WO2001093905A1 - Immunostimulatory oligodeoxynucleotides - Google Patents

Immunostimulatory oligodeoxynucleotides Download PDF

Info

Publication number
WO2001093905A1
WO2001093905A1 PCT/EP2001/006433 EP0106433W WO0193905A1 WO 2001093905 A1 WO2001093905 A1 WO 2001093905A1 EP 0106433 W EP0106433 W EP 0106433W WO 0193905 A1 WO0193905 A1 WO 0193905A1
Authority
WO
WIPO (PCT)
Prior art keywords
monothiophosphate
monophosphate
deoxyinosine
odn
deoxycytosine
Prior art date
Application number
PCT/EP2001/006433
Other languages
French (fr)
Inventor
Walter Schmidt
Karen Lingnau
Carola Schellack
Alena Egyed
Original Assignee
Intercell Biomedizinische Forschungs- Und Entwicklungs Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AT0100000A external-priority patent/AT410173B/en
Priority to DK01960277T priority Critical patent/DK1296713T3/en
Priority to SK1815-2002A priority patent/SK287689B6/en
Priority to IL15295901A priority patent/IL152959A0/en
Priority to EP20010960277 priority patent/EP1296713B1/en
Priority to BRPI0111639A priority patent/BRPI0111639B8/en
Priority to AU8181201A priority patent/AU8181201A/en
Priority to MXPA02012010A priority patent/MXPA02012010A/en
Priority to AT01960277T priority patent/ATE249839T1/en
Priority to CA2411575A priority patent/CA2411575C/en
Application filed by Intercell Biomedizinische Forschungs- Und Entwicklungs Ag filed Critical Intercell Biomedizinische Forschungs- Und Entwicklungs Ag
Priority to JP2002501476A priority patent/JP5271471B2/en
Priority to HU0301229A priority patent/HU228264B1/en
Priority to PL358982A priority patent/PL211036B1/en
Priority to AU2001281812A priority patent/AU2001281812B2/en
Priority to SI200130047T priority patent/SI1296713T1/en
Priority to DE2001600814 priority patent/DE60100814T2/en
Priority to US10/297,555 priority patent/US8568742B2/en
Publication of WO2001093905A1 publication Critical patent/WO2001093905A1/en
Priority to IS6627A priority patent/IS1993B/en
Priority to IL152959A priority patent/IL152959A/en
Priority to NO20025835A priority patent/NO329492B1/en
Priority to HK03109011A priority patent/HK1056678A1/en
Priority to US13/786,815 priority patent/US8945591B2/en
Priority to US14/585,740 priority patent/US9492537B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • 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
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/24Heterocyclic radicals containing oxygen or sulfur as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/18Type of nucleic acid acting by a non-sequence specific mechanism
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to immunostimulatory oligodeoxynucleic molecules (ODNs) and pharmaceutical compositions containing such ODNs .
  • ODNs immunostimulatory oligodeoxynucleic molecules
  • Vaccines can save more lives (and resources) than any other medical intervention (Nossal, 1998) . Owing to world-wide vaccination programs the incidence of many fatal diseases has been decreased drastically. Although this notion is valid for a whole panel of diseases, e.g. tuberculosis, diphtheria, pertussis, measles and tetanus, there are no effective vaccines for numerous infectious disease including most viral infections, such as AIDS. There are also no effective vaccines for other diseases, infectious or non-infectious claiming millions the lives of millions of patients per year including malaria or cancer. In addition, the rapid emergence of antibiotic-resistant bacteria and microorganisms calls for alternative treatments with vaccines being a logical choice. Finally, the great need for vaccines is also illustrated by the fact that infectious diseases, rather than cardiovascular disorders or cancer or injuries remain the largest cause of death and disability in the world (Bloom and Widdus, 1998) .
  • T cells which recognize molecules expressed on pathogen infected cells.
  • vaccines are designed that induce both T cells distinguishing diseased and/or infected cells from normal cells and, simultaneously, antibodies secreted by B cells recognising pathogens in extracellular compartments .
  • vaccines consist of live attenuated organism where the risk of reversion to the virulent wild-type strain exists. In particular in immunocompromised hosts this can be a live threatening scenario.
  • vaccines are administered as a combination of pathogen-derived antigens together with compounds that induce or enhance immune responses against these antigens (these compounds are commonly termed adjuvant) , since these subunit vaccines on their own are generally not effective.
  • Antigen presenting cells belong to the innate immune system, which has evolved as a first line host defence that limits infection early after exposure to microorganisms (Hoffmann et al . , 1999).
  • Cells of the innate immune sytem recognize patterns or relatively non-specific structures expressed on their targets rather than more sophisticated, specific structures which are recognised by the adaptive immune system (Hoffmann et al . , 1999).
  • Examples of cells of the innate immune system are macrophages and dendritic cells but also granulocytes (e.g. neutrophiles) , natural killer cells and others.
  • cells of the adaptive immune system recognize specific, antigenic structures, including peptides, in the case of T cells and peptides as well as three- dimensional structures in the case of B cells.
  • the adaptive immune system is much more specific and sophisticated than the innate immune system and improves upon repeat exposure to a given pathogen/antigen.
  • the innate immune system is much older and can be found already in very primitive organisms. Nevertheless, the innate immune system is critical during the initial phase of antigenic exposure since, in addition to containing pathogens, cells of the innate immune system, i.e. APCs, prime cells of the adaptive immune system and thus trigger specific immune responses leading to clearance of the intruders.
  • cells of the innate immune sytem and in particular APCs play a critical role during the induction phase of immune responses by a) containing infections by means of a primitive pattern recognition system and b) priming cells of the adaptive immune system leading to specific immune responses and memory resulting in clearance of intruding pathogens or of other targets (Roitt et al . , 1998). These mechanisms may also be important to clear or contain tumor cells . ,
  • cells of the innate immune system recognise patterns expressed on their respective targets .
  • examples are lipopolysaccharides ( PS) in the case of Gram-negative bacteria, mycobacterial glycolipids, lipoteichoic acids of Gram-positive bacteria, mannans of yeast and double stranded RNAs of viruses (Hoffmann et al . , 1999).
  • PS lipopolysaccharides
  • mycobacterial glycolipids lipoteichoic acids of Gram-positive bacteria
  • mannans of yeast double stranded RNAs of viruses
  • the immune system recognises lower organisms including bacteria probably due to structural and sequence usage differencies between pathogen and host DNA.
  • short stretches of DNA derived from non-vertebrates or in form of short synthetic ODNs containing nonmethylated cytosine-guanine dinucleotides (CpG) in a certain base context, are targeted (Krieg et al . , 1995) .
  • CpG motifs are found at the expected frequency in bacterial DNA but are much less frequent in vertebrate DNA (LipLord et al., 1998; Pisetsky, 1999).
  • non-vertebrate (i.e. bacterial) CpG motifs are not methylated whereas vertebrate CpG sequences are.
  • Natural CpG-containing DNA, ODNs, as well as thiophosphate-sub- stituted (exchange of thiophosphate residues for phosphate) ODNs containing CpG motifs are not only potent activators of immune cell proliferation and humoral immune responses (Krieg et al . , 1995), but also stimulate strong cellular immune responses (reviewed in Lipford et al . , 1998) .
  • DNA/ODNs containing non-methylated CpG motifs can directly activate monocytic cells (dendritic cells, macrophages) and B cells.
  • NK cells are not directly activated but respond to ono- cyte-derived IL-12 (interleukin 12) with a marked increase in their IFN- ⁇ production (Chace et al., 1997).
  • ono- cyte-derived IL-12 interleukin 12
  • CpG DNA promotes the induction of Thl-type responses and the development of cytotoxic T cells .
  • Ribonucleic acid based on inosine and cytosine like polyi- nosinic-polycytidylic acid (poly I:C), is known to promote Thl- specific immune responses. It is known to stimulate macrophages to produce cytokines such as IL-l ⁇ and IL-12 (Manetti et al . , 1995) , it is also known as a potent interferon type 1 inducer (Manetti et al . , 1995) and a potent NK cell stimulator (Cavanaugh et al. , 1996) .
  • ODNs containing non-methylated CpG motifs although being efficient in stimulating immune system, have essential disadvantages, especially with respect to specificity (high background) and induction of side effects, such as high systemic TNF- ⁇ generation.
  • High systemic TNF- ⁇ release is known to cause toxic shock syndrome, which can cause death of afflicted patients.
  • any X is 0 or S
  • NMP is a 2 ' deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2-amino-deoxyribosepu- rine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopen- • tenyl-deoxyadenosine-monophosphate or -monothiophosphat
  • NUC is a 2' deoxynucleoside, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythy
  • ODNs containing deoxyinosine residues show an immunostimulatory effect comparable or in many instances even better than ODNs containing CpG motifs.
  • ODNs according to the present invention produce more specific immune responses to a given antigen or antigen fragment than CpG ODNs.
  • ODNs according to the present invention reduced the induction of adverse side reactions, especially the induction of systemic TNF- ⁇ or IL-6.
  • the I-ODNs according to the present invention are - in contrast to ODNs based on the specific CpG motif - not dependent on a specific motif or a palindromic sequence as described for the CpG oligonucleotides (see e.g. EP 0 468 520 A2, WO96/02555, WO98/18810, W098/37919, WO98/40100, W098/52581, W099/51259 and W099/56755, all incoraliad herein by reference) .
  • one group of I-ODNs according to the present invention may preferably contain a CI motif (and therefore ODNs described in these incorporated references, wherein one or more guanosine residues are replaced with deoxyinosine residues are preferred embodiments of the present ODNs) . It is not necessary for its principle immunostimulatory property, since I-ODNs with an Inosine not placed in a CI or IC context exhibit immunostimulatory properties as well.
  • the I-ODN according to the present invention is therefore a DNA molecule containing a deoxyinosine residue which is preferably provided in single stranded form.
  • the I-ODN according to the present invention may be isolated through recombinant methods or chemically synthesized.
  • the I-ODN according to the present invention may also contain modified oligonucleotides which may be synthesized using standard chemical transformations, such as methylphosphon- ates or other phosphorous based modified oligonucleotides, such as phosphotriesters, phosphoamidates and phosphorodithiorates .
  • Other non-phosphorous based modified oligonucleotides can also be used (Stirchak et al . , MAR 17 (1989), 6129-6141), however, mono- phosphates or monothiophosphates being the preferred 2 'deoxynucleoside monophosphate to be used in the present invention.
  • the NMPs of the I-ODNs according to the present invention are preferably selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine- monophosphate or -monothiophosphate (as usual, the phosphate or thiophosphate group is 5' of the deoxyribose) .
  • this motif is unmethylated, this is surprisingly not the case for the ODNs according to the present invention, wherein e.g.
  • 2-methyl-deoxyinosine or 5-methyl-deoxycytosine residues have no general negative effect on immunostimulatory properties of the ODNs according to the present invention.
  • groups may be present at the 2-site of the ribose group, such as e.g. -F, -NH 2 , -CH , especially -CH .
  • -OH and SH groups are excluded for the I-ODNs according to the present invention to be present on the 2 '-site of the ribose, especially the ribose residue for the inosine NMP.
  • the length of the ODNs according to the present invention is in the range of the standard ODNs used according to the prior art. Therefore molecules with a total length under 4 and above 150 show gradually decreasing immunostimulatory potential.
  • Preferred ODNs contain between 10 and 60, especially between 15 and 40 bases (nucleosides) , implying that a + b in formula I is between 10 and 60, preferably between 15 and 40 in these preferred embodiments .
  • ribonucleic acid molecules containing inosine and cytidine described to be immunostimulatory in the prior art have been large and relatively undefined polynucleic acids with molecular weights far above 200,000 (a commercially available poly- inosinic-polycytidylic acid from Sigma Chemicals has a molecular weight ranging from 220,000 to 460,000 (at least 500-1000 C+I residues) .
  • the molecules according to the present invention are DNA molecules of much shorter length with a well defined length and composition, being highly reproducible in products.
  • the deoxyinosine containing NMP of the I-ODNs according to formula I is a monothiophosphate with one to four sulfur atoms and that also further NMPs, especially all further NMPs, are present as nucleoside monothiophosphates , because such ODNs display higher nuclease resistance (it is clear for the present invention that the "mono" in the "monothiophosphates" relates to the phosphate, i.e. that one phosphate group (one phosphor atom) is present in each NMP) .
  • at least one of X 1 and X2 is S and at least one of X3 and X4 is 0 in the
  • NMPs according to the present invention.
  • X and X are 0.
  • X may be (due to synthesis of the NMP) derived e.g. from the phosphate group or from the 3 '-group of the NMP-ribose) .
  • the ODNs according to the present invention contain the sequence hhh wdi dhh h nhh hhh wdi hhh hhh hhh wn, nhh wdi din hhh hdi ndi nh, nhh hhh wdi dhh hhh hhh wn or nhh wdi did hhh hdi ddi dh, wherein any n is a 2 '-deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxycytosine- or deoxythymidine-monophosphate or -monothiophosphate, any h is a 2 ' -deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxycytosine- or deoxythymidine-mono
  • a specific motif (such as CpG or a palindrome) is not necessary for the I-ODNs according to the present invention.
  • ODNs containing a CI motif are preferred so that in a preferred embodiment the ODN according to formula I contains at least one 2 'deoxycytosine-monophosphate or -monothiophosphate 3 '-adjacent to a 2 '-deoxyinosine-monophosphate or -monothiophosphate to form such a 5'-CI 3 '-motif.
  • Preferred ODNs according to the present invention contain one or more of the sequence
  • gacitt, iacitt, gaictt, iaictt wherein a is deoxyadenosine-monophosphate or -monothiophosphate, g is deoxyguanosine-monophosphate or -monothiophosphate, i is deoxyinosine-monophosphate or -monothiophosphate, c is deoxycytosine-monophosphate or -monothiophosphate and t is deoxythymidine-monophosphate or -monothiophosphate.
  • the I-ODNs according to the present invention are especially suitable for application in the pharmaceutical field, e.g. to be applied as a medicine to an animal or to humans . They are specifically adapted to act as an immunostimulatory agent, especially in or together with vaccine compositions.
  • the present invention also relates to a pharmaceutical composition comprising an ODN according to the present invention.
  • a preferred pharmaceutical composition according to the present invention is a vaccine
  • this composition should contain an antigen besides the ODN according to the present invention.
  • the potential of this antigen to raise a. protection/immune response of the vaccinated individual is strongly increased by combining it with the ODNs according to the present invention, especially due to their immunostimulatory activity.
  • a vaccine can contain a whole variety of different antigens. Examples of antigens are whole-killed organisms such as inactivated viruses or bacteria, fungi, protozoa or even cancer cells. Antigens may also consist of subfractions of these organisms/tissues, of proteins, or, in their most simple form, of peptides.
  • Antigens can also be recognised by the immune system in form of glycosy- lated proteins or peptides and may also be or contain polysaccha- rides or lipids.
  • Short peptides can be used since for example cytotoxic T cells (CTL) recognize antigens in form of short usually 8-11 amino acids long peptides in conjunction with major histocompatibility complex (MHC) (Rammensee et al . , Immunogenet- ics 41, (1995), 178-228).
  • CTL cytotoxic T cells
  • MHC major histocompatibility complex
  • B cells recognize longer peptides starting at around 15 amino acids (Harrow et al, Cold Spring Harbor: Cold Spring Harbor Laboratory, (1988)).
  • T cell epitopes the three dimensional structure of B cell antigens may also be important for recognition by antibodies .
  • adjuvants are helpful to trigger immune cascades that involve all cells of the immune system necessary.
  • adjuvants are acting, but are not restricted in their mode of action, on so-called antigen presenting cells (APCs) . These cells usually first encounter the antigen (s) followed by presentation of processed or unmodified antigen to immune effector. Intermediate cell types may also be involved. Only effector cells with the appropriate specificity are activated in a productive immune response.
  • the adjuvant may also locally retain antigens and co-injected other factors.
  • the adjuvant may act as a chemoattractant for other immune cells or may act locally and/or systemically as a stimulating agent for the immune system.
  • T cell epitopes are used as antigens .
  • a combination of T cell epitopes and B cell epitopes may also be preferred.
  • antigens to be used in the present compositions are not critical. Also mixtures of different antigens are of course possible to be used according to the present invention. Preferably, proteins or peptides derived from a viral or a bacterial pathogen or from fungi or parasites are used as such antigens (including derivatized antigens or glycosylated ' or lipidated antigens or polysaccharides or lipids) . Another preferred source of antigens are tumor antigens .
  • Preferred pathogens are selected from human immunodeficiency virus (HIV) , hepatitis A and B viruses, hepatitis C virus (HCV) , rous sarcoma virus (RSV) , Epstein Barr virus (EBV) Influenza virus, Rotavirus, Staphylococcus aureus, Chlamydia pneumonias, Chlamydia trachomatis, Mycobacterium tuberculosis, Streptococcus pneumonias, Bacillus anthracis, Vibrio cholerae, Plasmodium sp. (Pi. falciparum, Pi. vivax, etc.), As- pergillus sp . or Candida albicans .
  • HCV human immunodeficiency virus
  • HCV hepatitis A and B viruses
  • HCV hepatitis C virus
  • RSV rous sarcoma virus
  • EBV Epstein Barr virus
  • Influenza virus Influenza virus
  • Antigens may also be molecules expressed by cancer cells (tumor antigens) .
  • the derivation process may include the purification of a specific protein from the pathogen/cancer cells, the inactivation of the pathogen as well as the proteolytic or chemical derivatization or stabilisation of such a protein.
  • tumor antigens cancer vaccines
  • autoimmune antigens may be used in the pharmaceutical composition according to the present invention. With such compositions a tumor vaccination or a treatment for autoimmume diseases may be performed.
  • peptide antigens In the case of peptide antigens the use of peptide mimitopes/ago- nists/superagonists/antagonists or peptides changed in certain positions without affecting the immunologic properties or non- peptide mimitopes/agonists/superagonists/antagonists (reviewed in Sparbier and Walden, 1999) is included in the current invention.
  • Peptide antigens may also contain elongations either at the carboxy or at the amino terminus of the peptide antigen facilitating interaction with the polycationic compound(s) or the immunostimulatory compound(s) .
  • peptide antagonists may be applied for the treatment of autoimmune diseases.
  • Antigens may also be derivatized to include molecules enhancing antigen presentation and targeting of antigens to antigen presenting cells.
  • the pharmaceutical composition serves to confer tolerance to proteins or protein fragments and peptides which are involved in autoimmune diseases .
  • Antigens used in this embodiments serve to tolerize the immune system or down- regulate immune responses against epitopes involved in autoimmune processes .
  • the pharmaceutical composition according to the present invention especially in the form of a vaccine, further comprises a polycationic polymer, preferably a polycationic peptide, especially polyarginine, polylysine or an antimicrobial peptide.
  • the polycationic compound (s) to be used according to the present invention may be any polycationic compound which shows the characteristic effect according to the WO 97/30721.
  • Preferred polycationic compounds are selected from basic polypeptides, organic polycations, basic polyaminoacids or mixtures thereof. These polyaminoacids should have a. chain length of at least 4 amino acid residues (see: Tuftsin as described in Goldman et al (1983)).
  • substances containing peptidic bounds like polylysine, polyarginine and polypeptides containing more than 20%, especially more than 50% of basic amino acids in a range of more than 8, especially more than 20, amino acid residues or mixtures thereof.
  • polypeptides e.g. polyethyleneimine
  • WO 99/38528 e.g. polyethyleneimine
  • these polypeptides contain between 20 and 500 amino acid residues, especially between 30 and 200 residues.
  • polycationic compounds may be produced chemically or recom- binantly or may be derived from natural sources .
  • Cationic (poly) eptides may also be polycationic anti-bacterial microbial peptides with properties as reviewed in (Ganz and Le- hrer, 1999; Hancock, 1999). These (poly)peptides may be of pro- karyotic or animal or plant origin or may be produced chemically or recombinantly (Andreu and Rivas, 1998; Ganz and Lehrer, 1999; Simmaco et al . , 1998). Peptides may also belong to the class of defensins (Ganz, 1999; Ganz and Lehrer, 1999) . Sequences of such peptides can be, for example, be found in the Antimicrobial Sequences Database under the following internet address : http: //www.bbcm.univ. trieste.it/-tossi/pa ⁇ l .html
  • Such host defense peptides or defensives are also a preferred form of the polycationic polymer according to the present invention.
  • a compound allowing as an end product activation (or down-regulation) of the adaptive immune system, preferably mediated by APCs (including dendritic cells) is used as polycationic polymer.
  • cathelicidin derived antimicrobial peptides or derivatives thereof are especially preferred for use as polycationic substance in the present invention.
  • antimicrobial peptides derived from mammal cathelicidin preferably from human, bovine or mouse, or neuroac- tive compounds, such as (human) growth hormone.
  • Polycationic compound's derived from natural sources include HIV- REV or HIV-TAT (derived cationic peptides, antennapedia peptides, chitosan or other derivatives of chitin) or other peptides derived from these peptides or proteins by biochemical or recombi- nant production.
  • Other preferred polycationic compounds are cathelin or related or derived substances from cathelin.
  • mouse cathelin is a peptide which has the amino acid sequence NH -RLAGLLRKGGEKIGEKLKKIGOKIKNFFQKLVPQPE-COOH.
  • Related or derived cathelin substances contain the whole or parts of the cathelin sequence with at least 15-20 amino acid residues.
  • Derivations may include the substitution or modification of the natural amino acids by amino acids which are not among the 20 standard amino acids. Moreover, further cationic residues may be introduced into such cathelin molecules . These cathelin molecules are preferred to be combined with the antigen and the immunogenic ODN according to the present invention. However, these cathelin molecules surprisingly have turned out to be also effective as an adjuvant for a antigen without the addition of further adjuvants. It is therefore possible to use such cathelin molecules as efficient adjuvants in vaccine formulations with or without further immunactivating substances.
  • Another preferred polycationic substance to be used according to the present invention is a synthetic peptide containing at least 2 KLK-motifs separated by a linker of 3 to 7 hydrophobic amino acids (A 1789/2000, incorporated herein by reference) .
  • B and E in formula I are common groups for 5' and/or 3' ends of nucleic acid molecules .
  • Examples for such groups are readily available for the skilled man in the art (see e.g. "Oligonucleotides and Analogues - A Practical Approach” (1991) , ed. Eckstein, Oxford University Press) .
  • B and/or E are preferably selected independently from -H, -CH , -COCH , -OH, -CHO, a phosphate, thiophosphate, sulfate or a thiosulfate group, or a phosphoalkylgroup, especially with an alkyl length of C 1-C6 and/or with a terminal amino group (the amino group may e.g. be used for further labelling of the I-ODNs according to the present invention, e.g. -P0 (CH ) -NH or -PO - (CH 2) n-NH-Label) .
  • B Especially preferred as B are nucleosides, espe- cially the 2 'deoxynucleotides mentioned above (i.e. without the phosphate or thiophosphate group) .
  • these groups may also contain linker groups to other molecules, especially carrier molecules or labels .
  • these surfaces, particles, labels, etc. are then also part of the B and/or E groups .
  • the pharmaceutical composition according to the present invention may further comprise further active ingredients (pharmaceutically active substances), especially substances which are usable in a vaccine connection.
  • further active ingredients are cytokines, antiinflammatory substances, antimicrobial substances or combinations thereof.
  • composition according to the present invention may further contain auxiliary substances, especially a pharmaceutically acceptable carrier, buffer substances, stabilizers or combinations thereof.
  • the pharmaceutical composition according to the present invention preferably contains one or more ODNs according to the present invention, preferably 1 pg to 10 g, preferably 1 ng to 1 g, more preferred 100 ng to 10 mg, especially 10 mg to 1 mg.
  • the antigen as well as the polycationic polymer may be applied in similar dosages, a range of 1 to 10,000 mg antigen and 0.1 to 1,000 mg polycation per vaccination is preferred.
  • the present compositions may be applied to a patient, e.g. a vaccination candidate, in efficient amounts e.g. by weekly, biweekly or monthly intervals . Patients to be treated with the present compositions may also be vaccinated repeatedly or only once.
  • a preferred use of the present invention is the active immunisation, especially of humans or animals without protection against the specific antigen.
  • the route of application for the present composition is not critical, e.g. subcutaneous, intramuscular, intradermal or trans- dermal injection is suitable as well as oral uptake.
  • the present invention is therefore also directed to a kit comprising a composition containing the antigen and the polycationic polymer as one component and a composition containing dhe immunostimulating or chemotactic substance as a second component.
  • the components may be applied at the same site or time, however, an application at different sites or at a different time or for a different time period is also possible. It is also possible to vary the systemic or local applications of the composition or the components, respectively.
  • Fig. 1 shows the immune response against the ovalbumin-derived ginine (pR 60) and deoxyinosine I-containing oligodeoxynucleo- tides (I-ODN) or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. Four days later draining lymph node cells were ex vivo stimulated with OVAforce 25 c discipline7—2 n 6 r 4. The number of IFN-g-producing cells was determined 24 hours later using an ELISPOT assay. Results are expressed as the number of spots/lxlO 6 lymph node cells .
  • Fig. 2 shows the induction of systemic TNF-a production after the injection of OVA 257 _ 264 / poly-L-arginine (pR 60) and I-containing oligodeoxynucleotides (I-ODN) or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. One hour after injection blood was taken from the tail vein and serum was prepared. The concentration of TNF-a in the sera was determined using an ELISA.
  • Fig. 3 shows the immune response against the Ovalbumin-derived p ⁇ ep ⁇ tide OVA257-_264 after the inj-"ection of OVA justify25_ employ7- discipline26.
  • Mice were injected into the hind footpads with mixtures as indicated. Four days later, draining lymph node cells were ex vivo stimulated with OVA justify 25_ grasp7— haul264.
  • Fig. 4 shows the induction of systemic TNF-a production after the injection of OVA , poly-L-arginine (pR 60) and I-containing oligodeoxynucleotides (I-ODN), GpC or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. One hour after injection blood was taken from the tail vein and serum was prepared. The concentration of TNF-a and IL-6 in the sera was determined using cytokin-specific ELISAs.
  • Fig. 5 shows the immune response against the Ovalbumin-derived peptide OVA257—264 after the injection of TRP-2 , poly-L-arginine,
  • mice were injected into the hind footpads with mixtures as indicated. Four days later, draining lymph node cells were ex vivo stimu- . lated with TRP-2, an irrelevant peptide OVA 257— alone264 ' or pR 60. The number of IFN-g producing cells was determined 24 hours later using an ELISPOT assay. Results are expressed as the number of spots/lxlO 6 lymph node cells with standard deviation of triplicates .
  • Fig. 6 shows the combined injection of I-ODN and poly-L-arginine (pR 60) together with a Melanoma-derived peptide.
  • Fig. 7 shows that the combined injection of I-ODN and pR 60 together with a Melanoma-derived peptide reduces the induction of systemic TNF- ⁇ and IL-6.
  • Fig. 8 shows the combined injection of a random 10-mer I-ODN and pR 60 together with a Melanoma-derived peptide.
  • Fig. 9 shows that the combined application of ovalbumin (OVA) with oligo-dIC 2 6-mer and pR enhances production of OVA-specific IgG antibodies.
  • OVA ovalbumin
  • Mice were injected subcutaneously into the footpad with mixtures as indicated.
  • sera were collected and screened by ELISA for OVA-specific IgG2a (A) and IgGl (B) antibodies. The results are shown as the antibody titer.
  • thiophosphate-substituted ODNs (with thiophosphate residues substituting for phosphate, hereafter called "thiophosphate substituted oligodeoxynucleotides") were used since such ODNs display higher nuclease resistance (Ballas et al., 1996; Krieg et al . , 1995; Parronchi et al . , 1999).
  • Poly-L-arginine60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals
  • mice On day 0 mice were injected into each hind footpad with a total volume of 100 ml (50 ml per footpad) containing the above mentioned compounds. Animals, were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 mm cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl0 6 cells/ml in DMEM/5%/FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995). This method is.
  • Lymphocytes were stimulated ex vivo with medium background-control, OVA flood 25_ exert7— fashion264-peptide or Concanavalin A (Con A). Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO 6 cells are illustrated in Figure 1.
  • Peptide OVA 257 2 relieve64-Peptide (SIINFEKL) , a MHC class I
  • Poly-L-arginine 60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals Dose: lOO ⁇ g/mouse
  • CpG motif tec at ⁇ ac ⁇ ttc ctg atg ct, were synthesized by NAPS GmbH, G ⁇ ttingen.
  • mice On day 0 mice were injected into each hind footpad with a total volume of lOO ⁇ l (50 ⁇ l per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70um cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl0 6 cells/ml in DMEM/5%FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al., 1995). This method is a widely used procedure allowing the quantification of antigen-specific T cells .
  • Lymphocytes were stimulated ex vivo in triplicates with medium (background) , OVA 257264 -peptide, an irrelevant peptide TRP- 2 181—188 (murine tyrosinase related protein-2 , VYDFFVWL) , pR 60 and
  • Concanavalin A (Con A) . Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO 6 cells are illustrated in Figure 3, the standard deviation of ex vivo-stimulated triplicates are given. One hour after injection blood was taken from the tail vein and serum was prepared to determine the induction of systemic TNF-a and IL-6 using cytokine-specific ELISAs ( Figure 4) .
  • Example 3 Example 3 :
  • Poly-L-arginine 60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals Dose: lOO ⁇ g/mouse
  • CpG motif tec at ⁇ ac ⁇ ttc ctg atg ct, were synthesized by NAPS GmbH, G ⁇ ttingen.
  • TRP-2 + wdi + pR 60 10.TRP-2 + wdidin + pR 60 ll.TRP-2 + wdid + pR 60 12.TRP-2 + wdidid + pR 60
  • mice On day 0 mice were injected into each hind footpad with a total volume of lOO ⁇ l (50 ⁇ l per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 ⁇ m cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl0 6 cells/ml in DMEM/5%FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995).
  • Lymphocytes were stimulated ex vivo in triplicates with medium (background) , TRP-2-peptide, an irrelevant 0VA 257 _ 264 _ Pep- tide, pR 60 and Concanavalin A (Con A) . Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO 6 cells are illustrated in Figure 5, the standard deviation of ex vivo-stimu- lated triplicates are given.
  • mice On day 0 mice were injected into each hind footpad with a total volume of 100 ⁇ l (50 ⁇ l per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 ⁇ m cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3x10 cells/ml in DMEM/5%/FCS. An IFN- ⁇ ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995).
  • Lymphocytes were stimulated ex vivo in triplicates with medium background-control, TRP-2i8i-i88-peptide, an irrelevant OVA25 7 _254 ⁇ peptide and Concanavalin A (Con A) . Spots representing single IFN- ⁇ producing T cells were counted and the number of background spots was substracted from all samples . The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO cells are illustrated in Figure 6, the standard deviation of ex vivo- stimulated triplicates are given.
  • mice On day 0 mice were injected into each hind footpad with a total volume of 100 ⁇ l (50 ⁇ l per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 ⁇ m cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals). Cells were adjusted to 3xl0 6 cells/ml in DMEM/5%/FCS. An IFN- ⁇ ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995) .
  • This method is a widely used procedure allowing the quantification of antigen-specific T cells .
  • Lymphocytes were stimulated ex vivo in triplicates with medium background-control, TRP-2 ⁇ 8 ⁇ -i88-peptide, an irrelevant OVA 2 5 7 _2g4-peptide and Concanavalin A (Con A) .
  • Spots representing single IFN- ⁇ producing T cells were counted and the number of background spots was substracted from all samples .
  • the high number of spots detected after the stimulation with Con A indicate a good condition of the used lymphocytes.
  • the number of spots/lxl0 D cells are illustrated in Figure 8, the standard deviation of ex vivo- stimulated triplicates are given.
  • Poly-L-arginine60 (pR60] Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals
  • I-ODN 2 thiophosphate substituted ODNs containing deoxyinosine tec atg aci ttc ctg atg ct, were synthesized by NAPS GmbH, G ⁇ ttingen. Dose: 5nmol/mouse
  • oligo-deoxyIC 26 -mer and poly-L-arginine (pR) enhances the ovalbumin (OVA) -specific humoral response.
  • Ovalbumin Ovalbumin from chicken egg, grade V, SIGMA Chemicals, A-5503, Lot 54H7070 Dose: 50 ⁇ g/mouse
  • Poly-L-arginine (pR) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA Chemicals, P-4663, Lot 68H5903 Dose: 100 ⁇ g/mouse
  • mice were injected into each hind footpad with a total volume of lOO ⁇ l (50 ⁇ l per footpad) containing the above listed ⁇ compounds.
  • serum was collected and screened by ELISA for the presence of OVA-specific antibodies.
  • CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen. J Immunol 160, 870-876.
  • CpG DNA A potent signal for growth, activation, and maturation of human dendritic cells. Proc Natl Acad Sci U S A 96, 9305-9310.
  • Polyinosinic acid polycytidylic acid promotes T helper type 1-specific immune responses by stimulating macrophage production of interferon-a and interleukin-12.

Abstract

Described is an immunostimulatory oligodeoxynucleic acid molecule (ODN) having the structure according to formula (I), wherein any NMP is a 2' deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2-amino-deoxyribosepurine-, -6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N- isopentenyl-deoxyadenosine-monophosphate or -monothiophosphate, NUC is a 2' deoxynucleoside, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2-amino-deoxyribosepurine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopentenyl-deoxyadenosine, any X is O or S, a and b are integers from 0 to 100 with the proviso that a + b is between 4 and 150, B and E are common groups for 5' or 3' ends of nucleic acid molecules, as well as a pharmaceutical composition containing such ODNs.

Description

Immunostimulatory oligodeoxynucleotides
The present invention relates to immunostimulatory oligodeoxynucleic molecules (ODNs) and pharmaceutical compositions containing such ODNs .
Vaccines can save more lives (and resources) than any other medical intervention (Nossal, 1998) . Owing to world-wide vaccination programs the incidence of many fatal diseases has been decreased drastically. Although this notion is valid for a whole panel of diseases, e.g. tuberculosis, diphtheria, pertussis, measles and tetanus, there are no effective vaccines for numerous infectious disease including most viral infections, such as AIDS. There are also no effective vaccines for other diseases, infectious or non-infectious claiming millions the lives of millions of patients per year including malaria or cancer. In addition, the rapid emergence of antibiotic-resistant bacteria and microorganisms calls for alternative treatments with vaccines being a logical choice. Finally, the great need for vaccines is also illustrated by the fact that infectious diseases, rather than cardiovascular disorders or cancer or injuries remain the largest cause of death and disability in the world (Bloom and Widdus, 1998) .
From an immunological point of view one major problem in the field of vaccines today is that traditional vaccines (and/or the immune-modulating compounds contained within these preparations) are designed to induce high levels of antibodies (Harrow and Lane, 1988) . However, antibodies on their own are not effective in preventing a large number of diseases including most illnesses caused by viruses, intracellular bacteria, certain parasites and cancer. Examples for such diseases are, but are not restricted to, the above-mentioned HIV virus or Plasmodium spec, in case of malaria. In numerous experimental systems it has been shown that the cellular arm of the immune system, including T cells, rather than the humoral arm, is important for these indications. Therefore, novel, innovative technologies are needed to overcome the limitations of conventional vaccines. The focus must be on technologies that reliably induce the cellular immune system, including antigen specific T cells, which recognize molecules expressed on pathogen infected cells. Ideally, vaccines are designed that induce both T cells distinguishing diseased and/or infected cells from normal cells and, simultaneously, antibodies secreted by B cells recognising pathogens in extracellular compartments .
Several established vaccines consist of live attenuated organism where the risk of reversion to the virulent wild-type strain exists. In particular in immunocompromised hosts this can be a live threatening scenario. Alternatively, vaccines are administered as a combination of pathogen-derived antigens together with compounds that induce or enhance immune responses against these antigens (these compounds are commonly termed adjuvant) , since these subunit vaccines on their own are generally not effective.
Whilst there is no doubt that the above vaccines are valuable medical treatments, there is the disadvantage that, due to their complexity, severe side effects can be evoked, e.g. to antigens that are contained in the vaccine that display cross-reactivity with molecules expressed by cells of vaccinated individuals. In addition, existing requirements from regulatory authorities, e.g. the World Health Organization (WHO) , the Food and Drug Administration (FDA), and their European counterparts, for exact specification of vaccine composition and mechanisms of induction of immunity, are difficult to meet.
Antigen presenting cells belong to the innate immune system, which has evolved as a first line host defence that limits infection early after exposure to microorganisms (Hoffmann et al . , 1999). Cells of the innate immune sytem recognize patterns or relatively non-specific structures expressed on their targets rather than more sophisticated, specific structures which are recognised by the adaptive immune system (Hoffmann et al . , 1999). Examples of cells of the innate immune system are macrophages and dendritic cells but also granulocytes (e.g. neutrophiles) , natural killer cells and others. By contrast, cells of the adaptive immune system recognize specific, antigenic structures, including peptides, in the case of T cells and peptides as well as three- dimensional structures in the case of B cells. The adaptive immune system is much more specific and sophisticated than the innate immune system and improves upon repeat exposure to a given pathogen/antigen. Phylogenetically, the innate immune system is much older and can be found already in very primitive organisms. Nevertheless, the innate immune system is critical during the initial phase of antigenic exposure since, in addition to containing pathogens, cells of the innate immune system, i.e. APCs, prime cells of the adaptive immune system and thus trigger specific immune responses leading to clearance of the intruders. In sum, cells of the innate immune sytem and in particular APCs play a critical role during the induction phase of immune responses by a) containing infections by means of a primitive pattern recognition system and b) priming cells of the adaptive immune system leading to specific immune responses and memory resulting in clearance of intruding pathogens or of other targets (Roitt et al . , 1998). These mechanisms may also be important to clear or contain tumor cells . ,
As mentioned above, cells of the innate immune system recognise patterns expressed on their respective targets . Examples are lipopolysaccharides ( PS) in the case of Gram-negative bacteria, mycobacterial glycolipids, lipoteichoic acids of Gram-positive bacteria, mannans of yeast and double stranded RNAs of viruses (Hoffmann et al . , 1999). In addition they may recognise patterns such as altered glycosylations of proteins on tumor cells .
Recent findings describe DNAs of protozoan or lower eukaryotes as a further pattern recognised by the innate (but possibly also by the adaptive) immune system of mammals (and probably most if not all vertebrates) (Krieg, 1996; Lipford et al . , 1998).
The immune system recognises lower organisms including bacteria probably due to structural and sequence usage differencies between pathogen and host DNA. In particular short stretches of DNA, derived from non-vertebrates or in form of short synthetic ODNs containing nonmethylated cytosine-guanine dinucleotides (CpG) in a certain base context, are targeted (Krieg et al . , 1995) . CpG motifs are found at the expected frequency in bacterial DNA but are much less frequent in vertebrate DNA (LipLord et al., 1998; Pisetsky, 1999). In addition, non-vertebrate (i.e. bacterial) CpG motifs are not methylated whereas vertebrate CpG sequences are. These differences between bacterial DNA and verte- brate DNA allow vertebrates to recognise non-vertebrate DNA as a danger signal.
Natural CpG-containing DNA, ODNs, as well as thiophosphate-sub- stituted (exchange of thiophosphate residues for phosphate) ODNs containing CpG motifs (CpG-ODN) are not only potent activators of immune cell proliferation and humoral immune responses (Krieg et al . , 1995), but also stimulate strong cellular immune responses (reviewed in Lipford et al . , 1998) . DNA/ODNs containing non-methylated CpG motifs can directly activate monocytic cells (dendritic cells, macrophages) and B cells. Likely, natural killer (NK) cells are not directly activated but respond to ono- cyte-derived IL-12 (interleukin 12) with a marked increase in their IFN-γ production (Chace et al., 1997). In consequence, the induction of monocytes and NK cells by CpG DNA promotes the induction of Thl-type responses and the development of cytotoxic T cells .
Ribonucleic acid based on inosine and cytosine, like polyi- nosinic-polycytidylic acid (poly I:C), is known to promote Thl- specific immune responses. It is known to stimulate macrophages to produce cytokines such as IL-lα and IL-12 (Manetti et al . , 1995) , it is also known as a potent interferon type 1 inducer (Manetti et al . , 1995) and a potent NK cell stimulator (Cavanaugh et al. , 1996) .
This effect, however, was strictly restricted to ribonucleic acid containing inosine and cytidine residues (W098/16247) .
Investigations by the inventors of the present invention showed that ODNs containing non-methylated CpG motifs, although being efficient in stimulating immune system, have essential disadvantages, especially with respect to specificity (high background) and induction of side effects, such as high systemic TNF-α generation. High systemic TNF-α release is known to cause toxic shock syndrome, which can cause death of afflicted patients.
It is therefore an object of the present invention to provide suitable novel ODNs which do not have such drastic side effects as ODNs based on CpG sequences. It is a further object to reduce the side effects of pharmaceutical compositions containing known ODNs and to provide safe and efficient well-tolerable pharmaceutical compositions with efficient, immunostimulatory properties which are suitable for vaccination of animals, especially of mammals , including humans .
This object is solved' by immunostimulatory oligode'oxynucleic acid molecule (ODN) having the structure according to formula (I)
Figure imgf000007_0001
(I),
any X is 0 or S,
wherein any NMP is a 2 ' deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2-amino-deoxyribosepu- rine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopen- • tenyl-deoxyadenosine-monophosphate or -monothiophosphat, NUC is a 2' deoxynucleoside, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5- methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2- a ino-deoxyribosepurine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopentenyl-deoxyadenosine, a and b are integers from 0 to 100 with the proviso that a + b is between 4 and 150,
B and E are common groups for 5 ' or 3 ' ends of nucleic acid molecules . Surprisingly it turned out that ODNs containing deoxyinosine residues (I-ODNs) show an immunostimulatory effect comparable or in many instances even better than ODNs containing CpG motifs. Moreover, ODNs according to the present invention produce more specific immune responses to a given antigen or antigen fragment than CpG ODNs. In addition, ODNs according to the present invention reduced the induction of adverse side reactions, especially the induction of systemic TNF-α or IL-6.
Whereas certain immunostimulatory effects had been described for inosine containing RNA molecules, such as poly-IC or the molecules mentioned in W098/16247, it surprisingly turned out that deoxynucleic acid molecules containing deoxyinosine residues, may be good immunostimulating ODNs.
In addition, the I-ODNs according to the present invention are - in contrast to ODNs based on the specific CpG motif - not dependent on a specific motif or a palindromic sequence as described for the CpG oligonucleotides (see e.g. EP 0 468 520 A2, WO96/02555, WO98/18810, W098/37919, WO98/40100, W098/52581, W099/51259 and W099/56755, all incorpoarted herein by reference) . Therefore, one group of I-ODNs according to the present invention may preferably contain a CI motif (and therefore ODNs described in these incorporated references, wherein one or more guanosine residues are replaced with deoxyinosine residues are preferred embodiments of the present ODNs) . It is not necessary for its principle immunostimulatory property, since I-ODNs with an Inosine not placed in a CI or IC context exhibit immunostimulatory properties as well.
The I-ODN according to the present invention is therefore a DNA molecule containing a deoxyinosine residue which is preferably provided in single stranded form.
The I-ODN according to the present invention may be isolated through recombinant methods or chemically synthesized. In the latter case, the I-ODN according to the present invention may also contain modified oligonucleotides which may be synthesized using standard chemical transformations, such as methylphosphon- ates or other phosphorous based modified oligonucleotides, such as phosphotriesters, phosphoamidates and phosphorodithiorates . Other non-phosphorous based modified oligonucleotides can also be used (Stirchak et al . , MAR 17 (1989), 6129-6141), however, mono- phosphates or monothiophosphates being the preferred 2 'deoxynucleoside monophosphate to be used in the present invention.
The NMPs of the I-ODNs according to the present invention are preferably selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine- monophosphate or -monothiophosphate (as usual, the phosphate or thiophosphate group is 5' of the deoxyribose) . Whereas it is essential for the ODNs based on the CpG motif that this motif is unmethylated, this is surprisingly not the case for the ODNs according to the present invention, wherein e.g. 2-methyl-deoxyinosine or 5-methyl-deoxycytosine residues have no general negative effect on immunostimulatory properties of the ODNs according to the present invention. Alternatively, instead of the 2-deoxy- forms of the NMPs, also other, especially inert, groups may be present at the 2-site of the ribose group, such as e.g. -F, -NH2, -CH , especially -CH . Of course, -OH and SH groups are excluded for the I-ODNs according to the present invention to be present on the 2 '-site of the ribose, especially the ribose residue for the inosine NMP.
The length of the ODNs according to the present invention is in the range of the standard ODNs used according to the prior art. Therefore molecules with a total length under 4 and above 150 show gradually decreasing immunostimulatory potential. Preferred ODNs contain between 10 and 60, especially between 15 and 40 bases (nucleosides) , implying that a + b in formula I is between 10 and 60, preferably between 15 and 40 in these preferred embodiments .
Whereas the ribonucleic acid molecules containing inosine and cytidine described to be immunostimulatory in the prior art have been large and relatively undefined polynucleic acids with molecular weights far above 200,000 (a commercially available poly- inosinic-polycytidylic acid from Sigma Chemicals has a molecular weight ranging from 220,000 to 460,000 (at least 500-1000 C+I residues) . The molecules according to the present invention are DNA molecules of much shorter length with a well defined length and composition, being highly reproducible in products.
It is further preferred that the deoxyinosine containing NMP of the I-ODNs according to formula I is a monothiophosphate with one to four sulfur atoms and that also further NMPs, especially all further NMPs, are present as nucleoside monothiophosphates , because such ODNs display higher nuclease resistance (it is clear for the present invention that the "mono" in the "monothiophosphates" relates to the phosphate, i.e. that one phosphate group (one phosphor atom) is present in each NMP) . Preferably, at least one of X 1 and X2 is S and at least one of X3 and X4 is 0 in the
NMPs according to the present invention. Preferably, X and X are 0. (X may be (due to synthesis of the NMP) derived e.g. from the phosphate group or from the 3 '-group of the NMP-ribose) .
Preferably the ODNs according to the present invention contain the sequence hhh wdi dhh h nhh hhh wdi hhh hhh hhh wn, nhh wdi din hhh hdi ndi nh, nhh hhh wdi dhh hhh hhh wn or nhh wdi did hhh hdi ddi dh, wherein any n is a 2 '-deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxycytosine- or deoxythymidine-monophosphate or -monothiophosphate, any h is a 2 ' -deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxycytosine- or deoxythymidine-monophosphate or' -monothiophosphate i is deoxyinosi e-monophosphate or -monothiophosphate, any w is a 2 ' -deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine- or deoxythymidine-monophosphate or -monothiophosphate, and any d is a 2 ' -deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine- or deoxythymidine-monophosphate or -monothiophosphate. As outlined above, a specific motif (such as CpG or a palindrome) is not necessary for the I-ODNs according to the present invention. However, ODNs containing a CI motif are preferred so that in a preferred embodiment the ODN according to formula I contains at least one 2 'deoxycytosine-monophosphate or -monothiophosphate 3 '-adjacent to a 2 '-deoxyinosine-monophosphate or -monothiophosphate to form such a 5'-CI 3 '-motif.
Preferred ODNs according to the present invention contain one or more of the sequence
gacitt, iacitt, gaictt, iaictt, wherein a is deoxyadenosine-monophosphate or -monothiophosphate, g is deoxyguanosine-monophosphate or -monothiophosphate, i is deoxyinosine-monophosphate or -monothiophosphate, c is deoxycytosine-monophosphate or -monothiophosphate and t is deoxythymidine-monophosphate or -monothiophosphate.
The I-ODNs according to the present invention are especially suitable for application in the pharmaceutical field, e.g. to be applied as a medicine to an animal or to humans . They are specifically adapted to act as an immunostimulatory agent, especially in or together with vaccine compositions.
Therefore, the present invention also relates to a pharmaceutical composition comprising an ODN according to the present invention.
Since a preferred pharmaceutical composition according to the present invention is a vaccine, this composition should contain an antigen besides the ODN according to the present invention. The potential of this antigen to raise a. protection/immune response of the vaccinated individual is strongly increased by combining it with the ODNs according to the present invention, especially due to their immunostimulatory activity. A vaccine can contain a whole variety of different antigens. Examples of antigens are whole-killed organisms such as inactivated viruses or bacteria, fungi, protozoa or even cancer cells. Antigens may also consist of subfractions of these organisms/tissues, of proteins, or, in their most simple form, of peptides. Antigens can also be recognised by the immune system in form of glycosy- lated proteins or peptides and may also be or contain polysaccha- rides or lipids. Short peptides can be used since for example cytotoxic T cells (CTL) recognize antigens in form of short usually 8-11 amino acids long peptides in conjunction with major histocompatibility complex (MHC) (Rammensee et al . , Immunogenet- ics 41, (1995), 178-228). B cells recognize longer peptides starting at around 15 amino acids (Harrow et al, Cold Spring Harbor: Cold Spring Harbor Laboratory, (1988)). By contrast to T cell epitopes, the three dimensional structure of B cell antigens may also be important for recognition by antibodies . In order to obtain sustained, antigen-specific immune responses, adjuvants are helpful to trigger immune cascades that involve all cells of the immune system necessary. Primarily, adjuvants are acting, but are not restricted in their mode of action, on so-called antigen presenting cells (APCs) . These cells usually first encounter the antigen (s) followed by presentation of processed or unmodified antigen to immune effector. Intermediate cell types may also be involved. Only effector cells with the appropriate specificity are activated in a productive immune response. The adjuvant may also locally retain antigens and co-injected other factors. In addition the adjuvant may act as a chemoattractant for other immune cells or may act locally and/or systemically as a stimulating agent for the immune system.
According to a preferred embodiment, T cell epitopes are used as antigens . Alternatively, a combination of T cell epitopes and B cell epitopes may also be preferred.
The antigens to be used in the present compositions are not critical. Also mixtures of different antigens are of course possible to be used according to the present invention. Preferably, proteins or peptides derived from a viral or a bacterial pathogen or from fungi or parasites are used as such antigens (including derivatized antigens or glycosylated' or lipidated antigens or polysaccharides or lipids) . Another preferred source of antigens are tumor antigens . Preferred pathogens are selected from human immunodeficiency virus (HIV) , hepatitis A and B viruses, hepatitis C virus (HCV) , rous sarcoma virus (RSV) , Epstein Barr virus (EBV) Influenza virus, Rotavirus, Staphylococcus aureus, Chlamydia pneumonias, Chlamydia trachomatis, Mycobacterium tuberculosis, Streptococcus pneumonias, Bacillus anthracis, Vibrio cholerae, Plasmodium sp. (Pi. falciparum, Pi. vivax, etc.), As- pergillus sp . or Candida albicans . Antigens may also be molecules expressed by cancer cells (tumor antigens) . The derivation process may include the purification of a specific protein from the pathogen/cancer cells, the inactivation of the pathogen as well as the proteolytic or chemical derivatization or stabilisation of such a protein. In the same way also tumor antigens (cancer vaccines) or autoimmune antigens may be used in the pharmaceutical composition according to the present invention. With such compositions a tumor vaccination or a treatment for autoimmume diseases may be performed.
In the case of peptide antigens the use of peptide mimitopes/ago- nists/superagonists/antagonists or peptides changed in certain positions without affecting the immunologic properties or non- peptide mimitopes/agonists/superagonists/antagonists (reviewed in Sparbier and Walden, 1999) is included in the current invention. Peptide antigens may also contain elongations either at the carboxy or at the amino terminus of the peptide antigen facilitating interaction with the polycationic compound(s) or the immunostimulatory compound(s) . For the treatment of autoimmune diseases peptide antagonists may be applied.
Antigens may also be derivatized to include molecules enhancing antigen presentation and targeting of antigens to antigen presenting cells.
In one embodiment of the invention the pharmaceutical composition serves to confer tolerance to proteins or protein fragments and peptides which are involved in autoimmune diseases . Antigens used in this embodiments serve to tolerize the immune system or down- regulate immune responses against epitopes involved in autoimmune processes . Preferably the pharmaceutical composition according to the present invention, especially in the form of a vaccine, further comprises a polycationic polymer, preferably a polycationic peptide, especially polyarginine, polylysine or an antimicrobial peptide.
The polycationic compound (s) to be used according to the present invention may be any polycationic compound which shows the characteristic effect according to the WO 97/30721. Preferred polycationic compounds are selected from basic polypeptides, organic polycations, basic polyaminoacids or mixtures thereof. These polyaminoacids should have a. chain length of at least 4 amino acid residues (see: Tuftsin as described in Goldman et al (1983)). Especially preferred are substances containing peptidic bounds, like polylysine, polyarginine and polypeptides containing more than 20%, especially more than 50% of basic amino acids in a range of more than 8, especially more than 20, amino acid residues or mixtures thereof. Other preferred polycations and their pharmaceutical compositons are described in WO 97/30721 (e.g. polyethyleneimine) and WO 99/38528. Preferably these polypeptides contain between 20 and 500 amino acid residues, especially between 30 and 200 residues.
These polycationic compounds may be produced chemically or recom- binantly or may be derived from natural sources .
Cationic (poly) eptides may also be polycationic anti-bacterial microbial peptides with properties as reviewed in (Ganz and Le- hrer, 1999; Hancock, 1999). These (poly)peptides may be of pro- karyotic or animal or plant origin or may be produced chemically or recombinantly (Andreu and Rivas, 1998; Ganz and Lehrer, 1999; Simmaco et al . , 1998). Peptides may also belong to the class of defensins (Ganz, 1999; Ganz and Lehrer, 1999) . Sequences of such peptides can be, for example, be found in the Antimicrobial Sequences Database under the following internet address : http: //www.bbcm.univ. trieste.it/-tossi/paσl .html
Such host defense peptides or defensives are also a preferred form of the polycationic polymer according to the present invention. Generally, a compound allowing as an end product activation (or down-regulation) of the adaptive immune system, preferably mediated by APCs (including dendritic cells) is used as polycationic polymer.
Especially preferred for use as polycationic substance in the present invention are cathelicidin derived antimicrobial peptides or derivatives thereof (A 1416/2000, incorporated herein by reference) , especially antimicrobial peptides derived from mammal cathelicidin, preferably from human, bovine or mouse, or neuroac- tive compounds, such as (human) growth hormone.
Polycationic compound's derived from natural sources include HIV- REV or HIV-TAT (derived cationic peptides, antennapedia peptides, chitosan or other derivatives of chitin) or other peptides derived from these peptides or proteins by biochemical or recombi- nant production. Other preferred polycationic compounds are cathelin or related or derived substances from cathelin. For example, mouse cathelin is a peptide which has the amino acid sequence NH -RLAGLLRKGGEKIGEKLKKIGOKIKNFFQKLVPQPE-COOH. Related or derived cathelin substances contain the whole or parts of the cathelin sequence with at least 15-20 amino acid residues. Derivations may include the substitution or modification of the natural amino acids by amino acids which are not among the 20 standard amino acids. Moreover, further cationic residues may be introduced into such cathelin molecules . These cathelin molecules are preferred to be combined with the antigen and the immunogenic ODN according to the present invention. However, these cathelin molecules surprisingly have turned out to be also effective as an adjuvant for a antigen without the addition of further adjuvants. It is therefore possible to use such cathelin molecules as efficient adjuvants in vaccine formulations with or without further immunactivating substances.
Another preferred polycationic substance to be used according to the present invention is a synthetic peptide containing at least 2 KLK-motifs separated by a linker of 3 to 7 hydrophobic amino acids (A 1789/2000, incorporated herein by reference) .
It was very surprising that the immunostimulating effect of the pharmaceutical composition according to the present invention was - In significantly higher than it could be expected from the addition of the effects of each single component or even the addition of the effects of the ODN or the polycation with the antigen.
B and E in formula I are common groups for 5' and/or 3' ends of nucleic acid molecules . Examples for such groups are readily available for the skilled man in the art (see e.g. "Oligonucleotides and Analogues - A Practical Approach" (1991) , ed. Eckstein, Oxford University Press) . For the I-ODNs according to the present invention B and/or E are preferably selected independently from -H, -CH , -COCH , -OH, -CHO, a phosphate, thiophosphate, sulfate or a thiosulfate group, or a phosphoalkylgroup, especially with an alkyl length of C 1-C6 and/or with a terminal amino group (the amino group may e.g. be used for further labelling of the I-ODNs according to the present invention, e.g. -P0 (CH ) -NH or -PO - (CH 2) n-NH-Label) . Especially preferred as B are nucleosides, espe- cially the 2 'deoxynucleotides mentioned above (i.e. without the phosphate or thiophosphate group) . Alternatively these groups may also contain linker groups to other molecules, especially carrier molecules or labels . In such forms of ODNs wherein the ODNs are bound to solid surfaces or particles or labels, these surfaces, particles, labels, etc. are then also part of the B and/or E groups .
Of course, any ionised (salt) form or tautomeric forms of the molecules according to formula I are included in this formula I .
The pharmaceutical composition according to the present invention may further comprise further active ingredients (pharmaceutically active substances), especially substances which are usable in a vaccine connection. Preferred embodiments of such further active ingredients are cytokines, antiinflammatory substances, antimicrobial substances or combinations thereof.
Of course, the pharmaceutical composition according to the present invention may further contain auxiliary substances, especially a pharmaceutically acceptable carrier, buffer substances, stabilizers or combinations thereof.
The relative amounts of the ingredients in the present pharmaceu- tical composition are highly dependent on the necessities of the individual antigen and on the animal/human to which this composition should be applied to. Therefore, the pharmaceutical composition according to the present invention preferably contains one or more ODNs according to the present invention, preferably 1 pg to 10 g, preferably 1 ng to 1 g, more preferred 100 ng to 10 mg, especially 10 mg to 1 mg. The antigen as well as the polycationic polymer may be applied in similar dosages, a range of 1 to 10,000 mg antigen and 0.1 to 1,000 mg polycation per vaccination is preferred.
The present compositions may be applied to a patient, e.g. a vaccination candidate, in efficient amounts e.g. by weekly, biweekly or monthly intervals . Patients to be treated with the present compositions may also be vaccinated repeatedly or only once. A preferred use of the present invention is the active immunisation, especially of humans or animals without protection against the specific antigen.
The route of application for the present composition is not critical, e.g. subcutaneous, intramuscular, intradermal or trans- dermal injection is suitable as well as oral uptake.
It is also possible to apply the present composition separatedly e.g. by injecting the immunostimulating substance separatedly from the antigen/polycation composition. The present invention is therefore also directed to a kit comprising a composition containing the antigen and the polycationic polymer as one component and a composition containing dhe immunostimulating or chemotactic substance as a second component.
The components may be applied at the same site or time, however, an application at different sites or at a different time or for a different time period is also possible. It is also possible to vary the systemic or local applications of the composition or the components, respectively.
Details of the present invention are described by the following examples and the figures, but the invention is of course not limited thereto. Fig. 1 shows the immune response against the ovalbumin-derived
Figure imgf000018_0001
ginine (pR 60) and deoxyinosine I-containing oligodeoxynucleo- tides (I-ODN) or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. Four days later draining lymph node cells were ex vivo stimulated with OVA„ 25c„7—2 n6r4. The number of IFN-g-producing cells was determined 24 hours later using an ELISPOT assay. Results are expressed as the number of spots/lxlO6 lymph node cells .
Fig. 2 shows the induction of systemic TNF-a production after the injection of OVA 257_264/ poly-L-arginine (pR 60) and I-containing oligodeoxynucleotides (I-ODN) or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. One hour after injection blood was taken from the tail vein and serum was prepared. The concentration of TNF-a in the sera was determined using an ELISA.
Fig. 3 shows the immune response against the Ovalbumin-derived p ^ep ^tide OVA257-_264 after the inj-"ection of OVA„25_„7-„26. p ^olyJ-L-ar- ginine (pR60) and deoxyinosine -containing oligodeoxynucleotides (I-ODN), CpG 1668 or GpC . Mice were injected into the hind footpads with mixtures as indicated. Four days later, draining lymph node cells were ex vivo stimulated with OVA„ 25_„7—„264. an irrelevant peptide mTRP21 1o811—,18008 (murine tyrosinase related protein-2, VYDFFVWL) or pR 60. The number of IFN-g producing cells was determined 24 hours later using an ELISPOT assay. Results are expressed as the number of spots/lxlO6 lymph node cells with standard deviation of triplicates .
Fig. 4 shows the induction of systemic TNF-a production after the injection of OVA , poly-L-arginine (pR 60) and I-containing oligodeoxynucleotides (I-ODN), GpC or CpG 1668. Mice were injected into the hind footpads with mixtures as indicated. One hour after injection blood was taken from the tail vein and serum was prepared. The concentration of TNF-a and IL-6 in the sera was determined using cytokin-specific ELISAs.
Fig. 5 shows the immune response against the Ovalbumin-derived peptide OVA257—264 after the injection of TRP-2 , poly-L-arginine,
CpG 1668 or random 20-mer sequences containing deoxyinosine. Mice were injected into the hind footpads with mixtures as indicated. Four days later, draining lymph node cells were ex vivo stimu- . lated with TRP-2, an irrelevant peptide OVA 257—„264' or pR 60. The number of IFN-g producing cells was determined 24 hours later using an ELISPOT assay. Results are expressed as the number of spots/lxlO6 lymph node cells with standard deviation of triplicates .
Fig. 6 shows the combined injection of I-ODN and poly-L-arginine (pR 60) together with a Melanoma-derived peptide.
Fig. 7 shows that the combined injection of I-ODN and pR 60 together with a Melanoma-derived peptide reduces the induction of systemic TNF-α and IL-6.
Fig. 8 shows the combined injection of a random 10-mer I-ODN and pR 60 together with a Melanoma-derived peptide.
Fig. 9 shows that the combined application of ovalbumin (OVA) with oligo-dIC26-mer and pR enhances production of OVA-specific IgG antibodies. Mice were injected subcutaneously into the footpad with mixtures as indicated. At day 24 and 115 after injection, sera were collected and screened by ELISA for OVA-specific IgG2a (A) and IgGl (B) antibodies. The results are shown as the antibody titer.
E X AM P L E S
In all experiments thiophosphate-substituted ODNs (with thiophosphate residues substituting for phosphate, hereafter called "thiophosphate substituted oligodeoxynucleotides") were used since such ODNs display higher nuclease resistance (Ballas et al., 1996; Krieg et al . , 1995; Parronchi et al . , 1999).
Example 1
The combined injection of different I-ODNs and poly-L-arginine (pR 60) synergistically enhances the immune response against an Ovalbumin-derived peptide.
Mice C57BI/6 (Harlan/Olac) Peptide OVA_ 257-„264-Pep ^tide (SIINFEKL) , a MHC class I (H-2Kb) -restricted epitope of chicken ovalbumin (Rotzschke et al., 1991), was synthesized using standard solid phase F-moc chemistry synthesis, HPLC purified and analysed by mass spectroscopy for purity. Dose: 300 mg/mouse
Poly-L-arginine60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals
Dose: lOOmg/mouse
CpG-ODN 1668 thiophosphate substituted ODNs containing a CpG motif: tec atcr acσ ttc ctg atg ct, were synthesized by NAPS GmbH, Gδttin- gen.
Dose: 5 nmol/mouse
I-ODN 1 thiophosphate substituted ODNs containing deoxyinosine: tec ati aci ttc ctg atg ct, were synthesized by NAPS GmbH, Gottin- gen.
Dose: 5nmol/mouse
I-ODN 2 thiophosphate substituted ODNs containing deoxyinosine: tec atg aci ttc ctg atg ct, were synthesized by NAPS GmbH, Gδttin- gen. Dose: 5nmol/mouse I-ODN 3 thiophosphate substituted ODNs containing deoxyinosine: tec ati aci ttc cti ati ct, were synthesized by NAPS GmbH, Gottin- gen. Dose: 5nmol/mouse
Experimental groups (5 mice per group)
1 • OVA257-264
2 - O A257-264 + PR 6 0
3 - OVA257-264 + CPQ 1 6 6 8 - O A257-264 + τ ~0m 1
5 • OVA257-264 + X -0m 2
6 • OVA257-264 + IDN . 3
1 ■ OVA257-264 + CPG 1 6 6 8 + PR 6 0
8 . OVA257 264 + I-ODN 1 + pR 60
9 . OVA257 254 + I-ODN 2 + pR 60
10 . OVA 257— „264, + I-ODN 3 + pR 60
On day 0 mice were injected into each hind footpad with a total volume of 100 ml (50 ml per footpad) containing the above mentioned compounds. Animals, were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 mm cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl06cells/ml in DMEM/5%/FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995). This method is. a widely used procedure allowing the quantification of antigen-specific T cells. Lymphocytes were stimulated ex vivo with medium background-control, OVA„ 25_„7—„264-peptide or Concanavalin A (Con A). Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO6 cells are illustrated in Figure 1.
One hour after injection blood was taken from the tail vein and serum was prepared to determine the induction of systemic TNF-a using an ELISA (Figure 2) .
Example 2
The exchange of Guanosine by desoxy-Inosine converts the non-im- munogeneic GpC-sequence to a highly immunogeneic one, especially when combined with poly-L-arginine (pR60).
Mice C57B1/6 (Harlan/Olac)
Peptide OVA 257—2„64-Peptide (SIINFEKL) , a MHC class I
(H-2Kb) -restricted epitope of chicken ovalbumin (Rotzschke et al . , 1991), was synthesized using standard solid phase F-moc synthesis, HPLC purified and analysed by mass spectroscopy for purity. Dose: 300μg/mouse
Poly-L-arginine 60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals Dose: lOOμg/mouse
CpG-ODN 1668 thiophosphate substituted ODNs containing a
CpG motif: tec atσ acα ttc ctg atg ct, were synthesized by NAPS GmbH, Gδttingen.
Dose: 5nmol/mouse GpC-ODN thiophosphate substituted ODNs containing an non-immunogeneic GpC motif: tec atg age ttc ctg atg ct were synthesized by NAPS GmbH,
Gόttingen.
Dose: 5nmol/mouse I-ODN 9 thiophosphate substituted ODNs containing deoxyinosine: tec atg aic ttc ctg atg ct were synthesized by NAPS GmbH, Gόttingen.
Dose: 5nmol/mouse I-ODN 10 thiophosphate substituted ODNs containing deoxyinosine: tec ati aic ttc cti ati ct were synthesized by NAPS GmbH, Gδttingen. Dose: 5nmόl/mouse
Experimental groups (5 mice per group) OVA 257-264
OVA 257-264 + p ^R 60
OVA 257- _264 + Cp ^G 1668
OVA 2„5__7,-2 ne64 + Gp ^C
OVA 25C.7-2 64, + I-ODN 9
OVA 257-264 + I-ODN 10
OVA 257-264 + CpG 1668 _ + pR 60
OVAO 25C„7- „264, + Gp ^C + p ^R 60
OVA257-264 + IDN 9 + R 6 0
OVA 2,5„7-„264 + I-ODN 10 + pR 60
On day 0 mice were injected into each hind footpad with a total volume of lOOμl (50μl per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70um cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl06cells/ml in DMEM/5%FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al., 1995). This method is a widely used procedure allowing the quantification of antigen-specific T cells . Lymphocytes were stimulated ex vivo in triplicates with medium (background) , OVA257264-peptide, an irrelevant peptide TRP- 2 181—188 (murine tyrosinase related protein-2 , VYDFFVWL) , pR 60 and
Concanavalin A (Con A) . Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO6 cells are illustrated in Figure 3, the standard deviation of ex vivo-stimulated triplicates are given. One hour after injection blood was taken from the tail vein and serum was prepared to determine the induction of systemic TNF-a and IL-6 using cytokine-specific ELISAs (Figure 4) . Example 3 :
The combined injection of random 20-mer sequences containing deoxyinosine and a Melanoma-derived peptide induces a strong immune response against the peptide which can be further enhanced by the co-application of poly-L-arginine (pR 60) .
Mice C57B1/6 (Harlan/Olac)
Peptide TRP-2-peptide (VYDFFVWL) , a MHC class (H-2Kb) -restricted epitope of mouse tyrosinase related protein-2 (Bllo et al . , 1997) was synthesized by standard solid phase F-moc synthesis, HPLC purified and analyzed by mass spectroscopy for purity. Dose: 300μg/mouse
Poly-L-arginine 60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals Dose: lOOμg/mouse
CpG-ODN 1668 thiophosphate substituted ODNs containing a
CpG motif: tec atσ acα ttc ctg atg ct, were synthesized by NAPS GmbH, Gδttingen.
Dose: 5nmol/mouse wdi thiophosphate substituted ODNs: nhh hhh wdi nhh hhh hhh wn were synthesized by NAPS GmbH,
Gόttingen.
Dose: 5nmol/mouse
wdidin thiophosphate substituted ODNs : nhh hhh wdi nhh hhh hhh wn were synthesized by NAPS GmbH,
Gδttingen.
Dose: 5nmol/mouse
wdid thiophosphate substituted ODNs : nhh hhh wdi dhh hhh hhh wn were synthesized by NAPS GmbH,
Gόttingen.
Dose: 5nmol/mouse wdidid thiophosphate substituted ODNs : nhh wdi did hhh hdi ddi dh were synthesized by NAPS GmbH, Gόttingen. Dose: 5nmol/mouse
Experimental groups (5 mice per group)
1. TRP-2
2. TRP-2 + pR 60
3.. TRP-2 + CpG 1668 '4. TRP-2 + wdi
5. TRP-2 + wdidin
6. TRP-2 + wdid
7. TRP-2 + wdidid
8. TRP-2 + CpG 1668 + pR 60.
9. TRP-2 + wdi + pR 60 10.TRP-2 + wdidin + pR 60 ll.TRP-2 + wdid + pR 60 12.TRP-2 + wdidid + pR 60
On day 0 mice were injected into each hind footpad with a total volume of lOOμl (50μl per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70μm cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3xl06cells/ml in DMEM/5%FCS. An IFN-g ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995). This method is a widely used procedure allowing the quantification of antigen-specific T cells. Lymphocytes were stimulated ex vivo in triplicates with medium (background) , TRP-2-peptide, an irrelevant 0VA 257_264 _Pep- tide, pR 60 and Concanavalin A (Con A) . Spots representing single IFN-g producing T cells were counted and the number of background spots was substracted from all samples. The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO6 cells are illustrated in Figure 5, the standard deviation of ex vivo-stimu- lated triplicates are given. Example 4
The combined injection of I-ODN and poly-L-arginine (pR 60) syn- ergistically enhances the immune response against a Melanoma-derived peptide.
Experimental groups (5 mice per group)
Figure imgf000026_0001
3. TRP-2i8i-i88 + CpG 1668
4. TRP-2i8i-i88 + I-ODN 2
5. TRP-2i8i-i88 + CpG 1668 + pR 60
6. TRP-2i8i-i88 + I-ODN 2 + pR 60
On day 0 mice were injected into each hind footpad with a total volume of 100 μl (50 μl per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 μm cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals) . Cells were adjusted to 3x10 cells/ml in DMEM/5%/FCS. An IFN-γ ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995). This method is a widely used procedure allowing the quantification of antigen-specific T cells. Lymphocytes were stimulated ex vivo in triplicates with medium background-control, TRP-2i8i-i88-peptide, an irrelevant OVA257_254~peptide and Concanavalin A (Con A) . Spots representing single IFN-γ producing T cells were counted and the number of background spots was substracted from all samples . The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes . For each experimental group of mice the number of spots/lxlO cells are illustrated in Figure 6, the standard deviation of ex vivo- stimulated triplicates are given.
One hour after injection blood was taken from the tail vein and serum was prepared to determine the induction of systemic TNF-α and IL-6 using specific ELISAs (Figure 7) . Example 5
The combined injection of random 10-mer I-ODN and poly-L-arginine (pR 60) synergistically enhances the immune response against a Melanoma-derived peptide.
Experimental groups (5 mice per group)
Figure imgf000027_0001
3. TRP-2i8i-i88 + CpG 1668
Figure imgf000027_0002
5. TRP-2ιsι-i88 + CpG 1668 + pR 60
6. TRP-2i8i-i88 + ODN 17 + pR 60
On day 0 mice were injected into each hind footpad with a total volume of 100 μl (50 μl per footpad) containing the above mentioned compounds. Animals were sacrificed 4 days after injection and popliteal lymph nodes were harvested. Lymph nodes were passed through a 70 μm cell strainer and washed twice with DMEM medium (GIBCO BRL) containing 5% fetal calf serum (FCS, SIGMA chemicals). Cells were adjusted to 3xl06cells/ml in DMEM/5%/FCS. An IFN-γ ELISPOT assay was carried out in triplicates as described (Miyahira et al . , 1995) . This method is a widely used procedure allowing the quantification of antigen-specific T cells . Lymphocytes were stimulated ex vivo in triplicates with medium background-control, TRP-2ι8ι-i88-peptide, an irrelevant OVA257_2g4-peptide and Concanavalin A (Con A) . Spots representing single IFN-γ producing T cells were counted and the number of background spots was substracted from all samples . The high number of spots detected after the stimulation with Con A (data not shown) indicate a good condition of the used lymphocytes. For each experimental group of mice the number of spots/lxl0D cells are illustrated in Figure 8, the standard deviation of ex vivo- stimulated triplicates are given. Mice C57B1/6 (Harlan/Olac) Peptide TRP-2-peptide (VYDFFVWL) , a MHC class I (H-2Kb) -restricted epi- tope of mouse tyrosinase related protein-2 (Bllom et al . , 1997) was synthesized by standard solid phase F-moc synthesis, HPLC purified and analyzed by mass spec- troscopy for purity. Dose: lOOμg/mouse
Poly-L-arginine60 (pR60] Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals
Dose: lOOμg/mouse
CpG-ODN 1668 thiophosphate substituted ODNs containing a CpG motif: tec atg acg ttc ctg atg ct, were synthesized by NAPS GmbH, Gόttingen.
Dose: 5 nmol/mouse
ODN 17 thiophosphate substituted ODNs containing deoxyinosine: hhh wdi dhh h, were synthesized by NAPS GmbH, Gδttingen. (h = CAT, w = AT, d = GAT) Dose: 10 nmol/mouse
Mice C57B1/6 (Harlan/Olac) Peptide TRP-2-peptide (VYDFFVWL) , a MHC class I (H-2K-b) -restricted epi- tope of mouse tyrosinase related protein-2 (Bllom et al . , 1997) was synthesized by standard solid phase F-moc synthesis, HPLC purified and analyzed by mass spec- troscopy for purity. Dose: lOOμg/mouse Poly-L-arginine60 (pR60) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA chemicals
Dose: lOOμg/mouse
CpG-ODN 1668 thiophosphate substituted ODNs containing a CpG motif: tec atg acg ttc ctg atg ct, were synthesized by NAPS GmbH, Gόttingen.
Dose: 5 nmol/mouse
I-ODN 2 thiophosphate substituted ODNs containing deoxyinosine: tec atg aci ttc ctg atg ct, were synthesized by NAPS GmbH, Gδttingen. Dose: 5nmol/mouse
Example 6
The combined application of oligo-deoxyIC26-mer and poly-L-arginine (pR) enhances the ovalbumin (OVA) -specific humoral response.
Mice C57B1/6 (Harlan/Olac)
Ovalbumin (OVA) Ovalbumin from chicken egg, grade V, SIGMA Chemicals, A-5503, Lot 54H7070 Dose: 50 μg/mouse
Poly-L-arginine (pR) Poly-L-arginine with an average degree of polymerization of 60 arginine residues; SIGMA Chemicals, P-4663, Lot 68H5903 Dose: 100 μg/mouse
Oligo-deoxy IC, 26-mer oligo-dIC26-mer was synthesized by
(θligθ-dIC26-mer) standard phosphoamidide chemistry on a 4 μmol scale and purified by HPLC (NAPS Gόttingen, Germany) Dose: 5 nmol/mouse Experimental groups ( 4 mice per group)
1 . OVA + ol igo-dIC26-mer + pR
2 . OVA + oligo-dIC26-πιer
3 . OVA + pR
4 . OVA
On day 0, mice were injected into each hind footpad with a total volume of lOOμl (50μl per footpad) containing the above listed compounds. On day 24 after injection, serum was collected and screened by ELISA for the presence of OVA-specific antibodies. These results show that the injection of OVA in combination with oligo-dIC and pR enhanced the production of OVA-specific IgG antibodies when compared with injection of OVA with each of the substances alone (Figure 13A, B) . Interestingly, titers of both IgG2a and IgGl were increased upon one single injection of OVA with oligo-dlC/pR, implying that both Thl and Th2 cells were involved. However, after 115 days only the increased IgG2a levels were still detectable in sera of mice injected with OVA and oligo-dlC/pR.
These data demonstrate that the combined injection of OVA with oligo-dIC and pR enhances the OVA-specific humoral response. This response is characterized by the production of both Thl- and Th2- induced antibody isotypes in the early phase, but later, mainly by Thl-induced antibodies .
References
Andreu, D., and Rivas, L. (1998). Animal antimicrobial peptides: an overview. Biopolymers 47, 415-433.
Ballas, Z. K., Ras ussen, W. L., and Krieg, A. M. (1996). Induction of NK activity in urine and human cells by CpG motif in oligodeoxynucleotides and bacterial DNA. J Immunol 157, 1840- 1845.
Bloom, B. R., and Widdus, R. (1998). Vaccine visions and their global impact. Nat Med 4, 480-484.
Bloom, M. B., Perry-Lalley, D., Robbins, P. F., Li, Y. , el-Gamil, M. , Rosenberg, S. A., and Yang, J. C. (1997). Identification of tyrosinase-related protein 2 as a tumor rejection antigen for the B 16 melanoma. J Exp Med 185, 453-459.
Buschle, M., Schmidt, W. , Berger, M. , Schaffner, G., Kurzbauer, R. , Killisch, 1., Tiedemarm, J.K., Trska, B., Kirlappos, H., Mechtler, K., Schilcher, F., Gabler, C, and Birntsiel, M. L. (1998). Chemically defined, cell-free cancer vaccines: use of tumor antigen-derived peptides or polyepitope proteins for vaccination. Gene Ther. Mol. Biol . 1, 309-321
Buschle, M. , Schmidt, W. , Zauner, W. , Mechtler, K. , Trska, B., Kirlappos, H., and Birnstiel, M.L. (1997). Transloading of tumor antigen-derived peptides into antigen-presenting cells. Proc. Natl. Acad. Sci . USA 94, 3256-3261
Cavanaugh, P.F., Jr., Ho, Y-K, and Bardos, T.J. (1996). The activation of murine macrophages and natural killer cells by the Partially thiolated double stranded RNA poly (1) . mercapto poly(C) . Res .Comm.Mol .Pathol. Pharmacol. 91, 131-147
Chace, J. H. , Hooker, N. A., Mildenstein, K. L., Krieg, A. M. , and Cowdery, J. S. (1997). Bacterial DNA-induced NK cell IFN- gamma production is dependent on macrophage secretion of IL- 12. Clin Immunol Immunopathol 84, 185-193.
Davis, H. L., Weeranta, R. , Waldschmidt, T. J., Tygrett, L., Schorr, J. , and Krieg, A. M. (1998). CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen. J Immunol 160, 870-876.
Deng, G. M. , Nilsson, 1. M., Verdrengh, M. , Collins, L. V., and Tarkowski, A. (1999) . Intra-articularly localized bacterial DNA containing CpG motifs induces arthritis. Nat Med 5, 702-705.
Ganz, T. (1999) . Defensins and host defense [comment] . Science 286, 420-421.
Ganz, T., and Lehrer, R. 1. (1999). Antibiotic peptides from higher eukaryotes: biology and applications. Mol Med Today 5, 292-297.
Hancock, R. E. (1999). Host defence (cationic) peptides: what is their future clinical potential? Drugs 57, 469-473.
Harlow, E., and Lane, D. (1988). Antibodies: a laboratory manual (Cold Spring Harbor: Cold Spring Harbor Laboratory) .
Hartmann, G. , Weiner, G. J., and Krieg, A. M. (1999). CpG DNA: A potent signal for growth, activation, and maturation of human dendritic cells. Proc Natl Acad Sci U S A 96, 9305-9310.
Hoffmann, J. A., Kafatos, F. C, Janeway, C. A., and Ezekowitz, R. A. (1999) . Phylogenetic perspectives in innate immunity. Science 284, 1313-1318.
Klinman, D. M. , Yi, A. K., Beaucage, S. L., Conover, J. , and Krieg, A. M. (1996) . CpG motifs present in bacteria DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin 12, and interferon gamma. Proc Natl Acad Sci U S A 93, 2879-2883.
Krieg, A. M. (1999) . CpG DNA: a novel immunomodulator [letter] . Trends Microbiol 7, 64-5.
Krieg, A. M. (1996) . An innate immune defense mechanism based on the recognition of CpG motifs in microbial DNA. J Lab Clin Med 128, 128-133. Krieg, A. M. , Yi, A. K., Matson, S., Waldschmidt, T. J., Bishop, G. A., Teasdale, R. , Koretzky, G. A., and Klinman, D. M. (1995). CpG motifs in bacterial DNA trigger direct B-cell activation. Nature 374, 546-549.
Krieg, A. M. , Yi, A. K., Schorr, J. , and Davis, H. L. (1998). The role of CpG dinucleotides in DNA vaccines. Trends Microbiol 6, 23-27.
Lethe, B., van den Eynde, B., van Pel, A., Corradin, G. , and Boon, T. (1992) . Mouse tumor rejection antigens P815A and P815B: two epitopes carried by a single peptide. Eur J Immunol 22, 2283- 2288.
Liljeq ist, S.,' and Stahl, S. (1999). Production of recombinant subunit vaccines: protein immunogens, live delivery systems and nucleic acid vaccines. J Biotechnol 73, 1-33.
Lipford, G. B., Heeg, K. , and Wagner, H. (1998). Bacterial DNA as immune cell activator. Trends Microbiol 6, 496-500.
Manetti, R. , Annunziato, F., Tomasevic, L., Gianno, V., Parron- chi, P., Romagnani, S. and Maggi, E. (1995). Polyinosinic acid: polycytidylic acid promotes T helper type 1-specific immune responses by stimulating macrophage production of interferon-a and interleukin-12. Eur. J. Immunol. 25, 2656-2660
Mosmann, T. R. , Cherwinski, H., Bond, M. W. , Giedlin, M. A., and Coffman, R. L. (1986) . Two types of murine helper T cell clone. 1. Definition according to profiles of Iymphokine activities and secreted proteins. J Immunol 136, 2348-2357.
Nossal, G. (1998) . Living up to the legacy. Nat Med 4, 475-476
Oxenius, A., Martinic, M M. , Hengartner, H. , and Klenerman, P. (1999) . CpG-containing oligonucleotides are efficient adjuvants for induction of protective antiviral immune responses with T- cell peptide vaccines. J Virol 73, 4120-4126. Paillard, F. (1999) . CpG: the double-edged sword [comment] . Hum Gene Ther 10, 2089-2090.
Pamer, E. G., Harty, J. T., and Bevan, M. J. (1991). Precise prediction of a dominant class I MHC-restricted epitope of Listeria monocytogenes . Nature 353, 852-855.
Parronchi, P., Brugnolo, F., Annunziato, F., Manuelli, C, Sam- pognaro, S., Mavilia, C, Romagnani, S., and Maggi, E. (1999). Phosphorothioate oligodeoxynucleotides promote the in vitro development of human allergen-specific CD4+ T cells into Thl effectors. J Immunol 163. 5946-5953.
Pisetsky, D. S. (1997) . Immunostimulatory DNA: a clear and present danger? Nat Med 3, 829-831.
Pisetsky, D. S. (1999) . The influence of base sequence on the immunostimulatory properties of DNA. Immunol Res 19, 35-46.
Rammensee, H.G., Friede, T., Stevanoviic S. (1995), MHC ligands and peptide motifs: first listing. Immunogenetics 41, 178-228
Rodrigues, M. , Nussenzweig, R. S., Romero, P., and Zavala, F. (1992) . The in vivo cytotoxic activity of CD8+ T cell clones correlates with their levels of expression of adhesion molecules. J Exp Med 175, 895-905.
Roitt, 1., Brostoff, J., and Male, D. (1998). Immunology (London: Mosby International Ltd) .
Rotzschke, 0., Falk, K., Stevanovic, S., Jung, G. , Walden, P., and Rammensee, H. G. (1991) . Exact prediction of a natural T cell epitope. Eur J Immunol 21, 2891-2894.
Schmidt, W. , Buschle, M. , Zauner, W., Kirlappos, H. , Mechtler, K., Trska, B., and Bimstiel, M.L. (1997). Cell-free tumor antigen peptide-based cancer vaccines. Proc. Natl. Acad. Sci. USA 94, 3262-3267
Schwartz, D. A., Quinn, T. J., Thorne, P. S., Sayeed, S., Yi, A. K., and Krieg, A. M. (1997) . CpG motifs in bacterial DNA cause inflammation in the lower respiratory tract, J Clin Invest 100, 68-73.
Shimonkevitz, R., Colon, S., Kappler, J. W. , Marrack, P., and Grey, H. M. (1984) . Antigen recognition by H2-resctricted T cells 11. A tryptic ovalbumin peptide that substitutes for processed antigen. J Immunol 133, 2067-2074.
Simmaco, M. , Mignogna, G. , and Barra, D. (1998). Antimicrobial peptides from amphibian skin: what do they tell us? Biopolymers 47, 435-450.
Sparbier, K. , and Walden, P. (1999). T cell receptor specificity and mimotopes . Curr Opin Immunol 11, 214-218.
Sparwasser, T., Koch, E. S., Vabulas, R. M. , Heeg, K., Lipford, G. B., Ellwart, J. W. , and Wagner, H. (1998). Bacterial DNA and immunostimulatory CpG oligonucleotides trigger maturation and activation of murine dendritic cells. Eur J Immunol 28, 2045-2054.
Sparwasser, T., Miethke, T., Lipford, G., Borschert, K., Hacker, H., Heeg, K., and Wagner, H. (1997). Bacterial DNA causes septic shock [letter]. Nature 386, 336-337.
Sparwasser, T., Miethke, T., Lipford, G., Erdmann, A., Hacker, H., Heeg, K., and Wagner, H._ (1997). Macrophages sense pathogens via DNA mot)&: induction oftumor necrosis factor-alpha-mediated shock. EurJ Immunol 27, 1671-1679.
Weiner, G. J., Liu, H. M. , Wooldridge, J. E., Dahle, C. E., and Krieg, A. M. (1997) . Immunostimulatory oligodeoxynucleotides containing the CpG motif' are effective as immune adjuvants in tumor antigen immunization. Proc Natl Acad Sci U S A 94, 10833-10837.
Yew, N. S., Wang, K. X., Przybylska, M. , Bagley, R. G. , Stedman, M., Marshall, J., Scheule, R. K. , and Cheng, S. H. (1999). Contribution of plasmid DNA to inflammation in the lung after administration of cationic lipid:pDNA complexes. Hum Gene Ther 10, 223-234.

Claims

Claims :
1. Immunostimulatory oligodeoxynucleic acid molecule (ODN) having the structure according to formula (I)
Figure imgf000036_0001
(I) any X is 0 or S,
wherein any NMP is a 2 ' deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5-methyl-deoxycytosine-, deoxypseudouridine-, deoxyribosepurine-, 2-amino-deoxyribosepu- rine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopen- tenyl-deoxyadenosine-monophosphate or -monothiophosphat, NUC is a 2 ' deoxynucleoside, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2-methyl-deoxyinosine-, 5- methyl-deoxycytosine-, deoxypseudouridine-, eoxyribosepurine-, 2- amino-deoxyribosepurine-, 6-S-deoxyguanine-, 2-dimethyl-deoxyguanosine- or N-isopentenyl-deoxyadenosine, a and b are integers from 0 to 100 with the proviso that a + b is between 4 and 150,
B and E are common groups for 5' or 3 ' ends of nucleic acid molecules .
2. ODN according to claim 1, wherein any NMP is selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxyinosine-, deoxycytosine-, deoxyuridine-, deoxythymidine-, 2- methyl-deoxyinosine-, 5-methyl-deoxycytosine-monophosphate or -monothiophosphate .
3. ODN according to claim 1 or 2, characterized in that a + b is between 10 and 60, preferably between 15 and 40.
4. ODN according to any of claims 1 to 3, characterized in that at least one of X and 2 is S and at least one of X. and X4 is 0 and preferably any NMP is a nucleoside-monothiophosphate.
5. ODN according to any of claims 1 to 4, characterized in that it contains the sequence hhh wdi dhh h nhh hhh wdi nhh hhh hhh wn, i nhh wdi din hhh hdi ndi nh, nhh hhh wdi dhh hhh hhh wn or nhh wdi did hhh hdi ddi dh, wherein any n is a 2 '-deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine-, deoxycytosine- or deoxythymidine-monophosphate or -monothiophosphate, any h is a 2 '-deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxycytosine- or deoxythymidine-monophosphate or -monothiophosphate i is deoxyinosine-monophosphate or -monothiophosphate, any w is a 2 '-deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine- or deoxythymidine-monophosphate or -monothiophosphate, and any d is a 2 ' -deoxynucleoside monophosphate or monothiophosphate, selected from the group consisting of deoxyadenosine-, deoxyguanosine- or deoxythymidine-monophosphate or -monothiophosphate.
6. ODN according to any one of claims 1 to 5 , characterized in that it contains at least one 2 'deoxycytosine-monophosphate or -monothiophosphate 3 '-adjacent to a 2 '-deoxyinosine-monophosphate or -monothiophosphate.
7. ODN according to any one of claims 1 to 6, characterized in that it contains the sequence gacitt, iacitt, gaictt, iaictt, wherein a is deoxyadenosine- onophosphate or -monothiophosphate, g is deoxyguanosine-monophosphate or -monothiophosphate, i is deoxyinosine-monophosphate or -monothiophosphate, c is deoxycytosine-monophosphate or -monothiophosphate and t is deoxythymidine-monophosphate or -monothiophosphate.
8. ODN according to any one of claims 1 to 7, characterized in that it contains the sequence wdi , wdid, wdidin or, wdidid, wherein w,d, i and n are defined as above.
9. ODN according to any one of claims 1 to 8, characterized in that B and E are selected independently from the group consisting Of -H, -CH 3, -COH, -COCH3, -OH, -CHO, -PO4. -PSO3,, -PS2O2 , -PS3O,
-PS &„, -SO3, -PO4- (CH2) 1-6-NH2 or -PO4-(CH2)1-6-NH-Label .
10. Use of an ODN according to any one of claims 1 to 9 as a medicine, especially as an immunostimulatory agent.
11. Pharmaceutical composition comprising an ODN according to any one of claims 1 to 9.
12. Pharmaceutical composition comprising an ODN according to any one of claims 1 to 9 and an antigen.
13. Pharmaceutical composition according to claim 11 or 12, characterized in that it further comprises a polycationic polymer, preferably a polycationic peptide, especially polyarginine, polylysine or an antimicrobial peptide, especially a catheli- cidin-derived antimicrobial peptide, or a growth hormone, especially a human growth hormone.
14. Pharmaceutical composition according to any one of claims 11 to 13 further comprising further active ingredients, especially cytokines, antiinflammatory substances, antimicrobial substances or combinations thereof .
15. Pharmaceutical composition according to any one of claims 11 to 14, characterized in that it further contains auxiliary substances, especially a pharmaceutically acceptable carrier, buffer substances, stabilizers or combinations thereof.
16. Pharmaceutical composition according to any one of claims 11 to 15, characterized in that it contains 1 ng to 1 g, preferably 100 ng to 10 mg, especially 10 mg to 1 mg, of one or more ODNs according to any one of claims 1 to 9.
17. Use of an ODN according to any one of claims 1 to 9 for the preparation of a vaccine.
PCT/EP2001/006433 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides WO2001093905A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
JP2002501476A JP5271471B2 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
US10/297,555 US8568742B2 (en) 2000-06-08 2001-06-07 Methods and compositions involving immunostimulatory oligodeoxynucleotides
IL15295901A IL152959A0 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
EP20010960277 EP1296713B1 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
BRPI0111639A BRPI0111639B8 (en) 2000-06-08 2001-06-07 use of an immunostimulatory oligodeoxynucleic acid molecule and pharmaceutical composition.
AU8181201A AU8181201A (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
MXPA02012010A MXPA02012010A (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides.
AT01960277T ATE249839T1 (en) 2000-06-08 2001-06-07 IMMUNO-STIMULATING OLIGODEOXYNUCLEOTIDES
CA2411575A CA2411575C (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
SK1815-2002A SK287689B6 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
HU0301229A HU228264B1 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
DK01960277T DK1296713T3 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
SI200130047T SI1296713T1 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
AU2001281812A AU2001281812B2 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
PL358982A PL211036B1 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides
DE2001600814 DE60100814T2 (en) 2000-06-08 2001-06-07 IMMUNITIMULATING OLIGODEOXYNUCLEOTIDES
IS6627A IS1993B (en) 2000-06-08 2002-11-18 Immunostimulating phexoxia
IL152959A IL152959A (en) 2000-06-08 2002-11-20 Use of immunostimulatory oligodeoxynucleotides for the preparation of immunostimulatory pharmaceutical compositions
NO20025835A NO329492B1 (en) 2000-06-08 2002-12-04 Use of immunostimulatory oligodeoxynucleic acid molecules for the preparation of immunostimulatory, pharmaceutical compositions and pharmaceutical compositions containing the same.
HK03109011A HK1056678A1 (en) 2000-06-08 2003-12-11 Immunostimulatory oligodeoxynucleotides
US13/786,815 US8945591B2 (en) 2000-06-08 2013-03-06 Methods and compositions involving immunostimulatory oligodeoxynucleotides
US14/585,740 US9492537B2 (en) 2000-06-08 2014-12-30 Methods and compositions involving immunostimulatory oligodeoxynucleotides

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AT0100000A AT410173B (en) 2000-06-08 2000-06-08 ANTIQUE COMPOSITION
ATA1000/2000 2000-06-08
AT19732000 2000-11-23
ATA1973/2000 2000-11-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/297,555 A-371-Of-International US8568742B2 (en) 2000-06-08 2001-06-07 Methods and compositions involving immunostimulatory oligodeoxynucleotides
US13/786,815 Continuation US8945591B2 (en) 2000-06-08 2013-03-06 Methods and compositions involving immunostimulatory oligodeoxynucleotides

Publications (1)

Publication Number Publication Date
WO2001093905A1 true WO2001093905A1 (en) 2001-12-13

Family

ID=25608484

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/006433 WO2001093905A1 (en) 2000-06-08 2001-06-07 Immunostimulatory oligodeoxynucleotides

Country Status (26)

Country Link
US (3) US8568742B2 (en)
EP (1) EP1296713B1 (en)
JP (2) JP5271471B2 (en)
KR (1) KR100799788B1 (en)
CN (1) CN1309418C (en)
AT (1) ATE249839T1 (en)
AU (2) AU2001281812B2 (en)
BR (1) BRPI0111639B8 (en)
CA (1) CA2411575C (en)
CZ (1) CZ304195B6 (en)
DE (1) DE60100814T2 (en)
DK (1) DK1296713T3 (en)
ES (1) ES2206424T3 (en)
HK (1) HK1056678A1 (en)
HU (1) HU228264B1 (en)
IL (2) IL152959A0 (en)
IS (1) IS1993B (en)
MX (1) MXPA02012010A (en)
NO (1) NO329492B1 (en)
PL (1) PL211036B1 (en)
PT (1) PT1296713E (en)
RU (2) RU2293573C2 (en)
SI (1) SI1296713T1 (en)
SK (1) SK287689B6 (en)
TR (1) TR200302015T4 (en)
WO (1) WO2001093905A1 (en)

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002053184A2 (en) * 2001-01-05 2002-07-11 Intercell Ag Uses for polycationic compounds as vaccine adjuvants
WO2002053185A2 (en) * 2001-01-05 2002-07-11 Intercell Ag Anti-inflammatory use of polycationic compounds
WO2002069369A2 (en) * 2000-12-08 2002-09-06 Coley Pharmaceutical Gmbh Cpg-like nucleic acids and methods of use thereof
WO2002085283A2 (en) * 2001-04-25 2002-10-31 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Vaccine against tuberculosis
WO2002095027A2 (en) * 2001-05-21 2002-11-28 Intercell Ag Immunostimulatory oligodeoxynucleic molecules
WO2003035695A2 (en) * 2001-07-26 2003-05-01 Tanox, Inc. Agents that activate or inhibit toll-like receptor 9
WO2003047602A1 (en) * 2001-12-07 2003-06-12 Intercell Ag Immunostimulatory oligodeoxynucleotides
WO2004011650A2 (en) 2002-07-24 2004-02-05 Intercell Ag Antigens encoded by alternative reading frame from pathogenic viruses
WO2004084938A1 (en) 2003-03-24 2004-10-07 Intercell Ag Improved vaccines
WO2004084937A1 (en) * 2003-03-24 2004-10-07 Intercell Ag Use of alum and a th1 immune response inducing adjuvant for enhancing immune responses
JP2005530690A (en) * 2002-02-14 2005-10-13 ユナイテッド・バイオメディカル・インコーポレーテッド Stabilized synthetic immunogen delivery system
EP1616876A2 (en) 2001-01-26 2006-01-18 Intercell AG A method for identification, isolation and production of antigens to a specific pathogen
US7038029B2 (en) 2002-05-30 2006-05-02 Immunotech S.A. Immunostimulatory oligonucleotides and uses thereof
WO2006049454A1 (en) * 2004-11-05 2006-05-11 Bio Clue & Solution Co., Ltd Therapeutic use of cpg oligodeoxynucleotide for skin disease
US7244438B2 (en) 2001-01-05 2007-07-17 Intercell Ag Uses for polycationic compounds
US7378234B2 (en) 2002-09-13 2008-05-27 Intercell Ag Method for isolating hepatitis C virus peptides
WO2008135446A2 (en) * 2007-05-02 2008-11-13 Intercell Ag Klebsiella antigens
JP2009513542A (en) * 2003-07-11 2009-04-02 インターツェル・アクチェンゲゼルシャフト HCV vaccine
EP2053125A1 (en) 2003-03-04 2009-04-29 Intercell AG Streptococcus pyogenes antigens
US7635487B2 (en) 2003-04-15 2009-12-22 Intercell Ag S. pneumoniae antigens
EP2182065A2 (en) 2003-03-31 2010-05-05 Intercell AG Staphylococcus epidermidis antigens
WO2010089340A2 (en) 2009-02-05 2010-08-12 Intercell Ag Peptides protective against e. faecalis, methods and uses relating thereto
WO2010092176A2 (en) 2009-02-13 2010-08-19 Intercell Ag Nontypable haemophilus influenzae antigens
EP2267006A1 (en) 2003-05-30 2010-12-29 Intercell AG Enterococcus antigens
EP2266592A2 (en) 2007-01-12 2010-12-29 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2275435A2 (en) 2003-05-07 2011-01-19 Intercell AG Streptococcus agalactiae antigens I + II
EP2275434A1 (en) 2006-09-15 2011-01-19 Intercell AG Borrelia antigens
EP2287188A1 (en) 2006-07-07 2011-02-23 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2289544A2 (en) 2006-11-20 2011-03-02 Intercell AG Peptides protective against S. pneumoniae and compositions, methods and uses relating thereto
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
EP2308896A1 (en) 2009-10-09 2011-04-13 Sanofi-aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
WO2011042548A1 (en) 2009-10-09 2011-04-14 Sanofi-Aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
EP2314604A2 (en) 2002-10-15 2011-04-27 Intercell AG Nucleic acids coding for adhesion factors of group B streptococcus, adhesion factors of group B streptococcus and futher uses thereof
EP2319871A1 (en) 2009-11-05 2011-05-11 Sanofi-aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
WO2011101465A1 (en) 2010-02-19 2011-08-25 Intercell Ag Ic31 nanoparticles
EP2402024A1 (en) 2006-06-28 2012-01-04 Statens Serum Institut Expanding the t cell repertoire to include subdominant epitopes by vaccination with antigens delivered as protein fragments or peptide cocktails
WO2012020326A1 (en) 2010-03-18 2012-02-16 Novartis Ag Adjuvanted vaccines for serogroup b meningococcus
WO2012028741A1 (en) 2010-09-03 2012-03-08 Intercell Ag Isolated polypeptide of the toxin a and toxin b proteins of c. difficile and uses thereof
WO2012031760A1 (en) 2010-09-08 2012-03-15 Medigene Ag Parvovirus mutated structural proteins comprising cross - protective b - cell epitopes of a hpv l2 protein as well as products and methods relating thereto
US8241643B2 (en) 2008-03-17 2012-08-14 Intercell Ag Peptides protective against S. pneumoniae and compositions, methods and uses relating thereto
US8283328B2 (en) 2002-08-19 2012-10-09 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
EP2511291A2 (en) 2007-06-18 2012-10-17 Intercell AG Chlamydia antigens
US9624274B2 (en) 2007-05-31 2017-04-18 Medigene Ag Mutated structural protein of a parvovirus
US10226525B2 (en) 2015-05-04 2019-03-12 Pfizer Inc. Group B Streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
EP3527223A1 (en) 2018-02-16 2019-08-21 2A Pharma AB Mutated parvovirus structural protein
WO2019158636A1 (en) 2018-02-16 2019-08-22 2A Pharma Ab Parvovirus structural protein for the treatment of autoimmune diseases
US10392420B2 (en) 2012-12-20 2019-08-27 Pfizer Inc. Glycoconjugation process
US10408834B2 (en) 2007-05-31 2019-09-10 Medigene Ag Mutated parvovirus structural proteins as vaccines
US10751402B2 (en) 2016-11-09 2020-08-25 Pfizer Inc. Immunogenic compositions and uses thereof
WO2020234300A1 (en) 2019-05-20 2020-11-26 Valneva Se A subunit vaccine for treatment or prevention of a respiratory tract infection
WO2020240268A1 (en) 2019-05-31 2020-12-03 Universidad De Chile An immunogenic formulation that induces protection against shiga toxin-producing escherichia coli (stec)
WO2022074061A1 (en) 2020-10-07 2022-04-14 Valneva Sweden Ab Cholera vaccine formulation
WO2023083964A1 (en) 2021-11-11 2023-05-19 2A Pharma Ab Parvovirus structural protein against beta- and gamma-hpv
WO2023232901A1 (en) 2022-06-01 2023-12-07 Valneva Austria Gmbh Clostridium difficile vaccine

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6977245B2 (en) 1999-04-12 2005-12-20 The United States Of America As Represented By The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US8568742B2 (en) * 2000-06-08 2013-10-29 Valneva Austria Gmbh Methods and compositions involving immunostimulatory oligodeoxynucleotides
US7666674B2 (en) 2001-07-27 2010-02-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of sterically stabilized cationic liposomes to efficiently deliver CPG oligonucleotides in vivo
WO2003020884A2 (en) * 2001-08-14 2003-03-13 The Government Of The United States Of America As Represented By The Secretary Of Health And Human Services Method for rapid generation of mature dendritic cells
MX339524B (en) 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
US8466116B2 (en) 2001-12-20 2013-06-18 The Unites States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce epithelial cell growth
US7615227B2 (en) 2001-12-20 2009-11-10 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce angiogenesis
US7785608B2 (en) 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US8263091B2 (en) 2002-09-18 2012-09-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of treating and preventing infections in immunocompromised subjects with immunostimulatory CpG oligonucleotides
JP4817599B2 (en) * 2003-12-25 2011-11-16 独立行政法人科学技術振興機構 Immune activity enhancer and method for enhancing immune activity using the same
EP2471550A1 (en) 2005-10-07 2012-07-04 Health Protection Agency Proteins with improved solubility and methods for producing and using same
TW200806315A (en) 2006-04-26 2008-02-01 Wyeth Corp Novel formulations which stabilize and inhibit precipitation of immunogenic compositions
AR064642A1 (en) 2006-12-22 2009-04-15 Wyeth Corp POLINUCLEOTIDE VECTOR THAT INCLUDES IT RECOMBINATING CELL THAT UNDERSTANDS THE VECTOR POLYPEPTIDE, ANTIBODY, COMPOSITION THAT UNDERSTANDS THE POLINUCLEOTIDE, VECTOR, RECOMBINATING CELL POLYPEPTIDE OR ANTIBODY, USE OF THE COMPOSITION AND A COMPOSITION AND A METHOD
EP2650014A2 (en) 2008-06-20 2013-10-16 Wyeth LLC Compositions and methods of use of ORF1358 from beta-hemolytic streptococcal strains
PE20110065A1 (en) 2009-06-22 2011-01-31 Wyeth Llc COMPOSITIONS AND METHODS FOR PREPARING IMMUNOGENIC COMPOSITIONS OF STAPHYLOCOCCUS AUREUS SEROTYPES 5 AND 8 CAPSULAR POLYSACCHARIDE CONJUGATE
CN102481352A (en) 2009-06-22 2012-05-30 惠氏有限责任公司 Immunogenic compositions of staphylococcus aureus antigens
PT2575870T (en) 2010-06-04 2017-02-10 Wyeth Llc Vaccine formulations
PL3246044T3 (en) 2010-08-23 2021-08-23 Wyeth Llc Stable formulations of neisseria meningitidis rlp2086 antigens
ES2728282T3 (en) 2010-09-10 2019-10-23 Wyeth Llc Non-lipidated variants of ORF2086 antigens from Neisseria meningitidis
MX350170B (en) 2010-12-22 2017-08-28 Wyeth Llc Stable immunogenic compositions of staphylococcus aureus antigens.
PL2714908T3 (en) * 2011-05-26 2018-07-31 Intervet International B.V. Immunostimulatory oligodeoxynucleotides
RU2587633C2 (en) * 2011-05-26 2016-06-20 Интервет Интернэшнл Б.В. Immunostimulating oligodeoxynucleotides
ITMI20111182A1 (en) 2011-06-28 2012-12-29 Canio Buonavoglia VACCINE FOR CORONAVIRUS CANINO
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
EP3485906A1 (en) 2012-03-09 2019-05-22 Pfizer Inc Neisseria meningitidis compositions and methods thereof
WO2014136064A2 (en) 2013-03-08 2014-09-12 Pfizer Inc. Immunogenic fusion polypeptides
KR20210002757A (en) 2013-09-08 2021-01-08 화이자 인코포레이티드 Neisseria meningitidis compositions and methods thereof
WO2016130569A1 (en) 2015-02-09 2016-08-18 Mj Biologics, Inc. A composition comprising pedv antigens and methods for making and using the composition
CN106488929B (en) * 2014-05-09 2021-08-24 乌得勒支大学控股有限公司 Novel CATH2 derivatives
MX2017010705A (en) 2015-02-19 2017-12-04 Pfizer Neisseria meningitidis compositions and methods thereof.
IL303108B1 (en) 2017-01-31 2024-03-01 Pfizer Neisseria meningitidis compositions and methods thereof
WO2021211279A1 (en) 2020-04-17 2021-10-21 Regents Of The University Of Minnesota SARS-CoV-2 SPIKE RECEPTOR BINDING DOMAIN AND COMPOSITIONS AND METHODS THEREOF
AU2021332183A1 (en) 2020-08-26 2023-03-02 Pfizer Inc. Group B streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
WO2024069420A2 (en) 2022-09-29 2024-04-04 Pfizer Inc. Immunogenic compositions comprising an rsv f protein trimer

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3906092A (en) * 1971-11-26 1975-09-16 Merck & Co Inc Stimulation of antibody response
WO1990014424A1 (en) * 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1992011389A1 (en) * 1990-12-21 1992-07-09 F. Hoffmann-La Roche Ag Hla dqbeta dna typing
US5691136A (en) * 1991-10-23 1997-11-25 Baylor College Of Medicine Fingerprinting bacterial strains using repetitive DNA sequence amplification
WO1998016247A1 (en) * 1996-10-11 1998-04-23 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3725545A (en) 1971-02-03 1973-04-03 R Maes Enhancement of antibody production by nucleic acid-polycation complexes
AU635339B2 (en) 1989-10-11 1993-03-18 Merrell Dow Pharmaceuticals Inc. Inosine/guanosine derivatives as immunosuppressive agents
US5514577A (en) 1990-02-26 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide therapies for modulating the effects of herpes viruses
US5098437A (en) * 1991-02-13 1992-03-24 Pfizer Hospital Products Group, Inc. Acetabular cup positioning insert
US5646262A (en) * 1994-07-28 1997-07-08 Georgetown University Antisense oligonucleotides against hepatitis B viral replication
US20020081577A1 (en) * 1995-06-06 2002-06-27 Robert L. Kilkuskie Oligonucleotides speciific for hepatitis c virus
WO1996039535A1 (en) * 1995-06-06 1996-12-12 Jan Vijg Method of and apparatus for diagnostic dna testing
ZA964446B (en) * 1995-06-06 1996-12-06 Hoffmann La Roche Oligonucleotides specific for hepatitis c virus
WO1998014611A2 (en) 1996-10-02 1998-04-09 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Detection and identification of non-polio enteroviruses
AU753172B2 (en) 1997-06-06 2002-10-10 Dynavax Technologies Corporation Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
US5955443A (en) * 1998-03-19 1999-09-21 Isis Pharmaceuticals Inc. Antisense modulation of PECAM-1
US6001651A (en) * 1998-03-20 1999-12-14 Isis Pharmaceuticals Inc. Antisense modulation of LFA-3
IL126919A0 (en) * 1998-11-05 1999-09-22 Univ Ben Gurion Antisense oligomer
AU765940B2 (en) * 1998-06-24 2003-10-02 Innogenetics N.V. Particles of HCV envelope proteins: use for vaccination
EP1700603A3 (en) 1999-09-25 2007-06-13 Coley Pharmaceutical GmbH Immunostimulatory nucleic acids
CA2407942A1 (en) * 2000-05-01 2001-11-08 Hybridon, Inc. Modulation of oligonucleotide cpg-mediated immune stimulation by positional modification of nucleosides
AT410173B (en) * 2000-06-08 2003-02-25 Cistem Biotechnologies Gmbh ANTIQUE COMPOSITION
US8568742B2 (en) * 2000-06-08 2013-10-29 Valneva Austria Gmbh Methods and compositions involving immunostimulatory oligodeoxynucleotides
AU2004224746B2 (en) * 2003-03-24 2009-04-23 Valneva Austria Gmbh Improved vaccines
US7951845B2 (en) * 2006-01-19 2011-05-31 The Regents Of The University Of Michigan Composition and method of treating hearing loss

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3906092A (en) * 1971-11-26 1975-09-16 Merck & Co Inc Stimulation of antibody response
WO1990014424A1 (en) * 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1992011389A1 (en) * 1990-12-21 1992-07-09 F. Hoffmann-La Roche Ag Hla dqbeta dna typing
US5691136A (en) * 1991-10-23 1997-11-25 Baylor College Of Medicine Fingerprinting bacterial strains using repetitive DNA sequence amplification
WO1998016247A1 (en) * 1996-10-11 1998-04-23 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GAMPER, H.B. ET AL.: "G/C-modified oligodeoxynucleotides with selective complementarity: synthesis and hybridization properties", NUCLEIC ACIDS RES, vol. 24, no. 13, 1996, pages 2470 - 5, XP002180912 *
NAOYUKI MIURA ET AL: "USE OF THE DEOXYINOSINE-CONTAINING PROBE TO ISOLATE AND SEQUENCE CDNA ENCODING THE FUSION (F) GLYCOPROTEIN OF SENDAL VIRUS (HVJ)", GENE, ELSEVIER BIOMEDICAL PRESS. AMSTERDAM, NL, vol. 38, 1985, pages 271 - 274, XP000650050, ISSN: 0378-1119 *

Cited By (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002069369A2 (en) * 2000-12-08 2002-09-06 Coley Pharmaceutical Gmbh Cpg-like nucleic acids and methods of use thereof
EP1985702A3 (en) * 2000-12-08 2010-08-18 Coley Pharmaceutical GmbH CPG-like nucleic acids and methods of use thereof
WO2002069369A3 (en) * 2000-12-08 2003-06-26 Coley Pharm Gmbh Cpg-like nucleic acids and methods of use thereof
WO2002053185A2 (en) * 2001-01-05 2002-07-11 Intercell Ag Anti-inflammatory use of polycationic compounds
WO2002053184A3 (en) * 2001-01-05 2002-09-19 Intercell Biomedizinische Forschungs & Entwicklungs Gmbh Uses for polycationic compounds as vaccine adjuvants
WO2002053185A3 (en) * 2001-01-05 2002-10-31 Intercell Biomedizinische Forschungs & Entwicklungs Gmbh Anti-inflammatory use of polycationic compounds
US7244438B2 (en) 2001-01-05 2007-07-17 Intercell Ag Uses for polycationic compounds
WO2002053184A2 (en) * 2001-01-05 2002-07-11 Intercell Ag Uses for polycationic compounds as vaccine adjuvants
EP2412722A1 (en) 2001-01-26 2012-02-01 Intercell AG A method for identification, isolation and production of antigens to a specific pathogen
EP1616876A2 (en) 2001-01-26 2006-01-18 Intercell AG A method for identification, isolation and production of antigens to a specific pathogen
EP2322539A2 (en) 2001-01-26 2011-05-18 Intercell AG A method for identification, isolation and production of antigens to a specific pathogen
EP2319861A1 (en) 2001-01-26 2011-05-11 Intercell AG A method for identification, isolation and production of antigens to a specific pathogen
WO2002085283A2 (en) * 2001-04-25 2002-10-31 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Vaccine against tuberculosis
WO2002085283A3 (en) * 2001-04-25 2003-05-08 Intercell Biomedizinische Forschungs & Entwicklungs Gmbh Vaccine against tuberculosis
WO2002095027A3 (en) * 2001-05-21 2003-08-21 Intercell Ag Immunostimulatory oligodeoxynucleic molecules
US7858588B2 (en) 2001-05-21 2010-12-28 Intercell Ag Immunostimulatory oligodeoxynucleic molecules
WO2002095027A2 (en) * 2001-05-21 2002-11-28 Intercell Ag Immunostimulatory oligodeoxynucleic molecules
WO2003035695A3 (en) * 2001-07-26 2003-11-13 Tanox Inc Agents that activate or inhibit toll-like receptor 9
WO2003035695A2 (en) * 2001-07-26 2003-05-01 Tanox, Inc. Agents that activate or inhibit toll-like receptor 9
WO2003047602A1 (en) * 2001-12-07 2003-06-12 Intercell Ag Immunostimulatory oligodeoxynucleotides
JP2005530690A (en) * 2002-02-14 2005-10-13 ユナイテッド・バイオメディカル・インコーポレーテッド Stabilized synthetic immunogen delivery system
JP2010265291A (en) * 2002-02-14 2010-11-25 United Biomedical Inc Stabilized synthetic immunogen delivery system
US8084015B2 (en) 2002-02-14 2011-12-27 United Biomedical, Inc. Stabilized synthetic immunogen delivery system
US7038029B2 (en) 2002-05-30 2006-05-02 Immunotech S.A. Immunostimulatory oligonucleotides and uses thereof
US7943316B2 (en) 2002-05-30 2011-05-17 David Horn, Llc Immunostimulatory oligonucleotides and uses thereof
US7381807B2 (en) 2002-05-30 2008-06-03 Immunotech S.A. Immunostimulatory oligonucleotides and uses thereof
WO2004011650A2 (en) 2002-07-24 2004-02-05 Intercell Ag Antigens encoded by alternative reading frame from pathogenic viruses
US7528223B2 (en) 2002-07-24 2009-05-05 Intercell Ag Antigens encoded by alternative reading frames from pathogenic viruses
US8304396B2 (en) 2002-08-19 2012-11-06 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
US8283328B2 (en) 2002-08-19 2012-10-09 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
EP2402026A2 (en) 2002-09-13 2012-01-04 Intercell AG Method for isolating hepatitis C virus peptides
US7378234B2 (en) 2002-09-13 2008-05-27 Intercell Ag Method for isolating hepatitis C virus peptides
EP2314604A2 (en) 2002-10-15 2011-04-27 Intercell AG Nucleic acids coding for adhesion factors of group B streptococcus, adhesion factors of group B streptococcus and futher uses thereof
EP2314603A2 (en) 2002-10-15 2011-04-27 Intercell AG Nucleic acids coding for adhesion factors of group B streptococcus, adhesion factors of group B streptococcus and futher uses thereof
EP2287311A1 (en) 2003-03-04 2011-02-23 Intercell AG Streptococcus pyogenes antigens
EP2287314A1 (en) 2003-03-04 2011-02-23 Intercell AG Streptococcus pyogenes antigens
EP2287315A1 (en) 2003-03-04 2011-02-23 Intercell AG Streptococcus pyogenes antigens
US7638136B2 (en) 2003-03-04 2009-12-29 Intercell Ag Streptococcus pyogene antigens
EP2287313A1 (en) 2003-03-04 2011-02-23 Intercell AG Streptococcus pyogenes antigens
EP2287312A1 (en) 2003-03-04 2011-02-23 Intercell AG Streptococcus pyogenes antigens
EP2290082A1 (en) 2003-03-04 2011-03-02 Intercell AG Streptococcus pyogenes antigens
EP2053125A1 (en) 2003-03-04 2009-04-29 Intercell AG Streptococcus pyogenes antigens
EP2345420A1 (en) 2003-03-24 2011-07-20 Intercell AG Use of a TH1 immune response inducing adjuvant for enhancing immune responses
WO2004084938A1 (en) 2003-03-24 2004-10-07 Intercell Ag Improved vaccines
JP4676426B2 (en) * 2003-03-24 2011-04-27 インターツェル・アクチェンゲゼルシャフト Improved vaccine
WO2004084937A1 (en) * 2003-03-24 2004-10-07 Intercell Ag Use of alum and a th1 immune response inducing adjuvant for enhancing immune responses
US8784837B2 (en) 2003-03-24 2014-07-22 Valneva Austria Gmbh Vaccines comprising an immunostimulatory peptide and an immunostimulatory oligodeoxynucleic acid molecule
AU2004224746B2 (en) * 2003-03-24 2009-04-23 Valneva Austria Gmbh Improved vaccines
JP2006521320A (en) * 2003-03-24 2006-09-21 インターツェル・アクチェンゲゼルシャフト Improved vaccine
US7704514B2 (en) 2003-03-24 2010-04-27 Intercell Ag Vaccines
AU2009202420B2 (en) * 2003-03-24 2012-04-26 Valneva Austria Gmbh Improved Vaccines
JP2011042678A (en) * 2003-03-24 2011-03-03 Intercell Ag Improved vaccine
EP2182065A2 (en) 2003-03-31 2010-05-05 Intercell AG Staphylococcus epidermidis antigens
EP2311990A1 (en) 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
EP2311988A1 (en) 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
EP2333114A1 (en) 2003-04-15 2011-06-15 Intercell AG S. pneumoniae antigens
EP2336357A1 (en) 2003-04-15 2011-06-22 Intercell AG S. pneumoniae antigens
EP2314718A1 (en) 2003-04-15 2011-04-27 Intercell AG S. pneumoniae antigens
US8372411B2 (en) 2003-04-15 2013-02-12 Intercell Ag S. pneumoniae antigens
EP2314719A1 (en) 2003-04-15 2011-04-27 Intercell AG S. pneumoniae antigens
EP2311991A1 (en) 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
EP2311987A1 (en) 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
EP2311989A1 (en) 2003-04-15 2011-04-20 Intercell AG S. pneumoniae antigens
US7635487B2 (en) 2003-04-15 2009-12-22 Intercell Ag S. pneumoniae antigens
EP2298934A1 (en) 2003-04-15 2011-03-23 Intercell AG S. pneumoniae antigens
EP2289907A2 (en) 2003-05-07 2011-03-02 Intercell AG Streptococcus agalactiae antigens I + II
EP2275435A2 (en) 2003-05-07 2011-01-19 Intercell AG Streptococcus agalactiae antigens I + II
EP2287177A2 (en) 2003-05-07 2011-02-23 Intercell AG Streptococcus agalactiae antigens I + II
EP2267006A1 (en) 2003-05-30 2010-12-29 Intercell AG Enterococcus antigens
EP2327720A1 (en) 2003-05-30 2011-06-01 Intercell AG Enterococcus antigens
JP2009513542A (en) * 2003-07-11 2009-04-02 インターツェル・アクチェンゲゼルシャフト HCV vaccine
JP4734241B2 (en) * 2003-07-11 2011-07-27 インターツェル・アクチェンゲゼルシャフト HCV vaccine
US7935678B2 (en) 2004-11-05 2011-05-03 Bio Clue & Solutions Co, Ltd. Therapeutic use of CPG oligodeoxynucleotide for skin disease
WO2006049454A1 (en) * 2004-11-05 2006-05-11 Bio Clue & Solution Co., Ltd Therapeutic use of cpg oligodeoxynucleotide for skin disease
EP2402024A1 (en) 2006-06-28 2012-01-04 Statens Serum Institut Expanding the t cell repertoire to include subdominant epitopes by vaccination with antigens delivered as protein fragments or peptide cocktails
EP2402023A1 (en) 2006-06-28 2012-01-04 Statens Serum Institut Expanding the t cell repertoire to include subdominant epitopes by vaccination with antigens delivered as protein fragments or peptide cocktails
EP2287188A1 (en) 2006-07-07 2011-02-23 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2292648A2 (en) 2006-07-07 2011-03-09 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2292646A2 (en) 2006-07-07 2011-03-09 Intercell AG Small Streptococcus pyogenes antigens and their use
US8529911B2 (en) 2006-07-07 2013-09-10 Intercell Austria Ag Small Streptococcus pyogenes antigens and their use
EP2292645A2 (en) 2006-07-07 2011-03-09 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2292647A2 (en) 2006-07-07 2011-03-09 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2292644A2 (en) 2006-07-07 2011-03-09 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2319862A1 (en) 2006-07-07 2011-05-11 Intercell AG Small Streptococcus pyogenes antigens and their use
EP2287189A1 (en) 2006-07-07 2011-02-23 Intercell AG Small Streptococcus pyogenes antigens and their use
US8129165B2 (en) 2006-09-15 2012-03-06 Intercell Ag Borrelia antigens
EP2275434A1 (en) 2006-09-15 2011-01-19 Intercell AG Borrelia antigens
EP2287176A1 (en) 2006-09-15 2011-02-23 Intercell AG Borrelia antigens
US8303961B2 (en) 2006-09-15 2012-11-06 Intercell Ag Borrelia antigens
EP2289906A1 (en) 2006-09-15 2011-03-02 Intercell AG Borrelia antigens
EP2289544A2 (en) 2006-11-20 2011-03-02 Intercell AG Peptides protective against S. pneumoniae and compositions, methods and uses relating thereto
EP2298342A2 (en) 2006-11-20 2011-03-23 Intercell AG Peptides protective against s. pneumoniae and compositions, methods and uses relating thereto
US8795690B2 (en) 2007-01-12 2014-08-05 Valneva Austria Gmbh Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2269625A2 (en) 2007-01-12 2011-01-05 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2269624A2 (en) 2007-01-12 2011-01-05 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
US8343510B2 (en) 2007-01-12 2013-01-01 Intercell Ag Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2269626A2 (en) 2007-01-12 2011-01-05 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2266591A2 (en) 2007-01-12 2010-12-29 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2266592A2 (en) 2007-01-12 2010-12-29 Intercell AG Protective proteins of S. agalactiae, combinations thereof and methods of using the same
EP2360177A1 (en) 2007-05-02 2011-08-24 Intercell AG Klebsiella antigens
EP2497779A1 (en) 2007-05-02 2012-09-12 Intercell AG Klebsiella antigens
EP2338902A1 (en) 2007-05-02 2011-06-29 Intercell AG Klebsiella antigens
WO2008135446A2 (en) * 2007-05-02 2008-11-13 Intercell Ag Klebsiella antigens
US8637052B2 (en) 2007-05-02 2014-01-28 Valneva Austria Gmbh Klebsiella antigens
WO2008135446A3 (en) * 2007-05-02 2009-03-12 Intercell Ag Klebsiella antigens
US8236326B2 (en) 2007-05-02 2012-08-07 Intercell Ag Klebsiella antigens
EP2489673A3 (en) * 2007-05-02 2012-09-12 Intercell AG Klebsiella antigens
EP2489673A2 (en) 2007-05-02 2012-08-22 Intercell AG Klebsiella antigens
US10408834B2 (en) 2007-05-31 2019-09-10 Medigene Ag Mutated parvovirus structural proteins as vaccines
US9624274B2 (en) 2007-05-31 2017-04-18 Medigene Ag Mutated structural protein of a parvovirus
US10822378B2 (en) 2007-05-31 2020-11-03 Medigene Ag Mutated structural protein of a parvovirus
EP2511291A2 (en) 2007-06-18 2012-10-17 Intercell AG Chlamydia antigens
US8241643B2 (en) 2008-03-17 2012-08-14 Intercell Ag Peptides protective against S. pneumoniae and compositions, methods and uses relating thereto
US8445001B2 (en) 2008-03-17 2013-05-21 Intercell Ag Peptides protective against S. pneumoniae and compositions, methods and uses relating thereto
WO2010089340A2 (en) 2009-02-05 2010-08-12 Intercell Ag Peptides protective against e. faecalis, methods and uses relating thereto
WO2010092176A2 (en) 2009-02-13 2010-08-19 Intercell Ag Nontypable haemophilus influenzae antigens
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
EP2308896A1 (en) 2009-10-09 2011-04-13 Sanofi-aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
WO2011042548A1 (en) 2009-10-09 2011-04-14 Sanofi-Aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
EP2319871A1 (en) 2009-11-05 2011-05-11 Sanofi-aventis Polypeptides for binding to the "receptor for advanced glycation endproducts" as well as compositions and methods involving the same
US8765148B2 (en) 2010-02-19 2014-07-01 Valneva Austria Gmbh 1C31 nanoparticles
WO2011101465A1 (en) 2010-02-19 2011-08-25 Intercell Ag Ic31 nanoparticles
US9248180B2 (en) 2010-02-19 2016-02-02 Valneva Austria Gmbh IC31 nanoparticles
WO2012020326A1 (en) 2010-03-18 2012-02-16 Novartis Ag Adjuvanted vaccines for serogroup b meningococcus
US10821166B2 (en) 2010-09-03 2020-11-03 Valneva Austria Gmbh Isolated polypeptide of the toxin A and toxin B proteins of C. difficile and uses thereof
EP3167899A1 (en) 2010-09-03 2017-05-17 Valneva Austria GmbH Isolated polypeptide of the toxin a and toxin b proteins of c.difficile and uses thereof
US11478540B2 (en) 2010-09-03 2022-10-25 Valneva Austria Gmbh Isolated polypeptide of the toxin A and toxin B proteins of C. difficile and uses thereof
US10357557B2 (en) 2010-09-03 2019-07-23 Valneva Austria Gmbh Isolated polypeptide of the toxin A and toxin B proteins of C. difficile and uses thereof
EP3895728A1 (en) 2010-09-03 2021-10-20 Valneva Austria GmbH Isolated polypeptide of the toxin a and toxin b proteins of c. difficile and uses thereof
US9598472B2 (en) 2010-09-03 2017-03-21 Valneva Austria Gmbh Isolated polypeptide of the toxin A and toxin B proteins of C. difficile and uses thereof
WO2012028741A1 (en) 2010-09-03 2012-03-08 Intercell Ag Isolated polypeptide of the toxin a and toxin b proteins of c. difficile and uses thereof
US11357844B2 (en) 2010-09-03 2022-06-14 Valneva Austria Gmbh Isolated polypeptide of the toxin A and toxin B proteins of C. difficile and uses thereof
WO2012031760A1 (en) 2010-09-08 2012-03-15 Medigene Ag Parvovirus mutated structural proteins comprising cross - protective b - cell epitopes of a hpv l2 protein as well as products and methods relating thereto
US10745438B2 (en) 2012-12-20 2020-08-18 Pfizer Inc. Glycoconjugation process
US10392420B2 (en) 2012-12-20 2019-08-27 Pfizer Inc. Glycoconjugation process
US11603384B2 (en) 2012-12-20 2023-03-14 Pfizer Inc. Glycoconjugation process
US11117928B2 (en) 2012-12-20 2021-09-14 Pfizer Inc. Glycoconjugation process
US10946086B2 (en) 2015-05-04 2021-03-16 Pfizer Inc. Group B Streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
US10226525B2 (en) 2015-05-04 2019-03-12 Pfizer Inc. Group B Streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
US10751402B2 (en) 2016-11-09 2020-08-25 Pfizer Inc. Immunogenic compositions and uses thereof
US11147865B2 (en) 2016-11-09 2021-10-19 Pfizer Inc. Immunogenic compositions and uses thereof
WO2019158636A1 (en) 2018-02-16 2019-08-22 2A Pharma Ab Parvovirus structural protein for the treatment of autoimmune diseases
EP3527223A1 (en) 2018-02-16 2019-08-21 2A Pharma AB Mutated parvovirus structural protein
WO2020234300A1 (en) 2019-05-20 2020-11-26 Valneva Se A subunit vaccine for treatment or prevention of a respiratory tract infection
WO2020240268A1 (en) 2019-05-31 2020-12-03 Universidad De Chile An immunogenic formulation that induces protection against shiga toxin-producing escherichia coli (stec)
WO2022074061A1 (en) 2020-10-07 2022-04-14 Valneva Sweden Ab Cholera vaccine formulation
WO2023083964A1 (en) 2021-11-11 2023-05-19 2A Pharma Ab Parvovirus structural protein against beta- and gamma-hpv
WO2023232901A1 (en) 2022-06-01 2023-12-07 Valneva Austria Gmbh Clostridium difficile vaccine

Also Published As

Publication number Publication date
US20150125488A1 (en) 2015-05-07
JP5271471B2 (en) 2013-08-21
US20030171321A1 (en) 2003-09-11
MXPA02012010A (en) 2005-04-22
BR0111639A (en) 2003-03-25
AU8181201A (en) 2001-12-17
JP2003535146A (en) 2003-11-25
HK1056678A1 (en) 2004-02-27
BRPI0111639B8 (en) 2021-05-25
NO20025835D0 (en) 2002-12-04
CZ304195B6 (en) 2013-12-27
KR100799788B1 (en) 2008-01-31
NO20025835L (en) 2002-12-04
CN1434723A (en) 2003-08-06
EP1296713B1 (en) 2003-09-17
ATE249839T1 (en) 2003-10-15
DK1296713T3 (en) 2004-01-26
KR20030032964A (en) 2003-04-26
CN1309418C (en) 2007-04-11
BRPI0111639B1 (en) 2017-04-11
HU228264B1 (en) 2013-02-28
NO329492B1 (en) 2010-11-01
HUP0301229A3 (en) 2010-01-28
IL152959A (en) 2008-04-13
RU2413520C2 (en) 2011-03-10
CZ20024168A3 (en) 2003-09-17
PL358982A1 (en) 2004-08-23
US8945591B2 (en) 2015-02-03
SI1296713T1 (en) 2004-02-29
PL211036B1 (en) 2012-04-30
CA2411575C (en) 2015-04-07
JP5908851B2 (en) 2016-04-26
ES2206424T3 (en) 2004-05-16
DE60100814D1 (en) 2003-10-23
PT1296713E (en) 2004-02-27
US8568742B2 (en) 2013-10-29
IL152959A0 (en) 2003-06-24
IS1993B (en) 2005-03-15
CA2411575A1 (en) 2001-12-13
DE60100814T2 (en) 2004-07-01
HUP0301229A2 (en) 2003-08-28
JP2013121962A (en) 2013-06-20
IS6627A (en) 2002-11-18
RU2293573C2 (en) 2007-02-20
SK18152002A3 (en) 2003-08-05
SK287689B6 (en) 2011-06-06
US9492537B2 (en) 2016-11-15
AU2001281812B2 (en) 2005-04-07
EP1296713A1 (en) 2003-04-02
US20130183339A1 (en) 2013-07-18
TR200302015T4 (en) 2004-01-21
RU2007103151A (en) 2008-08-20

Similar Documents

Publication Publication Date Title
US9492537B2 (en) Methods and compositions involving immunostimulatory oligodeoxynucleotides
US7858588B2 (en) Immunostimulatory oligodeoxynucleic molecules
AU2001281812A1 (en) Immunostimulatory oligodeoxynucleotides
AU784403B2 (en) Pharmaceutical composition for immunomodulation and preparation of vaccines comprising an antigen and an immunogenic oligodeoxynucleotide and a polycationic polymer as adjuvants
ZA200209479B (en) Immunostimulatory oligodeoxynucleotides.
AU2002320762A1 (en) Immunostimulatory oligodeoxynucleic molecules

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 152959

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2002/09479

Country of ref document: ZA

Ref document number: 2001281812

Country of ref document: AU

Ref document number: 200209479

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2001960277

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 522908

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: PA/A/2002/012010

Country of ref document: MX

Ref document number: 2411575

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2002 501476

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 018107974

Country of ref document: CN

Ref document number: 10297555

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1020027016711

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: PV2002-4168

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 18152002

Country of ref document: SK

ENP Entry into the national phase

Ref country code: RU

Ref document number: RU A

Ref document number: 2003100409

Country of ref document: RU

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2001960277

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1020027016711

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 2001960277

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2002-4168

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 522908

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 522908

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 2001281812

Country of ref document: AU