WO1996015241A2 - A polynucleotide tuberculosis vaccine - Google Patents

A polynucleotide tuberculosis vaccine Download PDF

Info

Publication number
WO1996015241A2
WO1996015241A2 PCT/US1995/014899 US9514899W WO9615241A2 WO 1996015241 A2 WO1996015241 A2 WO 1996015241A2 US 9514899 W US9514899 W US 9514899W WO 9615241 A2 WO9615241 A2 WO 9615241A2
Authority
WO
WIPO (PCT)
Prior art keywords
dna
polynucleotide
functional equivalents
mycobacterial
antigen
Prior art date
Application number
PCT/US1995/014899
Other languages
French (fr)
Other versions
WO1996015241A3 (en
Inventor
Margaret A. Liu
Donna Montgomery
Jeffrey Ulmer
Jean Content
Kris Huygen
Original Assignee
Merck & Co., Inc.
N.V. Innogenetics S.A.
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 to CA 2205175 priority Critical patent/CA2205175C/en
Priority to AU41102/96A priority patent/AU715067B2/en
Application filed by Merck & Co., Inc., N.V. Innogenetics S.A. filed Critical Merck & Co., Inc.
Priority to NZ296477A priority patent/NZ296477A/en
Priority to SK597-97A priority patent/SK283254B6/en
Priority to PL95320091A priority patent/PL184839B1/en
Priority to JP51633096A priority patent/JP3881014B2/en
Priority to CZ19971451A priority patent/CZ289383B6/en
Priority to HU9701841A priority patent/HU222369B1/en
Priority to DK95939161T priority patent/DK0792358T3/en
Priority to DE69534250T priority patent/DE69534250T2/en
Priority to AT95939161T priority patent/ATE296882T1/en
Priority to EP95939161A priority patent/EP0792358B1/en
Publication of WO1996015241A2 publication Critical patent/WO1996015241A2/en
Publication of WO1996015241A3 publication Critical patent/WO1996015241A3/en
Priority to NO972196A priority patent/NO972196L/en
Priority to FI972034A priority patent/FI972034A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/04Mycobacterium, e.g. Mycobacterium tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • C07K14/35Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Mycobacteriaceae (F)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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

Definitions

  • CTLs cytotoxic T-lymphocytes
  • CTLs kill virally- or bacterially-infected cells when their T cell receptors recognize foreign peptides associated with MHC class I and/or class II molecules. These peptides can be derived from endogenously synthesized foreign proteins, regardless of the protein's location or function within the pathogen. By recognition of epitopes from conserved proteins, CTLs may provide heterologous protection. In the case of intracellular bacteria, proteins secreted by or released from the bacteria are processed and presented by MHC class I and II molecules, thereby generating T-cell responses that may play a role in reducing or eliminating infection. Most efforts to generate CTL responses have either used replicating vectors to produce the protein antigen within the cell [J.R. Bennink et al, ibid.
  • Retroviral vectors have restrictions on the size and structure of polypeptides that can be expressed as fusion proteins while maintaining the ability of the recombinant virus to replicate [A.D. Miller, Curr. Top. Microbiol. Immunol.
  • WO 93/17706 describes a method for vaccinating an animal against a virus, wherein carrier particles were coated with a gene construct and the coated particles are accelerated into cells of an animal.
  • Studies by Wolff et al. (supra) originally demonstrated that intramuscular injection of plasmid DNA encoding a reporter gene results in the expression of that gene in myocytes at and near the site of injection.
  • Recent reports demonstrated the successful immunization of mice against influenza by the injection of plasmids encoding influenza A hemagglutinin (Montgomery, D.L. et aj., 1993, Cell Biol., 12, pp.777- 783), or nucleoprotein (Montgomery, D.L.
  • Tuberculosis is a chronic infectious disease of the lung caused by the pathogen Mycobacterium tuberculosis. TB is one of the most clinically significant infections worldwide, with an incidence of 3 million deaths and 10 million new cases each year. It has been estimated that as much as one third of the world's population may be infected and, in developing countries, 55 million cases of active TB have been reported. Until the turn of the century, TB was the leading cause of death in the United States. But, with improved sanitary conditions and the advent of antimicrobial drugs, the incidence of mortality steadily declined to the point where it was predicted that the disease would be eradicated by the year 2000. However, in most developed countries, the number of cases of active TB has risen each year since the mid-1980's.
  • MDR multidrug-resistant
  • M. tuberculosis is an intracellular pathogen that infects macrophages and is able to survive within the harsh environment of the phagolysosome in this type of cell. Most inhaled bacilli are destroyed by activated alveolar macrophages. However, the surviving bacilli can multiply in macrophages and be released upon cell death, which signals the infiltration of lymphocytes, monocytes and macrophages to the site. Lysis of the bacilli-laden macrophages is mediated by delayed-type hypersensitivity (DTH) and results in the development of a solid caseous tubercle surrounding the area of infected cells. Continued DTH causes the tubercle to liquefy, thereby releasing entrapped bacilli. The large dose of extracellular bacilli triggers further DTH, causing damage to the bronchi and dissemination by lymphatic, hematogenous and bronchial routes, and eventually allowing infectious bacilli to be spread by respiration.
  • DTH
  • Immunity to TB involves several types of effector cells. Activation of macrophages by cytokines, such as interferon- ⁇ , is an effective means of minimizing intracellular mycobacterial multiplication. However, complete eradication of the bacilli by this means is often not achieved. Acquisition of protection against TB requires T lymphocytes. Among these, both CD8+ and CD4+ T cells seem to be important [Orme et al, 1993, J. Infect. Dis. 167, 1481]. These cell types secrete interferon- ⁇ in response to mycobacteria, indicative of a Thl immune response, and possess cytotoxic activity to mycobacteria-pulsed target cells.
  • Antigenic stimulation of T cells requires presentation by MHC molecules. In order for mycobacterial antigens to gain access to the antigen presentation pathway they must be released from the bacteria. In infected macrophages, this could be accomplished by secretion or bacterial lysis. Mycobacteria possess many potential T-cell antigens and several have now been identified [Andersen 1994, Dan. Med. Bull. 41, 205]. Some of these antigens are secreted by the bacteria. It is generally believed that immunity against TB is mediated by CD8 + and CD4+ T cells directed toward these secreted antigens.
  • T cell antigens are those proteins that are secreted by mycobacteria during their residence in macrophages, such as: i) the antigen 85 complex of proteins (85 A, 85B, 85C) [Wiker and Harboe, 1992, Microbiol. Rev. 56, 648], ii) a 6 kDa protein termed ESAT-6 [Andersen 1994, Infect. Immunity 62, 2536], iii) a 38 kDa lipoprotein with homology to PhoS [Young and Garbe, 1991, Res. Microbiol.
  • M.tb protein-coding DNA sequences were cloned into eukaryotic expression vectors. These DNA constructions elicit an immune response when injected into animals. Immunized animals are infected with mycobacteria to evaluate whether or not direct DNA immunization with the gene (or other M.tb genes) could protect them from disease. Nucleic acids, including DNA constructs and RNA transcripts, capable of inducing in vivo expression of M.tb proteins upon direct introduction into animal tissues via injection or otherwise are therefore disclosed.
  • nucleic acids may elicit immune responses which result in the production of cytotoxic T lymphocytes (CTLs) specific for M.tb antigens, as well as the generation of MJfr-specific helper T lymphocyte responses, which are protective upon subsequent challenge.
  • CTLs cytotoxic T lymphocytes
  • MJfr-specific helper T lymphocyte responses which are protective upon subsequent challenge.
  • These nucleic acids are useful as vaccines for inducing immunity to M.tb, which can prevent infection and/or ameliorate Jfr-related disease.
  • Fig. 1 General principle for cloning M.tb genes into expression vectors is shown.
  • FIG. 1 Vector map of VlJns.tPA85A.Cl is shown.
  • FIG. 3 Vector map of VUns.85A.C2 is shown.
  • FIG. 4 Vector map of VUns.85A.C3 is shown.
  • FIG. 5 Vector map of VUns.tPA85B.Cl is shown.
  • FIG. 6 Vector map of VUns.tPA85C.Cl is shown.
  • Fig. 9 Production of antigen 85A-specific antibodies in DNA- vaccinated mice is shown.
  • Fig. 10 IL-2 production in B ALB/c mice by a Tb DNA vaccine is shown.
  • Fig. 11 IL-2 production in C57BL/6 mice by a Tb DNA vaccine is shown.
  • Fig. 13 IFN- ⁇ production in C57BL/6 mice by a Tb DNA vaccine is shown.
  • Fig. 15 Lack of IL-6 production in mice by a Tb DNA vaccine is shown.
  • Fig. 16 Lack of IL-10 production in mice by a Tb DNA vaccine is shown.
  • Fig. 17 Reduction of BCG multiplication in lungs of C57BL/6 mice vaccinated with a Tb DNA vaccine is shown.
  • Fig. 18 Reduction of BCG multiplication in lungs of BALB/c mice vaccinated with a Tb DNA vaccine is shown.
  • Fig. 19 Reduction of BCG multiplication in spleens of BALB/c mice vaccinated with a Tb DNA vaccine is shown.
  • a polynucleotide is a nucleic acid which contains essential regulatory elements such that upon introduction into a living vertebrate cell, and is able to direct the cellular machinery to produce translation products encoded by the genes comprising the polynucleotide.
  • the polynucleotide is a polydeoxyribonucleic acid comprising Mycobacterium tuberculosis (M.tb ) genes operatively linked to a transcriptional promoter.
  • M.tb Mycobacterium tuberculosis
  • the polynucleotide vaccine comprises polyribonucleic acid encoding M.tb genes which are amenable to translation by the eukaryotic cellular machinery (ribosomes, tRNAs, and other translation factors).
  • the protein encoded by the polynucleotide is one which does not normally occur in that animal except in pathological conditions, (i.e. an heterologous protein) such as proteins associated with M.tb
  • the animals' immune system is activated to launch a protective immune response. Because these exogenous proteins are produced by the animals' own tissues, the expressed proteins are processed by the major histocompatibiHty system (MHC) in a fashion analogous to when an actual M.tb infection occurs.
  • MHC major histocompatibiHty system
  • the result is induction of immune responses against M.tb.
  • Polynucleotides for the purpose of generating immune responses to an encoded protein are referred to herein as polynucleotide vaccines or PNV.
  • the instant invention provides a method for using a polynucleotide which, upon introduction into mammalian tissue, induces the expression, in vivo, of the polynucleotide thereby producing the encoded protein. It is readily apparent to those skilled in the art that variations or derivatives of the nucleotide sequence encoding a protein can be produced which alter the amino acid sequence of the encoded protein. The altered expressed protein may have an altered amino acid sequence, yet still elicits immune responses which react with the mycobacterial protein, and are considered functional equivalents.
  • fragments of the full length genes which encode portions of the full length protein may also be constructed. These fragments may encode a protein or peptide which elicits antibodies which react with the mycobacterial protein, and are considered functional equivalents.
  • a gene encoding an M.tb gene product is incorporated in an expression vector.
  • the vector contains a transcriptional promoter recognized by eukaryotic RNA polymerase, and a transcriptional terminator at the end of the M.tb gene coding sequence.
  • the promoter is the cytomegalovirus promoter with the intron A sequence (CMV-intA), although those skilled in the art will recognize that any of a number of other known promoters such as the strong immunoglobulin, or other eukaryotic gene promoters may be used.
  • a preferred transcriptional terminator is the bovine growth hormone terminator. The combination of CMVintA-BGH terminator is preferred.
  • an antibiotic resistance marker is also optionally included in the expression vector under transcriptional control of a suitable prokaryotic promoter.
  • Ampicillin resistance genes, neomycin resistance genes or any other suitable antibiotic resistance marker may be used.
  • the antibiotic resistance gene encodes a gene product for neomycin/kanamycin resistance.
  • the vector to aid in the high level production of the polynucleotide by growth in prokaryotic organisms, it is advantageous for the vector to contain a prokaryotic origin of replication and be of high copy number. Any of a number of commercially available prokaryotic cloning vectors provide these elements.
  • these functionalities are provided by the commercially available vectors known as the pUC series. It may be desirable, however, to remove non- essential DNA sequences. Thus, the lacZ and lad coding sequences of pUC may be removed. It is also desirable that the vectors are not able to replicate in eukaryotic cells. This minimizes the risk of integration of polynucleotide vaccine sequences into the recipients' genome.
  • the expression vector pnRSV is used, wherein the Rous sarcoma virus (RSV) long terminal repeat (LTR) is used as the promoter.
  • RSV Rous sarcoma virus
  • LTR long terminal repeat
  • VI a mutated pBR322 vector into which the CMV promoter and the BGH transcriptional terminator were cloned is used.
  • the elements of VI and pUC19 have been been combined to produce an expression vector named V1J.
  • VlJtPA Into V1J, VlJtPA or another desirable expression vector is cloned an M.tb gene, such as one of the antigen 85 complex genes, or any other M.tb gene which can induce anti-M .tb immune responses (CTLs, helper T lymphocytes and antibodies).
  • the ampicillin resistance gene is removed from VI J and replaced with a neomycin resistance gene, to generate VlJ-neo, into which any of a number of different M.tb genes may be cloned for use according to this invention.
  • the vector is VUns, which is the same as VUneo except that a unique Sfil restriction site has been engineered into the single Kpnl site at position 2114 of VlJ-neo.
  • the incidence of Sfil sites in human genomic DNA is very low (approximately 1 site per 100,000 bases).
  • the vector is VI R. In this vector, as much non-essential DNA as possible is ''trimmed'' to produce a highly compact vector. This vector allows larger inserts to be used, with less concern that undesirable sequences are encoded and optimizes uptake by cells when the construct encoding specific vims genes is introduced into surrounding tissue.
  • the methods used in producing the foregoing vector modifications and development procedures may be accomplished according to methods known by those skilled in the art.
  • one of the utilities of the instant invention is to provide a system for in vivo as well as in vitro testing and analysis so that a correlation of M.tb sequence diversity with CTL and T-cell proliferative responses, as well as other parameters can be made.
  • the isolation and cloning of these various genes may be accomplished according to methods known to those skilled in the art.
  • This invention further provides a method for systematic identification of M.tb strains and sequences for vaccine production. Incorporation of genes from primary isolates of M.tb strains provides an immunogen which induces immune responses against clinical isolates of the organism and thus meets a need as yet unmet in the field. Furthermore, if the virulent isolates change, the immunogen may be modified to reflect new sequences as necessary.
  • a gene encoding an M.tb protein is directly linked to a transcriptional promoter.
  • tissue-specific promoters or enhancers for example the muscle creatine kinase (MCK) enhancer element may be desirable to limit expression of the polynucleotide to a particular tissue type.
  • myocytes are terminally differentiated cells which do not divide. Integration of foreign DNA into chromosomes appears to require both cell division and protein synthesis. Thus, limiting protein expression to non-dividing cells such as myocytes may be preferable.
  • use of the CMV promoter is adequate for achieving expression in many tissues into which the PNV is introduced.
  • M.tb and other genes are preferably ligated into an expression vector which has been specifically optimized for polynucleotide vaccinations.
  • Elements include a transcriptional promoter, immunogenic epitopes, and additional cistrons encoding immunoenhancing or immunomodulatory genes, with their own promoters, transcriptional terminator, bacterial origin of replication and antibiotic resistance gene, as described herein.
  • the vector may contain internal ribosome entry sites (IRES) for the expression of polycistronic mRNA.
  • IRS internal ribosome entry sites
  • RNA polymerase promoters as the T7 or SP6 promoters
  • T7 or SP6 promoters RNA polymerase promoters
  • T7 or SP6 promoters RNA polymerase promoters
  • T7 or SP6 promoters RNA polymerase promoters
  • the protective efficacy of polynucleotide M.tb immunogens against subsequent challenge is demonstrated by immunization with the DNA of this invention. This is advantageous since no infectious agent is involved, no assembly/replication of bacteria is required, and determinant selection is permitted. Furthermore, because the sequence of mycobacterial gene products may be conserved among various strains of M.tb, protection against subsequent challenge by another strain of M.tb is obtained.
  • the injection of a DNA expression vector encoding antigen 85A, B or C may result in the generation of significant protective immunity against subsequent challenge.
  • specific CTLs and helper T lymphocyte responses may be produced.
  • M.tb gene products exhibit a high degree of conservation among the various strains of M.tb and because immune responses may be generated in response to intracellular expression and MHC processing, it is expected that many different M.tb PNV constructs may give rise to cross reactive immune responses.
  • the invention offers a means to induce heterologous protective immunity without the need for self-replicating agents or adjuvants.
  • the generation of high titer antibodies against expressed proteins after injection of viral protein and human growth hormone DNA [Tang et al.. Nature 356, 152, 1992], indicates this is a facile and highly effective means of making antibody-based vaccines, either separately or in combination with cytotoxic T-lymphocyte and helper T lymphocyte vaccines targeted towards conserved antigens.
  • the ease of producing and purifying DNA constructs compares favorably with traditional protein purification, facilitating the generation of combination vaccines.
  • multiple constructs for example encoding antigen 85 complex genes and any other M.tb gene also including non-M.tb genes may be prepared, mixed and co- administered.
  • the amount of expressible DNA or transcribed RNA to be introduced into a vaccine recipient will have a very broad dosage range and may depend on the strength of the transcriptional and translational promoters used.
  • the magnitude of the immune response may depend on the level of protein expression and on the immunogenicity of the expressed gene product.
  • an effective dose ranges of about 1 ng to 5 mg, lOOng to 2.5 mg, l ⁇ g to 750 ⁇ g, and preferably about 10 ⁇ g to 300 ⁇ g of DNA is administered directly into muscle tissue.
  • Subcutaneous injection, intradermal introduction, impression through the skin, and other modes of administration such as intraperitoneal, intravenous, or inhalation delivery are also suitable. It is also contemplated that booster vaccinations may be provided. Following vaccination with M.tb polynucleotide immunogen, boosting with M.tb protein immunogens such as the antigen 85 complex gene products is also contemplated.
  • Parenteral administration such as intravenous, intramuscular, subcutaneous or other means of administration of interleukin-12 protein (or other cytokines, e.g. GM- CSF), concurrently with or subsequent to. parenteral introduction of the PNV of this invention may be advantageous.
  • the polynucleotide may be naked, that is, unassociated with any proteins, adjuvants or other agents which affect the recipients' immune system.
  • it is desirable for the polycucleotide to be in a physiologically acceptable solution such as, but not limited to, sterile saline or sterile buffered saline.
  • the DNA may be associated with liposomes, such as lecithin liposomes or other liposomes known in the art, as a DNA-liposome mixture, or the DNA may be associated with an adjuvant known in the art to boost immune responses, such as a protein or other carrier.
  • Agents which assist in the cellular uptake of DNA such as, but not limited to, calcium ions, may also be used.
  • transfection facilitating reagents and pharmaceutically acceptable carriers.
  • Techniques for coating microprojectiles coated with polynucleotide are known in the art and are also useful in connection with this invention.
  • DNA intended for human use it may be useful to have the final DNA product in a pharmaceutically acceptable carrier or buffer solution.
  • Pharmaceutically acceptable carriers or buffer solutions are known in the art and include those described in a variety of texts such as Remington's Pharmaceutical Sciences.
  • the invention is a polynucleotide which comprises contiguous nucleic acid sequences capable of being expressed to produce a gene product upon introduction of said polynucleotide into eukaryotic tissues in vivo.
  • the encoded gene product preferably either acts as an immunostimulant or as an antigen capable of generating an immune response.
  • the nucleic acid sequences in this embodiment encode an M.tb immunogenic epitope, and optionally a cytokine or a T-cell costimulatory element, such as a member of the B7 family of proteins.
  • the first is the relative simplicity with which native or nearly native antigen can be presented to the immune system. Mammalian proteins expressed recombinantly in bacteria, yeast, or even mammalian cells often require extensive treatment to insure appropriate antigenicity.
  • a second advantage of DNA immunization is the potential for the immunogen to enter the MHC class I pathway and evoke a cytotoxic T cell response. Immunization of mice with DNA encoding the influenza A nucleoprotein (NP) elicited a CD8+ response to NP that protected mice against challenge with heterologous strains of flu. (Montgomery, D.L. et aj . ., supra; Ulmer, J.
  • Immunization by DNA injection also allows, as discussed above, the ready assembly of multicomponent subunit vaccines.
  • the vaccines of the present invention are useful for administration to domesticated or agricultural animals, as well as humans.
  • Vaccines of the present invention may be used to prevent and/or combat infection of any agricultural animals, including but not limited to, dairy cattle, which are susceptible to Mycobacterial infection.
  • the techniques for administering these vaccines to animals and humans are known to those skilled in the veterinary and human health fields, respectively.
  • the expression vector VI was constructed from pCMVIE- AKI-DHFR [Y. Whang et al, J. Virol. 61 , 1796 (1987)].
  • the AKI and DHFR genes were removed by cutting the vector with EcoR I and self- ligating. This vector does not contain intron A in the CMV promoter, so it was added as a PCR fragment that had a deleted internal Sac I site [at 1855 as numbered in B.S. Chapman et al, Nuc. Acids Res. 19, 3979 (1991)].
  • the template used for the PCR reactions was pCMVintA-Lux, made by ligating the Hind IH and Nhe I fragment from pCMV6al20 [see B.S.
  • the primers that spanned intron A are: 5' primer, SEQ. ID:1 :
  • the primers used to remove the Sac I site are: sense primer, SEQ ID:3:
  • the PCR fragment was cut with Sac I and Bgl II and inserted into the vector which had been cut with the same enzymes.
  • VU The purpose in creating VU. was to remove the promoter and transcription termination elements from vector V 1 in order to place them within a more defined context, create a more compact vector, and to improve plasmid purification yields.
  • VI J is derived from vectors VI and pUC18, a commercially available plasmid.
  • V 1 was digested with Sspl and EcoRI restriction enzymes producing two fragments of DNA. The smaller of these fragments, containing the CMVintA promoter and Bovine Growth Hormone (BGH) transcription termination elements which control the expression of heterologous genes, was purified from an agarose electrophoresis gel. The ends of this DNA fragment were then "blunted” using the T4 DNA polymerase enzyme in order to facilitate its ligation to another "blunt-ended" DNA fragment.
  • pUC18 was chosen to provide the "backbone" of the expression vector.
  • Eaml 1051 restriction enzymes The remaining plasmid was purified by agarose gel electrophoresis, blunt-ended with T4 DNA polymerase, and then treated with calf intestinal alkaline phosphatase.
  • the commercially available kanr gene derived from transposon 903 and contained within the pUC4K plasmid, was excised using the PstI restriction enzyme, purified by agarose gel electrophoresis, and blunt-ended with T4 DNA polymerase. This fragment was ligated with the VI J backbone and plasmids with the kanr gene in either orientation were derived which were designated as VUneo #'s 1 and 3.
  • VUneo#3 referred to as VUneo hereafter, was selected which contains the kanr gene in the same orientation as the ampr gene in VU as the expression construct.
  • V neo was linearized with Kpn I, gel purified, blunted by T4 DNA polymerase, and ligated to the blunt Sfi I linker. Clonal isolates were chosen by restriction mapping and verified by sequencing through the linker. The new vector was designated VlJns. Expression of heterologous genes in VlJns (with Sfi I) was comparable to expression of the same genes in VUneo (with Kpn I).
  • VlJns was modified to include the human tissue-specific plasminogen activator (tPA) leader.
  • tPA tissue-specific plasminogen activator
  • Two synthetic complementary oligomers were annealed and then ligated into VUn which had been Bgl ⁇ digested.
  • the sense and antisense oligomers were 5 -GATC ACC ATG GAT GCA ATG AAG AGA GGG CTC TGC TGT GTG CTG CTG CTG TGT GGA GCA GTC TTC GTT TCG CCC AGC GA-3 ⁇ SEQ.
  • an Sfil restriction site was placed at the Kpnl site within the BGH terminator region of VUn-tPA by blunting the Kpnl site with T4 DNA polymerase followed by ligation with an Sfil linker (catalogue #1138, New England Biolabs). This modification was verified by restriction digestion and agarose gel electrophoresis.
  • X any antigenic gene
  • the murine B7 gene was PCR amplified from the B lymphoma cell line CHI (obtained from the ATCC).
  • B7 is a member of a family of proteins which provide essential costimulation T cell activation by antigen in the context of major histocompatibiHty complexes I and II.
  • CHI cells provide a good source of B7 mRNA because they have the phenotype of being constitutively activated and B7 is expressed primarily by activated antigen presenting cells such as B cells and macrophages. These cells were further stimulated in vitro using cAMP or IL-4 and mRNA prepared using standard guanidinium thiocyanate procedures. cDNA synthesis was performed using this mRNA using the GeneAmp RNA PCR kit (Perkin -Elmer Cetus) and a priming oligomer (5'-GTA CCT CAT GAG CCA CAT AAT ACC ATG-3', SEQ. ID:7:) specific for B7 located downstream of the B7 translational open reading frame.
  • B7 was amplified by PCR using the following sense and antisense PCR oligomers: 5'-GGT ACA AGA TCT ACC ATG GCT TGC AAT TGT CAG TTG ATG C-3', SEQ. ID:8:, and 5 -CCA CAT AGA TCT CCA TGG GAA CTA AAG GAA GAC GGT CTG TTC-3', SEQ. ID:9:, respectively.
  • These oligomers provide Bgi ⁇ restriction enzyme sites at the ends of the insert as well as a Kozak translation initiation sequence containing an Ncol restriction site and an additional Ncol site located immediately prior to the 3'-terminal Bglll site.
  • Ncol digestion yielded a fragment suitable for cloning into pGEM- 3 -IRES which had been digested with Ncol.
  • the resulting vector, pGEM-3-IRES-B7 contains an IRES-B7 cassette which can easily be transferred to VUns-X, where X represents an antigen-encoding gene.
  • This vector contains a cassette analogous to that described in item C above except that the gene for the immuno stimulatory cytokine, GM-CSF, is used rather than B7.
  • GM-CSF is a macrophage differentiation and stimulation cytokine which has been shown to elicit potent anti-tumor T cell activities in vivo [G. Dranoff et al, Proc. Natl. Acad. Sci. USA, 90, 3539 (1993).
  • This vector contains a cassette analogous to that described in item C above except that the gene for the immunostimulatory cytokine, IL-12, is used rather than B7.
  • IL- 12 has been demonstrated to have an influential role in shifting immune responses towards cellular, T cell-dominated pathways as opposed to humoral responses [L. Alfonso et al, Science, 263, 235, 1994].
  • V1R Vector V 1 R Preparation
  • V1R a derivative of VlJns, designated V1R.
  • the purpose for this vector construction was to obtain a minimum-sized vaccine vector without unneeded DNA sequences, which still retained the overall optimized heterologous gene expression characteristics and high plasmid yields that VI J and VlJns afford. It was determined from the literature as well as by experiment that (1) regions within the pUC backbone comprising the E.
  • VI R was constructed by using PCR to synthesize three segments of DNA from VlJns representing the CMVintA promoter/BGH terminator, origin of replication, and kanamycin resistance elements, respectively.
  • Restriction enzymes unique for each segment were added to each segment end using the PCR oligomers: Sspl and Xhol for CMVintA/BGH; EcoRV and BamHI for the kan r gene; and, Bell and Sail for the ori r. These enzyme sites were chosen because they allow directional ligation of each of the PCR-derived DNA segments with subsequent loss of each site: EcoRV and Sspl leave blunt- ended DNAs which are compatible for ligation while BamHI and Bell leave complementary overhangs as do Sail and Xhol. After obtaining these segments by PCR each segment was digested with the appropriate restriction enzymes indicated above and then ligated together in a single reaction mixture containing all three DNA segments.
  • the 5'-end of the ori r was designed to include the T2 rho independent terminator sequence that is normally found in this region so that it could provide termination information for the kanamycin resistance gene.
  • M.tb antigens RD cells human rhabdomyosarcoma ATCC CCL 1366 were grown at 37°C, 5% C ⁇ 2 in Dulbecco's modified Eagle's medium
  • DMEM fetal bovine serum
  • 20mM HEPES 20mM HEPES
  • 4mM L-glutamine 100 ⁇ g/mL each of penicillin and streptomycin.
  • Cells were seeded at 1.5x10 ⁇ cells/100 mm ⁇ plate and grown for 1 hours.
  • Cell were transfected with lO ⁇ g/plate of the TB construct and 10 ⁇ g of co-transfected Cat construct using the
  • cell pellets were lysed in 50 ⁇ L of Single Detergent Lysis Buffer (50 mM Tris-Cl, pH 8.0, 150 mM NaCl, 0.02% NaN3, l %Nonidet P-40, 100 mM PMSF, 2 ⁇ g/mL aprotinin, 2 ⁇ g/mL leupeptin, and 1 ⁇ g/mL Pepstatin A) and sonicated on ice (2-15 second bursts). Lysates were centrifuged at 13,000xg, 4°C, for 10 minutes.
  • Single Detergent Lysis Buffer 50 mM Tris-Cl, pH 8.0, 150 mM NaCl, 0.02% NaN3, l %Nonidet P-40, 100 mM PMSF, 2 ⁇ g/mL aprotinin, 2 ⁇ g/mL leupeptin, and 1 ⁇ g/mL Pepstatin A
  • Lysates were centrifuged at
  • Protein concentration was determined by the Bradford method and 20 ⁇ g of cell extract protein per lane was applied to a 10% TRIS-glycine polyacrylamide gel (Novex), then transferred to Immobilon P (Millipore) membrane. Immunoblots were reacted overnight with a 1 :20 dilution of the mouse monoclonal antibody TD 17-4 [Huygen et al, 1994, Infect. Immunity 62, 363], followed by a 1.5 hours reaction with a 1 : 1000 dilution of goat anti-mouse IgGFc peroxidase (Jackson). The blots were developed using the ECL kit (Amersham).
  • VI Jns-tPA-85A contains mature Ag85A with tPA signal sequence
  • sense 85A.C1 primer [SEQ.ID.NO.:16] GG AAG ATC TTT TCC CGG CCG GGC TTG CCG Bgl D
  • antisense 85 A primer [SEQ.ID.NO.:17] GGAAGATCTTGTCTGTTCGGAGCTAGGC.
  • the Ag85A from M. tuberculosis was amplified from plasmid p85A.tub, which was prepared by ligating an 800 bp Hindlll fragment to a 1600 bp Hindlll-SphI fragment from Figure 2 of Borremans et al, 1989 [Infect. Immunity 57, 3123].
  • the resulting 2400 bp insert was subcloned in the Hindlll and SphI sites of the BlueScribe M13+.
  • the entire coding sequence and flanking regions in BlueScribe Ml 3+ (VCS/Stratagene) were amplified by PCR with the indicated primers in the following conditions.
  • Each 100 ⁇ l reaction contains 2.5 Units Cloned Pfu DNA Polymerase (Stratagene), 200 mM dNTP, 0.5 ⁇ g of each primer and 250 ng of template DNA in the reaction buffer supplied with the enzyme (Stratagene).
  • the Hybaid Thermal Reactor was programmed as follows: 5 minutes denaturation at 94°C followed by 25 cycles (1 minute at 94°C, 2 minutes at 55°C and 3 minutes at 72°C) ending with 10 minutes extension at 72°C. Amplified DNA was digested with 50 ⁇ g/ml Proteinase K
  • VUns-85A [C2] (contains mature Ag85A with no signal sequence) was done using the following primers:
  • Antisense 85 A [SEQ.ID.NO.:17] GGAAGATCTTGCTGTTCGGAGCTAGGC.
  • VlJns-85A [C3] contains Ag85A with its own signal sequence
  • Sense 85A C3 [SEQ.ID.NO.:19] GGAAGATCTACC ATG GCA CAG CTT GTT GAC AGG GTT
  • Antisense 85 A [SEQ.JD.NO.:17] GGAAGATCTTGCTGTTCGGAGCTAGGC.
  • VUns-tPA-85B [Cl] (contains Ag85B with tPA signal sequence) was done using the following primers:
  • GGAAG ATC TCC TTC TCC CGG CCG GGG CTG CCG GTC GAG
  • Antisense 85B [SEQ.ID.NO.:21] GGAAGATCTAACCTTCGGTTGATCCCGTCAGCC.
  • Antisense 85C [SEQ.ID.NO.:23] GGAAGATCTTGCCGATGCTGGCTTGCTGGCTCAGGC. The same procedure as 1 above was followed, except that the template for PCR was p85C.tub.
  • Antisense 85B [SEQ.JD.NO.:21]
  • Antisense 85C [SEQ.ID.NO.:23] GGAAGATCTTGCCGATGCTGGCTTGCTGGCTCAGGC.
  • the plasmid constructions were characterized by restriction mapping and sequence analysis of the vector-insert junctions (see Figures 1 -6). Results were consistent with published M.tb sequence data and showed that the initiation codon was intact for each construct ( Figure 7). Also shown are the various additional amino acid residues unrelated to M.tb Ag85 that were inserted as a result of cloning.
  • Rhabdomyosarcoma cells (ATCC CCL136) were planted one day before use at a density of 1.2 X106 cells per 9.5 cm2 well in six-well tissue culture clusters in high glucose DMEM supplemented with 10% heat-inactivated fetal calf serum, 2 mM L-glutamine, 25 mM HEPES, 50 U/ml penicillin and 50 ⁇ g/ml streptomycin.
  • Phenol chloroform extracted cesium chloride purified plasmid DNA was precipitated with calcium phosphate using Pharmacia CellPhect reagents according to the kit instmctions except that 5 - 15 ⁇ g is used for each 9.5 cm2 well of RD cells. Cultures were glycerol shocked six hours post addition of calcium phosphate-DNA precipate; after refeeding, cultures were incubated for two days prior to harvest.
  • Lysates of transfected cultures were prepared in IX RIPA (0.5% SDS, 1.0% TRITON X-100, 1% sodium deoxycholate, ImM EDTA, 150mM NaCl, 25 mM TRIS-HC1 pH 7.4) supplemented with l ⁇ M leupeptin, l ⁇ M pepstatin, 300nM aprotinin, and lO ⁇ M TLCK, and sonicated briefly to reduce viscosity. Lysates were resolved by electrophoresis on 10% Tricine gels (Novex) and then transferred to nitrocellulose membranes. Immunoblots were processed with M.tb monoclonal antibodies 17/4 and 32/15 [Huygen et al, 1994, Infect. Immunity 62, 363] and developed with the ECL detection kit (Amersham).
  • mice Five- to six-week-old female BALB/c and C57BL/6 mice were anesthetized by intraperitoneal (i.p.) injection of a mixture of 5 mg ketamine HC1 (Aveco, Fort Dodge, IA) and 0.5 mg xylazine
  • FIG. 9 shows specific immunoblot reactivity of sera from Ag85 DNA-injected mice (Cl) but not from mice that received a control DNA not containing a gene insert (V1J). Reactivity was detected to a serum dilution of at least 1 : 160 against 300 ng of purified antigen 85A (Figure 9b). This demonstrates that injection of Ag85 DNA resulted in Ag85 expression in vivo such that it was available for the generation of antibody responses in both BALB/c and C57BL/6 (B6) mice.
  • Spleen cells from vaccinated mice were analyzed for cytokine secretion in response to specific antigen restimulation as described in Huygen et al, 1992 [Infect. Immunity 60, 2880]. Specifically, spleen cells were incubated with culture filtrate (CF) proteins from M. bovis BCG purified antigen 85A or a 20-mer peptide (p25) corresponding to a known T-cell epitope for C57BL/6 mice (amino acids 241-260). Mice were immunized with VUns.tPA85A (Cl ) (100 ⁇ g) three times with three week intervals and analyzed 17 days after the final injection.
  • CF culture filtrate
  • Cytokines were assayed using bio-assays for IL-2, interferon- ⁇ (IFN- ⁇ ) and IL-6, and by ELISA for IL-4 and IL-10. Substantial IL-2 and IFN- ⁇ production was observed in both BALB/c and C57BL/6 mice vaccinated with VUns.tPA85A (Cl) ( Figures 10- 13). Furthermore, C57BL/6 mice also reacted to the H-2b-restricted T- cell epitope ( Figure 13). IL-4, IL-6 and IL-10 levels were not increased in VUns.tPA85 A- vaccinated mice ( Figures 14-16). These results indicate that a Thl type of helper T-cell response was generated by the DNA vaccine.
  • mice were challenged with an intravenous injection of live M. bovis BCG (0.5 mg) and BCG multiplication was analyzed in the spleens and lungs.
  • BCG multiplication was measured in challenged naive mice (primary infection) and challenged mice that were vaccinated with BCG at the time of DNA injection (secondary infection).
  • the number of colony-forming units (CFU) in lungs of VUns.tPA85A (Cl)-vaccinated mice was substantially reduced compared to mice with primary infection or mice vaccinated with control DNA VU.
  • (l) APPLICANT CONTENT, JEAN HUYGEN, KRIS LIU, MARGARET A. MONTGOMER , DONNA ULMER, JEFFREY
  • MOLECULE TYPE DNA (genomic! (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1 : CTATATAAGC AGAGCTCGTT TAG 23
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)

Abstract

Genes encoding Mycobacterium tuberculosis (M.tb, proteins were cloned into eukaryotic expression vectors to express the encoded proteins in mammalian muscle cells in vivo. Animals were immunized by injection of these DNA constructs, termed polynucleotide vaccines or PNV, into their muscles. Immune antisera was produced against M.tb antigens. Specific T-cell responses were detected in spleen cells of vaccinated mice and the profile of cytokine secretion in response to antigen (85) was indicative of a Th1 type of helper T-cell response (i.e., high IL-2 and IFN-η). Protective efficacy of an M.tb DNA vaccine was demonstrated in mice after challenge with M.bovis BCG, as measured by a reduction in mycobacterial multiplication in the spleens and lungs of M.tb DNA-vaccinated mice compared to control DNA-vaccinated mice or primary infection in naive mice.

Description

TITLE OF THE INVENTION
A POLYNUCLEOTIDE TUBERCULOSIS VACCINE
BACKGROUND OF THE INVENTION A major obstacle to the development of vaccines against viruses and bacteria, particularly those with multiple serotypes or a high rate of mutation, against which elicitation of neutralizing antibodies and/or protective cell-mediated immune responses is desirable, is the diversity of the external proteins among different isolates or strains. Since cytotoxic T-lymphocytes (CTLs) in both mice and humans are capable of recognizing epitopes derived from conserved internal viral proteins [J.W. Yewdell et al, Proc Natl. Acad. Sci. (USA) 82, 1785 (1985); A.R.M. Townsend, et al, Cell 44, 959 (1986); A.J. McMichael et al, J. Gen. Virol. 67, 719 (1986); J. Bastin et al., J. Exp. Med. 165, 1508 (1987); A.R.M. Townsend and H. Bodmer, Annu. Rev. Immunol 7, 601 (1989)], and are thought to be important in the immune response against viruses [Y.-L. Lin and B.A. Askonas, J. Exp. Med. 154, 225 (1981); I. Gardner et al, Eur. J. Immunol. 4, 68 (1974); K.L. Yap and G.L. Ada, Nature 273, 238 (1978); AJ. McMichael et al, New Engl. J. Med. 309, 13 (1983); P.M. Taylor and B.A. Askonas, Immunol. 58, 417 (1986)], efforts have been directed towards the development of CTL vaccines capable of providing heterologous protection against different viral strains.
It is known that CTLs kill virally- or bacterially-infected cells when their T cell receptors recognize foreign peptides associated with MHC class I and/or class II molecules. These peptides can be derived from endogenously synthesized foreign proteins, regardless of the protein's location or function within the pathogen. By recognition of epitopes from conserved proteins, CTLs may provide heterologous protection. In the case of intracellular bacteria, proteins secreted by or released from the bacteria are processed and presented by MHC class I and II molecules, thereby generating T-cell responses that may play a role in reducing or eliminating infection. Most efforts to generate CTL responses have either used replicating vectors to produce the protein antigen within the cell [J.R. Bennink et al, ibid. 311, 578 (1984); J.R. Bennink and J.W. Yewdell, Curr. Top. Microbiol Immunol. 163, 153 (1990); C.K. Stover et al, Nature 351, 456 (1991); A. Aldovini and R.A. Young, Nature 351 , 479 (1991); R. Schafer et al, J. Immunol. 149, 53 (1992); CS. Hahn et al, Proc. Natl Acad. Sci. (USA) 89, 2679 (1992)], or they have focused upon the introduction of peptides into the cytosol [F.R. Carbone and M.J. Bevan, J. Exp. Med. 169, 603 (1989); K. Deres et al, Nature 342, 561 (1989); H. Takahashi et al, ibid. 344, 873 (1990); D.S. Collins et al, J. Immunol 148, 3336 (1992); M.J. Newman et al, ibid. 148, 2357 (1992)]. Both of these approaches have limitations that may reduce their utility as vaccines. Retroviral vectors have restrictions on the size and structure of polypeptides that can be expressed as fusion proteins while maintaining the ability of the recombinant virus to replicate [A.D. Miller, Curr. Top. Microbiol. Immunol. 158, 1 (1992)], and the effectiveness of vectors such as vaccinia for subsequent immunizations may be compromised by immune responses against vaccinia [E.L. Cooney et al, Lancet 337, 567 (1991)]. Also, viral vectors and modified pathogens have inherent risks that may hinder their use in humans [R.R. Redfield et al, New Engl. J. Med. 316, 673 (1987); L. Mascola et al, Arch. Intern. Med. 149, 1569 (1989)]. Furthermore, the selection of peptide epitopes to be presented is dependent upon the structure of an individual's MHC antigens and, therefore, peptide vaccines may have limited effectiveness due to the diversity of MHC haplotypes in outbred populations.
Benvenisty, N., and Reshef, L. [PNAS 83, 9551 -9555, (1986)] showed that CaCl2 precipitated DNA introduced into mice intraperitoneally (i.p.), intravenously (i.v.) or intramuscularly (i.m.) could be expressed. The intramuscular (i.m.) injection of DNA expression vectors in mice has been demonstrated to result in the uptake of DNA by the muscle cells and expression of the protein encoded by the DNA [J.A. Wolff et al, Science 247, 1465 (1990); G. Ascadi et al, Nature 352, 815 (1991)]. The plasmids were shown to be maintained episomally and did not replicate. Subsequently, persistent expression has been observed after i.m. injection in skeletal muscle of rats, fish and primates, and cardiac muscle of rats [H. Lin et al, Circulation 82, 2217 (1990); R.N. Kitsis et al, Proc. Natl. Acad. Sci. (USA) 88, 4138 (1991); E. Hansen et al, FEBS Lett. 290, 73 (1991); S. Jiao et al, Hum. Gene Therapy 3, 21 (1992); J.A. Wolff et al, Human Mol. Genet. 1, 363 (1992)]. The technique of using nucleic acids as therapeutic agents was reported in WO90/11092 (4 October 1990), in which naked polynucleotides were used to vaccinate vertebrates. Recently, the coordinate roles of B7 and the major histocompatibility complex (MHC) presentation of epitopes on the surface of antigen presenting cells in activating CTLs for the elimination of tumors was reviewed [Edgington, Biotechnology 11 , 1117-1119, 1993]. Once the MHC molecule on the surface of an antigen presenting cell (APC) presents an epitope to a T-cell receptor (TCR), B7 expressed on the surface of the same APC acts as a second signal by binding to CTLA-4 or CD28. The result is rapid division of CD4+ helper T-cells which signal CD8+ T-cells to proliferate and kill the APC. It is not necessary for the success of the method that immunization be intramuscular. Thus, Tang et al, [Nature, 356, 152- 154 (1992)] disclosed that introduction of gold microprojectiles coated with DNA encoding bovine growth hormone (BGH) into the skin of mice resulted in production of anti-BGH antibodies in the mice. Furth et al, [Analytical Biochemistry, 205, 365-368, (1992)] showed that a jet injector could be used to transfect skin, muscle, fat, and mammary tissues of living animals. Various methods for introducing nucleic acids was recently reviewed [Friedman, T., Science, 244, 1275-1281 (1989)]. See also Robinson et al, [Abstracts of Papers Presented at the 1992 meeting on Modern Approaches to New Vaccines, Including Prevention of AIDS, Cold Spring Harbor, p92; Vaccine 1 1, 957 (1993)], where the im, ip, and iv administration of avian influenza DNA into chickens was alleged to have provided protection against lethal challenge. Intravenous injection of a DNA ationic liposome complex in mice was shown by Zhu et al, [Science 261, 209-211 (9 July 1993); see also WO93/24640, 9 Dec. 1993] to result in systemic expression of a cloned transgene. Recently, Ulmer et al, [Science 259, 1745-1749, (1993)] reported on the heterologous protection against influenza virus infection by injection of DNA encoding influenza virus proteins.
Wang et al, [P.N.A.S. USA 90, 4156-4160 (May, 1993)] reported on elicitation of immune responses in mice against HIV by intramuscular inoculation with a cloned, genomic (unspliced) HIV gene. However, the level of immune responses achieved was very low, and the system utilized portions of the mouse mammary tumor vims (MMTV) long terminal repeat (LTR) promoter and portions of the simian virus 40 (SV40) promoter and terminator. SV40 is known to transform cells, possibly through integration into host cellular DNA. Thus, the system described by Wang et al, is wholly inappropriate for administration to humans, which is one of the objects of the instant invention.
WO 93/17706 describes a method for vaccinating an animal against a virus, wherein carrier particles were coated with a gene construct and the coated particles are accelerated into cells of an animal. Studies by Wolff et al. (supra) originally demonstrated that intramuscular injection of plasmid DNA encoding a reporter gene results in the expression of that gene in myocytes at and near the site of injection. Recent reports demonstrated the successful immunization of mice against influenza by the injection of plasmids encoding influenza A hemagglutinin (Montgomery, D.L. et aj., 1993, Cell Biol., 12, pp.777- 783), or nucleoprotein (Montgomery, D.L. et aj., supra; Ulmer, J.B. et ah, 1 93, Science, 259, pp.1745- 1749). The first use of DNA immunization for a herpes virus has been reported (Cox et al., 1993, J.Virol., 67, pp.5664-5667). Injection of a plasmid encoding bovine herpesvirus 1 (BHV-1) glycoprotein g IV gave rise to anti-g IV antibodies in mice and calves. Upon intranasal challenge with BHV-1 , immunized calves showed reduced symptoms and shed substantially less virus than controls. Tuberculosis (TB) is a chronic infectious disease of the lung caused by the pathogen Mycobacterium tuberculosis. TB is one of the most clinically significant infections worldwide, with an incidence of 3 million deaths and 10 million new cases each year. It has been estimated that as much as one third of the world's population may be infected and, in developing countries, 55 million cases of active TB have been reported. Until the turn of the century, TB was the leading cause of death in the United States. But, with improved sanitary conditions and the advent of antimicrobial drugs, the incidence of mortality steadily declined to the point where it was predicted that the disease would be eradicated by the year 2000. However, in most developed countries, the number of cases of active TB has risen each year since the mid-1980's. Part of this resurgence has been attributed to immigration and the growing number of immunocompromised, HIV-infected individuals. If left unabated, it is predicted that TB will claim more than 30 million human lives in the next ten years. As alarming as these figures may seem, it is of even greater concern that multidrug-resistant (MDR) strains of M. tuberculosis have arisen. These MDR strains are not tractable by traditional drug therapy and have been responsible for several recent outbreaks of TB, particularly in urban centers.
Therefore, one of the key components in the management of TB in the long-term will be an effective vaccine [for review see Bloom and Murray, 1993, Science 257, 1055].
M. tuberculosis is an intracellular pathogen that infects macrophages and is able to survive within the harsh environment of the phagolysosome in this type of cell. Most inhaled bacilli are destroyed by activated alveolar macrophages. However, the surviving bacilli can multiply in macrophages and be released upon cell death, which signals the infiltration of lymphocytes, monocytes and macrophages to the site. Lysis of the bacilli-laden macrophages is mediated by delayed-type hypersensitivity (DTH) and results in the development of a solid caseous tubercle surrounding the area of infected cells. Continued DTH causes the tubercle to liquefy, thereby releasing entrapped bacilli. The large dose of extracellular bacilli triggers further DTH, causing damage to the bronchi and dissemination by lymphatic, hematogenous and bronchial routes, and eventually allowing infectious bacilli to be spread by respiration.
Immunity to TB involves several types of effector cells. Activation of macrophages by cytokines, such as interferon-γ, is an effective means of minimizing intracellular mycobacterial multiplication. However, complete eradication of the bacilli by this means is often not achieved. Acquisition of protection against TB requires T lymphocytes. Among these, both CD8+ and CD4+ T cells seem to be important [Orme et al, 1993, J. Infect. Dis. 167, 1481]. These cell types secrete interferon-γ in response to mycobacteria, indicative of a Thl immune response, and possess cytotoxic activity to mycobacteria-pulsed target cells. In recent studies using β-2 microglobulin- and CD8-deficient mice, CTL responses have been shown to be critical in providing protection against M. tuberculosis [Flynn et al, 1992, Proc. Natl. Acad. Sci. USA 89, 12013; Flynn et al, 1993, J. Exp. Med. 178, 2249; Cooper et al, 1993, J. Exp. Med. 178, 2243]. In contrast, B lymphocytes do not seem to be involved, and passive transfer of anti-mycobacterial antibodies does not provide protection. Therefore, effective vaccines against TB must generate cell- mediated immune responses.
Antigenic stimulation of T cells requires presentation by MHC molecules. In order for mycobacterial antigens to gain access to the antigen presentation pathway they must be released from the bacteria. In infected macrophages, this could be accomplished by secretion or bacterial lysis. Mycobacteria possess many potential T-cell antigens and several have now been identified [Andersen 1994, Dan. Med. Bull. 41, 205]. Some of these antigens are secreted by the bacteria. It is generally believed that immunity against TB is mediated by CD8+ and CD4+ T cells directed toward these secreted antigens. In mouse and guinea pig models of TB, protection from bacterial challenge, as measured by reduced weight loss, has been achieved using a mixture of secreted mycobacterial antigens [Pal and Horowitz. 1992 Infect. Immunity 60, 4781; Andersen 1994, Infect. Immunity 62, 2536; Collins, 1994, Veterin. Microbiol. 40, 95].
Several potentially protective T cell antigens have been identified in M. tuberculosis and some of these are being investigated as vaccine targets. Recent work has indicated that the predominant T-cell antigens are those proteins that are secreted by mycobacteria during their residence in macrophages, such as: i) the antigen 85 complex of proteins (85 A, 85B, 85C) [Wiker and Harboe, 1992, Microbiol. Rev. 56, 648], ii) a 6 kDa protein termed ESAT-6 [Andersen 1994, Infect. Immunity 62, 2536], iii) a 38 kDa lipoprotein with homology to PhoS [Young and Garbe, 1991, Res. Microbiol. 142, 55; Andersen, 1992, J. Infect. Dis. 166, 874], iv) the 65 kDa GroEL heat-shock protein [Siva and Lowrie, 1994, Immunol. 82, 244], v) a 55 kDa protein rich in proline and threonine [Romain et al, 1993, Proc. Natl. Acad. Sci. USA 90, 5322], and vi) a 19 kDa lipoprotein [Faith et al, 1991, Immunol. 74,
1].
The genes for each of the three antigen 85 proteins (A, B, and C) have been cloned and sequenced [Borremans et al, 1989, Infect.
Immunity 57, 3123; Content et al, Infect. Immunity 59, 3205; DeWit et al 1994, DNA Seq. 4, 267]. In addition, these structurally-related proteins are targets for strong T-cell responses after both infection and vaccination [Huygen et al, 1988, Scand. J. Immunol. 27, 187; Launois et al, 1991, Clin. Exp. Immunol. 86, 286; Huygen et al, 1992, Infect.
Immunity 60, 2880; Munk et al, 1994, Infect. Immunity 62, 726; Launois et al, 1994, Infect. Immunity 62, 3679]. Therefore, the antigen
85 proteins are considered to be good vaccine targets.
SUMMARY OF THE INVENTION
To test the efficacy of DNA immunization in the prevention of M.tb disease, M.tb protein-coding DNA sequences were cloned into eukaryotic expression vectors. These DNA constructions elicit an immune response when injected into animals. Immunized animals are infected with mycobacteria to evaluate whether or not direct DNA immunization with the gene (or other M.tb genes) could protect them from disease. Nucleic acids, including DNA constructs and RNA transcripts, capable of inducing in vivo expression of M.tb proteins upon direct introduction into animal tissues via injection or otherwise are therefore disclosed. Injection of these nucleic acids may elicit immune responses which result in the production of cytotoxic T lymphocytes (CTLs) specific for M.tb antigens, as well as the generation of MJfr-specific helper T lymphocyte responses, which are protective upon subsequent challenge. These nucleic acids are useful as vaccines for inducing immunity to M.tb, which can prevent infection and/or ameliorate Jfr-related disease.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1. General principle for cloning M.tb genes into expression vectors is shown.
Fig. 2. Vector map of VlJns.tPA85A.Cl is shown.
Fig. 3. Vector map of VUns.85A.C2 is shown.
Fig. 4. Vector map of VUns.85A.C3 is shown.
Fig. 5. Vector map of VUns.tPA85B.Cl is shown.
Fig. 6. Vector map of VUns.tPA85C.Cl is shown.
Fig. 7 N-Terminal sequence verification of constructs is shown.
Fig. 8 Expression of M.tb proteins in tissue culture is shown.
Fig. 9 Production of antigen 85A-specific antibodies in DNA- vaccinated mice is shown. Fig. 10 IL-2 production in B ALB/c mice by a Tb DNA vaccine is shown.
Fig. 11 IL-2 production in C57BL/6 mice by a Tb DNA vaccine is shown.
Fig. 12 IFN-γ production in B ALB/c mice by a Tb DNA vaccine is shown.
Fig. 13 IFN-γ production in C57BL/6 mice by a Tb DNA vaccine is shown.
Fig. 14 Lack of IL-4 production in B ALB/c mice by a Tb
DNA vaccine is shown.
Fig. 15 Lack of IL-6 production in mice by a Tb DNA vaccine is shown.
Fig. 16 Lack of IL-10 production in mice by a Tb DNA vaccine is shown.
Fig. 17 Reduction of BCG multiplication in lungs of C57BL/6 mice vaccinated with a Tb DNA vaccine is shown.
Fig. 18 Reduction of BCG multiplication in lungs of BALB/c mice vaccinated with a Tb DNA vaccine is shown.
Fig. 19 Reduction of BCG multiplication in spleens of BALB/c mice vaccinated with a Tb DNA vaccine is shown.
Fig. 20 Reduction of BCG multiplication in spleens of C57BL/6 mice vaccinated with a Tb DNA vaccine is shown.
DETAILED DESCRIPTION OF THE INVENTION This invention provides polynucleotides which, when directly introduced into a vertebrate in vivo, including mammals such as humans, induces the expression of encoded proteins within the animal. As used herein, a polynucleotide is a nucleic acid which contains essential regulatory elements such that upon introduction into a living vertebrate cell, and is able to direct the cellular machinery to produce translation products encoded by the genes comprising the polynucleotide. In one embodiment of the invention, the polynucleotide is a polydeoxyribonucleic acid comprising Mycobacterium tuberculosis (M.tb ) genes operatively linked to a transcriptional promoter. In another embodiment of the invention the polynucleotide vaccine comprises polyribonucleic acid encoding M.tb genes which are amenable to translation by the eukaryotic cellular machinery (ribosomes, tRNAs, and other translation factors). Where the protein encoded by the polynucleotide is one which does not normally occur in that animal except in pathological conditions, (i.e. an heterologous protein) such as proteins associated with M.tb, the animals' immune system is activated to launch a protective immune response. Because these exogenous proteins are produced by the animals' own tissues, the expressed proteins are processed by the major histocompatibiHty system (MHC) in a fashion analogous to when an actual M.tb infection occurs. The result, as shown in this disclosure, is induction of immune responses against M.tb. Polynucleotides for the purpose of generating immune responses to an encoded protein are referred to herein as polynucleotide vaccines or PNV.
There are many embodiments of the instant invention which those skilled in the art can appreciate from the specification. Thus, different transcriptional promoters, terminators, carrier vectors or specific gene sequences may be used successfully. The instant invention provides a method for using a polynucleotide which, upon introduction into mammalian tissue, induces the expression, in vivo, of the polynucleotide thereby producing the encoded protein. It is readily apparent to those skilled in the art that variations or derivatives of the nucleotide sequence encoding a protein can be produced which alter the amino acid sequence of the encoded protein. The altered expressed protein may have an altered amino acid sequence, yet still elicits immune responses which react with the mycobacterial protein, and are considered functional equivalents. In addition, fragments of the full length genes which encode portions of the full length protein may also be constructed. These fragments may encode a protein or peptide which elicits antibodies which react with the mycobacterial protein, and are considered functional equivalents.
In one embodiment of this invention, a gene encoding an M.tb gene product is incorporated in an expression vector. The vector contains a transcriptional promoter recognized by eukaryotic RNA polymerase, and a transcriptional terminator at the end of the M.tb gene coding sequence. In a preferred embodiment, the promoter is the cytomegalovirus promoter with the intron A sequence (CMV-intA), although those skilled in the art will recognize that any of a number of other known promoters such as the strong immunoglobulin, or other eukaryotic gene promoters may be used. A preferred transcriptional terminator is the bovine growth hormone terminator. The combination of CMVintA-BGH terminator is preferred. In addition, to assist in preparation of the polynucleotides in prokaryotic cells, an antibiotic resistance marker is also optionally included in the expression vector under transcriptional control of a suitable prokaryotic promoter. Ampicillin resistance genes, neomycin resistance genes or any other suitable antibiotic resistance marker may be used. In a preferred embodiment of this invention, the antibiotic resistance gene encodes a gene product for neomycin/kanamycin resistance. Further, to aid in the high level production of the polynucleotide by growth in prokaryotic organisms, it is advantageous for the vector to contain a prokaryotic origin of replication and be of high copy number. Any of a number of commercially available prokaryotic cloning vectors provide these elements. In a preferred embodiment of this invention, these functionalities are provided by the commercially available vectors known as the pUC series. It may be desirable, however, to remove non- essential DNA sequences. Thus, the lacZ and lad coding sequences of pUC may be removed. It is also desirable that the vectors are not able to replicate in eukaryotic cells. This minimizes the risk of integration of polynucleotide vaccine sequences into the recipients' genome.
In another embodiment, the expression vector pnRSV is used, wherein the Rous sarcoma virus (RSV) long terminal repeat (LTR) is used as the promoter. In yet another embodiment, VI, a mutated pBR322 vector into which the CMV promoter and the BGH transcriptional terminator were cloned is used. In a preferred embodiment of this invention, the elements of VI and pUC19 have been been combined to produce an expression vector named V1J.
Into V1J, VlJtPA or another desirable expression vector is cloned an M.tb gene, such as one of the antigen 85 complex genes, or any other M.tb gene which can induce anti-M .tb immune responses (CTLs, helper T lymphocytes and antibodies). In another embodiment, the ampicillin resistance gene is removed from VI J and replaced with a neomycin resistance gene, to generate VlJ-neo, into which any of a number of different M.tb genes may be cloned for use according to this invention. In yet another embodiment, the vector is VUns, which is the same as VUneo except that a unique Sfil restriction site has been engineered into the single Kpnl site at position 2114 of VlJ-neo. The incidence of Sfil sites in human genomic DNA is very low (approximately 1 site per 100,000 bases). Thus, this vector allows careful monitoring for expression vector integration into host DNA, simply by Sfil digestion of extracted genomic DNA. In a further embodiment, the vector is VI R. In this vector, as much non-essential DNA as possible is ''trimmed'' to produce a highly compact vector. This vector allows larger inserts to be used, with less concern that undesirable sequences are encoded and optimizes uptake by cells when the construct encoding specific vims genes is introduced into surrounding tissue. The methods used in producing the foregoing vector modifications and development procedures may be accomplished according to methods known by those skilled in the art.
From this work those skilled in the art will recognize that one of the utilities of the instant invention is to provide a system for in vivo as well as in vitro testing and analysis so that a correlation of M.tb sequence diversity with CTL and T-cell proliferative responses, as well as other parameters can be made. The isolation and cloning of these various genes may be accomplished according to methods known to those skilled in the art. This invention further provides a method for systematic identification of M.tb strains and sequences for vaccine production. Incorporation of genes from primary isolates of M.tb strains provides an immunogen which induces immune responses against clinical isolates of the organism and thus meets a need as yet unmet in the field. Furthermore, if the virulent isolates change, the immunogen may be modified to reflect new sequences as necessary.
In one embodiment of this invention, a gene encoding an M.tb protein is directly linked to a transcriptional promoter. The use of tissue-specific promoters or enhancers, for example the muscle creatine kinase (MCK) enhancer element may be desirable to limit expression of the polynucleotide to a particular tissue type. For example, myocytes are terminally differentiated cells which do not divide. Integration of foreign DNA into chromosomes appears to require both cell division and protein synthesis. Thus, limiting protein expression to non-dividing cells such as myocytes may be preferable. However, use of the CMV promoter is adequate for achieving expression in many tissues into which the PNV is introduced.
M.tb and other genes are preferably ligated into an expression vector which has been specifically optimized for polynucleotide vaccinations. Elements include a transcriptional promoter, immunogenic epitopes, and additional cistrons encoding immunoenhancing or immunomodulatory genes, with their own promoters, transcriptional terminator, bacterial origin of replication and antibiotic resistance gene, as described herein. Optionally, the vector may contain internal ribosome entry sites (IRES) for the expression of polycistronic mRNA. Those skilled in the art will appreciate that RNA which has been transcribed in vitro to produce multi-cistronic mRNAs encoded by the DNA counterparts is within the scope of this invention. For this purpose, it is desirable to use as the transcriptional promoter such powerful RNA polymerase promoters as the T7 or SP6 promoters, and performing in vitro run-on transcription with a linearized DNA template. These methods are well known in the art. The protective efficacy of polynucleotide M.tb immunogens against subsequent challenge is demonstrated by immunization with the DNA of this invention. This is advantageous since no infectious agent is involved, no assembly/replication of bacteria is required, and determinant selection is permitted. Furthermore, because the sequence of mycobacterial gene products may be conserved among various strains of M.tb, protection against subsequent challenge by another strain of M.tb is obtained.
The injection of a DNA expression vector encoding antigen 85A, B or C may result in the generation of significant protective immunity against subsequent challenge. In particular, specific CTLs and helper T lymphocyte responses may be produced.
Because each of the M.tb gene products exhibit a high degree of conservation among the various strains of M.tb and because immune responses may be generated in response to intracellular expression and MHC processing, it is expected that many different M.tb PNV constructs may give rise to cross reactive immune responses.
The invention offers a means to induce heterologous protective immunity without the need for self-replicating agents or adjuvants. The generation of high titer antibodies against expressed proteins after injection of viral protein and human growth hormone DNA, [Tang et al.. Nature 356, 152, 1992], indicates this is a facile and highly effective means of making antibody-based vaccines, either separately or in combination with cytotoxic T-lymphocyte and helper T lymphocyte vaccines targeted towards conserved antigens. The ease of producing and purifying DNA constructs compares favorably with traditional protein purification, facilitating the generation of combination vaccines. Thus, multiple constructs, for example encoding antigen 85 complex genes and any other M.tb gene also including non-M.tb genes may be prepared, mixed and co- administered. Additionally, protein expression is maintained following DNA injection [H. Lin et al, Circulation 82, 2217 (1990); R.N. Kitsis et al, Proc. Natl. Acad. Sci. (USA) 88, 4138 (1991); E. Hansen et al, FEBS Lett. 290, 73 (1991); S. Jiao et al, Hum. Gene Therapy 3, 21 (1992); J.A. Wolff et al, Human Mol. Genet. 1 , 363 (1992)], the persistence of B- and T-cell memory may be enhanced [D. Gray and P. Matzinger, J. Exp. Med. 174, 969 (1991); S. Oehen et al, ibid. 176, 1273 (1992)], thereby engendering long-lived humoral and cell- mediated immunity. The amount of expressible DNA or transcribed RNA to be introduced into a vaccine recipient will have a very broad dosage range and may depend on the strength of the transcriptional and translational promoters used. In addition, the magnitude of the immune response may depend on the level of protein expression and on the immunogenicity of the expressed gene product. In general, an effective dose ranges of about 1 ng to 5 mg, lOOng to 2.5 mg, lμg to 750 μg, and preferably about 10 μg to 300 μg of DNA is administered directly into muscle tissue. Subcutaneous injection, intradermal introduction, impression through the skin, and other modes of administration such as intraperitoneal, intravenous, or inhalation delivery are also suitable. It is also contemplated that booster vaccinations may be provided. Following vaccination with M.tb polynucleotide immunogen, boosting with M.tb protein immunogens such as the antigen 85 complex gene products is also contemplated. Parenteral administration, such as intravenous, intramuscular, subcutaneous or other means of administration of interleukin-12 protein (or other cytokines, e.g. GM- CSF), concurrently with or subsequent to. parenteral introduction of the PNV of this invention may be advantageous.
The polynucleotide may be naked, that is, unassociated with any proteins, adjuvants or other agents which affect the recipients' immune system. In this case, it is desirable for the polycucleotide to be in a physiologically acceptable solution, such as, but not limited to, sterile saline or sterile buffered saline. Alternatively, the DNA may be associated with liposomes, such as lecithin liposomes or other liposomes known in the art, as a DNA-liposome mixture, or the DNA may be associated with an adjuvant known in the art to boost immune responses, such as a protein or other carrier. Agents which assist in the cellular uptake of DNA, such as, but not limited to, calcium ions, may also be used. These agents are generally referred to herein as transfection facilitating reagents and pharmaceutically acceptable carriers. Techniques for coating microprojectiles coated with polynucleotide are known in the art and are also useful in connection with this invention. For DNA intended for human use it may be useful to have the final DNA product in a pharmaceutically acceptable carrier or buffer solution. Pharmaceutically acceptable carriers or buffer solutions are known in the art and include those described in a variety of texts such as Remington's Pharmaceutical Sciences.
In another embodiment, the invention is a polynucleotide which comprises contiguous nucleic acid sequences capable of being expressed to produce a gene product upon introduction of said polynucleotide into eukaryotic tissues in vivo. The encoded gene product preferably either acts as an immunostimulant or as an antigen capable of generating an immune response. Thus, the nucleic acid sequences in this embodiment encode an M.tb immunogenic epitope, and optionally a cytokine or a T-cell costimulatory element, such as a member of the B7 family of proteins.
There are several advantages of immunization with a gene rather than its gene product. The first is the relative simplicity with which native or nearly native antigen can be presented to the immune system. Mammalian proteins expressed recombinantly in bacteria, yeast, or even mammalian cells often require extensive treatment to insure appropriate antigenicity. A second advantage of DNA immunization is the potential for the immunogen to enter the MHC class I pathway and evoke a cytotoxic T cell response. Immunization of mice with DNA encoding the influenza A nucleoprotein (NP) elicited a CD8+ response to NP that protected mice against challenge with heterologous strains of flu. (Montgomery, D.L. et aj.., supra; Ulmer, J. et ah, supra) There is strong evidence that cell-mediated immunity is important in controlling M.tb infection [Orme et al, 1993, J. Infect. Dis. 167, 1481; Cooper et al 1993, J. Exp. Med. 178, 2243; Flynn et al, 1993, J. Exp. Med. 178, 2249; Orme et al, 1993, J. Immunol. 151, 518]. Since DNA immunization can evoke both humoral and cell-mediated immune responses, its greatest advantage may be that it provides a relatively simple method to survey a large number of M.tb genes for their vaccine potential.
Immunization by DNA injection also allows, as discussed above, the ready assembly of multicomponent subunit vaccines.
Simultaneous immunization with multiple influenza genes has recently been reported. (Donnelly, J. et al., 1994, Vaccines, pp 55-59). The inclusion in an M.tb vaccine of genes whose products activate different arms of the immune system may also provide thorough protection from subsequent challenge.
The vaccines of the present invention are useful for administration to domesticated or agricultural animals, as well as humans. Vaccines of the present invention may be used to prevent and/or combat infection of any agricultural animals, including but not limited to, dairy cattle, which are susceptible to Mycobacterial infection. The techniques for administering these vaccines to animals and humans are known to those skilled in the veterinary and human health fields, respectively.
The following examples are provided to illustrate the present invention without, however, limiting the same thereto.
EXAMPLE 1
Vectors for Vaccine Production A) V I Expression Vector
The expression vector VI was constructed from pCMVIE- AKI-DHFR [Y. Whang et al, J. Virol. 61 , 1796 (1987)]. The AKI and DHFR genes were removed by cutting the vector with EcoR I and self- ligating. This vector does not contain intron A in the CMV promoter, so it was added as a PCR fragment that had a deleted internal Sac I site [at 1855 as numbered in B.S. Chapman et al, Nuc. Acids Res. 19, 3979 (1991)]. The template used for the PCR reactions was pCMVintA-Lux, made by ligating the Hind IH and Nhe I fragment from pCMV6al20 [see B.S. Chapman et al, ibid.,] which includes hCMV-EEl enhancer/promoter and intron A, into the Hind HI and Xba I sites of pBL3 to generate pCMVIntBL. The 1881 base pair luciferase gene fragment (Hind Bl-Sma I Klenow filled-in) from RSV-Lux [J.R. de Wet et al, Mol. Cell Biol. 7, 725, 1987] was cloned into the Sal I site of pCMVIntBL, which was Klenow filled-in and phosphatase treated.
The primers that spanned intron A are: 5' primer, SEQ. ID:1 :
5 -CTATATAAGCAGAG CTCGTTTAG-3'; The 3' primer, SEQ ID:2: 5 -GTAGCAA AG ATCTAAGG ACGGTG A CTGC AG-3'.
The primers used to remove the Sac I site are: sense primer, SEQ ID:3:
5-GTATGTGTCTGAAAATGAGCQIGGAGATTGGGCTCGCAC-3' and the antisense primer, SEQ ID:4:
5 -GTGCGAGCCCAATCTCCACGCTCATTTTCAGACACA TAC-3'.
The PCR fragment was cut with Sac I and Bgl II and inserted into the vector which had been cut with the same enzymes.
B) VI J Expression Vector
The purpose in creating VU.was to remove the promoter and transcription termination elements from vector V 1 in order to place them within a more defined context, create a more compact vector, and to improve plasmid purification yields.
VI J is derived from vectors VI and pUC18, a commercially available plasmid. V 1 was digested with Sspl and EcoRI restriction enzymes producing two fragments of DNA. The smaller of these fragments, containing the CMVintA promoter and Bovine Growth Hormone (BGH) transcription termination elements which control the expression of heterologous genes, was purified from an agarose electrophoresis gel. The ends of this DNA fragment were then "blunted" using the T4 DNA polymerase enzyme in order to facilitate its ligation to another "blunt-ended" DNA fragment. pUC18 was chosen to provide the "backbone" of the expression vector. It is known to produce high yields of plasmid, is we 11 -characterized by sequence and function, and is of small size. The entire lac operon was removed from this vector by partial digestion with the Haeϋ restriction enzyme. The remaining plasmid was purified from an agarose electrophoresis gel, blunt-ended with the T4 DNA polymerase treated with calf intestinal alkaline phosphatase, and ligated to the CMVintA/BGH element described above. Plasmids exhibiting either of two possible orientations of the promoter elements within the pUC backbone were obtained. One of these plasmids gave much higher yields of DNA in E. coli and was designated V1J. This vector's structure was verified by sequence analysis of the junction regions and was subsequently demonstrated to give comparable or higher expression of heterologous genes compared with VI.
C) VUneo Expression Vector
It was necessary to remove the ampr gene used for antibiotic selection of bacteria harboring VI J because ampicillin may not be desirable in large-scale fermenters. The ampr gene from the pUC backbone of VI J was removed by digestion with Sspl and
Eaml 1051 restriction enzymes. The remaining plasmid was purified by agarose gel electrophoresis, blunt-ended with T4 DNA polymerase, and then treated with calf intestinal alkaline phosphatase. The commercially available kanr gene, derived from transposon 903 and contained within the pUC4K plasmid, was excised using the PstI restriction enzyme, purified by agarose gel electrophoresis, and blunt-ended with T4 DNA polymerase. This fragment was ligated with the VI J backbone and plasmids with the kanr gene in either orientation were derived which were designated as VUneo #'s 1 and 3. Each of these plasmids was confirmed by restriction enzyme digestion analysis, DNA sequencing of the junction regions, and was shown to produce similar quantities of plasmid as VI J. Expression of heterologous gene products was also comparable to VI J for these VUneo vectors. VUneo#3, referred to as VUneo hereafter, was selected which contains the kanr gene in the same orientation as the ampr gene in VU as the expression construct.
D) VUns Expression Vector
An Sfi I site was added to VUneo to facilitate integration studies. A commercially available 13 base pair Sfi I linker (New
England BioLabs) was added at the Kpn I site within the BGH sequence of the vector. V neo was linearized with Kpn I, gel purified, blunted by T4 DNA polymerase, and ligated to the blunt Sfi I linker. Clonal isolates were chosen by restriction mapping and verified by sequencing through the linker. The new vector was designated VlJns. Expression of heterologous genes in VlJns (with Sfi I) was comparable to expression of the same genes in VUneo (with Kpn I).
E) VUns-tPA In order to provide an heterologous leader peptide sequence to secreted and/or membrane proteins, VlJns was modified to include the human tissue-specific plasminogen activator (tPA) leader. Two synthetic complementary oligomers were annealed and then ligated into VUn which had been Bglϋ digested. The sense and antisense oligomers were 5 -GATC ACC ATG GAT GCA ATG AAG AGA GGG CTC TGC TGT GTG CTG CTG CTG TGT GGA GCA GTC TTC GTT TCG CCC AGC GA-3\ SEQ. JD:5:, and 5 -GAT CTC GCT GGG CGA AAC GAA GAC TGC TCC ACA CAG CAG CAG CAC ACA GCA GAG CCC TCT CTT CAT TGC ATC CAT GGT-3', SEQ. ID:6. The Kozak sequence is underlined in the sense oligomer. These oligomers have overhanging bases compatible for ligation to Bglll-cleaved sequences. After ligation the upstream Bgiπ site is destroyed while the downstream Bglll is retained for subsequent ligations. Both the junction sites as well as the entire tPA leader sequence were verified by DNA sequencing. Additionally, in order to conform with the consensus optimized vector VlJns (=VUneo with an Sfil site), an Sfil restriction site was placed at the Kpnl site within the BGH terminator region of VUn-tPA by blunting the Kpnl site with T4 DNA polymerase followed by ligation with an Sfil linker (catalogue #1138, New England Biolabs). This modification was verified by restriction digestion and agarose gel electrophoresis.
F) pGEM-3-X-IRES-B7 (where X = any antigenic gene) As an example of a dicistronic vaccine construct which provides coordinate expression of a gene encoding an immunogen and a gene encoding an immuno- stimulatory protein, the murine B7 gene was PCR amplified from the B lymphoma cell line CHI (obtained from the ATCC). B7 is a member of a family of proteins which provide essential costimulation T cell activation by antigen in the context of major histocompatibiHty complexes I and II. CHI cells provide a good source of B7 mRNA because they have the phenotype of being constitutively activated and B7 is expressed primarily by activated antigen presenting cells such as B cells and macrophages. These cells were further stimulated in vitro using cAMP or IL-4 and mRNA prepared using standard guanidinium thiocyanate procedures. cDNA synthesis was performed using this mRNA using the GeneAmp RNA PCR kit (Perkin -Elmer Cetus) and a priming oligomer (5'-GTA CCT CAT GAG CCA CAT AAT ACC ATG-3', SEQ. ID:7:) specific for B7 located downstream of the B7 translational open reading frame. B7 was amplified by PCR using the following sense and antisense PCR oligomers: 5'-GGT ACA AGA TCT ACC ATG GCT TGC AAT TGT CAG TTG ATG C-3', SEQ. ID:8:, and 5 -CCA CAT AGA TCT CCA TGG GAA CTA AAG GAA GAC GGT CTG TTC-3', SEQ. ID:9:, respectively. These oligomers provide Bgiπ restriction enzyme sites at the ends of the insert as well as a Kozak translation initiation sequence containing an Ncol restriction site and an additional Ncol site located immediately prior to the 3'-terminal Bglll site. Ncol digestion yielded a fragment suitable for cloning into pGEM- 3 -IRES which had been digested with Ncol. The resulting vector, pGEM-3-IRES-B7, contains an IRES-B7 cassette which can easily be transferred to VUns-X, where X represents an antigen-encoding gene.
G) pGEM-3-X-IRES-GM-CSF
(where X = any antigenic gene) This vector contains a cassette analogous to that described in item C above except that the gene for the immuno stimulatory cytokine, GM-CSF, is used rather than B7. GM-CSF is a macrophage differentiation and stimulation cytokine which has been shown to elicit potent anti-tumor T cell activities in vivo [G. Dranoff et al, Proc. Natl. Acad. Sci. USA, 90, 3539 (1993).
H) pGEM-3-X-IRES-IL-12
(where X = any antigenic gene) This vector contains a cassette analogous to that described in item C above except that the gene for the immunostimulatory cytokine, IL-12, is used rather than B7. IL- 12 has been demonstrated to have an influential role in shifting immune responses towards cellular, T cell-dominated pathways as opposed to humoral responses [L. Alfonso et al, Science, 263, 235, 1994].
EXAMPLE 2
Vector V 1 R Preparation In an effort to continue to optimize the basic vaccination vector, a derivative of VlJns, designated V1R, was prepared. The purpose for this vector construction was to obtain a minimum-sized vaccine vector without unneeded DNA sequences, which still retained the overall optimized heterologous gene expression characteristics and high plasmid yields that VI J and VlJns afford. It was determined from the literature as well as by experiment that (1) regions within the pUC backbone comprising the E. coli origin of replication could be removed without affecting plasmid yield from bacteria; (2) the 3'-region of the kan* gene following the kanamycin open reading frame could be removed if a bacterial terminator was inserted in its place; and, (3) ~300 bp from the 3'- half of the BGH terminator could be removed without affecting its regulatory function (following the original Kpnl restriction enzyme site within the BGH element). VI R was constructed by using PCR to synthesize three segments of DNA from VlJns representing the CMVintA promoter/BGH terminator, origin of replication, and kanamycin resistance elements, respectively. Restriction enzymes unique for each segment were added to each segment end using the PCR oligomers: Sspl and Xhol for CMVintA/BGH; EcoRV and BamHI for the kan r gene; and, Bell and Sail for the ori r. These enzyme sites were chosen because they allow directional ligation of each of the PCR-derived DNA segments with subsequent loss of each site: EcoRV and Sspl leave blunt- ended DNAs which are compatible for ligation while BamHI and Bell leave complementary overhangs as do Sail and Xhol. After obtaining these segments by PCR each segment was digested with the appropriate restriction enzymes indicated above and then ligated together in a single reaction mixture containing all three DNA segments. The 5'-end of the ori r was designed to include the T2 rho independent terminator sequence that is normally found in this region so that it could provide termination information for the kanamycin resistance gene. The ligated product was confirmed by restriction enzyme digestion (>8 enzymes) as well as by DNA sequencing of the ligation junctions. DNA plasmid yields and heterologous expression using viral genes within VI R appear similar to VlJns. The net reduction in vector size achieved was 1346 bp (VlJns = 4.86 kb; V1R = 3.52 kb).
PCR oligomer sequences used to synthesize V1R (restriction enzyme sites are underlined and identified in brackets following sequence):
(1) 5 -GGT ACA AAT ATT GG CTA TTG GCC ATT GCA TAC G- 3 ' [SspI], SEQ.ID:10:,
(2) 5 -CCA CAT CTC GAG GAA CCG GGT CAA TTC TTC AGC ACC-3' [Xhol], SEQ.ID:11 : (for CMVintA/BGH segment)
(3)5-GGTACA GATATCGGA AAGCCA CGTTGTGTCTCA AAATC-3'[EcoRV], SEQ.ID:12: (4) 5-CCA CATGGA TCCG TAATGCTCTGCCAGTGTTACA ACC-3' [BamHI],SEQ.ID:13:
(for kanamycin resistance gene segment)
(5) 5'-GGT ACA TGA TCA CGT AGA AAA GAT CAA AGG ATC TTC TTG-3*[BclI], SEQ.ID: 14:,
(6) 5 -CCA CAT GTC GAC CC GTA AAA AGG CCG CGT TGC
TGG-3' [Sail], SEQ.ID.15:
(for E. coli origin of replication)
EXAMPLE 3
Cell Culture and Transfection
For preparation of stably transfected cell lines expressing M.tb antigens RD cells (human rhabdomyosarcoma ATCC CCL 136) were grown at 37°C, 5% Cθ2 in Dulbecco's modified Eagle's medium
(DMEM) supplemented with 10% heat inactivated fetal bovine serum, 20mM HEPES, 4mM L-glutamine, and 100 μg/mL each of penicillin and streptomycin. Cells were seeded at 1.5x10^ cells/100 mm^ plate and grown for 1 hours. Cell were transfected with lOμg/plate of the TB construct and 10 μg of co-transfected Cat construct using the
CellPhect kit (Pharmacia), and glycerol shocked (15% glycerol in PBS, pH 7.2 for 2.5 min) 5 hours after DNA was added to the cells. Cultures were harvested 72 hours after transfection by washing the plates 2x-10 mL of cold PBS, pH 7.2, adding 5 mL of cold TEN buffer (40 mM TRIS-Cl, pH 7.5, 1 mM EDTA, 150 mM NaCl) and scraping. For analysis of protein expression, cell pellets were lysed in 50 μL of Single Detergent Lysis Buffer (50 mM Tris-Cl, pH 8.0, 150 mM NaCl, 0.02% NaN3, l %Nonidet P-40, 100 mM PMSF, 2 μg/mL aprotinin, 2 μg/mL leupeptin, and 1 μg/mL Pepstatin A) and sonicated on ice (2-15 second bursts). Lysates were centrifuged at 13,000xg, 4°C, for 10 minutes. Protein concentration was determined by the Bradford method and 20 μg of cell extract protein per lane was applied to a 10% TRIS-glycine polyacrylamide gel (Novex), then transferred to Immobilon P (Millipore) membrane. Immunoblots were reacted overnight with a 1 :20 dilution of the mouse monoclonal antibody TD 17-4 [Huygen et al, 1994, Infect. Immunity 62, 363], followed by a 1.5 hours reaction with a 1 : 1000 dilution of goat anti-mouse IgGFc peroxidase (Jackson). The blots were developed using the ECL kit (Amersham).
EXAMPLE 4
Cloning and DNA preparation
1. Construction of VI Jns-tPA-85A (contains mature Ag85A with tPA signal sequence) was done using the following primers:
sense 85A.C1 primer [SEQ.ID.NO.:16] GG AAG ATC TTT TCC CGG CCG GGC TTG CCG Bgl D
antisense 85 A primer [SEQ.ID.NO.:17] GGAAGATCTTGTCTGTTCGGAGCTAGGC.
The Ag85A from M. tuberculosis was amplified from plasmid p85A.tub, which was prepared by ligating an 800 bp Hindlll fragment to a 1600 bp Hindlll-SphI fragment from Figure 2 of Borremans et al, 1989 [Infect. Immunity 57, 3123]. The resulting 2400 bp insert was subcloned in the Hindlll and SphI sites of the BlueScribe M13+. The entire coding sequence and flanking regions in BlueScribe Ml 3+ (VCS/Stratagene) were amplified by PCR with the indicated primers in the following conditions. Each 100 μl reaction contains 2.5 Units Cloned Pfu DNA Polymerase (Stratagene), 200 mM dNTP, 0.5 μg of each primer and 250 ng of template DNA in the reaction buffer supplied with the enzyme (Stratagene). The Hybaid Thermal Reactor was programmed as follows: 5 minutes denaturation at 94°C followed by 25 cycles (1 minute at 94°C, 2 minutes at 55°C and 3 minutes at 72°C) ending with 10 minutes extension at 72°C. Amplified DNA was digested with 50 μg/ml Proteinase K
(Boehringer Mannheim) for 30 minutes at 37°C, heated 10 minutes at 95 °C followed by 2 phenol (Chloroform-Isoamyl alcohol) extractions and precipitated with 1 volume of isopropanol, washed twice with 70% ethanol, dried and dissolved in 20 μl H2θ. 3 μg of amplified DNA was digested with 40 Units of Bgl II (Boehringer Mannheim) and the 907 bp fragment (in the case of 85A-C1) was isolated on a 1 % agarose gel and extracted on "Prep a Gene" (BioRad) following the manufacturer's instructions.
Fifty ng of this fragment was ligated to 20 ng of the Bgl II digested and dephosphorylated VUns.tPA vector in a 10 μl reaction containing 2.5 Units T4 DNA ligase (Amersham) in ligation buffer for 16 hours at 14°C, transformed into competent DH5 E. coli (BRL) and plated on Kanamycin (50 μg/ml) containing LB Agar medium. Transformants were picked up and their plasmidic DNA was restricted with Bgl II (to confirm the presence of insert) and with Pvu II to define its orientation.
2. Construction of VUns-85A [C2] (contains mature Ag85A with no signal sequence) was done using the following primers:
Sense 85 A C2 [SEQ.ID.NO.:18]
GGAAGATCTACC ATG GGC TTT TCC CGG CCG GGC TTG C
Antisense 85 A [SEQ.ID.NO.:17] GGAAGATCTTGCTGTTCGGAGCTAGGC.
The same procedure as 1 above was followed, except that cloning was in VlJns. 3. Construction of VlJns-85A [C3] (contains Ag85A with its own signal sequence) was done using the primers:
Sense 85A C3 [SEQ.ID.NO.:19] GGAAGATCTACC ATG GCA CAG CTT GTT GAC AGG GTT
Antisense 85 A [SEQ.JD.NO.:17] GGAAGATCTTGCTGTTCGGAGCTAGGC.
The same procedure as 1 above was followed, except that cloning was in VlJns.
4. Construction of VUns-tPA-85B [Cl] (contains Ag85B with tPA signal sequence) was done using the following primers:
Sense 85B [Cl] [SEQ.ID.NO.:20]
GGAAG ATC TCC TTC TCC CGG CCG GGG CTG CCG GTC GAG
Antisense 85B [SEQ.ID.NO.:21] GGAAGATCTAACCTTCGGTTGATCCCGTCAGCC.
The same procedure as 1 above was followed, except that the template for PCR was p85B.tub.
5. Construction of V Uns-tPA-85C [Cl ] (contains Ag85C with tPA signal sequence) was done using the following primers:
Sense 85C [Cl] [SEQ.ID.NO.:22]
GGAAG ATC TCC TTC TCT AGG CCC GGT CTT CCA
Antisense 85C [SEQ.ID.NO.:23] GGAAGATCTTGCCGATGCTGGCTTGCTGGCTCAGGC. The same procedure as 1 above was followed, except that the template for PCR was p85C.tub.
6. Construction of VlJns-85B [C2] (contains Ag85B with no signal sequence) is done using the following primers:
Sense 85B [C2] [SEQ.ID.NO.:24]
GGA AGA TCT ACC ATG GGC TTC TCC CGG CCG GGG CTG C
Antisense 85B [SEQ.JD.NO.:21]
GGAAGATCTAACCTCGGTTGATCCCGTCAGCC.
The same procedure as 1 above is followed, except that template for PCR is p85B.tub and that cloning is in VlJns.
7. Construction of VI Jns-85C [C2] (contains Ag85C with no signal sequence) is done using the following primers:
Sense 85C [C2] [SEQ.ID.NO.:25] GGA AGA TCT ACC ATG GGC TTC TCT AGG CCC GGT CTT C
Antisense 85C [SEQ.ID.NO.:23] GGAAGATCTTGCCGATGCTGGCTTGCTGGCTCAGGC.
The same procedure as 1 above is followed, except that template for PCR is p85C.tub and that cloning is in VlJns.
After restriction analysis all of the constructions are partially sequenced across the vector junctions. Large scale DNA preparation was essentially as described (Montgomery, D.L. et al., supra).
The plasmid constructions were characterized by restriction mapping and sequence analysis of the vector-insert junctions (see Figures 1 -6). Results were consistent with published M.tb sequence data and showed that the initiation codon was intact for each construct (Figure 7). Also shown are the various additional amino acid residues unrelated to M.tb Ag85 that were inserted as a result of cloning.
EXAMPLE 5
Expression of M.tb proteins from VUns.tPA plasmids
Rhabdomyosarcoma cells (ATCC CCL136) were planted one day before use at a density of 1.2 X106 cells per 9.5 cm2 well in six-well tissue culture clusters in high glucose DMEM supplemented with 10% heat-inactivated fetal calf serum, 2 mM L-glutamine, 25 mM HEPES, 50 U/ml penicillin and 50 μg/ml streptomycin. (All from BRL-Gibco) Phenol : chloroform extracted cesium chloride purified plasmid DNA was precipitated with calcium phosphate using Pharmacia CellPhect reagents according to the kit instmctions except that 5 - 15 μg is used for each 9.5 cm2 well of RD cells. Cultures were glycerol shocked six hours post addition of calcium phosphate-DNA precipate; after refeeding, cultures were incubated for two days prior to harvest. Lysates of transfected cultures were prepared in IX RIPA (0.5% SDS, 1.0% TRITON X-100, 1% sodium deoxycholate, ImM EDTA, 150mM NaCl, 25 mM TRIS-HC1 pH 7.4) supplemented with lμM leupeptin, lμM pepstatin, 300nM aprotinin, and lOμM TLCK, and sonicated briefly to reduce viscosity. Lysates were resolved by electrophoresis on 10% Tricine gels (Novex) and then transferred to nitrocellulose membranes. Immunoblots were processed with M.tb monoclonal antibodies 17/4 and 32/15 [Huygen et al, 1994, Infect. Immunity 62, 363] and developed with the ECL detection kit (Amersham).
Expression of M.tb antigen 85 complex genes was demonstrated by transient transfection of RD cells. Lysates of transfected or mock transfected cells were fractionated by SDS PAGE and analyzed by immunoblotting. Figure 8 shows that V Uns.tPA- 85A(C1), VUns.tPA-85A(C2), VUns.tPA-85A(C3), and V Uns.tPA- 85B(C1 ) transfected RD cells express an immunoreactive protein with an apparent molecular weight of approximately 30-32kDa.
EXAMPLE 6
Immunization with PNV and Expression of Antigen 85 Proteins In Vivo
Five- to six-week-old female BALB/c and C57BL/6 mice were anesthetized by intraperitoneal (i.p.) injection of a mixture of 5 mg ketamine HC1 (Aveco, Fort Dodge, IA) and 0.5 mg xylazine
(Mobley Corp., Shawnee, KS.) in saline. The hind legs were washed with 70% ethanol. Animals were injected three times with 100 μl of DNA (2 mg/ml) suspended in saline: 50 μl each leg. At 17-18 days after immunization, serum samples were collected and analyzed for the presence of anti-Ag85 antibodies. Figure 9 shows specific immunoblot reactivity of sera from Ag85 DNA-injected mice (Cl) but not from mice that received a control DNA not containing a gene insert (V1J). Reactivity was detected to a serum dilution of at least 1 : 160 against 300 ng of purified antigen 85A (Figure 9b). This demonstrates that injection of Ag85 DNA resulted in Ag85 expression in vivo such that it was available for the generation of antibody responses in both BALB/c and C57BL/6 (B6) mice.
EXAMPLE 7
Antigen 85-Specific T-Cell Responses
Spleen cells from vaccinated mice were analyzed for cytokine secretion in response to specific antigen restimulation as described in Huygen et al, 1992 [Infect. Immunity 60, 2880]. Specifically, spleen cells were incubated with culture filtrate (CF) proteins from M. bovis BCG purified antigen 85A or a 20-mer peptide (p25) corresponding to a known T-cell epitope for C57BL/6 mice (amino acids 241-260). Mice were immunized with VUns.tPA85A (Cl ) (100 μg) three times with three week intervals and analyzed 17 days after the final injection. Cytokines were assayed using bio-assays for IL-2, interferon-γ (IFN-γ) and IL-6, and by ELISA for IL-4 and IL-10. Substantial IL-2 and IFN-γ production was observed in both BALB/c and C57BL/6 mice vaccinated with VUns.tPA85A (Cl) (Figures 10- 13). Furthermore, C57BL/6 mice also reacted to the H-2b-restricted T- cell epitope (Figure 13). IL-4, IL-6 and IL-10 levels were not increased in VUns.tPA85 A- vaccinated mice (Figures 14-16). These results indicate that a Thl type of helper T-cell response was generated by the DNA vaccine.
EXAMPLE 8
Protection from Mycobacterial Challenge
To test the efficacy of an M.tb DNA vaccine, mice were challenged with an intravenous injection of live M. bovis BCG (0.5 mg) and BCG multiplication was analyzed in the spleens and lungs. As controls, BCG multiplication was measured in challenged naive mice (primary infection) and challenged mice that were vaccinated with BCG at the time of DNA injection (secondary infection). The number of colony-forming units (CFU) in lungs of VUns.tPA85A (Cl)-vaccinated mice was substantially reduced compared to mice with primary infection or mice vaccinated with control DNA VU. In C57BL/6 mice, CFU were reduced by 83% on day 8 after challenge (Figure 17) and in BALB/c mice CFU was reduced by 65% on day 20 (Figure 18). In spleen, CFU was reduced by approximately 40% at day 20 after challenge in BALB/c mice (Figure 19) and day 8 in C57BL/6 mice (Figure 20). Therefore, the immune responses observed after injection of an M.tb DNA vaccine provided protection in a live M. bovis challenge model. SEQUENCE LISTING
(1) GENERAL INFORMATION:
(l) APPLICANT: CONTENT, JEAN HUYGEN, KRIS LIU, MARGARET A. MONTGOMER , DONNA ULMER, JEFFREY
(11) TITLE OF INVENTION: A POLYNUCLEOTIDE TUBERCULOSIS VACCINE
(in) NUMBER OF SEQUENCES: 25
(IV) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: JACK L. TRIBBLE
(B) STREET: 126 E. LINCOLN AVE. , P.O. BOX 2000
(C) CITY: RAHWAY
(D) STATE: NEW JERSEY
(E) COUNTRY: USA
(F) ZIP: 07065-0907
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/338,992
(B) FILING DATE: 14-NOV-1994
(C) CLASSIFICATION:
(vin) ATTORNEY/AGENT INFORMATION:
(A) NAME: TRIBBLE, JACK L.
(B) REGISTRATION NUMBER: 32,633
(C) REFERENCE/DOCKET NUMBER: 19342
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (908) 594-5321
(B) TELEFAX: (908) 594-4720
(2) INFORMATION FOR SEQ ID NO: 1 :
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic! (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1 : CTATATAAGC AGAGCTCGTT TAG 23
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2 : GTAGCAAAGA TCTAAGGACG GTGACTGCAG 30
(2) INFORMATION FOR SEQ ID NO:3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3 : GTATGTGTCT GAAAATGAGC GTGGAGATTG GGCTCGCAC 39
(2) INFORMATION FOR SEQ ID NO: :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: : GTGCGAGCCC AATCTCCACG CTCATTTTCA GACACATAC 39
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 78 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5 : GATCACCATG GATGCAATGA AGAGAGGGCT CTGCTGTGTG CTGCTGCTGT GTGGAGCAGT 60 CTTCGTTTCG CCCAGCGA 78
(2) INFORMATION FOR SEQ ID NO: 6 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 78 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: GATCTCGCTG GGCGAAACGA AGACTGCTCC ACACAGCAGC AGCACACAGC AGAGCCCTCT 60 CTTCATTGCA TCCATGGT 78
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7 : GTACCTCATG AGCCACATAA TACCATG 27
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 40 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single !D^ TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: GGTACAAGAT CTACCATGGC TTGCAATTGT CAGTTGATGC 40
(2) INFORMATION FOR SEQ ID NO:9 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: CCACATAGAT CTCCATGGGA ACTAAAGGAA GACGGTCTGT TC 42
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10: GGTACAAATA TTGGCTATTG GCCATTGCAT ACG 33
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: CCACATCTCG AGGAACCGGG TCAATTCTTC AGCACC 36 (2) INFORMATION FOR SEQ ID NO:12:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 38 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(n) MOLECULE TYPE: DNA (genomic)
(XI) SEQUENCE DESCRIPTION: SEQ ID NO:12: GGTACAGATA TCGGAAAGCC ACGTTGTGTC TCAAAATC 38
(2) INFORMATION FOR SEQ ID NO: 13:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 37 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: CCACATGGAT CCGTAATGCT CTGCCAGTGT TACAACC 37
(2) INFORMATION FOR SEQ ID NO:14:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14: GGTACATGAT CACGTAGAAA AGATCAAAGG ATCTTCTTG 39
(2) INFORMATION FOR SEQ ID NO:15:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 base pairs
(B) TYPE: nucleic acid ( STRANDEDNESS: single (D> TOPOLOGY: linear (11) MOLECULE TYPE: DNA (genomic)
(Xl) SEQUENCE DESCRIPTION: SEQ ID NO:15: CCACATGTCG ACCCGTAAAAA GGCCGCGTTG CTGG 35
(2) INFORMATION FOR SEQ ID NO:16:
(1) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(Xl) SEQUENCE DESCRIPTION: SEQ ID NO:16: GGAAGATCTT TTCCCGGCCG GGCTTGCCG 29
(2) INFORMATION FOR SEQ ID NO:17:
(1) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 28 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17: GGAAGATCTT GTCTGTTCGG AGCTAGGC 28
(2) INFORMATION FOR SEQ ID NO:18:
( l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 37 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(Xl) SEQUENCE DESCRIPTION: SEQ ID NO: 18: GGAAGATCTA CCATGGGCTT TTCCCGGCCG GGCTTGC 37 (2) INFORMATION FOR SEQ ID NO:19:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: GGAAGATCTA CCATGGCACA GCTTGTTGAC AGGGTT 36
(2) INFORMATION FOR SEQ ID NO:20:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 38 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20: GGAAGATCTC CTTCTCCCGG CCGGGGCTGC CGGTCGAG 38
(2) INFORMATION FOR SEQ ID NO:21:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(n) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION. SEQ ID NO:21: GGAAGATCTA ACCTTCGGTT GATCCCGTCA GCC 33
(2) INFORMATION FOR SEQ ID NO:22:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: GGAAGATCTC CTTCTCTAGG CCCGGTCTTC CA 32
(2) INFORMATION FOR SEQ ID NO:23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: GGAAGATCTT GCCGATGCTG GCTTGCTGGC TCAGGC 36
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 37 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: GGAAGATCTA CCATGGGCTT CTCCCGGCCG GGGCTGC 37
(2) INFORMATION FOR SEQ ID NO:25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 37 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25: GGAAGATCTA CCATGGGCTT CTCTAGGCCC GGTCTTC 37

Claims

WHAT IS CLAIMED IS:
1. A polynucleotide which induces upon introduction into vertebrate tissue, one or more anti-Mycobacterial immune responses selected from antibodies, CTL, helper T lymphocyte responses and protective immune responses, wherein said polynucleotide comprises one or more genes encoding one or more Mycobacterial proteins or functional equivalents thereof, said genes being operably linked to a transcription promoter.
2. The polynucleotide of Claim 1, wherein said gene encodes a Mycobacterium tuberculosis protein, and functional equivalents thereof.
3. The polynucleotide of Claim 2, wherein said gene encodes a protein selected from a group consisting of antigen 85A, B, and/or C, and functional equivalents thereof.
4. A method for inducing immune responses in a vertebrate against Mycobacterial epitopes, comprising introducing between 1 ng and 5 mg of a polynucleotide according to Claim 1 into a tissue of a vertebrate.
5. The method of Claim 4, wherein said gene encodes a Mycobacterium tuberculosis protein, and functional equivalents thereof.
6. The method of Claim 5, wherein said gene encodes a protein selected from a group consisting of antigen 85A, B, and C, and functional equivalents thereof.
7. A vaccine for inducing immune responses against Mycobacterial antigens, comprising the polynucleotide of Claim 1 and a pharmaceutically acceptable carrier.
8. The vaccine of Claim 7, wherein said antigen is a Mycobacterium tuberculosis antigen, and functional equivalents thereof.
9. The vaccine of Claim 8, wherein said antigen is a protein selected from a group consisting of antigen 85A, B, and C, and functional equivalents thereof.
10. A method for inducing immune responses against mycobacterial antigens, comprising introducing into a tissue of a vertebrate one or more isolated and purified mycobacterial genes eliciting an immune response which prevents mycobacterial infection and/or ameliorates mycobacterial disease.
1 1. The method of Claim 10, wherein said gene encodes a Mycobacterium tuberculosis protein, and functional equivalents thereof.
12. The method of Claim 1 1 , wherein said gene encodes a protein selected from a group consisting of antigen 85 A, B, and C, and functional equivalents thereof.
13. A polynucleotide comprising: a) a eukaryotic transcription promoter; b) an open reading frame operably linked to said promoter encoding one or more mycobacterial epitopes, and a translation termination signal; and c) optionally containing one or more operably linked IRES, one or more open reading frames encoding one or more additional genes, and one or more transcription termination signals.
14. The polynucleotide of Claim 13 wherein said additional genes of c) are immunomodulatory or immunostimulatory genes selected from a group consisting of GM-CSF, IL-12, interferon, and a member of the B7 family of T-cell costimulatory proteins.
15. The polynucleotide of Claim 13 wherein said mycobacterial gene of a) encodes a Mycobacterium tuberculosis protein, and functional equivalents thereof.
16. The polynucleotide of Claim 15 wherein said mycobacterial gene of a) encodes a Mycobacterium tuberculosis protein selected from a group consisting of antigen 85 A, B, and C, and functional equivalents thereof.
17. The polynucleotide of Claim 13 wherein said additional genes of c) are Mycobacterium tuberculosis genes selected from a group consisting of antigen 85 A, B, and C, and functional equivalents thereof.
18. A method of treating a patient in need of such treatment with a polynucleotide which induces upon introduction into vertebrate tissue, one or more anti-mycobacterial immune responses selected from antibodies, CTL, helper T lymphocyte responses and protective immune responses, wherein said polynucleotide comprises a gene encoding one or more mycobacterial proteins or functional equivalents thereof, said gene being operably linked to a transcription promoter.
19. The method of Claim 18, wherein said gene encodes a Mycobacterium tuberculosis protein, and functional equivalents thereof.
20. The method of claim 19 wherein said gene encodes one or more proteins selected from a group consisting of antigen 85A, B, and C, and functional equivalents thereof.
21. The method of claim 10 wherein said patient is a domestic animal or livestock.
22. A vaccine for inducing immune responses against Mycobacterial infection in domesticated or agricultural animals comprising the polynucleotide of Claim 1 and a pharmaceutically acceptable carrier.
PCT/US1995/014899 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine WO1996015241A2 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
DK95939161T DK0792358T3 (en) 1994-11-14 1995-11-13 Polynucleotide tuberculosis vaccine
HU9701841A HU222369B1 (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine
NZ296477A NZ296477A (en) 1994-11-14 1995-11-13 Polynucleotides and tuberculosis vaccines
AU41102/96A AU715067B2 (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine
PL95320091A PL184839B1 (en) 1994-11-14 1995-11-13 Polynucleotidic vaccine against tuberculosis
JP51633096A JP3881014B2 (en) 1994-11-14 1995-11-13 Polynucleotide tuberculosis vaccine
DE69534250T DE69534250T2 (en) 1994-11-14 1995-11-13 POLYNUCLEOTIDE VACCINE AGAINST TBC
CA 2205175 CA2205175C (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine
SK597-97A SK283254B6 (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine
CZ19971451A CZ289383B6 (en) 1994-11-14 1995-11-13 Polynucleotide and polynucleotide tuberculosis vaccine
AT95939161T ATE296882T1 (en) 1994-11-14 1995-11-13 POLYNUCLEOTIDE VACCINE AGAINST TB
EP95939161A EP0792358B1 (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine
NO972196A NO972196L (en) 1994-11-14 1997-05-13 Polynucleotide vaccine for tuberculosis
FI972034A FI972034A (en) 1994-11-14 1997-05-13 Polynucleotide tuberculosis vaccine

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US338,992 1994-11-14
US08/338,992 US5736524A (en) 1994-11-14 1994-11-14 Polynucleotide tuberculosis vaccine

Publications (2)

Publication Number Publication Date
WO1996015241A2 true WO1996015241A2 (en) 1996-05-23
WO1996015241A3 WO1996015241A3 (en) 1996-11-07

Family

ID=23326996

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/014899 WO1996015241A2 (en) 1994-11-14 1995-11-13 A polynucleotide tuberculosis vaccine

Country Status (23)

Country Link
US (2) US5736524A (en)
EP (1) EP0792358B1 (en)
JP (1) JP3881014B2 (en)
KR (1) KR970707281A (en)
CN (1) CN1171814A (en)
AT (1) ATE296882T1 (en)
AU (1) AU715067B2 (en)
CZ (1) CZ289383B6 (en)
DE (1) DE69534250T2 (en)
DK (1) DK0792358T3 (en)
ES (1) ES2242193T3 (en)
FI (1) FI972034A (en)
HU (1) HU222369B1 (en)
IL (1) IL115883A0 (en)
MX (1) MX9703606A (en)
NO (1) NO972196L (en)
NZ (1) NZ296477A (en)
PL (1) PL184839B1 (en)
PT (1) PT792358E (en)
RU (1) RU2186109C2 (en)
SK (1) SK283254B6 (en)
WO (1) WO1996015241A2 (en)
ZA (1) ZA959608B (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998016646A2 (en) * 1996-10-11 1998-04-23 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
WO1998035029A1 (en) * 1997-02-07 1998-08-13 Vanderbilt University Synthetic genes for recombinant mycobacterium proteins
JP2000515522A (en) * 1996-07-19 2000-11-21 メリアル Transdermal bovine polynucleotide vaccine formulation
US6338852B1 (en) 1995-09-01 2002-01-15 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6384018B1 (en) * 1994-11-14 2002-05-07 Merck & Co., Inc. Polynucleotide tuberculosis vaccine
US6458366B1 (en) 1995-09-01 2002-10-01 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6465633B1 (en) 1998-12-24 2002-10-15 Corixa Corporation Compositions and methods of their use in the treatment, prevention and diagnosis of tuberculosis
US6592877B1 (en) 1995-09-01 2003-07-15 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US7022320B1 (en) 1999-02-09 2006-04-04 Powderject Vaccines, Inc. Mycobacterium tuberculosis immunization
EP1817061A2 (en) * 2004-12-01 2007-08-15 Aeras Global Tuberculosis Vaccine Foundation Electroporation of mycobacterium and overexpression of antigens in mycobacteria
JP2009149608A (en) * 1996-07-19 2009-07-09 Merial Sas Polynucleotide vaccine preparation for bird
US8012467B2 (en) 2004-11-16 2011-09-06 Crucell Holland B.V. Multivalent vaccines comprising recombinant viral vectors

Families Citing this family (302)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2711670B1 (en) 1993-10-22 1996-01-12 Pasteur Institut Nucleotide vector, composition containing it and vaccine for immunization against hepatitis.
US6995008B1 (en) * 1994-03-07 2006-02-07 Merck & Co., Inc. Coordinate in vivo gene expression
US6727230B1 (en) 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
FR2732895B1 (en) * 1995-04-11 1997-05-16 Pasteur Merieux Serums Vacc USE OF A CATIONIC AMPHIPATHIC COMPOUND AS A TRANSFECTING AGENT, AS A VACCINE ADDITIVE, OR AS A MEDICINAL PRODUCT
US20070021364A1 (en) * 1995-12-13 2007-01-25 Hans Herweijer Methods for genetic immunization
US20020165183A1 (en) * 1999-11-29 2002-11-07 Hans Herweijer Methods for genetic immunization
ZA973642B (en) * 1996-04-26 1997-11-25 Merck & Co Inc DNA vaccine formulations.
US5846946A (en) * 1996-06-14 1998-12-08 Pasteur Merieux Serums Et Vaccins Compositions and methods for administering Borrelia DNA
JP2001500738A (en) * 1996-09-17 2001-01-23 カイロン コーポレイション Compositions and methods for treating intracellular diseases
US20060002949A1 (en) 1996-11-14 2006-01-05 Army Govt. Of The Usa, As Rep. By Secretary Of The Office Of The Command Judge Advocate, Hq Usamrmc. Transcutaneous immunization without heterologous adjuvant
US6797276B1 (en) * 1996-11-14 2004-09-28 The United States Of America As Represented By The Secretary Of The Army Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
US5980898A (en) * 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
US20060002959A1 (en) * 1996-11-14 2006-01-05 Government Of The United States Skin-sctive adjuvants for transcutaneous immuization
US6261281B1 (en) * 1997-04-03 2001-07-17 Electrofect As Method for genetic immunization and introduction of molecules into skeletal muscle and immune cells
US20040258703A1 (en) * 1997-11-14 2004-12-23 The Government Of The Us, As Represented By The Secretary Of The Army Skin-active adjuvants for transcutaneous immunization
JP2002511396A (en) * 1998-04-14 2002-04-16 メルク エンド カムパニー インコーポレーテッド Needleless administration of polynucleotide preparations
BR9915055A (en) * 1998-11-04 2001-08-07 Isis Innovation Tuberculosis diagnostic test
US8197461B1 (en) 1998-12-04 2012-06-12 Durect Corporation Controlled release system for delivering therapeutic agents into the inner ear
JP4932086B2 (en) * 1999-04-08 2012-05-16 インターセル ユーエスエイ、インコーポレイテッド Dry formulation for transcutaneous immunization
US20040259803A1 (en) * 1999-04-15 2004-12-23 Monash University Disease prevention by reactivation of the thymus
US20040241842A1 (en) * 1999-04-15 2004-12-02 Monash University Stimulation of thymus for vaccination development
US20040258672A1 (en) * 1999-04-15 2004-12-23 Monash University Graft acceptance through manipulation of thymic regeneration
US20070274946A1 (en) * 1999-04-15 2007-11-29 Norwood Immunoloty, Ltd. Tolerance to Graft Prior to Thymic Reactivation
US20050020524A1 (en) * 1999-04-15 2005-01-27 Monash University Hematopoietic stem cell gene therapy
US20040265285A1 (en) * 1999-04-15 2004-12-30 Monash University Normalization of defective T cell responsiveness through manipulation of thymic regeneration
AUPR074500A0 (en) * 2000-10-13 2000-11-09 Monash University Treatment of t cell disorders
WO2001000663A2 (en) * 1999-06-28 2001-01-04 Oklahoma Medical Research Foundation Catalytically active recombinant memapsin and methods of use thereof
US6514948B1 (en) 1999-07-02 2003-02-04 The Regents Of The University Of California Method for enhancing an immune response
US20050100928A1 (en) * 1999-09-16 2005-05-12 Zycos Inc., A Delaware Corporation Nucleic acids encoding polyepitope polypeptides
CA2389633A1 (en) * 1999-11-01 2001-05-10 Chiron Corporation Expression vectors, transfection systems, and method of use thereof
CA2386341A1 (en) 1999-11-18 2001-05-25 Epimmune Inc. Heteroclitic analogs and related methods
CA2398207A1 (en) * 1999-12-22 2001-06-28 The Ohio State University Research Foundation Methods for protecting against lethal infection with bacillus anthracis
FR2804028B1 (en) * 2000-01-21 2004-06-04 Merial Sas IMPROVED DNA VACCINES FOR PENSION ANIMALS
US6552006B2 (en) 2000-01-31 2003-04-22 The Regents Of The University Of California Immunomodulatory polynucleotides in treatment of an infection by an intracellular pathogen
US6613751B2 (en) 2000-02-23 2003-09-02 The Regents Of The University Of California Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
US20030130217A1 (en) * 2000-02-23 2003-07-10 Eyal Raz Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
JP2003531865A (en) * 2000-04-28 2003-10-28 アメリカ合衆国 Improving immunogenicity using a combination of DNA and vaccinia virus vector vaccines
US6590087B1 (en) * 2000-05-25 2003-07-08 Johns Hopkins University whmD, an essential cell division gene from mycobacteria
US7288261B2 (en) * 2000-07-10 2007-10-30 Colorado State University Research Foundation Mid-life vaccine and methods for boosting anti-mycobacterial immunity
EP2075582A3 (en) 2000-07-12 2010-01-06 Agensys, Inc. Novel tumor antigen useful in diagnosis and therapy of bladder, ovary, lung and kidney cancers
WO2002018578A2 (en) 2000-08-28 2002-03-07 Agensys, Inc. Nucleic acid and corresponding protein entitled 85p1b3 useful in treatment and detection of cancer
US20060088512A1 (en) * 2001-10-15 2006-04-27 Monash University Treatment of T cell disorders
CA2425648A1 (en) 2000-10-19 2002-04-19 Epimmune Inc. Hla class i and ii binding peptides and their uses
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US7829084B2 (en) 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
DE60227157D1 (en) * 2001-02-13 2008-07-31 Us Gov Sec Army IMPREGENTS FOR TRANSCUTANEOUS IMMUNIZATION AGAINST TRAVEL DIARRHOUS
US7491394B2 (en) 2001-02-15 2009-02-17 The Board Of Trustees Of The University Of Illinois Cytotoxic factors for modulating cell death
US6924358B2 (en) 2001-03-05 2005-08-02 Agensys, Inc. 121P1F1: a tissue specific protein highly expressed in various cancers
US7271240B2 (en) 2001-03-14 2007-09-18 Agensys, Inc. 125P5C8: a tissue specific protein highly expressed in various cancers
US8034791B2 (en) 2001-04-06 2011-10-11 The University Of Chicago Activation of Egr-1 promoter by DNA damaging chemotherapeutics
US20040242523A1 (en) * 2003-03-06 2004-12-02 Ana-Farber Cancer Institue And The Univiersity Of Chicago Chemo-inducible cancer gene therapy
ATE541937T1 (en) * 2001-04-06 2012-02-15 Univ Chicago CHEMOTHERAPEUTIC INTRODUCTION OF EGR-1 PROMOTER ACTIVITY IN GENE THERAPY
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20030191073A1 (en) 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US7013940B2 (en) * 2001-04-19 2006-03-21 Michelin Recherche Et Technique S.A. Device for attenuating cavity noise in a tire and wheel
WO2002095002A2 (en) 2001-05-22 2002-11-28 University Of Chicago N4 virion single-stranded dna dependent rna polymerase
WO2003015716A2 (en) * 2001-08-13 2003-02-27 Ige Therapeutics, Inc. Immunoglobulin e vaccines and methods of use thereof
EP2287186B1 (en) 2001-09-06 2014-12-31 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
AU2002348417B9 (en) * 2001-10-26 2010-02-04 Baylor College Of Medicine A composition and method to alter lean body mass and bone properties in a subject
BR0214869A (en) 2001-12-11 2005-03-08 Advisys Inc Plasmid-mediated supplementation for treatment of chronically ill individuals
IL162844A0 (en) * 2002-01-18 2005-11-20 Inovio As Use of an expression vector for preparing pharmaceutical compositions
CA2475003A1 (en) 2002-02-01 2003-08-07 Sequitur, Inc. Double-stranded oligonucleotides
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20050267025A1 (en) * 2002-02-01 2005-12-01 Ho John L Compositions and methods for treatment of infectious and inflammatory diseases
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
WO2003063798A2 (en) * 2002-02-01 2003-08-07 Cornell Research Foundation, Inc. Compositions and methods for treatment of infectious and inflammatory diseases
EP2258712A3 (en) * 2002-03-15 2011-05-04 Multicell Immunotherapeutics, Inc. Compositions and Methods to Initiate or Enhance Antibody and Major-histocompatibility Class I or Class II-restricted T Cell Responses by Using Immunomodulatory, Non-coding RNA Motifs
US20070037769A1 (en) * 2003-03-14 2007-02-15 Multicell Immunotherapeutics, Inc. Compositions and methods to treat and control tumors by loading antigen presenting cells
JP2005526778A (en) * 2002-03-15 2005-09-08 アストラル,インコーポレイテッド Compositions and methods for initiating or enhancing antibodies and major histocompatibility class I restricted or class II restricted T cell responses using immunomodulatory non-coding RNA motifs
US7078037B2 (en) * 2002-04-19 2006-07-18 The Arizona Board Of Regents On Behalf Of The University Of Arizona Peptides and DNA encoding the peptides useful for immunizations against Coccidioides spp. infections
AU2003301841A1 (en) * 2002-05-01 2004-06-07 National Institutes Of Health Immunotherapy regimens in hiv-infected patients
CN100418981C (en) 2002-06-10 2008-09-17 瓦西尼斯公司 Gene differentially expressed in breast and bladder cancer and encoded polypeptides
US20050175625A1 (en) * 2002-07-12 2005-08-11 The Johns Hopkins University Mesothelin vaccines and model systems
US20090110702A1 (en) 2002-07-12 2009-04-30 The Johns Hopkins University Mesothelin Vaccines and Model Systems and Control of Tumors
US9200036B2 (en) 2002-07-12 2015-12-01 The Johns Hopkins University Mesothelin vaccines and model systems
US6916474B2 (en) 2002-07-15 2005-07-12 Board Of Regents, The University Of Texas System Antibodies with increased affinities for anthrax antigens
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs
KR101170653B1 (en) 2002-08-12 2012-08-03 제네렉스, 인코포레이티드 Methods and compositions concerning poxviruses and cancer
US20040081653A1 (en) 2002-08-16 2004-04-29 Raitano Arthur B. Nucleic acids and corresponding proteins entitled 251P5G2 useful in treatment and detection of cancer
WO2004037175A2 (en) * 2002-10-21 2004-05-06 Mgi Pharma Biologics, Inc. Compositions and methods for treating human papillomavirus-mediated disease
JP2006508163A (en) 2002-11-27 2006-03-09 アジェンシス, インコーポレイテッド Nucleic acids and corresponding proteins referred to as 24P4C12 useful in the treatment and detection of cancer
EP1903056A3 (en) 2002-12-10 2008-05-07 Idm Pharma, Inc. HLA-A1, -A2 -A3, -A24, -B7, and -B44 binding peptides comprising tumor associated antigen epitopes, and compositions thereof
EP2343315A3 (en) 2003-02-10 2011-11-23 Agensys, Inc. Nucleic acid and corresponding protein named 158P1D7 useful in the treatment and detection of bladder and other cancers
US7262027B2 (en) * 2003-03-14 2007-08-28 Medical College Of Ohio Polypeptide and DNA immunization against Coccidioides spp. infections
CN1795274A (en) * 2003-03-26 2006-06-28 多单元免疫治疗公司 Selected rna motifs to include cell death and/or apoptosis
TW200424214A (en) * 2003-04-21 2004-11-16 Advisys Inc Plasmid mediated GHRH supplementation for renal failures
US20070037151A1 (en) * 2003-04-28 2007-02-15 Babe Lilia M Cd4+ human papillomavirus (hpv) epitopes
EP2319524B1 (en) 2003-05-30 2013-08-21 Agensys, Inc. Prostate stem cell antigen (PSCA) variants and subsequences thereof
WO2004110482A1 (en) * 2003-06-13 2004-12-23 Isis Innovation Limited Improved vaccines
CN1894581B (en) 2003-07-09 2012-02-01 生命技术公司 Method for assaying protein-protein interaction
US20070224615A1 (en) * 2003-07-09 2007-09-27 Invitrogen Corporation Methods for assaying protein-protein interactions
AU2004271951B2 (en) * 2003-09-05 2008-08-21 Genencor International, Inc. HPV CD8+ T-cell epitopes
US20080279812A1 (en) * 2003-12-05 2008-11-13 Norwood Immunology, Ltd. Disease Prevention and Vaccination Prior to Thymic Reactivation
CA2555013C (en) * 2004-02-11 2013-10-15 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Carcinoembryonic antigen fusions and uses thereof
CA2559586A1 (en) * 2004-03-18 2005-09-29 Fred Hutchinson Cancer Research Center Methods and compositions involving s-ship promoter regions
JP4651663B2 (en) 2004-05-28 2011-03-16 アジェンシス,インコーポレイテッド Antibodies and related molecules that bind to PSCA proteins
US20090221440A1 (en) * 2004-07-12 2009-09-03 Board Of Regents, The University Of Texas System Methods and compositions related to identifying protein-protein interactions
US7572600B2 (en) 2004-08-04 2009-08-11 Chemocentryx, Inc. Enzymatic activities in chemokine-mediated inflammation
EP2302054B1 (en) 2004-11-12 2014-07-16 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
WO2006086284A2 (en) * 2005-02-11 2006-08-17 Merck & Co., Inc. Adenovirus serotype 26 vectors, nucleic acid and viruses produced thereby
PL2325305T3 (en) 2005-02-25 2014-07-31 Oncotherapy Science Inc Peptide vaccines for lung cancers expressing TTK, URLC10 or KOC1 polypeptides
WO2006093030A1 (en) 2005-02-28 2006-09-08 Oncotherapy Science, Inc. Epitope peptides derived from vascular endothelial growth factor receptor 1 and vaccines containing these peptides
RU2413735C2 (en) 2005-03-31 2011-03-10 Эдженсис, Инк. Antibodies and related molecules binding with proteins 161p2f10b
SI2298815T1 (en) 2005-07-25 2015-08-31 Emergent Product Development Seattle, Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
PL1910839T3 (en) 2005-07-27 2016-11-30 Colon cancer related gene tom34
EP1922083A2 (en) * 2005-08-10 2008-05-21 Oklahoma Medical Research Foundation Truncated memapsin 2 for use for treating alzheimer's disease
US8980246B2 (en) 2005-09-07 2015-03-17 Sillajen Biotherapeutics, Inc. Oncolytic vaccinia virus cancer therapy
CA2621982C (en) * 2005-09-07 2017-11-28 Jennerex Biotherapeutics Ulc Systemic treatment of metastatic and/or systemically-disseminated cancers using gm-csf-expressing poxviruses
US7919258B2 (en) * 2005-10-07 2011-04-05 Arizona Board Of Regents On Behalf Of The University Of Arizona Rapid tuberculosis detection method
EP1998800A2 (en) 2006-01-18 2008-12-10 University Of Chicago Compositions and methods related to staphylococcal bacterium proteins
EP2253957B1 (en) 2006-03-14 2013-05-15 Oregon Health and Science University Methods for producing an immune response to tuberculosis.
GB0605474D0 (en) * 2006-03-17 2006-04-26 Isis Innovation Clinical correlates
CA3149553C (en) 2006-06-12 2023-11-21 Aptevo Research And Development Llc Single-chain multivalent binding proteins with effector function
ATE543097T1 (en) 2006-07-27 2012-02-15 Univ Maryland CELLULAR RECEPTOR FOR ANTIPROLIFERATIVE FACTOR
US8481023B2 (en) 2006-09-15 2013-07-09 Ottawa Hospital Research Institute Oncolytic rhabdovirus
SG10201502098YA (en) 2006-10-17 2015-05-28 Oncotherapy Science Inc Peptide Vaccines For Cancers Expressing MPHOSPH1 OR DEPDC1 Polypeptides
MX2009004221A (en) * 2006-10-20 2009-09-10 Univ Arizona State Modified cyanobacteria.
ATE555128T1 (en) 2006-11-30 2012-05-15 Res Dev Foundation IMPROVED IMMUNOLOBULIN LIBRARIES
TWI596109B (en) 2007-02-21 2017-08-21 腫瘤療法 科學股份有限公司 Peptide vaccines for cancers expressing tumor-associated antigens
KR20080084528A (en) 2007-03-15 2008-09-19 제네렉스 바이오테라퓨틱스 인크. Oncolytic vaccinia virus cancer therapy
TW201425333A (en) 2007-04-11 2014-07-01 Oncotherapy Science Inc TEM8 peptides and vaccines comprising the same
AU2008247819B2 (en) 2007-05-01 2013-02-14 Research Development Foundation Immunoglobulin Fc libraries
CN101990439A (en) * 2007-07-06 2011-03-23 特鲁比昂药品公司 Binding peptides having a c-terminally disposed specific binding domain
EP2666784B1 (en) 2007-08-31 2017-04-05 University Of Chicago Methods and compositions related to immunizing against staphylococcal lung diseases and conditions
EP3085707B1 (en) 2007-11-01 2019-02-27 Mayo Foundation for Medical Education and Research Hla-dr binding peptides and their uses
BRPI0906429B1 (en) 2008-01-10 2021-08-03 Research Development Foundation METHOD OF IDENTIFYING AN E. CHAFFEENSIS INFECTION IN AN INDIVIDUAL, USE OF ONE OR MORE SYNTHETIC POLYPEPTIDE AND KIT
KR20100128281A (en) * 2008-01-23 2010-12-07 릭스하스피탈렛 Classification of individuals suffering from cardiovascular diseases according to survival prognoses as found by measuring the levels of biomarker ykl-40
MX2010008168A (en) 2008-01-25 2011-02-24 P53 Inc P53 biomarkers.
CA2718904C (en) 2008-03-17 2017-01-03 The Scripps Research Institute Combined chemical and genetic approaches for generation of induced pluripotent stem cells
WO2009117134A2 (en) * 2008-03-21 2009-09-24 National Institutes Of Health Aerosolized genetic vaccines and methods of use
MX340204B (en) * 2008-04-11 2016-06-30 Emergent Product Dev Seattle Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof.
EP2853269B1 (en) 2008-05-19 2019-05-01 Advaxis, Inc. Dual delivery system for heterologous antigens comprising a recombinant Listeria strain attenuated by mutation of dal/dat and deletion of ActA comprising a nucleic acid molecule encoding an listeriolysin O - prostate specific anigen fusion protein
US9017660B2 (en) 2009-11-11 2015-04-28 Advaxis, Inc. Compositions and methods for prevention of escape mutation in the treatment of Her2/neu over-expressing tumors
US9650639B2 (en) 2008-05-19 2017-05-16 Advaxis, Inc. Dual delivery system for heterologous antigens
EP3279314A1 (en) 2008-06-04 2018-02-07 Cellular Dynamics International, Inc. Methods for the production of ips cells using non-viral approach
MY184182A (en) * 2008-07-25 2021-03-24 Dept Of Biotechnology India Constructing a dna chimera for vaccine development against leishmaniasis and tuberculosis
WO2010019569A1 (en) 2008-08-12 2010-02-18 Cellular Dynamics International. Inc. Methods for the production of ips cells
US8912146B2 (en) 2008-08-18 2014-12-16 University Of Maryland, Baltimore Derivatives of APF and methods of use
EP2329044B1 (en) 2008-08-27 2016-05-18 Oncotherapy Science, Inc. Prmt1 for target genes of cancer therapy and diagnosis
EP2341929B1 (en) 2008-10-06 2017-01-25 University Of Chicago Compositions and methods related to bacterial emp proteins
WO2010048252A1 (en) * 2008-10-23 2010-04-29 Intervet International B.V. Lawsonia intracellularis vaccines
TWI500932B (en) 2008-12-05 2015-09-21 Oncotherapy Science Inc Wdrpuh epitope peptides and vaccines containing the same
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
BRPI0922572A2 (en) 2008-12-17 2019-09-24 Scripps Research Inst method for culturing pluripotent cells, pluripotent mammalian cell culture, cell culture medium, isolated pluripotent animal cell, and method for increasing pluripotence of a mammalian cell.
EP2379720B1 (en) 2009-01-20 2016-08-17 Alona Zilberberg Mir-21 promoter driven targeted cancer therapy
TWI469791B (en) 2009-02-18 2015-01-21 Oncotherapy Science Inc Foxm1 peptides and vaccines containing the same
CN102356155B (en) 2009-03-18 2016-02-24 肿瘤疗法科学股份有限公司 NEIL3 peptide and comprise its vaccine
SI3281947T1 (en) 2009-04-03 2020-07-31 The University Of Chicago Compositions and methods related to protein a (spa) variants
US8748381B2 (en) 2009-04-28 2014-06-10 Vanderbilt University Compositions and methods for the treatment of disorders involving epithelial cell apoptosis
TWI507204B (en) 2009-05-26 2015-11-11 Oncotherapy Science Inc Cdc45l peptides and vaccines including the same
KR101813464B1 (en) 2009-06-05 2018-01-30 셀룰러 다이내믹스 인터내셔널, 인코포레이티드 Reprogramming T cells and hematopoietic cells
US20110053803A1 (en) 2009-08-26 2011-03-03 Xin Ge Methods for creating antibody libraries
KR101838472B1 (en) 2009-09-14 2018-03-15 신라젠(주) Oncolytic vaccinia virus combination cancer therapy
WO2011038290A2 (en) 2009-09-25 2011-03-31 The U. S. A., As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to hiv-1 and their use
CA2775720A1 (en) * 2009-09-30 2011-04-07 Saint Louis University Peptides for inducing heterosubtypic influenza t cell responses
MX337982B (en) 2009-10-16 2016-03-30 Scripps Research Inst Induction of pluripotent cells.
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
MX365946B (en) 2009-12-10 2019-06-19 Turnstone Lp Oncolytic rhabdovirus.
TW201136604A (en) 2009-12-14 2011-11-01 Oncotherapy Science Inc TMEM22 peptides and vaccines including the same
WO2011100508A2 (en) 2010-02-12 2011-08-18 Arizona Board Of Regents For And On Behalf Of Arizona State University Methods and compositions related to glycoprotein-immunoglobulin fusions
EP2538943B1 (en) 2010-02-25 2016-03-30 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
US20130052221A1 (en) 2010-02-26 2013-02-28 The Govt. of the U.S, as represented by The Sec. of The Dept. of Health and Human Services Dna-protein vaccination protocols
WO2011108930A1 (en) 2010-03-04 2011-09-09 Interna Technologies Bv A MiRNA MOLECULE DEFINED BY ITS SOURCE AND ITS DIAGNOSTIC AND THERAPEUTIC USES IN DISEASES OR CONDITIONS ASSOCIATED WITH EMT
BR112012022641A2 (en) 2010-03-11 2017-02-14 Oncotherapy Science Inc hjurp peptides and vaccines that include the same
EP3199623B1 (en) 2010-03-31 2021-07-28 The Scripps Research Institute Reprogramming cells
TWI538685B (en) 2010-04-02 2016-06-21 腫瘤療法 科學股份有限公司 Ect2 peptides and vaccines including the same
JP2013523818A (en) 2010-04-05 2013-06-17 ザ・ユニバーシティー・オブ・シカゴ Compositions and methods relating to protein A (SpA) antibodies as enhancers of immune responses
WO2011126976A1 (en) 2010-04-07 2011-10-13 Vanderbilt University Reovirus vaccines and methods of use therefor
CA2796601C (en) 2010-04-19 2019-03-26 Research Development Foundation Rtef-1 variants and uses thereof
WO2011156588A1 (en) 2010-06-09 2011-12-15 Dana-Farber Cancer Institute, Inc. A mek 1 mutation conferring resistance to raf and mek inhibitors
EP2580320B1 (en) 2010-06-14 2018-08-01 The Scripps Research Institute Reprogramming of cells to a new fate
ES2670842T3 (en) 2010-06-15 2018-06-01 Cellular Dynamics International, Inc. Generation of induced pluripotent stem cells from small volumes of peripheral blood
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
JP6002128B2 (en) 2010-07-02 2016-10-05 ザ・ユニバーシティ・オブ・シカゴThe University Of Chicago Compositions and methods related to protein A (SpA) variants
NZ719520A (en) 2010-07-06 2017-07-28 Int Tech Bv Mirna and its diagnostic and therapeutic uses in diseases or conditions associated with melanoma, or in diseases or conditions associated with activated braf pathway
CA2804595C (en) 2010-07-07 2018-11-13 Cellular Dynamics International, Inc. Endothelial cell production by programming
CA2806858C (en) 2010-08-04 2021-06-15 Cellular Dynamics International, Inc. Reprogramming immortalized b cells
US9095540B2 (en) 2010-09-09 2015-08-04 The University Of Chicago Methods and compositions involving protective staphylococcal antigens
US9084746B2 (en) 2010-09-22 2015-07-21 The Regents Of The University Of Colorado, A Body Corporate Therapeutic applications of SMAD7
WO2012138377A2 (en) 2010-10-01 2012-10-11 Trustees Of The University Of Pennsylvania The use of listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
AU2011308567B2 (en) 2010-10-01 2015-09-03 Fundacion Centro Nacional De Investigaciones Oncologicas, Carlos Iii Manipulation of stem cell function by p53 isoforms
EP2655601A4 (en) 2010-12-22 2014-09-10 Fate Therapeutics Inc Cell culture platform for single cell sorting and enhanced reprogramming of ipscs
JP6121910B2 (en) 2011-01-04 2017-04-26 シラジェン バイオセラピューティクス インコーポレイテッド Generation of antibodies against tumor antigens and tumor-specific complement-dependent cytotoxicity by administration of oncolytic vaccinia virus
EP2474617A1 (en) 2011-01-11 2012-07-11 InteRNA Technologies BV Mir for treating neo-angiogenesis
RU2539035C2 (en) * 2011-01-13 2015-01-10 Амир Закиевич Максютов Preventive or therapeutic polyepitopic anti-tuberculosis vaccine construction providing induction of cellular immune response of cd4+ or cd8+ t-lymphocytes
EP2673299B1 (en) 2011-02-07 2017-05-10 Research Development Foundation Engineered immunoglobulin fc polypeptides
US9574179B2 (en) 2011-02-08 2017-02-21 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
AU2012229218B2 (en) 2011-03-11 2017-03-02 Advaxis, Inc. Listeria-based adjuvants
US9085631B2 (en) 2011-04-08 2015-07-21 Nov Vac APS Proteins and nucleic acids useful in vaccines targeting Staphylococcus aureus
US8945588B2 (en) 2011-05-06 2015-02-03 The University Of Chicago Methods and compositions involving protective staphylococcal antigens, such as EBH polypeptides
ES2931180T3 (en) 2011-05-19 2022-12-27 Fund Publica Andaluza Progreso Y Salud Highly inducible dual promoter lentiviral Tet-on type system
US9364532B2 (en) 2011-06-08 2016-06-14 Children's Hospital Of Eastern Ontario Research Institute Inc. Compositions and methods for glioblastoma treatment
US20130040302A1 (en) 2011-07-11 2013-02-14 Thomas J. Burke Methods for cell reprogramming and genome engineering
EP2738255B1 (en) 2011-07-29 2016-11-23 Tokushima University Erap1-derived peptide and use thereof
WO2013024582A1 (en) 2011-08-12 2013-02-21 Oncotherapy Science, Inc. Mphosph1 peptides and vaccines including the same
JP6317670B2 (en) 2011-08-15 2018-04-25 ザ・ユニバーシティ・オブ・シカゴThe University Of Chicago Compositions and methods related to antibodies to staphylococcal protein A
WO2013026015A1 (en) 2011-08-18 2013-02-21 Dana-Farber Cancer Institute, Inc. Muc1 ligand traps for use in treating cancers
JP2013046596A (en) * 2011-08-29 2013-03-07 Alpha-Nano-Medica Co Ltd New complex, medicine including the same and method of treatment for cancer
EP2766388A1 (en) 2011-10-12 2014-08-20 Møller, Niels Iversen Peptides derived from campylobacter jejuni and their use in vaccination
CA3122778A1 (en) 2011-10-28 2013-05-02 Oncotherapy Science, Inc. Topk peptides and vaccines including the same
EP3369818B1 (en) 2011-12-22 2021-06-09 InteRNA Technologies B.V. Mirna for treating head and neck cancer
SG10201700392UA (en) 2012-03-12 2017-03-30 Advaxis Inc Suppressor cell function inhibition following listeria vaccine treatment
BR112014026861A2 (en) 2012-04-26 2018-05-15 Univ Chicago coagulase staphylococcal antigens and methods of use
CN104703622B (en) 2012-04-26 2017-05-24 芝加哥大学 Compositions and methods related to antibodies that neutralize coagulase activity during staphylococcus aureus disease
WO2014010232A1 (en) 2012-07-10 2014-01-16 Oncotherapy Science, Inc. Ly6k epitope peptides for th1 cells and vaccines containing the same
WO2014010231A1 (en) 2012-07-10 2014-01-16 Oncotherapy Science, Inc. Kif20a epitope peptides for th1 cells and vaccines containing the same
JP6283861B2 (en) 2012-09-11 2018-02-28 オンコセラピー・サイエンス株式会社 UBE2T peptide and vaccine containing the same
WO2014065945A1 (en) 2012-10-23 2014-05-01 The Board Of Regents Of The University Of Texas System Antibodies with engineered igg fc domains
EP2917348A1 (en) 2012-11-06 2015-09-16 InteRNA Technologies B.V. Combination for use in treating diseases or conditions associated with melanoma, or treating diseases or conditions associated with activated b-raf pathway
WO2014116721A1 (en) 2013-01-22 2014-07-31 The Arizona Board Of Regents For And On Behalf Of Arizona State University Geminiviral vector for expression of rituximab
CN105121631B (en) 2013-02-20 2019-04-19 瑞泽恩制药公司 The genetic modification of rat
RU2684211C2 (en) 2013-02-21 2019-04-04 Тёрнстоун Лимитед Партнершип Vaccine composition
US20140242595A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US10086093B2 (en) 2013-02-28 2018-10-02 The General Hospital Corporation miRNA profiling compositions and methods of use
EP2964774B1 (en) 2013-03-08 2020-05-06 The Regents of The University of Colorado, A Body Corporate Ptd-smad7 therapeutics
TWI658049B (en) 2013-03-12 2019-05-01 腫瘤療法 科學股份有限公司 Kntc2 peptides and vaccines containing the same
EP2968506B1 (en) * 2013-03-15 2019-07-31 Université de Genève Anti-mycobacterial vaccines
WO2014153636A1 (en) 2013-03-27 2014-10-02 Immunovaccine Technologies Inc. Method for improving the efficacy of a survivin vaccine in the treatment of cancer
RU2520078C1 (en) * 2013-04-25 2014-06-20 Федеральное государственное бюджетное учреждение "Научно-исследовательский институт эпидемиологии и микробиологии имени почетного академика Н.Ф. Гамалеи" Министерства здравоохранения Российской Федерации (ФГБУ "НИИЭМ им. Н.Ф. Гамалеи" Минздрава России) METHOD FOR OBTAINING IMMUNOGENIC COMPOSITION BASED ON Ag85A-DBD HYBRID PROTEIN AND DEXTRANE; pAg85A-DBD RECOMBINANT PLASMIDE; Escherichia coli [pREP4, pAg85A-DBD] STRAIN; Ag85A-DBD CHIMERIC PROTEIN
US11819555B2 (en) 2013-09-09 2023-11-21 Figene, Llc Gene therapy for the regeneration of chondrocytes or cartilage type cells
US10028902B2 (en) 2013-11-08 2018-07-24 Baylor Research Institute Nuclear localization of GLP-1 stimulates myocardial regeneration and reverses heart failure
WO2015070009A2 (en) 2013-11-08 2015-05-14 The Board Of Regents Of The University Of Texas System Vh4 antibodies against gray matter neuron and astrocyte
EP4227685A3 (en) 2013-12-03 2024-02-28 Evaxion Biotech A/S Proteins and nucleic acids useful in vaccines targeting staphylococcus aureus
MX2016009954A (en) 2014-01-29 2017-02-23 Dana Farber Cancer Inst Inc Antibodies against the muc1-c/extracellular domain (muc1-c/ecd).
EP3105332A4 (en) 2014-02-14 2018-01-10 University of Utah Research Foundation Methods and compositions for inhibiting retinopathy of prematurity
US20170037091A1 (en) 2014-02-25 2017-02-09 Research Development Foundation Sty peptides for inhibition of angiogenesis
KR102340553B1 (en) 2014-03-04 2021-12-21 페이트 세러퓨틱스, 인코포레이티드 Improved reprogramming methods and cell culture platforms
US20170107486A1 (en) 2014-04-21 2017-04-20 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
MX2017001650A (en) 2014-08-04 2017-04-27 Oncotherapy Science Inc Cdca1-derived peptide and vaccine containing same.
EP3590954A3 (en) 2014-08-04 2020-03-25 OncoTherapy Science, Inc. Koc1-derived peptide and vaccine including same
KR20220165831A (en) 2014-08-04 2022-12-15 온코세라피 사이언스 가부시키가이샤 Urlc10-derived peptide and vaccine containing same
WO2016025510A1 (en) 2014-08-12 2016-02-18 Rappolee Daniel A Systems and methods to detect stem cell stress and uses thereof
US20180169211A1 (en) 2014-11-13 2018-06-21 Evaxion Biotech Aps Peptides derived from acinetobacter baumannii and their use in vaccination
CA2972635A1 (en) 2015-01-06 2016-07-14 Immunovaccine Technologies Inc. Lipid a mimics, methods of preparation, and uses thereof
US10434162B2 (en) 2015-01-12 2019-10-08 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting Klebsiella pneumoniae
CN107438665A (en) 2015-01-28 2017-12-05 沙特基础全球技术有限公司 For efficiently producing the method and composition of bio-fuel and/or biomass
AU2016219511B2 (en) 2015-02-09 2020-11-12 Research Development Foundation Engineered immunoglobulin Fc polypeptides displaying improved complement activation
US11421229B2 (en) 2015-02-20 2022-08-23 Baylor College Of Medicine p63 inactivation for the treatment of heart failure
WO2016183420A1 (en) 2015-05-13 2016-11-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods and compositions for inducing an immune response using conserved element constructs
WO2016196366A1 (en) 2015-05-29 2016-12-08 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Extension of replicative lifespan in diseases of premature aging using p53 isoforms
WO2017005670A1 (en) 2015-07-04 2017-01-12 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting pseudomonas aeruginosa
US10526408B2 (en) 2015-08-28 2020-01-07 Research Development Foundation Engineered antibody FC variants
WO2017053469A2 (en) 2015-09-21 2017-03-30 Aptevo Research And Development Llc Cd3 binding polypeptides
EP3360886B1 (en) 2015-10-08 2023-01-25 Oncotherapy Science, Inc. Foxm1-derived peptide, and vaccine including same
EP3362570A4 (en) 2015-10-16 2019-03-20 Fate Therapeutics, Inc. Platform for the induction & maintenance of ground state pluripotency
US10865381B2 (en) 2015-10-20 2020-12-15 FUJIFILM Cellular Dynamics, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
CA3003145A1 (en) 2015-10-30 2017-05-04 Gay M. Crooks Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
SI3370733T1 (en) 2015-11-02 2021-11-30 Board Of Regents The University Of Texas System Methods of cd40 activation and immune checkpoint blockade
CN108884159A (en) 2015-11-07 2018-11-23 茂体外尔公司 The composition use for cancer treatment blocked comprising tumor suppressor gene treatment and immunologic test point
AU2016354440B2 (en) 2015-11-09 2023-09-28 The Children's Hospital Of Philadelphia Glypican 2 as a cancer marker and therapeutic target
US10946084B2 (en) 2016-02-22 2021-03-16 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting Staphylococcus aureus
US20190177391A1 (en) 2016-03-31 2019-06-13 Baylor Research Institute Angiopoietin-like protein 8 (angptl8)
WO2017216384A1 (en) 2016-06-17 2017-12-21 Evaxion Biotech Aps Vaccination targeting ichthyophthirius multifiliis
US11091775B2 (en) * 2016-06-22 2021-08-17 Oregon Health And Science University Recombinant cytomegalovirus vectors as vaccines for tuberculosis
WO2017220787A1 (en) 2016-06-24 2017-12-28 Evaxion Biotech Aps Vaccines against aearomonas salmonicida infection
JP7099967B2 (en) 2016-07-01 2022-07-12 リサーチ ディベロップメント ファウンデーション Elimination of Proliferative Cells from Stem Cell-Derived Grafts
EP3889167A1 (en) 2016-07-22 2021-10-06 Evaxion Biotech ApS Chimeric proteins for inducing immunity towards infection with s. aureus
WO2018035429A1 (en) 2016-08-18 2018-02-22 Wisconsin Alumni Research Foundation Peptides that inhibit syndecan-1 activation of vla-4 and igf-1r
CA3038690A1 (en) 2016-10-05 2018-04-12 FUJIFILM Cellular Dynamics, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
EP3523423A1 (en) 2016-10-05 2019-08-14 FUJIFILM Cellular Dynamics, Inc. Methods for directed differentiation of pluripotent stem cells to hla homozygous immune cells
WO2018111902A1 (en) 2016-12-12 2018-06-21 Multivir Inc. Methods and compositions comprising viral gene therapy and an immune checkpoint inhibitor for treatment and prevention of cancer and infectious diseases
US11718648B2 (en) 2017-01-05 2023-08-08 Evaxion Biotech A/S Vaccines targeting Pseudomonas aeruginosa
DK3612557T3 (en) 2017-04-18 2022-04-19 Fujifilm Cellular Dynamics Inc ANTIGEN-SPECIFIC IMMUNE EFFECTOR CELLS
US11497762B2 (en) 2017-11-03 2022-11-15 Interna Technologies B.V. MiRNA molecule, equivalent, antagomir, or source thereof for treating and/or diagnosing a condition and/or a disease associated with neuronal deficiency or for neuronal (re)generation
US11612618B2 (en) 2017-11-14 2023-03-28 Henry Ford Health System Compositions for use in the treatment and prevention of cardiovascular disorders resulting from cerebrovascular injury
MX2020007011A (en) 2018-01-05 2020-12-03 Ottawa Hospital Res Inst Modified vaccinia vectors.
WO2019145399A1 (en) 2018-01-24 2019-08-01 Evaxion Biotech Aps Vaccines for prophylaxis of s. aureus infections
WO2020036635A2 (en) 2018-03-19 2020-02-20 Multivir Inc. Methods and compositions comprising tumor suppressor gene therapy and cd122/cd132 agonists for the treatment of cancer
WO2019186274A2 (en) 2018-03-30 2019-10-03 University Of Geneva Micro rna expression constructs and uses thereof
TW202023581A (en) 2018-08-02 2020-07-01 日商腫瘤療法 科學股份有限公司 Cdca1-derived peptide and vaccine containing same
CN113164556A (en) 2018-08-30 2021-07-23 特纳亚治疗股份有限公司 Cardiac cell reprogramming with cardiac myoprotein and ASCL1
US20220000932A1 (en) 2018-09-28 2022-01-06 Henry Ford Health System Use of extracellular vesicles in combination with tissue plasminogen activator and/or thrombectomy to treat stroke
US20220111031A1 (en) 2018-10-22 2022-04-14 Evaxion Biotech Aps Vaccines targeting M. catharrhalis
EP3889264A4 (en) 2018-11-30 2022-10-12 Tokushima University Therapeutic agent for breast caner comprising big3-phb2 interaction-inhibiting peptide derived from phb2
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
WO2020174044A1 (en) 2019-02-27 2020-09-03 Evaxion Biotech Aps Vaccines targeting h. influenzae
EP3997226A1 (en) 2019-07-11 2022-05-18 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
AU2020317009A1 (en) 2019-07-19 2022-02-03 The Board Of Trustees Of The Leland Stanford Junior University Chimeric antigen receptors containing Glypican 2 binding domains
US20210147525A1 (en) 2019-10-18 2021-05-20 The Regents Of The University Of California Methods and compositions for treating pathogenic blood vessel disorders
KR102237349B1 (en) 2019-10-23 2021-04-07 한국과학기술연구원 Pharmaceutical composition for the treatment or prevention of nicotine addiction and withdrawal comprising miRNA
WO2021113644A1 (en) 2019-12-05 2021-06-10 Multivir Inc. Combinations comprising a cd8+ t cell enhancer, an immune checkpoint inhibitor and radiotherapy for targeted and abscopal effects for the treatment of cancer
WO2021140123A1 (en) 2020-01-06 2021-07-15 Evaxion Biotech Aps Vaccines targeting neisseria gonorrhoeae
CN115552022A (en) 2020-03-02 2022-12-30 特纳亚治疗股份有限公司 MicroRNA-controlled gene vector expressed by cardiac muscle cells
JP2023520359A (en) 2020-03-25 2023-05-17 エラスムス・ユニヴァーシティ・メディカル・センター・ロッテルダム Reporter system for radionuclide imaging
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
IL298254A (en) 2020-05-29 2023-01-01 Fujifilm Cellular Dynamics Inc Bilayer of retinal pigmented epithelium and photoreceptors and use thereof
CN116033912A (en) 2020-05-29 2023-04-28 富士胶片细胞动力公司 Retinal pigment epithelium and photoreceptor double cell aggregates and methods of use thereof
WO2022053130A1 (en) 2020-09-09 2022-03-17 Sid Alex Group, S.R.O. Antago-mir-155 for treatment of v-src, c-src-tyrosine kinase-induced cancers
CA3202849A1 (en) 2021-02-09 2022-08-18 University Of Houston System Oncolytic virus for systemic delivery and enhanced anti-tumor activities
WO2022175815A1 (en) 2021-02-19 2022-08-25 Pfizer Inc. Methods of protecting rna
CN117242173A (en) 2021-05-03 2023-12-15 安斯泰来再生医药协会 Method for producing mature corneal endothelial cells
TW202309268A (en) 2021-05-07 2023-03-01 安斯泰來再生醫藥協會 Methods of generating mature hepatocytes
AU2022280051A1 (en) 2021-05-26 2023-11-23 FUJIFILM Cellular Dynamics, Inc. Methods to prevent rapid silencing of genes in pluripotent stem cells
CA3224564A1 (en) 2021-07-05 2023-01-12 Andreas Holm MATTSSON Vaccines targeting neisseria gonorrhoeae
WO2023089556A1 (en) 2021-11-22 2023-05-25 Pfizer Inc. Reducing risk of antigen mimicry in immunogenic medicaments
WO2023144779A1 (en) 2022-01-28 2023-08-03 Pfizer Inc. Coronavirus antigen variants
US20230295661A1 (en) 2022-03-16 2023-09-21 University Of Houston System Persistent hsv gene delivery system
GB202205265D0 (en) 2022-04-11 2022-05-25 Mogrify Ltd Cell conversion
WO2023213393A1 (en) 2022-05-04 2023-11-09 Evaxion Biotech A/S Staphylococcal protein variants and truncates
GB202206507D0 (en) 2022-05-04 2022-06-15 Antion Biosciences Sa Expression construct
WO2023239940A1 (en) 2022-06-10 2023-12-14 Research Development Foundation Engineered fcriib selective igg1 fc variants and uses thereof
WO2024006911A1 (en) 2022-06-29 2024-01-04 FUJIFILM Holdings America Corporation Ipsc-derived astrocytes and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994021797A1 (en) * 1993-03-18 1994-09-29 Merck & Co., Inc. Nucleic acid pharmaceuticals

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5591632A (en) * 1987-03-02 1997-01-07 Beth Israel Hospital Recombinant BCG
US5807830A (en) * 1987-12-30 1998-09-15 Cytoven J.V. Method for treatment of purulent inflammatory diseases
DE69032284T2 (en) * 1989-03-21 1998-10-08 Vical Inc EXPRESSION OF EXOGENOUS POLYNUCLEOTIDE SEQUENCES IN VERTEBLE
IL95742A (en) * 1989-09-19 2000-12-06 Innogenetics Nv Recombinant polypeptides and peptides nucleic acids coding for the same and use of these polypeptides and peptides in the diagnosis of tuberculosis
EP0499003A1 (en) * 1991-02-14 1992-08-19 N.V. Innogenetics S.A. Polypeptides and peptides, particularly recombinant polypeptides and peptides, nucleic acids coding for the same and use of these polypeptides and peptides in the diagnosis of tuberculosis
US5643578A (en) * 1992-03-23 1997-07-01 University Of Massachusetts Medical Center Immunization by inoculation of DNA transcription unit
US5955077A (en) * 1993-07-02 1999-09-21 Statens Seruminstitut Tuberculosis vaccine
IL113817A (en) * 1994-06-30 2001-03-19 Merck & Co Inc Polynucleotide vaccne for papillomavirus
US5736524A (en) * 1994-11-14 1998-04-07 Merck & Co.,. Inc. Polynucleotide tuberculosis vaccine
WO1997000067A1 (en) * 1995-06-15 1997-01-03 University Of Victoria Innovation And Development Mycobacterium tuberculosis dna sequences encoding immunostimulatory peptides
US6160093A (en) * 1996-08-29 2000-12-12 Genesis Researth And Development Corporation Limited Compounds and methods for treatment and diagnosis of mycobacterial infections

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994021797A1 (en) * 1993-03-18 1994-09-29 Merck & Co., Inc. Nucleic acid pharmaceuticals

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
9th International Congress of Immunology, July 23-29, 1995, San Francisco, California, abstract 4843 XP002012393 *
AIDS RESEARCH AND HUMAN RETROVIRUS, vol. 10, no. Suppl. 2, 1994, pages S43-S45, XP002012391 J.R. HAYNES ET AL.: "Accell Particle-mediated DNA immunization elicits humoral, cytotoxic and protective immune responses" *
INFECTION AND IMMUNITY, vol. 57, no. 10, 1989, pages 3123-3130, XP002012388 M. BORREMANS ET AL.: "Cloning, sequence determination nand expression of a 32 kDa-protein gene of Mycobacterium tuberculosis" *
INFECTION AND IMMUNITY, vol. 59, no. 9, 1991, pages 3205-3212, XP002012386 J. CONTENT ET AL.: "The genes coding for the antigen 85 complexes of Mycobacterium tuberculosis and Mycobnacterium bovis BCG are members of a gene family: cloning, sequence determination and genomic organization of the gene coding for antigen 85-C of mycobacterium tuberculosis" *
INTERNATIONAL JOURNAL OF LEPROSY, vol. 61, no. 4, 1993, page 108A XP002012385 LEILA DE MENDONCA LIMA: "Cloning and characterisation of the genes coding for the 85-complex antigens of mycobacterium leprae" *
NUCLEIC ACIDS RESEARCH, vol. 18, no. 13, 1990, page 3995 XP002012387 LUK DE WIT ET AL.: "Nucleotide sequence of the 32 hDa-protein gene (antigen 85 A) of mycobacterium bovis BCG" *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES USA., vol. 90, 1993, pages 11478-11482, XP002012390 E.F. FYNAN ET AL.: "DNA vaccines: protective immunizations by parenteral, mucosal and gene-gun inoculations" *
TROPICAL MEDICINE AND PARASITOLOGY, vol. 41, 1990, pages 331-332, XP002012389 J. DE BRUYN ET AL.: "The 32 kDa protein antigen of M. bovis BCG and M. tuberculosis H37Rv*see the whole article*" *
VACCINES , vol. 94, 1994, pages 65-70, XP002012392 J.R HAYNES ET AL: "Gene-gun-mediated DNA immunization elicits humoral, cytotoxic and protective immune responses" *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6384018B1 (en) * 1994-11-14 2002-05-07 Merck & Co., Inc. Polynucleotide tuberculosis vaccine
US6949246B2 (en) 1995-09-01 2005-09-27 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US7122196B2 (en) 1995-09-01 2006-10-17 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6290969B1 (en) 1995-09-01 2001-09-18 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US6338852B1 (en) 1995-09-01 2002-01-15 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6458366B1 (en) 1995-09-01 2002-10-01 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6962710B2 (en) 1995-09-01 2005-11-08 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US6592877B1 (en) 1995-09-01 2003-07-15 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US7238358B2 (en) 1995-09-01 2007-07-03 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
JP2000515522A (en) * 1996-07-19 2000-11-21 メリアル Transdermal bovine polynucleotide vaccine formulation
JP2009149608A (en) * 1996-07-19 2009-07-09 Merial Sas Polynucleotide vaccine preparation for bird
WO1998016646A3 (en) * 1996-10-11 1998-10-08 Corixa Corp Compounds and methods for immunotherapy and diagnosis of tuberculosis
WO1998016646A2 (en) * 1996-10-11 1998-04-23 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
WO1998035029A1 (en) * 1997-02-07 1998-08-13 Vanderbilt University Synthetic genes for recombinant mycobacterium proteins
US6465633B1 (en) 1998-12-24 2002-10-15 Corixa Corporation Compositions and methods of their use in the treatment, prevention and diagnosis of tuberculosis
US7022320B1 (en) 1999-02-09 2006-04-04 Powderject Vaccines, Inc. Mycobacterium tuberculosis immunization
US8012467B2 (en) 2004-11-16 2011-09-06 Crucell Holland B.V. Multivalent vaccines comprising recombinant viral vectors
US8202723B2 (en) 2004-11-16 2012-06-19 Crucell Holland B.V. Multivalent vaccines comprising recombinant viral vectors
US8609402B2 (en) 2004-11-16 2013-12-17 Aeras Global Tb Vaccine Foundation Multivalent vaccines comprising recombinant viral vectors
EP1817061A2 (en) * 2004-12-01 2007-08-15 Aeras Global Tuberculosis Vaccine Foundation Electroporation of mycobacterium and overexpression of antigens in mycobacteria
EP1817061A4 (en) * 2004-12-01 2009-07-08 Aeras Global Tuberculosis Vacc Electroporation of mycobacterium and overexpression of antigens in mycobacteria

Also Published As

Publication number Publication date
JPH10508753A (en) 1998-09-02
DK0792358T3 (en) 2005-08-29
RU2186109C2 (en) 2002-07-27
WO1996015241A3 (en) 1996-11-07
US6384018B1 (en) 2002-05-07
IL115883A0 (en) 1996-01-31
CN1171814A (en) 1998-01-28
US5736524A (en) 1998-04-07
AU715067B2 (en) 2000-01-13
PT792358E (en) 2005-09-30
AU4110296A (en) 1996-06-06
US20020032162A1 (en) 2002-03-14
DE69534250T2 (en) 2006-05-04
NO972196D0 (en) 1997-05-13
EP0792358A2 (en) 1997-09-03
PL184839B1 (en) 2002-12-31
CZ289383B6 (en) 2002-01-16
DE69534250D1 (en) 2005-07-07
SK283254B6 (en) 2003-04-01
SK59797A3 (en) 1998-01-14
FI972034A0 (en) 1997-05-13
CZ145197A3 (en) 1997-10-15
FI972034A (en) 1997-07-11
KR970707281A (en) 1997-12-01
HU222369B1 (en) 2003-06-28
NZ296477A (en) 1999-04-29
ES2242193T3 (en) 2005-11-01
HUT77028A (en) 1998-03-02
ZA959608B (en) 1996-05-29
PL320091A1 (en) 1997-09-15
MX9703606A (en) 1998-07-31
ATE296882T1 (en) 2005-06-15
EP0792358B1 (en) 2005-06-01
JP3881014B2 (en) 2007-02-14
NO972196L (en) 1997-07-11

Similar Documents

Publication Publication Date Title
EP0792358B1 (en) A polynucleotide tuberculosis vaccine
AU743616B2 (en) Synthetic HIV gag genes
US6696291B2 (en) Synthetic HIV gag genes
US20020136737A1 (en) Isolated nucleic acids comprising Listeria dal and dat genes
US20060018881A1 (en) Coordinate in vivo gene expression
US7122180B2 (en) DNA vectors containing mutated HIV proviruses
JP2007332149A (en) Vaccine against mycobacterial infection
CA2247091A1 (en) Recombinant live feline immunodeficiency virus and proviral dna vaccines
US7094767B2 (en) Polynucleotide herpes virus vaccine
AU708460B2 (en) A polynucleotide herpes virus vaccine
CA2205175C (en) A polynucleotide tuberculosis vaccine
CA2267645A1 (en) A polynucleotide herpes virus vaccine
AU738835B2 (en) A polynucleotide herpes virus vaccine
PT969862E (en) Synthetic hiv gag genes
MXPA99007248A (en) Synthetic hiv gag

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 95197250.2

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AU BB BG BR BY CA CN CZ EE FI GE HU IS JP KG KR KZ LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TT UA US UZ

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref document number: 296477

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2205175

Country of ref document: CA

Ref document number: 2205175

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1995939161

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 59797

Country of ref document: SK

Ref document number: PV1997-1451

Country of ref document: CZ

Ref document number: 1019970703204

Country of ref document: KR

Ref document number: 972034

Country of ref document: FI

WWP Wipo information: published in national office

Ref document number: 1995939161

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1997-1451

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1019970703204

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: PV1997-1451

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 1995939161

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1019970703204

Country of ref document: KR