 publication Critical patent/US20090081256A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8242Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits
    • C12N15/8257Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits for the production of primary gene products, e.g. pharmaceutical products, interferon
    • C12N15/8258Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits for the production of primary gene products, e.g. pharmaceutical products, interferon for the production of oral vaccines (antigens) or immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • C07K14/28Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Vibrionaceae (F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8216Methods for controlling, regulating or enhancing expression of transgenes in plant cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/88Lyases (4.)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin
    • 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
    • C12N2720/12311Rotavirus, e.g. rotavirus A
    • C12N2720/12334Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • Patent Application No. 60/082,688, titled “Plant Vaccines Against Autoimmune Disease,” filed April 22, 1998 and 2) is a continuation-in-part of U.S. patent application Ser. No. 09/167,493, titled “Expression of Cholera Toxin B Subunit in Transgenic Plants and Efficacy Thereof in Oral Vaccines,” filed Oct. 7, 1998, now abandoned, which claims the benefit of U.S. Provisional Patent Application No. 60/061,265, titled “Cholera Toxin in Food Plants,” filed Oct. 7, 1997; and U.S. patent application No. 11/370,697 is a continuation of U.S. patent application Ser. No.
  • Acute infectious enteric diseases such as acute gastroenteritis
  • Cholera, rotavirus and enterotoxigenic E. coli are the three major causative agents of acute gastroenteritis.
  • Human rotavirus for example, is the most important cause of infantile gastroenteritis worldwide. This virus has a tremendous public health impact worldwide, infecting nearly every child in the first few years of life. Rotavirus infection is responsible for approximately 1 million deaths each year and an estimated 18 million hospitalizations. 20% to 40% of the hospitalizations are for childhood diarrhea, which makes the rotavirus the most important single cause of diarrheal mortality among children.
  • Treatment for acute gastroenteritis includes antibiotics and metabolic support. However, adequate treatment is often not available, particularly in lesser developed areas where the incidence of acute gastroenteritis is highest. Prevention of acute gastroenteritis would be preferable to treatment. However, preventative measures, such as the provision of safe drinking water, are often inadequate or unavailable.
  • a DNA construct that encodes, upon expression in a plant cell, a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease.
  • the first immunogenic antigen can be a rotavirus antigen.
  • the first immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • the fusion protein encoded by the DNA construct can further comprise a second cholera toxin subunit.
  • the second cholera toxin subunit can be cholera toxin A2 subunit.
  • the fusion protein encoded by the DNA construct can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease.
  • the second immunogenic antigen can be a rotavirus antigen.
  • the second immunogenic antigen can also be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • a DNA construct that encodes, upon expression in a plant cell, a fusion protein comprising a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a first mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease.
  • the first immunogenic antigen can be a rotavirus antigen.
  • the second immunogenic antigen can be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • an expression vector comprising a DNA construct of the present invention, a transgenic plant cell transformed with a DNA construct of the present invention, a transgenic plant seed transformed with the DNA construct of the present invention, and a transgenic plant transformed with the DNA construct of the present invention.
  • a method of producing an immunogen in a plant comprising cultivating a transgenic plant of the present invention under conditions effective to express the fusion protein.
  • a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of a plant of the present invention.
  • the present invention also includes means for producing, in a plant cell, a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease.
  • the means can comprise a DNA construct that encodes, upon expression in the plant cell, a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease.
  • the first immunogenic antigen can be a rotavirus antigen.
  • the first immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • the fusion protein can further comprise a second cholera toxin subunit, such as cholera toxin A2 subunit.
  • the fusion protein can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease.
  • the second immunogenic antigen can be a rotavirus antigen.
  • the second immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • the present invention also includes means for producing, in a plant cell, a fusion protein comprising a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a first mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease.
  • the first immunogenic antigen can be a rotavirus antigen.
  • the second immunogenic antigen can be an enterotoxigenic E. coli antigen.
  • an expression vector comprising the means of the present invention, a transgenic plant cell transformed with means of the present invention, a transgenic plant seed transformed with the means of the present invention, and a transgenic plant transformed with the means of the present invention.
  • the present invention also includes a method of producing an immunogen in a plant comprising cultivating the transgenic plant of the present invention under conditions effective to express the fusion protein.
  • the present invention further includes a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of a plant of the present invention.
  • a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a mammalian disease.
  • the first immunogenic antigen can be a rotavirus antigen.
  • the first immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • the fusion protein can further comprise a second cholera toxin subunit.
  • the second cholera toxin subunit can be cholera toxin A2 subunit.
  • the fusion protein can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease.
  • the second immunogenic antigen can be a rotavirus antigen.
  • the second immunogenic antigen can also be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • the fusion protein comprises a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease.
  • a fusion protein encoded by the DNA construct of the present invention is provided.
  • a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of the fusion protein of the present invention.
  • FIG. 1 is a diagram of the vector pPCV701FM4-CTB:NSP4;
  • FIG. 2 is a diagram of the vector pPCV701CFA/I-CTB-NSP4.
  • a method of inducing partial or complete immunity to an infectious disease such as gastroenteritis, in a mammal by administering to the mammal a portion of a transgenic plant comprising a fusion protein, where the fusion protein comprises at least one cholera toxin subunit and an immunogenic antigen from a causal factor of the disease.
  • the fusion protein comprises at least two cholera toxin subunits, at least one of which functions as an antigen, in addition to functioning as an adjuvant for the immunogenic antigen.
  • the fusion protein comprises at least two immunogenic antigens, each fused to a cholera toxin subunit.
  • the fusion protein By fusing the immunogenic antigen to the cholera toxin subunit, the fusion protein more specifically targets the appropriate immune system tissue upon administration. This increased specificity compensates for the low level of production of the protein in the transgenic plant and increases the response of the mammal's immune system.
  • the fusion protein comprises the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit (CTB).
  • CTB cholera toxin B subunit
  • the fusion protein comprises the enterotoxigenic E. coli (ETEC) fimbrial colonization factor CFA/I fused to the cholera toxin A2 subunit (CTA2).
  • the fusion protein comprises both the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit, and the fusion protein comprises the enterotoxigenic E. coli fimbrial colonization factor CFA/I fused to the cholera toxin A2 subunit.
  • transgenic plant producing a fusion protein comprising the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit.
  • the transgenic plant can be administered to a mammal to immunize the mammal against cholera and rotavirus infection simultaneously.
  • FIG. 1 there is shown a diagram of the vector used to prepare the transgenic plant.
  • the vector contained four genes located within the transferred DNA (T-DNA) sequence flanked by the right and left border (RB and LB), and 25 bp direct repeats of the borders required for integration of the transferred DNA into plant genomic DNA.
  • the four genes were the CTBH:NSP4(114-135):SEKDEL coding sequence under control of the mas P2 promoter; the bacterial luciferase AB fusion gene (luxF) under control of the mas P1 promoter used as a detectable marker; an NPT II expression cassette used for resistance to kanamycin in plants; and a ⁇ -lactamase cassette for resistance to ampicillin in E.
  • the g7pA polyadenylation signal was from the A. tumefaciens T L -DNA gene 7.
  • the OcspA polyadenylation signal is from the octopine synthase gene.
  • Pnos was the promoter of the nopaline synthase gene.
  • g4pA was the polyadenylation signal from TL-DNA gene 4.
  • OriT was the origin of transfer derived from pRK2.
  • OriV was the wide host range origin of replication for multiplication of the plasmid in A. tumefaciens derived from pRK2.
  • Ori pBR322 was the replication origin of pBR322 for maintenance of the plasmid in E. coli.
  • the vector pPCV701FM4-CTB:NSP4 was constructed as follows.
  • the plant expression vector pPCV701FM4 a derivative of plasmid pPCV701
  • the oligonucleotide 5′ primer (5′-gctctagagccaccatgattaaattaaaatttggtg-3′), SEQ ID NO:2, and the 3′ primer (5′-ctggagctcgggccccggcccatttgccatactaattgcgg-3′), SEQ ID NO:3, were synthesized with XbaI and SacI restriction endonuclease recognition sites (bold) for amplification and cloning of the CTB-hinge coding sequence, SEQ ID NO:4, in a model 394 DNA/RNA Synthesizer (Applied Biosystems, Inc. Foster City, Calif. US)
  • the DNA sequence, SEQ ID NO:5, encoding the 21 amino acid leader peptide of the CTB was retained to direct the nascent CTB fusion peptide into the lumen of the ER.
  • the 3′ primer SEQ ID NO:3, was designed to contain a nucleotide sequence encoding a Gly-Pro box (Gly-Pro-Gly-Pro) with relatively less frequently used codons in plants to allow the ribosomes to halt for proper folding of CTB moiety before translation of the downstream message sequence.
  • Gly-Pro box Gly-Pro box
  • An additional function of the Gly-Pro box was to act as a flexible hinge between CTB and the conjugated peptide.
  • the methods for cloning the CTBH fusion gene, SEQ ID NO:4, into the multiple cloning site immediately downstream of the mas P2 promoter and the DNA sequence confirmation were as follows. PCR amplification was performed using a Gene Amp PCR System 9600, (The Perkin Elmer Corporation, Norwalk, Conn. US) according to the following reaction parameters; 94° C., 45 sec.: 55° C. for 60 sec.: 72° C. for 45 sec., 30 cycles total. The ligated vector and PCRed fragment (T4 ligase at 16° C.
  • Escherichia coli strain HB101 250 ⁇ FD, 200 ⁇ , and 2,500 volts; BioRad® Gene Pulser II unit (Bio Rad Laboratories, Inc., Hercules, Calif. US) and ampicillin resistant colonies were isolated after overnight growth at 37° C.
  • the plasmid was isolated from individual colonies of transformants and subjected to DNA sequence analysis with the forward primer (5′-accaatacattacactagcatctg-3′), SEQ ID NO:6, specific for the mas P2 promoter and the reverse primer (5′-gactgagtgcgatattatgtgtaatac-3′), SEQ ID NO:7, specific for the gene 7 poly(A) signal (model 373A DNA Sequencer, Applied Biosystems, Inc.).
  • This plant transformation vector was designated as pPCV701FM4-CTBH.
  • the 5′ oligonucleotide, SEQ ID NO:9, 5′-gccgagctcgataagttgactactagggagattgagcaagttgagttgttgaagaggatt-3′ and the 3′ oligonucleotide, SEQ ID NO: 10, 5′-gccgagctcacaactcatccttctcagaagtcaacttatcgtaaatcctcttcaacaact-3′ were designed to contain 17 bp complementary sequence for the thermostable vent DNA polymerase (New England Biolabs, Beverly, Mass. US ) attachment site for the initial cycle of the PCR reaction.
  • the 3′ oligonucletide, SEQ ID NO: 10, contained the DNA sequence encoding endoplasmic reticulum retention signal (SEKDEL) with codons most frequently found in potato plants. Both oligonucleotides contains SacI recognition sites (bold) to clone the synthetic gene fragment into SacI site immediately downstream of the hinge sequence of the vector to create vector pPCV701FM4-CTBH:NSP4.
  • SEKDEL endoplasmic reticulum retention signal
  • the shuttle vector was transferred into A. tumefaciens recipient strain GV3101 pMP90RK by the same electroporation conditions used for E. coli transformation.
  • A. tumefaciens transformants were grown at 29° C. on YEB solid medium containing the antibiotics carbenicillin (100 ⁇ g/ml), rifampicin (100 ⁇ g/ml), kanamycin (25 ⁇ g/ml), and gentamycin (25 ⁇ g/ml) for selection of transformants.
  • the plasmid was isolated from an A. tumefaciens transformant and transferred back into E. coli HB101 by electroporation, and restriction endonuclease analysis was used to confirm that no significant deletion had occurred in the vector. Structural confirmation of the plasmid was required because recombination events within the rec + A. tumefaciens strain could alter the T-DNA sequence. Transfer of the plasmid from A. tumefaciens back to the E. coli host was necessary because significant amounts of plasmid are difficult to isolate directly from A. tumefaciens . Agrobacteria carrying the plant expression vector were grown on YEB solid medium containing all four antibiotics for 48 hours at 29° C. and directly used for transformation of sterile potato leaf explants.
  • Sterile potato plants S. tuberosum cv. Bintje were grown in Magenta boxes (Sigma Chemical Co., St. Louis, Mo. US) on solid Murashige and Skoog (MS) complete organic medium (JRH Biosciences, Lenexa, KS US) containing 3.0% sucrose and 0.2% gelrite.
  • Leaf explants excised from the young plants were laterally bisected in a 9 cm diameter culture dish containing an overnight culture of A. tumefaciens suspension (1 ⁇ 10 10 cell/ml) harboring pPCV701FM4-CTBH:NSP4.
  • the bacterial suspension was supplemented with acetosyringone (370 ⁇ M) to increase transformation efficiency.
  • the explants were incubated in the bacterial suspension for 5 minutes, blotted on sterile filter paper, and transferred to MS solid medium, pH 5.7, containing 0.1 ⁇ g/ml naphthalene acetic acid (NAA) and 1.0 ⁇ g/ml trans-zeatin.
  • the leaf explants were then incubated for 48 hours at room temperature on MS solid medium to permit T-DNA transfer into the plant genome.
  • the leaf explants were transferred to MS solid medium containing the antibiotics kanamycin (100 ⁇ g/ml) and claforan (300 ⁇ g/ml).
  • Transformed plant cells formed calli on the selective medium after continuous incubation for 2 to 3 weeks at room temperature in a light room under cool white fluorescent tubes on a 12-hour photoperiod regime.
  • transformed calli grew to between 5 mm and 10 mm in diameter
  • the leaf tissue was transferred to MS medium containing 1.0 ⁇ g/ml trans-zeatin, 50 ⁇ g/ml kanamycin and 400 ⁇ g/ml claforan for shoot induction.
  • Regenerated shoots were excised and transferred to MS solid medium without plant hormones or antibiotics to stimulate root formation. Plantlets were allowed to grow and form microtubers under sterile conditions to characterization.
  • Luciferase activity was detected in transformed A. tumefaciens and transgenic plants as follows. The presence of the plant expression plasmid in agrobacteria, luxF gene expression under control of the mas P1 promoter was monitored by low-light image analysis. To perform the bioluminescent assay, bacterial culture grown for 24 hours on YEB solid culture medium was covered with a glass culture plate lid swabbed with substrate n-decyl aldehyde and analyzed by the Argus-100 intensified camera system (Hamamatsu Photonics UK Ltd., Bridgewater, N.J. US).
  • luxF gene was also monitored to confirm the presence of the CTBH:NSP4(114-135):SEKDEL, SEQ ID NO: 11, in the plant genome and to estimate the level of CTB fusion gene expression by mas P2 promoter.
  • Leaves excised from putative transformants were wounded by scalpel blade followed by incubation on MS solid medium containing naphthalene acetic acid (5 ⁇ g/ml) and 2,4-dichlorophenoxy acetic acid (6 ⁇ g/ml) for 48 hours. Light emission from the wounded leaf tissues was detected as described for agrobacteria.
  • More than forty independent kanamycin-resistant plants were regenerated from Agrobacterium mediated transformation of potato leaf explants with the plant expression vector pPCV701FM4-CTBH:NSP4. Three of the forty plants were found to express luciferase activities above background levels from untransformed plants. No luciferase activity was detected in leaves of untransformed potato plants.
  • the three transformed potato plants showing luciferase activities were analyzed for the presence of the fusion gene in plant genomic DNA isolated from young leaf tissues as follows. Genomic DNA was isolated from the transformed potato leaf tissues. Presence of the CTB fusion gene was determined by PCR analysis using the oligonucleotide primers specific for the T-DNA sequence. Transformed plant genomic DNA (500 ng) was used as a template to detect the CTB gene by PCR amplification (94° C. for 45 sec.: 55° C. for 60 sec.: 62° C. for 60 sec. for a total of 30 cycles). A 650 bp DNA fragment including both 5′ and 3′ flanking sequences of the fusion gene, was amplified. The PCR amplification was very specific probably due to high specificity of the primers used for the PCR reaction.
  • the DNA fragments amplified from plasmid vector pPCV701FM4-CTBH:NSP4 and from transgenic plant genomic DNA were identical in molecular weight. Although, identical amounts of template genomic DNA (500 ng) was used for the PCR reaction, the plant exhibiting the highest luciferase activity also demonstrated the highest level of PCR amplification.
  • the presence of the CTB fusion protein was detected in transformed potato tissues as follows.
  • Transgenic potato leaf and microtuber tissues were analyzed for the CTB fusion gene expression by immunoblot analysis.
  • Callus tissues were derived from leaf or tuber tissues incubated for 4 weeks on MS solid medium containing 5.0 mg/l NAA and 6.0 mg/1 2,4-D.
  • Tissues were homogenized by grinding by a mortar and pestle at 4° C. in extraction buffer (1:1 w/v) (200 mM Tris-Cl, pH 8.0, 100 mM NaCl, 400 mM sucrose, 10 mM EDTA, 14 mM 2-mercaptoethanol, 1 mM phenylmethylsulfonyl fluoride, 0.05% Tween-20).
  • tissue homogenate was centrifuged at 17,000 ⁇ g in a Beckman GS-15R centrifuge for 15 minutes at 4° C. to remove insoluble cell debris.
  • An aliquot of supernatant containing 100 ⁇ g of total soluble protein, as determined by Bradford protein assay (Bio Rad Laboratories, Inc.) was separated by 15% sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE) at 125 volts for 30 to 45 minutes in Tris-glycine buffer (25 mM Tris, 250 mM glycine, pH 8.3, 0.1% SDS). Samples were either loaded directly on the gel or boiled for 5 minutes prior to electrophoresis.
  • the separated protein bands were transferred from the gel to approximately 80 cm 2 Immun-Lite membranes (Bio Rad Laboratories, Inc.) by electroblotting on a semi-dry blotter (Labconco, Kansas City, Mo. US) for 60 minutes at 15 V and 100 mA.
  • Nonspecific antibody reactions were blocked by incubation of the membrane in 25 ml of 5% non-fat dry milk in TBS buffer (20 mM Tris pH 7.5 and 500 mM NaCl) for 1 hour with gentle agitation on a rotary shaker (40 rpm), followed by washing in TBS buffer for 5 minutes.
  • the membrane was incubated overnight at room temperature with gentle agitation in a 1:5,000 dilution of rabbit anti-cholera antiserum (Sigma C-3062) in TTBS antibody dilution buffer (TBS with 0.05% Tween-20 and 1% non-fat dry milk) followed by washing three times in TBST washing buffer (TBS with 0.05% Tween-20).
  • the membrane was incubated for 1 hour at room temperature with gentle agitation in a 1: 10,000 dilution of mouse anti-rabbit IgG conjugated with alkaline phosphatase (Sigma A-2556) in antibody dilution buffer.
  • the membrane was washed three times in TTBS buffer as before and once with TBS buffer, followed by incubation in 1 ⁇ chemiluminescent substrate CSPDTM (Bio Rad Laboratories, Inc.) for 5 minutes at room temperature with gentle agitation.
  • the membrane was then wrapped with transparent plastic membrane and placed in a photocassette on Kodak X-OMAT film (cat# 1651454).
  • the membrane was also used to image chemiluminescent light intensity in both the numerical and graphic form by the Argus-100 video image analysis.
  • the film was subjected to 1-10 minutes exposure and developed in a Kodak M35A X-OMAT Processor.
  • transgenic potato tuber tissues from the three transformed plants were shown to contain CTB fusion protein ( ⁇ 60 kDa) that strongly reacted with anti-cholera toxin antibody which predominantly recognized pentameric form of cholera toxin or its B subunit.
  • Potato plants transformed with the plant expression vector only which did not contain the CTBH:NSP4(114-135):SEKDEL sequence, SEQ ID NO:11, did not show this protein band.
  • One plant, designated Plant #1 showed approximately 3 to 5 fold higher chimeric protein level than the other two plants, Plant #2 and Plant #3.
  • Potato-synthesized CTB-NSP4 fusion peptide exhibited higher molecular weight than both pentameric bacterial CTB subunit (45 kDa) and potato-synthesized pentameric CTB subunit with ER retention signal (50 kDa).
  • the level of CTB fusion protein in the in tubers of transgenic Plant #1 was quantified using both chemiluminescent G M1 -ELISA and chemiluminescent immunoblot assays as follows. Pentameric CTB fusion protein levels in transgenic potato plants and its affinity for G M1 -ganglioside were evaluated by quantitative chemiluminescent G M1 -ELISA assays. The microtiter plate was coated with 100 ⁇ l/well of monosialoganglioside G M1 (3.0 ⁇ g/ml) (Sigma G-7641) in bicarbonate buffer, pH 9.6 (15 mM Na 2 CO 3 , 35mM NaHCO 3 ) and incubated at 4° C. overnight.
  • Wells were loaded with 100 ⁇ l/well of 10-fold serial dilutions of total soluble potato leaf or tuber protein in phosphate buffered saline (PBS) and incubated overnight at 4° C. The plate was washed three times in PBST (PBS containing 0.05% Tween-20). The wells were blocked by adding 300 ⁇ l/well of 1% bovine serum albumin (BSA) in PBS and incubated at 37° C. for 2 hours followed by washing three times with PBST.
  • PBS phosphate buffered saline
  • the wells were loaded with 100 ⁇ l/well of 1:5,000 dilution of rabbit anti-cholera toxin antibody (Sigma C-3062) and incubated for 2 hours at 37° C., followed by washing the wells three times with PBST.
  • the plate was incubated with 100 ⁇ l/well of 1:50,000 dilution of alkaline phosphatase-conjugated anti-rabbit IgG (Sigma A-2556) for 2 hours at 37° C. and washed three times with PBST.
  • the plate was finally incubated with 100 ⁇ l/well of Lumi-Phos® Plus (Lumigen, Inc. P-701) for 30 minutes at 37° C. and the enzyme-substrate reaction was measured in a MicroliteTM ML3000 Microtiter® Plate Luminometer (Dynatech Laboratories).
  • the amount of plant CTB fusion protein was measured by comparison of chemiluminescent intensities from a known amount of bacterial CTB protein-antibody complex with that emitted from a known amount of transformed plant soluble protein.
  • Two standard curves (1% and 0.1%) were generated based on the relative light units (RLU) measured for different amount of bacterial CTB.
  • the RLU generated from serial dilutions of transgenic potato plant homogenates were plotted into the graph, and found to reside within the 0. 1% and 0.01% curves, indicating that the fusion protein level in the transgenic potato tissue is slightly less than 0.1%.
  • chemiluminescent intensities of bacterial and plant CTB protein bands blotted on Immun-Lite membranes after SDS-PAGE were measured by the Argus-100 low-light imager Data Analysis Program.
  • the number of photons emitted from either bacterial CTB or plant CTB or plant CTB-NSP4 fusion protein bands were quantified, and their values compared to provide a semi-quantitative estimate of the amount of plant synthesized CTB fusion protein. Based on the amount of light emission detected from a known amount of bacterial CTB protein (100 ng), the amount of plant CTB fusion protein was calculated to be approximately 100 ng.
  • the percent of chimeric protein in the plant was calculated based on the amount of soluble plant protein (100 ⁇ g) used in the assay. Based on this method, the percent of plant CTB protein was found to be approximately 0.1% of total soluble plant protein, a value in close agreement with measurements made by the chemiluminescent G M1 -ELISA method. Based on the results of the chemiluminescent ELISA and immunoblot assays, 1 g of callus tissues (fresh weight) obtained from auxin-induced potato leaves contained 10 ⁇ g of recombinant plant CTB-NSP4 fusion protein.
  • Pentamerization of CTB subunits is essential for its affinity for the natural receptor.
  • plant-produced chimeric protein and bacterial CTB demonstrated a strong affinity for G M1 -ganglioside but not for BSA, which was the bases of protein production level measurement.
  • the ability of plant-derived CTB to bind G M1 -ganglioside indicates that the specific protein-ganglioside binding interactions between amino acid residues forming the G M1 binding sites and the oligosaccharide moiety of G M1 -ganglioside are conserved.
  • the strong binding efficiency of plant CTB conjugate for G M1 indicate that molecular configurations of CTB moiety is well conserved.
  • a transgenic plant producing a fusion protein comprising the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit and the ETEC fimbrial antigen CFA/I fused to the cholera toxin A2 subunit.
  • the immunodominant epitope of the murine rotavirus enterotoxin NSP4, the cholera toxin B subunit and the ETEC fimbrial antigen CFA/I function as antigens.
  • the cholera toxin B subunit functions as an antigen and as an adjuvant.
  • the cholera toxin A2 subunit functions as an adjuvant.
  • the transgenic plant can be administered to a mammal to immunize the mammal against cholera, rotavirus and enterotoxigenic E. coli infection simultaneously.
  • cholera toxin fusion proteins expressed in transformed potato tuber tissues assembled into a cholera holo-toxin-like oligomeric structure, which retained enterocyte membrane receptor G M1 -ganglioside binding affinity.
  • Both serum and intestinal antibodies against NSP4, CFA/I and CTB were induced in orally immunized mice.
  • Fluorescent antibody based cell sorting (FACS) analysis of immunized mouse spleen cells showed an increase in CD4+but not CD8+memory cell populations. Following rotavirus challenge, passively immunized mouse pups showed a 50% reduction of diarrhea symptoms.
  • the vector pPCV701CFA/I-CTB-NSP4 contained four genes located within the transferred DNA (T-DNA) sequence flanked by the right and left border (RB and LB), and 25 bp direct repeats required for integration of the T-DNA into plant genomic DNA.
  • the four genes were the CTBH:NSP4(114-135):SEKDEL coding sequence, SEQ ID NO: 11, under control of the mas P2 promoter; the CFA/I:CTA2 (SEQ ID NO: 12 and SEQ ID NO: 13) coding sequence under control of the mas P1 promoter; an NPT II expression cassette in the T-DNA to provide resistance to kanamycin in plants for selection of transformed plants; and a ⁇ -lactamase cassette for resistance to ampicillin in E. coli and carbenicillin in A. tumefaciens .
  • the g7pA polyadenylation signal was from the A. tumefaciens T L -DNA gene 7.
  • the OcspA polyadenylation signal is from the octopine synthase gene.
  • Each cholera toxin fusion gene contains its own leader sequence and an ER retention signal.
  • GPGP glycine-proline
  • the expression vector pPCV701CFA/I-CTB-NSP4 was assembled from the parental plasmid pPCV701 in the following manner.
  • the CTB gene, SEQ ID NO: 1 were amplified by polymerase chain reaction (PCR) from the cholera toxin (ctxAB) operon in plasmid pPt42.
  • the CTB 3′ primer, SEQ ID NO:3, was designed to contain an oligonucleotide encoding the tetrapeptide hinge (Gly-Pro-Gly-Pro) to incorporate a degree of flexibility between the CTB and NSP4 peptides.
  • a synthesized DNA fragment, SEQ ID NO:8, encoding the rotavirus enterotoxin NSP4 (114-135), epitope was inserted in frame between the CTB-hinge and the SEKDEL sequences.
  • the CTA leader sequence, SEQ ID NO: 14, and the CTA2 gene were amplified by PCR from the ctxAB operon and cloned into pPCv701 downstream of the mas P1 promoter region.
  • the whole CTA leader-CFA/I-CTA2 fusion gene is given as SEQ ID NO: 15.
  • the resultant plant expression vector pPCV701CFA/I-CTB-NSP4 was introduced into Agrobacterium tumefaciens strain GV3101 pMP90RK. From sterile plants grown in culture medium in a light room, potato (Solanum tuberosum cv. Bintje) leaf tissue explants were transformed with A. tumefaciens harboring the plant expression vector pPCV701 CFA/I-CTB-NSP4. Transformed plants were regenerated from the explants on selection medium containing kanamycin.
  • transgenic tubers Prior to analysis of antigen gene expression, transgenic tubers were stimulated to produce high levels of the antigen proteins by incubation of tuber slices on growth medium containing auxin 2,4-D (2,4 dichlorophenoxy acetic acid) for 4 days at room temperature.
  • the presence of the CFA/I and CTB-NSP4 fusion proteins in the transformed plants were detected by immunoblot techniques as follows. Protein extracts from auxin stimulated transformed potato tubers containing 100 mg of total soluble protein (TSP) were loaded on a 10-15% SDS-PAGE gel with or without 5 minutes boiling prior to electrophoresis. The separated protein bands were transferred to nitrocellulose membrane by electroblotting on a semi-dry blotter (Sigma) at 30V, 60 mA for one and a half hours.
  • TSP total soluble protein
  • CTB, NSP4 and CFA/I proteins were identified by incubation of the blot in rabbit anti-CTB antiserum (Sigma 1:5000 dilution) overnight at room temperature followed by incubation in alkaline phosphatase-conjugated mouse anti-rabbit IgG (Sigma, at 1:10,000 dilution) for 2 hours at room temperature. Finally the membrane was incubated in the substrate BCIP/NPT (Sigma) for 10 min. The color reaction was stopped by washing the membrane several times in distilled water.
  • the bacterial CTB assembled into an oligomeric structure with a molecular weight of 45 kD, characteristic of the CTB pentamer.
  • the transgenic plant produced CTB-NSP4 fusion peptide formed a 50 kDa oligomeric structure.
  • the 5 kDa increase in molecular mass is consistent with the presence of the additional NSP4 peptide and the 6 amino acid SEKDEL signal.
  • the plant sample containing both CFA/I-CTA2 and CTB-NSP4 fusion proteins showed the presence of a 70 kDa protein band, indicative of the insertion of CFA/I-CTA2 peptide into the CTB-NSP4 pentamer.
  • the untransformed plant showed no cross reaction with the cholera toxin antibody.
  • the bacterial CTB monomer has a molecular mass of 11 kDa.
  • the plant derived CTB-NSP4 multimer dissociated into an 18 kDa monomer which is consistent with the molecular mass of CTB plus NSP4.
  • CD-1 female mice each were fed 3 g transgenic potato tuber tissues containing a total of 7 mg of the recombinant fusion proteins previously determined by chemiluminescent ELISA on day 0, 5, 15, 23 and 56.
  • a group of 5 CD-1 mice each were fed 3 g of untransformed potato tuber tissues as a negative control.
  • CD1 mice 5 per group were gavaged with pure NSP4 peptide with or without pure bacterial CTB (adjuvant) according to the same oral inoculation schedule.
  • blood was taken from each mouse for serum antibody titer determination.
  • mice per group were euthanized at three different time points: 13, 34 and 68 days after the fifth immunization. Intestinal washings were collected for mucosal antibody detection. Spleen cells from both immunized and negative control mice (3 ⁇ 10 6 cells/well) were suspended in RPMI 1640 medium containing 10% fetal calf serum in duplicate samples, in 24 well tissue culture plates. After incubation for 72 hours at 37° C. in a humidified, 5% CO 2 incubator, supernatants from the spleen cell cultures were collected for assessment of IL-2, IL-4 and INFg secretion.
  • mice Following the five oral inoculations with transgenic potato tuber tissues, blood samples were collected and the serum anti CTB, NSP4 and CFA/I IgG titers were measured by ELISA methods used in our laboratory. Out of 10 mice, 8 generated serum IgG against CTB with a mean titer of 312.5 ⁇ 81.3. Of the 10 immunized mice, 8 developed serum IgG against NSP4 with a mean titer of 125 ⁇ 61.23. Out of the 10 immunized mice, 10 developed serum IgG against CFA/I with a mean titer of 84 ⁇ 44.2.
  • Intestinal IgG and IgA antibody titers against the three antigens were analyzed by chemiluminescent ELISA method used in our laboratory. Out of 10 immunized mice, 5 generated measurable intestinal anti-CTB antibody titers; 5 were found to have measurable intestinal anti-NSP4 antibody titers and 6 were found to have significant intestinal anti-CFA/I antibody titers. Negative control mice fed untransformed potato tuber tissues did not develop detectable specific serum or mucosal antibodies.
  • Adjuvant and carrier functions of CTB in the CTB-NSP4 fusion protein were determined by measuring serum anti-NSP4 antibody titers in mice from different vaccination groups. Mice fed the NSP4 peptide alone generated the lowest anti-NSP4 titer. Immunization with 7 mg of bacterial CTB (the same amount detected in the plant derived CTB-NSP4 fusion protein) increased the serum anti-NSP4 IgG titer approximately two fold. Mice fed 3 g transformed potato tuber tissues containing the CTB-NSP4 fusion protein developed the highest anti-NSP4 titer.
  • Directly linking small antigens with CTB subunit not only results in specific targeting of the antigens to the mucosal immune system via specific enterocyte attachment but also increases the local antigen concentration at the mucosal surface, which may explain our detection of the strongest immune response directed against the CTB-NSP4 fusion protein.
  • T lymphocyte populations in immunized mice were analyzed in immunized mice as follows.
  • IL-2, IL-4 and INFg produced in the spleen cell culture supernatants were assayed by ELISA.
  • Spleen lymphocytes were stained with fluorochrome-labeled monoclonal antibodies (mAb) for immunophenotyping.
  • mAb fluorochrome-labeled monoclonal antibodies
  • Two monoclonal antibody panels were constructed for three color analysis (fluoresceinisothiocanate (FITC), phycoerythrin (PE), and Cy-Chrome).
  • the first combination used CD62L*FITC/CD4*PE/CD44*Cy-Chrome designates naive and memory T helper cells.
  • the second combination CD62L*FITC/CD8b.2*PE/CD44*Cy-Chrome designates naive and memory cytotoxic T cells.
  • the spleen cells were resuspended at 10 6 cells/ml in PBS and stained with fluorochrome-labeled mAbs.
  • the labeled cells were analyzed by fluorescene activated cell sorting (FACS) to determine the T lymphocyte memory cell sub-populations.
  • FACS fluorescene activated cell sorting
  • the II-2 and the INFg expression levels in spleen cells dramatically increased, reaching the highest level 34 days after the fifth immunization and decreasing to basal levels by 68 days after vaccination. Throughout this time period IL-4 levels remained low equivalent to that found in unimmunized mice.
  • a cytokine expression pattern clearly indicated a Th1 lymphocyte mediated immune response generated by feeding mice the plant derived cholera toxin fusion antigens. Therefore, the overall cytokine secretion pattern of this multicomponent plant vaccine indicates a strong Th1 response.
  • mice Protection against rotavirus was evaluated as follows.
  • Adult female CD-1 mice (five per group) were fed 3 g of untransformed or transgenic potato tuber slices once a week for four weeks.
  • the mice were mated with uninfected males.
  • mouse pups were born to the immunized dams.
  • each pup received one oral dose of simian rotavirus SA-11 in 50 ul PBS that contained 15 DD 50 (the virus dose determined empirically to cause diarrhea in 50% of the mouse pups).
  • the mice were examined for the presence of diarrhea daily for 5 days following inoculation by gentle palpation of their abdomen to produce fecal pellets. The diarrhea score and the proportion of mice showing diarrhea symptoms in each study group were recorded.
  • a method of inducing partial or complete immunity to an infectious disease in a mammal comprises providing to the mammal for oral consumption an effective amount of a fusion protein according to the present invention.
  • the fusion protein is made in a transgenic plant.
  • the fusion protein comprises a multimeric a cholera toxin B subunit and a first immunogenic antigen from a causal factor of the disease.
  • the fusion protein additionally comprises a second immunogenic antigen from a causal factor of a mammalian disease fused to a cholera toxin subunit, such as cholera toxin subunit A2.
  • the cholera toxin subunits act as adjuvants for the immunogenic antigens and, in the case of cholera toxin B subunit, also act as an immunogenic antigen against cholera infection.
  • the fusion protein can be provided to the mammal in a dose and frequency sufficient to render the mammal partially or completely immune from the first infectious disease, the second infection disease, cholera or a combination of the preceding.
  • the specific dose and frequency are determined by well-known techniques as will be understood by those with skill in the art with reference to this disclosure.

Abstract

A method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of a protein complex comprising five monomeric fusion proteins.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • The present application is a divisional of U.S. patent application Ser. No. 11/370,697, titled “Transgenic Plant-Based Vaccines,” filed Mar. 7, 2006, now allowed, which is a continuation-in-part of U.S. patent application Ser. No. 10/920,648, titled “Methods and Substances for Preventing and Treating Autoimmune Disease,” filed Aug. 17, 2004, now abandoned, which is a continuation of U.S. patent application Ser. No. 09/296,981, titled “Methods and Substances for Preventing and Treating Autoimmune Disease,” filed Apr. 22, 1999, now U.S. Pat. No. 6,777,546, issued on Aug. 17, 2004, which: 1) claims the benefit of U.S. Patent Application No. 60/082,688, titled “Plant Vaccines Against Autoimmune Disease,” filed April 22, 1998 and 2) is a continuation-in-part of U.S. patent application Ser. No. 09/167,493, titled “Expression of Cholera Toxin B Subunit in Transgenic Plants and Efficacy Thereof in Oral Vaccines,” filed Oct. 7, 1998, now abandoned, which claims the benefit of U.S. Provisional Patent Application No. 60/061,265, titled “Cholera Toxin in Food Plants,” filed Oct. 7, 1997; and U.S. patent application No. 11/370,697 is a continuation of U.S. patent application Ser. No. 11/001,153, titled “Transgenic Plant-based Vaccines,” filed Nov. 30, 2004, now abandoned, which is a continuation of U.S. patent application Ser. No. 10/245,749, titled “Transgenic Plant-based Vaccines,” filed Sep. 16, 2002, now abandoned, which is a divisional of U.S. patent application Ser. No. 09/771,536, filed Jan. 29, 2001, now abandoned, which claims the benefit of U.S. Provisional Patent Application No. 60/178,403, titled “Production of a Cholera Toxin B Subunit-rotavirus NSP4 Enterotoxin Fusion Protein in Potato,” filed Jan. 27, 2000; the contents of which are incorporated by reference herein in their entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • This invention was made with United States Government support under subcontract number 010-FY97-LLU-LANGRIDGE with the National Medical Test Bed, United States Department of the Army. The United States Government has certain rights in this invention.
  • BACKGROUND
  • Acute infectious enteric diseases, such as acute gastroenteritis, are second only to acute respiratory diseases as a cause of human death worldwide. Cholera, rotavirus and enterotoxigenic E. coli are the three major causative agents of acute gastroenteritis. Human rotavirus, for example, is the most important cause of infantile gastroenteritis worldwide. This virus has a tremendous public health impact worldwide, infecting nearly every child in the first few years of life. Rotavirus infection is responsible for approximately 1 million deaths each year and an estimated 18 million hospitalizations. 20% to 40% of the hospitalizations are for childhood diarrhea, which makes the rotavirus the most important single cause of diarrheal mortality among children.
  • Treatment for acute gastroenteritis includes antibiotics and metabolic support. However, adequate treatment is often not available, particularly in lesser developed areas where the incidence of acute gastroenteritis is highest. Prevention of acute gastroenteritis would be preferable to treatment. However, preventative measures, such as the provision of safe drinking water, are often inadequate or unavailable.
  • Therefore, it would be useful to have a new method for the prevention of acute gastroenteritis. Further, it would be particularly useful to have a method for the prevention of acute gastroenteritis which would prevent multiple types of acute gastroenteritis simultaneously.
  • SUMMARY
  • According to one embodiment of the present invention, there is provided a DNA construct that encodes, upon expression in a plant cell, a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease. The first immunogenic antigen can be a rotavirus antigen. The first immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • The fusion protein encoded by the DNA construct can further comprise a second cholera toxin subunit. The second cholera toxin subunit can be cholera toxin A2 subunit.
  • The fusion protein encoded by the DNA construct can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease. The second immunogenic antigen can be a rotavirus antigen. The second immunogenic antigen can also be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • According to another embodiment of the present invention, there is provided a DNA construct that encodes, upon expression in a plant cell, a fusion protein comprising a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a first mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease. The first immunogenic antigen can be a rotavirus antigen. The second immunogenic antigen can be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • According to another embodiment of the present invention, there is provided an expression vector comprising a DNA construct of the present invention, a transgenic plant cell transformed with a DNA construct of the present invention, a transgenic plant seed transformed with the DNA construct of the present invention, and a transgenic plant transformed with the DNA construct of the present invention.
  • According to yet another embodiment of the present invention, there is provided a method of producing an immunogen in a plant comprising cultivating a transgenic plant of the present invention under conditions effective to express the fusion protein.
  • According to another embodiment of the present invention, there is provided a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of a plant of the present invention.
  • The present invention also includes means for producing, in a plant cell, a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease. The means can comprise a DNA construct that encodes, upon expression in the plant cell, a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a first mammalian disease. The first immunogenic antigen can be a rotavirus antigen. The first immunogenic antigen can also be an enterotoxigenic E. coli antigen. The fusion protein can further comprise a second cholera toxin subunit, such as cholera toxin A2 subunit. The fusion protein can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease. The second immunogenic antigen can be a rotavirus antigen. The second immunogenic antigen can also be an enterotoxigenic E. coli antigen.
  • The present invention also includes means for producing, in a plant cell, a fusion protein comprising a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a first mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease. The first immunogenic antigen can be a rotavirus antigen. The second immunogenic antigen can be an enterotoxigenic E. coli antigen.
  • According to another embodiment of the present invention, there is provided an expression vector comprising the means of the present invention, a transgenic plant cell transformed with means of the present invention, a transgenic plant seed transformed with the means of the present invention, and a transgenic plant transformed with the means of the present invention.
  • The present invention also includes a method of producing an immunogen in a plant comprising cultivating the transgenic plant of the present invention under conditions effective to express the fusion protein. The present invention further includes a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of a plant of the present invention.
  • According to another embodiment of the present invention, there is provided a fusion protein comprising a multimeric cholera toxin B subunit and a first immunogenic antigen from a causal factor of a mammalian disease. The first immunogenic antigen can be a rotavirus antigen. The first immunogenic antigen can also be an enterotoxigenic E. coli antigen. The fusion protein can further comprise a second cholera toxin subunit. The second cholera toxin subunit can be cholera toxin A2 subunit. The fusion protein can further comprise a second immunogenic antigen from a causal factor of a second mammalian disease. The second immunogenic antigen can be a rotavirus antigen. The second immunogenic antigen can also be an enterotoxigenic E. coli antigen. Either the first mammalian disease or the second mammalian disease or both can be an infectious enteric disease.
  • In one embodiment, the fusion protein comprises a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease.
  • According to another embodiment of the present invention, there is provided a fusion protein encoded by the DNA construct of the present invention.
  • According to another embodiment of the present invention, there is provided a method of inducing partial or complete immunity to an infectious disease in a mammal comprising providing to the mammal for oral consumption an effective amount of the fusion protein of the present invention.
  • FIGURES
  • The features, aspects and advantages of the present invention will become better understood with regard to the following description, appended claims and accompanying figures where:
  • FIG. 1 is a diagram of the vector pPCV701FM4-CTB:NSP4; and
  • FIG. 2 is a diagram of the vector pPCV701CFA/I-CTB-NSP4.
  • DESCRIPTION
  • According to one embodiment of the present invention, there is provided a method of inducing partial or complete immunity to an infectious disease, such as gastroenteritis, in a mammal by administering to the mammal a portion of a transgenic plant comprising a fusion protein, where the fusion protein comprises at least one cholera toxin subunit and an immunogenic antigen from a causal factor of the disease. In a preferred embodiment, the fusion protein comprises at least two cholera toxin subunits, at least one of which functions as an antigen, in addition to functioning as an adjuvant for the immunogenic antigen. In another preferred embodiment, the fusion protein comprises at least two immunogenic antigens, each fused to a cholera toxin subunit. By fusing the immunogenic antigen to the cholera toxin subunit, the fusion protein more specifically targets the appropriate immune system tissue upon administration. This increased specificity compensates for the low level of production of the protein in the transgenic plant and increases the response of the mammal's immune system.
  • In one embodiment, the fusion protein comprises the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit (CTB). In another embodiment, the fusion protein comprises the enterotoxigenic E. coli (ETEC) fimbrial colonization factor CFA/I fused to the cholera toxin A2 subunit (CTA2). In yet another embodiment, the fusion protein comprises both the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit, and the fusion protein comprises the enterotoxigenic E. coli fimbrial colonization factor CFA/I fused to the cholera toxin A2 subunit.
  • Though the method is described in the context of preventing gastroenteritis by way of example, it will be understood by those with skill in the art with reference to this disclosure, that the present method can be used to prevent other enteric infectious diseases and other non-enteric infectious diseases such as respiratory diseases. The method will now be described in more detail.
  • 1) Construction of a transgenic plant producing a fusion protein comprising the immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit and confirmation of transformation.
  • According to one embodiment of the present invention, there is provided a transgenic plant producing a fusion protein comprising the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit. The transgenic plant can be administered to a mammal to immunize the mammal against cholera and rotavirus infection simultaneously.
  • Referring now to FIG. 1, there is shown a diagram of the vector used to prepare the transgenic plant. As can be seen, the vector contained four genes located within the transferred DNA (T-DNA) sequence flanked by the right and left border (RB and LB), and 25 bp direct repeats of the borders required for integration of the transferred DNA into plant genomic DNA. The four genes were the CTBH:NSP4(114-135):SEKDEL coding sequence under control of the mas P2 promoter; the bacterial luciferase AB fusion gene (luxF) under control of the mas P1 promoter used as a detectable marker; an NPT II expression cassette used for resistance to kanamycin in plants; and a β-lactamase cassette for resistance to ampicillin in E. coli and carbenicillin in A. tumefaciens. The g7pA polyadenylation signal was from the A. tumefaciens TL-DNA gene 7. The OcspA polyadenylation signal is from the octopine synthase gene. Pnos was the promoter of the nopaline synthase gene. g4pA was the polyadenylation signal from TL-DNA gene 4. OriT was the origin of transfer derived from pRK2. OriV was the wide host range origin of replication for multiplication of the plasmid in A. tumefaciens derived from pRK2. Ori pBR322 was the replication origin of pBR322 for maintenance of the plasmid in E. coli.
  • The vector pPCV701FM4-CTB:NSP4 was constructed as follows. The plant expression vector pPCV701FM4, a derivative of plasmid pPCV701, was digested with XbaI and SacI restriction endonucleases within the multiple cloning site to insert a gene encoding the cholera toxin B subunit and its leader sequence, SEQ ID NO: 1, from plasmid pRT42 containing the ctxAB operon. The oligonucleotide 5′ primer (5′-gctctagagccaccatgattaaattaaaatttggtg-3′), SEQ ID NO:2, and the 3′ primer (5′-ctggagctcgggccccggcccatttgccatactaattgcgg-3′), SEQ ID NO:3, were synthesized with XbaI and SacI restriction endonuclease recognition sites (bold) for amplification and cloning of the CTB-hinge coding sequence, SEQ ID NO:4, in a model 394 DNA/RNA Synthesizer (Applied Biosystems, Inc. Foster City, Calif. US)
  • The oligonucleotide sequence surrounding the translation initiation codon of the CTB gene, SEQ ID NO: 1, was altered to a preferred nucleotide context for translation in eukaryotic cells, (5′-gccacc-3′)and a putative Shine-Dalgarno sequence (AGGA) present in the ctxAB operon in plasmid pPt42 was removed. The DNA sequence, SEQ ID NO:5, encoding the 21 amino acid leader peptide of the CTB was retained to direct the nascent CTB fusion peptide into the lumen of the ER.
  • The 3′ primer, SEQ ID NO:3, was designed to contain a nucleotide sequence encoding a Gly-Pro box (Gly-Pro-Gly-Pro) with relatively less frequently used codons in plants to allow the ribosomes to halt for proper folding of CTB moiety before translation of the downstream message sequence. An additional function of the Gly-Pro box was to act as a flexible hinge between CTB and the conjugated peptide.
  • The methods for cloning the CTBH fusion gene, SEQ ID NO:4, into the multiple cloning site immediately downstream of the mas P2 promoter and the DNA sequence confirmation were as follows. PCR amplification was performed using a Gene Amp PCR System 9600, (The Perkin Elmer Corporation, Norwalk, Conn. US) according to the following reaction parameters; 94° C., 45 sec.: 55° C. for 60 sec.: 72° C. for 45 sec., 30 cycles total. The ligated vector and PCRed fragment (T4 ligase at 16° C. for 20 hrs.) were electroporated into Escherichia coli strain HB101 (250 μFD, 200 Ω, and 2,500 volts; BioRad® Gene Pulser II unit (Bio Rad Laboratories, Inc., Hercules, Calif. US) and ampicillin resistant colonies were isolated after overnight growth at 37° C.
  • To confirm the presence of the correct CTBH fusion gene sequence, SEQ ID NO:4, in transformed E. coli cells, the plasmid was isolated from individual colonies of transformants and subjected to DNA sequence analysis with the forward primer (5′-accaatacattacactagcatctg-3′), SEQ ID NO:6, specific for the mas P2 promoter and the reverse primer (5′-gactgagtgcgatattatgtgtaatac-3′), SEQ ID NO:7, specific for the gene 7 poly(A) signal (model 373A DNA Sequencer, Applied Biosystems, Inc.). This plant transformation vector was designated as pPCV701FM4-CTBH.
  • To insert the rotavirus enterotoxin NSP4(114-135) epitope gene, SEQ ID NO:8, two overlapping primer sequences were synthesized and equimolar amounts of both single-stranded deoxyribonucleotide fragments were subjected to PCR amplification (94° C. 45 sec.: 55° C. for 60 sec.: 72° C. for 60 sec.: 30 cycles total) to create double stranded 103 bp length synthetic gene. The 5′ oligonucleotide, SEQ ID NO:9, 5′-gccgagctcgataagttgactactagggagattgagcaagttgagttgttgaagaggatt-3′ and the 3′ oligonucleotide, SEQ ID NO: 10, 5′-gccgagctcacaactcatccttctcagaagtcaacttatcgtaaatcctcttcaacaact-3′ were designed to contain 17 bp complementary sequence for the thermostable vent DNA polymerase (New England Biolabs, Beverly, Mass. US ) attachment site for the initial cycle of the PCR reaction. The 3′ oligonucletide, SEQ ID NO: 10, contained the DNA sequence encoding endoplasmic reticulum retention signal (SEKDEL) with codons most frequently found in potato plants. Both oligonucleotides contains SacI recognition sites (bold) to clone the synthetic gene fragment into SacI site immediately downstream of the hinge sequence of the vector to create vector pPCV701FM4-CTBH:NSP4.
  • Following confirmation of the correct fusion gene sequence, CTBH:NSP4(114-135): SEKDEL, SEQ ID NO: 11, the shuttle vector was transferred into A. tumefaciens recipient strain GV3101 pMP90RK by the same electroporation conditions used for E. coli transformation. A. tumefaciens transformants were grown at 29° C. on YEB solid medium containing the antibiotics carbenicillin (100 μg/ml), rifampicin (100 μg/ml), kanamycin (25 μg/ml), and gentamycin (25 μg/ml) for selection of transformants.
  • The plasmid was isolated from an A. tumefaciens transformant and transferred back into E. coli HB101 by electroporation, and restriction endonuclease analysis was used to confirm that no significant deletion had occurred in the vector. Structural confirmation of the plasmid was required because recombination events within the rec+ A. tumefaciens strain could alter the T-DNA sequence. Transfer of the plasmid from A. tumefaciens back to the E. coli host was necessary because significant amounts of plasmid are difficult to isolate directly from A. tumefaciens. Agrobacteria carrying the plant expression vector were grown on YEB solid medium containing all four antibiotics for 48 hours at 29° C. and directly used for transformation of sterile potato leaf explants.
  • Sterile potato plants S. tuberosum cv. Bintje were grown in Magenta boxes (Sigma Chemical Co., St. Louis, Mo. US) on solid Murashige and Skoog (MS) complete organic medium (JRH Biosciences, Lenexa, KS US) containing 3.0% sucrose and 0.2% gelrite. Leaf explants excised from the young plants were laterally bisected in a 9 cm diameter culture dish containing an overnight culture of A. tumefaciens suspension (1×1010 cell/ml) harboring pPCV701FM4-CTBH:NSP4. The bacterial suspension was supplemented with acetosyringone (370 μM) to increase transformation efficiency. The explants were incubated in the bacterial suspension for 5 minutes, blotted on sterile filter paper, and transferred to MS solid medium, pH 5.7, containing 0.1 μg/ml naphthalene acetic acid (NAA) and 1.0 μg/ml trans-zeatin. The leaf explants were then incubated for 48 hours at room temperature on MS solid medium to permit T-DNA transfer into the plant genome. For selection of transformed plant cells and for counter selection against continued Agrobacterium growth, the leaf explants were transferred to MS solid medium containing the antibiotics kanamycin (100 μg/ml) and claforan (300 μg/ml).
  • Transformed plant cells formed calli on the selective medium after continuous incubation for 2 to 3 weeks at room temperature in a light room under cool white fluorescent tubes on a 12-hour photoperiod regime. When transformed calli grew to between 5 mm and 10 mm in diameter, the leaf tissue was transferred to MS medium containing 1.0 μg/ml trans-zeatin, 50 μg/ml kanamycin and 400 μg/ml claforan for shoot induction. Regenerated shoots were excised and transferred to MS solid medium without plant hormones or antibiotics to stimulate root formation. Plantlets were allowed to grow and form microtubers under sterile conditions to characterization.
  • Luciferase activity was detected in transformed A. tumefaciens and transgenic plants as follows. The presence of the plant expression plasmid in agrobacteria, luxF gene expression under control of the mas P1 promoter was monitored by low-light image analysis. To perform the bioluminescent assay, bacterial culture grown for 24 hours on YEB solid culture medium was covered with a glass culture plate lid swabbed with substrate n-decyl aldehyde and analyzed by the Argus-100 intensified camera system (Hamamatsu Photonics UK Ltd., Bridgewater, N.J. US).
  • Expression of luxF gene was also monitored to confirm the presence of the CTBH:NSP4(114-135):SEKDEL, SEQ ID NO: 11, in the plant genome and to estimate the level of CTB fusion gene expression by mas P2 promoter. Leaves excised from putative transformants were wounded by scalpel blade followed by incubation on MS solid medium containing naphthalene acetic acid (5 μg/ml) and 2,4-dichlorophenoxy acetic acid (6 μg/ml) for 48 hours. Light emission from the wounded leaf tissues was detected as described for agrobacteria.
  • More than forty independent kanamycin-resistant plants were regenerated from Agrobacterium mediated transformation of potato leaf explants with the plant expression vector pPCV701FM4-CTBH:NSP4. Three of the forty plants were found to express luciferase activities above background levels from untransformed plants. No luciferase activity was detected in leaves of untransformed potato plants.
  • The three transformed potato plants showing luciferase activities were analyzed for the presence of the fusion gene in plant genomic DNA isolated from young leaf tissues as follows. Genomic DNA was isolated from the transformed potato leaf tissues. Presence of the CTB fusion gene was determined by PCR analysis using the oligonucleotide primers specific for the T-DNA sequence. Transformed plant genomic DNA (500 ng) was used as a template to detect the CTB gene by PCR amplification (94° C. for 45 sec.: 55° C. for 60 sec.: 62° C. for 60 sec. for a total of 30 cycles). A 650 bp DNA fragment including both 5′ and 3′ flanking sequences of the fusion gene, was amplified. The PCR amplification was very specific probably due to high specificity of the primers used for the PCR reaction.
  • The DNA fragments amplified from plasmid vector pPCV701FM4-CTBH:NSP4 and from transgenic plant genomic DNA were identical in molecular weight. Although, identical amounts of template genomic DNA (500 ng) was used for the PCR reaction, the plant exhibiting the highest luciferase activity also demonstrated the highest level of PCR amplification.
  • The presence of the CTB fusion protein was detected in transformed potato tissues as follows. Transgenic potato leaf and microtuber tissues were analyzed for the CTB fusion gene expression by immunoblot analysis. Callus tissues were derived from leaf or tuber tissues incubated for 4 weeks on MS solid medium containing 5.0 mg/l NAA and 6.0 mg/1 2,4-D. Tissues were homogenized by grinding by a mortar and pestle at 4° C. in extraction buffer (1:1 w/v) (200 mM Tris-Cl, pH 8.0, 100 mM NaCl, 400 mM sucrose, 10 mM EDTA, 14 mM 2-mercaptoethanol, 1 mM phenylmethylsulfonyl fluoride, 0.05% Tween-20). The tissue homogenate was centrifuged at 17,000×g in a Beckman GS-15R centrifuge for 15 minutes at 4° C. to remove insoluble cell debris. An aliquot of supernatant containing 100 μg of total soluble protein, as determined by Bradford protein assay (Bio Rad Laboratories, Inc.), was separated by 15% sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE) at 125 volts for 30 to 45 minutes in Tris-glycine buffer (25 mM Tris, 250 mM glycine, pH 8.3, 0.1% SDS). Samples were either loaded directly on the gel or boiled for 5 minutes prior to electrophoresis.
  • The separated protein bands were transferred from the gel to approximately 80 cm2 Immun-Lite membranes (Bio Rad Laboratories, Inc.) by electroblotting on a semi-dry blotter (Labconco, Kansas City, Mo. US) for 60 minutes at 15 V and 100 mA. Nonspecific antibody reactions were blocked by incubation of the membrane in 25 ml of 5% non-fat dry milk in TBS buffer (20 mM Tris pH 7.5 and 500 mM NaCl) for 1 hour with gentle agitation on a rotary shaker (40 rpm), followed by washing in TBS buffer for 5 minutes. The membrane was incubated overnight at room temperature with gentle agitation in a 1:5,000 dilution of rabbit anti-cholera antiserum (Sigma C-3062) in TTBS antibody dilution buffer (TBS with 0.05% Tween-20 and 1% non-fat dry milk) followed by washing three times in TBST washing buffer (TBS with 0.05% Tween-20). The membrane was incubated for 1 hour at room temperature with gentle agitation in a 1: 10,000 dilution of mouse anti-rabbit IgG conjugated with alkaline phosphatase (Sigma A-2556) in antibody dilution buffer. The membrane was washed three times in TTBS buffer as before and once with TBS buffer, followed by incubation in 1×chemiluminescent substrate CSPD™ (Bio Rad Laboratories, Inc.) for 5 minutes at room temperature with gentle agitation. The membrane was then wrapped with transparent plastic membrane and placed in a photocassette on Kodak X-OMAT film (cat# 1651454). (The membrane was also used to image chemiluminescent light intensity in both the numerical and graphic form by the Argus-100 video image analysis.) The film was subjected to 1-10 minutes exposure and developed in a Kodak M35A X-OMAT Processor.
  • Using this method, transgenic potato tuber tissues from the three transformed plants were shown to contain CTB fusion protein (˜60 kDa) that strongly reacted with anti-cholera toxin antibody which predominantly recognized pentameric form of cholera toxin or its B subunit. Potato plants transformed with the plant expression vector only, which did not contain the CTBH:NSP4(114-135):SEKDEL sequence, SEQ ID NO:11, did not show this protein band. One plant, designated Plant #1, showed approximately 3 to 5 fold higher chimeric protein level than the other two plants, Plant #2 and Plant #3. Potato-synthesized CTB-NSP4 fusion peptide exhibited higher molecular weight than both pentameric bacterial CTB subunit (45 kDa) and potato-synthesized pentameric CTB subunit with ER retention signal (50 kDa).
  • The level of CTB fusion protein in the in tubers of transgenic Plant #1 was quantified using both chemiluminescent GM1-ELISA and chemiluminescent immunoblot assays as follows. Pentameric CTB fusion protein levels in transgenic potato plants and its affinity for GM1-ganglioside were evaluated by quantitative chemiluminescent GM1-ELISA assays. The microtiter plate was coated with 100 μl/well of monosialoganglioside GM1 (3.0 μg/ml) (Sigma G-7641) in bicarbonate buffer, pH 9.6 (15 mM Na2CO3, 35mM NaHCO3) and incubated at 4° C. overnight. Wells were loaded with 100 μl/well of 10-fold serial dilutions of total soluble potato leaf or tuber protein in phosphate buffered saline (PBS) and incubated overnight at 4° C. The plate was washed three times in PBST (PBS containing 0.05% Tween-20). The wells were blocked by adding 300 μl/well of 1% bovine serum albumin (BSA) in PBS and incubated at 37° C. for 2 hours followed by washing three times with PBST. The wells were loaded with 100 μl/well of 1:5,000 dilution of rabbit anti-cholera toxin antibody (Sigma C-3062) and incubated for 2 hours at 37° C., followed by washing the wells three times with PBST. The plate was incubated with 100 μl/well of 1:50,000 dilution of alkaline phosphatase-conjugated anti-rabbit IgG (Sigma A-2556) for 2 hours at 37° C. and washed three times with PBST. The plate was finally incubated with 100 μl/well of Lumi-Phos® Plus (Lumigen, Inc. P-701) for 30 minutes at 37° C. and the enzyme-substrate reaction was measured in a Microlite™ ML3000 Microtiter® Plate Luminometer (Dynatech Laboratories).
  • In the chemiluminescent GM1-ELISA method, the amount of plant CTB fusion protein was measured by comparison of chemiluminescent intensities from a known amount of bacterial CTB protein-antibody complex with that emitted from a known amount of transformed plant soluble protein. Two standard curves (1% and 0.1%) were generated based on the relative light units (RLU) measured for different amount of bacterial CTB. The RLU generated from serial dilutions of transgenic potato plant homogenates were plotted into the graph, and found to reside within the 0. 1% and 0.01% curves, indicating that the fusion protein level in the transgenic potato tissue is slightly less than 0.1%.
  • In the chemiluminescent immunoblot method, luminescent intensities of bacterial and plant CTB protein bands blotted on Immun-Lite membranes after SDS-PAGE were measured by the Argus-100 low-light imager Data Analysis Program. The number of photons emitted from either bacterial CTB or plant CTB or plant CTB-NSP4 fusion protein bands were quantified, and their values compared to provide a semi-quantitative estimate of the amount of plant synthesized CTB fusion protein. Based on the amount of light emission detected from a known amount of bacterial CTB protein (100 ng), the amount of plant CTB fusion protein was calculated to be approximately 100 ng. The percent of chimeric protein in the plant was calculated based on the amount of soluble plant protein (100 μg) used in the assay. Based on this method, the percent of plant CTB protein was found to be approximately 0.1% of total soluble plant protein, a value in close agreement with measurements made by the chemiluminescent GM1-ELISA method. Based on the results of the chemiluminescent ELISA and immunoblot assays, 1 g of callus tissues (fresh weight) obtained from auxin-induced potato leaves contained 10 μg of recombinant plant CTB-NSP4 fusion protein.
  • Pentamerization of CTB subunits is essential for its affinity for the natural receptor. In GM1-ELISA binding assays, plant-produced chimeric protein and bacterial CTB demonstrated a strong affinity for GM1-ganglioside but not for BSA, which was the bases of protein production level measurement. The ability of plant-derived CTB to bind GM1-ganglioside indicates that the specific protein-ganglioside binding interactions between amino acid residues forming the GM1 binding sites and the oligosaccharide moiety of GM1-ganglioside are conserved. The strong binding efficiency of plant CTB conjugate for GM1 indicate that molecular configurations of CTB moiety is well conserved. In addition, the absence of a monomeric form of chimera by immunoblot analysis indicates that predominant molecular species of chimeric protein is in the pentameric form, because monomeric CTB is unable to bind to GM1-ganglioside. Therefore, the monomeric B subunit fusion polypeptide accumulated within the lumen of the ER of plant cells and self-assembly into pentameric GM1 binding forms took place.
  • 2) Method of Construction of a transgenic plant producing a fusion protein comprising the immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit and the ETEC fimbrial antigen CFA/I fused to the cholera toxin A2 subunit and confirmation of transformation.
  • According to another embodiment of the present invention, there is provided a transgenic plant producing a fusion protein comprising the twenty-two amino acid immunodominant epitope of the murine rotavirus enterotoxin NSP4 fused to the cholera toxin B subunit and the ETEC fimbrial antigen CFA/I fused to the cholera toxin A2 subunit. The immunodominant epitope of the murine rotavirus enterotoxin NSP4, the cholera toxin B subunit and the ETEC fimbrial antigen CFA/I function as antigens. The cholera toxin B subunit functions as an antigen and as an adjuvant. The cholera toxin A2 subunit functions as an adjuvant. The transgenic plant can be administered to a mammal to immunize the mammal against cholera, rotavirus and enterotoxigenic E. coli infection simultaneously.
  • As disclosed in greater detail below, the cholera toxin fusion proteins expressed in transformed potato tuber tissues assembled into a cholera holo-toxin-like oligomeric structure, which retained enterocyte membrane receptor GM1-ganglioside binding affinity. Both serum and intestinal antibodies against NSP4, CFA/I and CTB were induced in orally immunized mice. Analysis of IL-2, IL-4 and INFg cytokine levels in spleen cells isolated from immunized mice indicated the presence of a strong Th1 immune response to the plant synthesized antigens. Fluorescent antibody based cell sorting (FACS) analysis of immunized mouse spleen cells showed an increase in CD4+but not CD8+memory cell populations. Following rotavirus challenge, passively immunized mouse pups showed a 50% reduction of diarrhea symptoms.
  • Referring now to FIG. 2, there is shown a diagram of the vector used to prepare the transgenic plant. As can be seen, the vector pPCV701CFA/I-CTB-NSP4 contained four genes located within the transferred DNA (T-DNA) sequence flanked by the right and left border (RB and LB), and 25 bp direct repeats required for integration of the T-DNA into plant genomic DNA. The four genes were the CTBH:NSP4(114-135):SEKDEL coding sequence, SEQ ID NO: 11, under control of the mas P2 promoter; the CFA/I:CTA2 (SEQ ID NO: 12 and SEQ ID NO: 13) coding sequence under control of the mas P1 promoter; an NPT II expression cassette in the T-DNA to provide resistance to kanamycin in plants for selection of transformed plants; and a β-lactamase cassette for resistance to ampicillin in E. coli and carbenicillin in A. tumefaciens. The g7pA polyadenylation signal was from the A. tumefaciens TL-DNA gene 7. The OcspA polyadenylation signal is from the octopine synthase gene. Each cholera toxin fusion gene contains its own leader sequence and an ER retention signal. To increase the flexibility of the fusion protein, a four amino acid glycine-proline (GPGP) hinge region was inserted between the CTB and NSP4 peptides.
  • The expression vector pPCV701CFA/I-CTB-NSP4 was assembled from the parental plasmid pPCV701 in the following manner. A nucleotide sequence encoding the endoplasmic reticulum (ER) retention signal, SEKDEL, was first cloned into the plant expression vector pPCV701on the P2 site of the mannopine synthase (mas) dual P1, P2 promoter. The CTB gene, SEQ ID NO: 1, were amplified by polymerase chain reaction (PCR) from the cholera toxin (ctxAB) operon in plasmid pPt42. The CTB 3′ primer, SEQ ID NO:3, was designed to contain an oligonucleotide encoding the tetrapeptide hinge (Gly-Pro-Gly-Pro) to incorporate a degree of flexibility between the CTB and NSP4 peptides. A synthesized DNA fragment, SEQ ID NO:8, encoding the rotavirus enterotoxin NSP4 (114-135), epitope was inserted in frame between the CTB-hinge and the SEKDEL sequences. The CTA leader sequence, SEQ ID NO: 14, and the CTA2 gene were amplified by PCR from the ctxAB operon and cloned into pPCv701 downstream of the mas P1 promoter region. A DNA fragment, (431 bp), SEQ ID NO: 12, encoding the enterotoxigenic E. coli colonization factor CFA/I, was amplified from plasmid pIGx15A, and was inserted in frame between the CTA leader sequence, SEQ ID NO: 14, and the CTA2 gene, SEQ ID NO: 13. The whole CTA leader-CFA/I-CTA2 fusion gene is given as SEQ ID NO: 15.
  • The resultant plant expression vector pPCV701CFA/I-CTB-NSP4, was introduced into Agrobacterium tumefaciens strain GV3101 pMP90RK. From sterile plants grown in culture medium in a light room, potato (Solanum tuberosum cv. Bintje) leaf tissue explants were transformed with A. tumefaciens harboring the plant expression vector pPCV701 CFA/I-CTB-NSP4. Transformed plants were regenerated from the explants on selection medium containing kanamycin. Prior to analysis of antigen gene expression, transgenic tubers were stimulated to produce high levels of the antigen proteins by incubation of tuber slices on growth medium containing auxin 2,4-D (2,4 dichlorophenoxy acetic acid) for 4 days at room temperature.
  • The presence of the CFA/I and CTB-NSP4 fusion proteins in the transformed plants were detected by immunoblot techniques as follows. Protein extracts from auxin stimulated transformed potato tubers containing 100 mg of total soluble protein (TSP) were loaded on a 10-15% SDS-PAGE gel with or without 5 minutes boiling prior to electrophoresis. The separated protein bands were transferred to nitrocellulose membrane by electroblotting on a semi-dry blotter (Sigma) at 30V, 60 mA for one and a half hours. The location of CTB, NSP4 and CFA/I proteins were identified by incubation of the blot in rabbit anti-CTB antiserum (Sigma 1:5000 dilution) overnight at room temperature followed by incubation in alkaline phosphatase-conjugated mouse anti-rabbit IgG (Sigma, at 1:10,000 dilution) for 2 hours at room temperature. Finally the membrane was incubated in the substrate BCIP/NPT (Sigma) for 10 min. The color reaction was stopped by washing the membrane several times in distilled water.
  • The bacterial CTB assembled into an oligomeric structure with a molecular weight of 45 kD, characteristic of the CTB pentamer. The transgenic plant produced CTB-NSP4 fusion peptide formed a 50 kDa oligomeric structure. The 5 kDa increase in molecular mass is consistent with the presence of the additional NSP4 peptide and the 6 amino acid SEKDEL signal. The plant sample containing both CFA/I-CTA2 and CTB-NSP4 fusion proteins showed the presence of a 70 kDa protein band, indicative of the insertion of CFA/I-CTA2 peptide into the CTB-NSP4 pentamer. The untransformed plant showed no cross reaction with the cholera toxin antibody. Immersion of the samples in boiling water for 5 minutes resulted in dissociation of the multimeric structures into monomers. The bacterial CTB monomer has a molecular mass of 11 kDa. The plant derived CTB-NSP4 multimer dissociated into an 18 kDa monomer which is consistent with the molecular mass of CTB plus NSP4.
  • 3) Method of Immunizing a Mammal Against Infectious Diseases and Analysis of the Results.
  • A group of 10 CD-1 female mice each were fed 3 g transgenic potato tuber tissues containing a total of 7 mg of the recombinant fusion proteins previously determined by chemiluminescent ELISA on day 0, 5, 15, 23 and 56. Using the same feeding schedule, a group of 5 CD-1 mice each were fed 3 g of untransformed potato tuber tissues as a negative control. To evaluate the adjuvant effect of the CTB protein in the CTB-NSP4 fusion, CD1 mice (5 per group) were gavaged with pure NSP4 peptide with or without pure bacterial CTB (adjuvant) according to the same oral inoculation schedule. On day 13 after the final immunization, blood was taken from each mouse for serum antibody titer determination. Three mice per group were euthanized at three different time points: 13, 34 and 68 days after the fifth immunization. Intestinal washings were collected for mucosal antibody detection. Spleen cells from both immunized and negative control mice (3×106 cells/well) were suspended in RPMI 1640 medium containing 10% fetal calf serum in duplicate samples, in 24 well tissue culture plates. After incubation for 72 hours at 37° C. in a humidified, 5% CO2 incubator, supernatants from the spleen cell cultures were collected for assessment of IL-2, IL-4 and INFg secretion.
  • Following the five oral inoculations with transgenic potato tuber tissues, blood samples were collected and the serum anti CTB, NSP4 and CFA/I IgG titers were measured by ELISA methods used in our laboratory. Out of 10 mice, 8 generated serum IgG against CTB with a mean titer of 312.5±81.3. Of the 10 immunized mice, 8 developed serum IgG against NSP4 with a mean titer of 125±61.23. Out of the 10 immunized mice, 10 developed serum IgG against CFA/I with a mean titer of 84±44.2.
  • Intestinal IgG and IgA antibody titers against the three antigens were analyzed by chemiluminescent ELISA method used in our laboratory. Out of 10 immunized mice, 5 generated measurable intestinal anti-CTB antibody titers; 5 were found to have measurable intestinal anti-NSP4 antibody titers and 6 were found to have significant intestinal anti-CFA/I antibody titers. Negative control mice fed untransformed potato tuber tissues did not develop detectable specific serum or mucosal antibodies. Since the CTB pentamer can bind to GM1 ganglioside located on the mucosal epithelial cell surface, induction of both systemic and mucosal antibodies in the immunized mice indicated the successful delivery of the cholera toxin fusion proteins to the GALT.
  • Adjuvant and carrier functions of CTB in the CTB-NSP4 fusion protein were determined by measuring serum anti-NSP4 antibody titers in mice from different vaccination groups. Mice fed the NSP4 peptide alone generated the lowest anti-NSP4 titer. Immunization with 7 mg of bacterial CTB (the same amount detected in the plant derived CTB-NSP4 fusion protein) increased the serum anti-NSP4 IgG titer approximately two fold. Mice fed 3 g transformed potato tuber tissues containing the CTB-NSP4 fusion protein developed the highest anti-NSP4 titer.
  • Small soluble proteins like the NSP4 22 amino acid epitope that are highly imunogenic by parenteral routes are frequently ineffective when administered orally unless a large dose of the protein is used. This result can be attributed to intestinal digestion and lack of tropism of the peptide for the gut associated lymphoid tissues. Either cholera holotoxin or the CTB subunit, which function as mucosal adjuvants can stimulate an immune response against co-administered protein antigens. Directly linking small antigens with CTB subunit not only results in specific targeting of the antigens to the mucosal immune system via specific enterocyte attachment but also increases the local antigen concentration at the mucosal surface, which may explain our detection of the strongest immune response directed against the CTB-NSP4 fusion protein.
  • The T lymphocyte populations in immunized mice were analyzed in immunized mice as follows. IL-2, IL-4 and INFg produced in the spleen cell culture supernatants were assayed by ELISA. Spleen lymphocytes were stained with fluorochrome-labeled monoclonal antibodies (mAb) for immunophenotyping. Two monoclonal antibody panels were constructed for three color analysis (fluoresceinisothiocanate (FITC), phycoerythrin (PE), and Cy-Chrome).
  • The first combination used, CD62L*FITC/CD4*PE/CD44*Cy-Chrome designates naive and memory T helper cells. The second combination, CD62L*FITC/CD8b.2*PE/CD44*Cy-Chrome designates naive and memory cytotoxic T cells. The spleen cells were resuspended at 106 cells/ml in PBS and stained with fluorochrome-labeled mAbs. The labeled cells were analyzed by fluorescene activated cell sorting (FACS) to determine the T lymphocyte memory cell sub-populations.
  • Following multiple oral immunizations, the II-2 and the INFg expression levels in spleen cells dramatically increased, reaching the highest level 34 days after the fifth immunization and decreasing to basal levels by 68 days after vaccination. Throughout this time period IL-4 levels remained low equivalent to that found in unimmunized mice. Thus, a cytokine expression pattern clearly indicated a Th1 lymphocyte mediated immune response generated by feeding mice the plant derived cholera toxin fusion antigens. Therefore, the overall cytokine secretion pattern of this multicomponent plant vaccine indicates a strong Th1 response. FACS analysis of spleen cells collected on day 13, 34 and 68 after the last immunization showed an elevated population of CD4+ memory cells in comparison with the unimmunized mice through the two months after immunization. The CD4+ memory cell subpopulation (CD62 CD44+, gate R4) detected in the immunized mice was observed to be significantly higher than the CD4+ memory cell subset in unimmunized mice. Thus, the generation of a significantly increased T helper memory cell population in the immunized mice indicated successful protective immunization mediated by the plant delivered antigens. The existence of increased numbers of memory cells provided the ability to mount a strong immune response following a second encounter with the same pathogen. The CD8+ memory cell population detected in immunized mice did not show any significant increase over the unimmunized mouse negative control group.
  • Protection against rotavirus was evaluated as follows. Adult female CD-1 mice (five per group) were fed 3 g of untransformed or transgenic potato tuber slices once a week for four weeks. Immediately following the fourth immunization at maximum anti-NSP4 antibody titer, the mice were mated with uninfected males. After a 19-20 day gestation period, mouse pups were born to the immunized dams. On day 6 post parturition, each pup received one oral dose of simian rotavirus SA-11 in 50 ul PBS that contained 15 DD50 (the virus dose determined empirically to cause diarrhea in 50% of the mouse pups). The mice were examined for the presence of diarrhea daily for 5 days following inoculation by gentle palpation of their abdomen to produce fecal pellets. The diarrhea score and the proportion of mice showing diarrhea symptoms in each study group were recorded.
  • The number of pups which developed diarrhea symptoms and the duration of the diarrhea was significantly reduced in the pups passively immunized with CTB-NSP4 fusion protein in comparison with pups born to unimmunized dams. On day 3 after rotavirus challenge, a 50% reduction of diarrhea symptoms was detected in the immunized pups. Complete resolution of diarrhea symptoms occurred 4 days after virus challenge in pups from immunized dams. To exclude the possibility of diarrhea reduction due to the presence of anti-CTB antibodies, pups born to dams immunized with plant derived CTB only were also challenged with an identical dose of rotavirus SA11. No reduction of diarrhea symptoms was detected in mice immunized with plant derived CTB alone. This experiment demonstrated that anti-NSP4 antibodies generated in orally immunized mice were passed on to the pups and protected them from the onset of rotavirus infection as well as significantly reducing the duration of the virus infection.
  • Therefore, according to one embodiment of the present invention, there is provided a method of inducing partial or complete immunity to an infectious disease in a mammal. The method comprises providing to the mammal for oral consumption an effective amount of a fusion protein according to the present invention. Preferably, the fusion protein is made in a transgenic plant. Further preferably, the fusion protein comprises a multimeric a cholera toxin B subunit and a first immunogenic antigen from a causal factor of the disease. In a preferred embodiment, the fusion protein additionally comprises a second immunogenic antigen from a causal factor of a mammalian disease fused to a cholera toxin subunit, such as cholera toxin subunit A2. The cholera toxin subunits act as adjuvants for the immunogenic antigens and, in the case of cholera toxin B subunit, also act as an immunogenic antigen against cholera infection.
  • The fusion protein can be provided to the mammal in a dose and frequency sufficient to render the mammal partially or completely immune from the first infectious disease, the second infection disease, cholera or a combination of the preceding. The specific dose and frequency are determined by well-known techniques as will be understood by those with skill in the art with reference to this disclosure.
  • Although the present invention has been discussed in considerable detail with reference to certain preferred embodiments, other embodiments are possible. Therefore, the scope of the appended claims should not be limited to the description of preferred embodiments contained in this disclosure. All references cited herein are incorporated by reference in their entirety.

Claims (30)

1. A method of producing an immunogen in a plant comprising cultivating a transgenic plant transformed with a DNA construct that encodes, upon expression in a plant cell, a protein complex comprising a multimeric cholera toxin B subunit and a an immunogenic antigen from a causal factor of a first mammalian disease under conditions effective to express the protein complex.
2. A method of producing an immunogen in a plant comprising cultivating a transgenic plant transformed with a DNA construct that encodes, upon expression in a plant cell, a protein complex comprising a cholera toxin A2 subunit, a multimeric cholera toxin B subunit, a first immunogenic antigen from a causal factor of a first mammalian disease, and a second immunogenic antigen from a causal factor of a second mammalian disease under conditions effective to express the protein complex.
3. The method of claim 1, where the multimeric cholera toxin B subunit comprises five monomeric fusion proteins.
4. The method of claim 3, where the immunogenic antigen is NSP4 antigen from rotavirus, and where each fusion protein further comprises a cholera toxin B subunit linked to an NSP4 antigen from rotavirus.
5. The method of claim 3, where the protein complex further comprises ETEC fimbrial antigen CFA/1 from enterotoxigenic E. coli.
6. The method of claim 1, where the DNA construct comprises a bidirectional promoter controlling transcription of the DNA construct.
7. A plant produced according to the method of claim 1.
8. A plant produced according to the method of claim 3.
9. A plant produced according to the method of claim 4.
10. A plant produced according to the method of claim 5.
11. A plant produced according to the method of claim 6.
12. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 7.
13. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 8.
14. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 9.
15. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 10.
16. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 11.
17. The method of claim 2, where the multimeric cholera toxin B subunit comprises five monomeric fusion proteins.
18. The method of claim 17, where the first immunogenic antigen is NSP4 antigen from rotavirus, and where each fusion protein further comprises a cholera toxin B subunit linked to an NSP4 antigen from rotavirus.
19. The method of claim 17, where the second immuogenic antigen is ETEC fimbrial antigen CFA/1 from enterotoxigenic E. coli.
20. The method of claim 2, where the DNA construct comprises a bidirectional promoter controlling transcription of the DNA construct.
21. A plant produced according to the method of claim 2.
22. A plant produced according to the method of claim 17.
23. A plant produced according to the method of claim 18.
24. A plant produced according to the method of claim 19.
25. A plant produced according to the method of claim 20.
26. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 21.
27. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 22.
28. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 23.
29. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 24.
30. A method for the prevention of multiple types of acute gastroenteritis simultaneously in a mammal comprising providing to the mammal for oral consumption an effective amount of the plant of claim 25.
US12/190,383 1997-10-07 2008-08-12 Transgenic plant-based vaccines Abandoned US20090081256A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/190,383 US20090081256A1 (en) 1997-10-07 2008-08-12 Transgenic plant-based vaccines

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US6126597P 1997-10-07 1997-10-07
US8268898P 1998-04-22 1998-04-22
US16749398A 1998-10-07 1998-10-07
US09/296,981 US6777546B2 (en) 1997-10-07 1999-04-22 Methods and substances for preventing and treating autoimmune disease
US17840300P 2000-01-27 2000-01-27
US09/771,536 US20030165543A1 (en) 2000-01-27 2001-01-29 Transgenic plant-based vaccines
US10/245,749 US20030021803A1 (en) 2000-01-27 2002-09-16 Transgenic plant-based vaccines
US10/920,648 US20050044588A1 (en) 1997-10-07 2004-08-17 Methods and substances for preventing and treating autoimmune disease
US11/001,153 US20050186219A1 (en) 1997-10-07 2004-11-30 Transgenic plant-based vaccines
US11/370,697 US7422747B2 (en) 1997-10-07 2006-03-07 Transgenic plant-based vaccines
US12/190,383 US20090081256A1 (en) 1997-10-07 2008-08-12 Transgenic plant-based vaccines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/370,697 Division US7422747B2 (en) 1997-10-07 2006-03-07 Transgenic plant-based vaccines

Publications (1)

Publication Number Publication Date
US20090081256A1 true US20090081256A1 (en) 2009-03-26

Family

ID=46324005

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/370,697 Expired - Fee Related US7422747B2 (en) 1997-10-07 2006-03-07 Transgenic plant-based vaccines
US12/190,383 Abandoned US20090081256A1 (en) 1997-10-07 2008-08-12 Transgenic plant-based vaccines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/370,697 Expired - Fee Related US7422747B2 (en) 1997-10-07 2006-03-07 Transgenic plant-based vaccines

Country Status (1)

Country Link
US (2) US7422747B2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110076513A1 (en) * 2009-09-28 2011-03-31 National Taiwan University Transparent conductive films and fabrication methods thereof
US10160789B2 (en) 2011-03-14 2018-12-25 University Of Louisville Research Foundation, Inc. Polypeptides having immunoactivating activity and methods of producing the same
CN109096403A (en) * 2018-07-04 2018-12-28 浙江工商大学 A kind of protein carrier and its preparation method and application for protein transduction
US11524062B2 (en) 2015-06-29 2022-12-13 University Of Louisville Research Foundation, Inc. Compositions and methods for treating cancer and promoting wound healing

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014047625A1 (en) * 2012-09-24 2014-03-27 Montana State University Recombinant lactococcus lactis expressing escherichia coli colonization factor antigen i (cfa/i) fimbriae and their methods of use

Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4879113A (en) * 1987-10-27 1989-11-07 Smith Clyde K Vaccine for immunologically mediated nutritional disturbances associated with plant protein antigens
US4956282A (en) * 1985-07-29 1990-09-11 Calgene, Inc. Mammalian peptide expression in plant cells
US5079165A (en) * 1988-06-29 1992-01-07 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit in S. typhi
US5098998A (en) * 1987-04-29 1992-03-24 President And Fellows Of Harvard College Cholera vaccines and peptides
US5268276A (en) * 1988-09-16 1993-12-07 Jan Holmgren Recombinant systems for expression of cholera B-sub-unit with the aid of foreign promoters and/or leader peptides
US5308835A (en) * 1984-07-09 1994-05-03 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit
US5484719A (en) * 1991-08-26 1996-01-16 Edible Vaccines, Inc. Vaccines produced and administered through edible plants
US5589384A (en) * 1991-06-11 1996-12-31 Glaxo Wellcome Inc. Fusion proteins
US5612487A (en) * 1991-08-26 1997-03-18 Edible Vaccines, Inc. Anti-viral vaccines expressed in plants
US5628994A (en) * 1983-03-04 1997-05-13 The University Of Maryland Vibrio cholerae strain CVD103Hgr, method of making same, and vaccines derived therefrom
US5654184A (en) * 1988-09-06 1997-08-05 Washington University Oral immunization by transgenic plants
US5681571A (en) * 1993-10-08 1997-10-28 Duotol Ab Immunological tolerance-inducing agent
US5698416A (en) * 1995-06-02 1997-12-16 The United States Of America As Represented By The Secretary Of The Army Methods for production of antigens under control of temperature-regulated promotors in enteric bacteria
US5789245A (en) * 1993-09-15 1998-08-04 Chiron Corporation Alphavirus structural protein expression cassettes
US5869057A (en) * 1995-06-07 1999-02-09 Rock; Edwin P. Recombinant vaccines to break self-tolerance
US5891432A (en) * 1997-07-29 1999-04-06 The Immune Response Corporation Membrane-bound cytokine compositions comprising GM=CSF and methods of modulating an immune response using same
US5914114A (en) * 1995-06-02 1999-06-22 The United States Of America As Represented By The Secretary Of The Army Method of raising antibodies against E. coli of the family CS4-CFA/I
US5917026A (en) * 1996-02-05 1999-06-29 Loewenadler; Bjoern Fusion proteins of immunopotentiating activity
US6030624A (en) * 1996-08-16 2000-02-29 Uab Research Foundation Mucosal immunogens for novel vaccines
US6036953A (en) * 1996-11-29 2000-03-14 The General Hospital Corporation Heterologous antigens in live cell V. cholerae strains
US6110724A (en) * 1996-10-18 2000-08-29 The Research Foundation For Microbial Diseases Of Osaka University Rotavirus antigen, vaccine and diagnostic agent for rotavirus infections, and a method for producing the antigen
US6194560B1 (en) * 1994-10-24 2001-02-27 Texas A & M University System Oral immunization with transgenic plants
US6210682B1 (en) * 1995-06-14 2001-04-03 Baylor College Of Medicine Rotavirus enterotoxin NSP4 and methods of using same
US6395964B1 (en) * 1995-10-24 2002-05-28 The Texas A&M University System Oral immunization with transgenic plants
US6777546B2 (en) * 1997-10-07 2004-08-17 Loma Linda University Methods and substances for preventing and treating autoimmune disease

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR910014790A (en) 1990-01-10 1991-08-31 한태희 Function Selection Method and Interface Circuit of Laptop Computer
NO175188C (en) 1990-06-27 1994-09-14 Sjur Olsnes Process for Preparing a Peptide Conjugate with the ability to penetrate cell cytosol
WO1993010246A1 (en) 1991-11-15 1993-05-27 Board Of Regents, The University Of Texas System Membrane expression of heterologous genes
GB9319429D0 (en) 1993-09-21 1993-11-03 London Health Ass Methods and products for controlling immune responses in mammals
GB9319722D0 (en) 1993-09-24 1993-11-10 John Innes Foundation Plant genetic manipulation
WO1996011708A1 (en) 1994-10-18 1996-04-25 Board Of Regents, The University Of Texas System Membrane expression of heterologous genes
DE69637130T2 (en) 1995-02-20 2008-02-21 The Walter And Eliza Hall Institute Of Medical Research, Parkville IMMUNOREACTIVE AND IMMUNOTHERAPEUTIC MOLECULES THAT INTERACT IN INDIVIDUALS WITH INSULIN-DEPENDENT DIABETES MELLITUS
AU750623B2 (en) 1998-04-22 2002-07-25 Loma Linda University Methods and substances for preventing and treating autoimmune disease
US6589529B1 (en) * 1998-10-30 2003-07-08 Children's Hospital Medical Center Rotavirus subunit vaccine

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5628994A (en) * 1983-03-04 1997-05-13 The University Of Maryland Vibrio cholerae strain CVD103Hgr, method of making same, and vaccines derived therefrom
US5308835A (en) * 1984-07-09 1994-05-03 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit
US4956282A (en) * 1985-07-29 1990-09-11 Calgene, Inc. Mammalian peptide expression in plant cells
US5098998A (en) * 1987-04-29 1992-03-24 President And Fellows Of Harvard College Cholera vaccines and peptides
US4879113A (en) * 1987-10-27 1989-11-07 Smith Clyde K Vaccine for immunologically mediated nutritional disturbances associated with plant protein antigens
US5079165A (en) * 1988-06-29 1992-01-07 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit in S. typhi
US5679880A (en) * 1988-09-06 1997-10-21 Washington University Oral immunization by transgenic plants
US5686079A (en) * 1988-09-06 1997-11-11 Washington University Oral immunization by transgenic plants
US5654184A (en) * 1988-09-06 1997-08-05 Washington University Oral immunization by transgenic plants
US5268276A (en) * 1988-09-16 1993-12-07 Jan Holmgren Recombinant systems for expression of cholera B-sub-unit with the aid of foreign promoters and/or leader peptides
US5589384A (en) * 1991-06-11 1996-12-31 Glaxo Wellcome Inc. Fusion proteins
US5612487A (en) * 1991-08-26 1997-03-18 Edible Vaccines, Inc. Anti-viral vaccines expressed in plants
US5484719A (en) * 1991-08-26 1996-01-16 Edible Vaccines, Inc. Vaccines produced and administered through edible plants
US6015694A (en) * 1993-09-15 2000-01-18 Chiron Corporation Method for stimulating an immune response utilizing recombinant alphavirus particles
US5789245A (en) * 1993-09-15 1998-08-04 Chiron Corporation Alphavirus structural protein expression cassettes
US5681571A (en) * 1993-10-08 1997-10-28 Duotol Ab Immunological tolerance-inducing agent
US6194560B1 (en) * 1994-10-24 2001-02-27 Texas A & M University System Oral immunization with transgenic plants
US5698416A (en) * 1995-06-02 1997-12-16 The United States Of America As Represented By The Secretary Of The Army Methods for production of antigens under control of temperature-regulated promotors in enteric bacteria
US5914114A (en) * 1995-06-02 1999-06-22 The United States Of America As Represented By The Secretary Of The Army Method of raising antibodies against E. coli of the family CS4-CFA/I
US5869057A (en) * 1995-06-07 1999-02-09 Rock; Edwin P. Recombinant vaccines to break self-tolerance
US6210682B1 (en) * 1995-06-14 2001-04-03 Baylor College Of Medicine Rotavirus enterotoxin NSP4 and methods of using same
US6395964B1 (en) * 1995-10-24 2002-05-28 The Texas A&M University System Oral immunization with transgenic plants
US5917026A (en) * 1996-02-05 1999-06-29 Loewenadler; Bjoern Fusion proteins of immunopotentiating activity
US6030624A (en) * 1996-08-16 2000-02-29 Uab Research Foundation Mucosal immunogens for novel vaccines
US6110724A (en) * 1996-10-18 2000-08-29 The Research Foundation For Microbial Diseases Of Osaka University Rotavirus antigen, vaccine and diagnostic agent for rotavirus infections, and a method for producing the antigen
US6036953A (en) * 1996-11-29 2000-03-14 The General Hospital Corporation Heterologous antigens in live cell V. cholerae strains
US5891432A (en) * 1997-07-29 1999-04-06 The Immune Response Corporation Membrane-bound cytokine compositions comprising GM=CSF and methods of modulating an immune response using same
US6777546B2 (en) * 1997-10-07 2004-08-17 Loma Linda University Methods and substances for preventing and treating autoimmune disease

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110076513A1 (en) * 2009-09-28 2011-03-31 National Taiwan University Transparent conductive films and fabrication methods thereof
US8329253B2 (en) * 2009-09-28 2012-12-11 National Taiwan University Method for forming a transparent conductive film by atomic layer deposition
US10160789B2 (en) 2011-03-14 2018-12-25 University Of Louisville Research Foundation, Inc. Polypeptides having immunoactivating activity and methods of producing the same
US11524062B2 (en) 2015-06-29 2022-12-13 University Of Louisville Research Foundation, Inc. Compositions and methods for treating cancer and promoting wound healing
CN109096403A (en) * 2018-07-04 2018-12-28 浙江工商大学 A kind of protein carrier and its preparation method and application for protein transduction

Also Published As

Publication number Publication date
US7422747B2 (en) 2008-09-09
US20060277635A1 (en) 2006-12-07

Similar Documents

Publication Publication Date Title
Arakawa et al. Expression of cholera toxin B subunit oligomers in transgenic potato plants
Gunasekaran et al. A review on edible vaccines and their prospects
Arakawa et al. A plant-based cholera toxin B subunit–insulin fusion protein protects against the development of autoimmune diabetes
Arakawa et al. Efficacy of a food plant-based oral cholera toxin B subunit vaccine
US5686079A (en) Oral immunization by transgenic plants
Waheed et al. Plastid expression of a double‐pentameric vaccine candidate containing human papillomavirus‐16 L1 antigen fused with LTB as adjuvant: transplastomic plants show pleiotropic phenotypes
US20050241024A1 (en) Transgenic plant-based vaccines
JP2008161199A (en) Method for stimulating immunological response by administering host organism expressing intimin singly or as fused protein with one or more other antigen
Arakawa et al. Synthesis of a cholera toxin B subunit-rotavirus NSP4 fusion protein in potato
Kim et al. Assembly of cholera toxin B subunit full-length rotavirus NSP4 fusion protein oligomers in transgenic potato
Kim et al. HIV-1 gp120 V3 cholera toxin B subunit fusion gene expression in transgenic potato
US7422747B2 (en) Transgenic plant-based vaccines
AU2001234592B2 (en) Transgenic plant-based vaccines
Arakawa et al. Food plant-delivered cholera toxin B subunit for vaccination and immunotolerization
Choi et al. Synthesis and assembly of a cholera toxin B subunit-rotavirus VP7 fusion protein in transgenic potato
US20030191076A1 (en) Prime-boost vaccination strategy
Yu et al. Novel approaches to oral vaccines: delivery of antigens by edible plants
WO1999018225A1 (en) Expression of cholera toxin b subunit in transgenic plants and efficacy thereof in oral vaccines
JP2008500008A (en) Chimeric carrier molecules for mucosal vaccine generation
Kim et al. Synthesis of an HIV-1 Tat transduction domain-rotavirus enterotoxin fusion protein in transgenic potato
Karimi et al. Immunogenicity of EIT chimeric protein expressed in transplastomic tobacco plants towards development of an oral vaccine against Escherichia coli O157: H7
US20050186219A1 (en) Transgenic plant-based vaccines
Lee et al. Plant-synthesized E. coli CFA/I fimbrial protein protects Caco-2 cells from bacterial attachment
JP4769977B2 (en) Vaccine gene introduction rice
Kim et al. Synthesis and assembly of SIV mac Gag p27 capsid protein cholera toxin B subunit fusion protein in transgenic potato

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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