WO2004053104A2 - 5’ cpg nucleic acids and methods of use - Google Patents

5’ cpg nucleic acids and methods of use Download PDF

Info

Publication number
WO2004053104A2
WO2004053104A2 PCT/US2003/039775 US0339775W WO2004053104A2 WO 2004053104 A2 WO2004053104 A2 WO 2004053104A2 US 0339775 W US0339775 W US 0339775W WO 2004053104 A2 WO2004053104 A2 WO 2004053104A2
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
cancer
nucleotides
subject
cytosine
Prior art date
Application number
PCT/US2003/039775
Other languages
French (fr)
Other versions
WO2004053104A3 (en
Inventor
Arthur M. Krieg
Marion Jurk
Jorg Vollmer
Eugen Uhlmann
Original Assignee
Coley Pharmaceutical Group, Inc.
Coley Pharmaceutical Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Coley Pharmaceutical Group, Inc., Coley Pharmaceutical Gmbh filed Critical Coley Pharmaceutical Group, Inc.
Priority to AU2003300919A priority Critical patent/AU2003300919A1/en
Priority to CA002502015A priority patent/CA2502015A1/en
Priority to JP2005511961A priority patent/JP2006512927A/en
Priority to EP03813010A priority patent/EP1578954A4/en
Publication of WO2004053104A2 publication Critical patent/WO2004053104A2/en
Publication of WO2004053104A3 publication Critical patent/WO2004053104A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • 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

Definitions

  • the present invention relates generally to immunostimulatory nucleic acids, compositions thereof and methods of using the immunostimulatory nucleic acids.
  • Bacterial DNA has immune stimulatory effects to activate B cells and natural killer cells, but vertebrate DNA does not (Tokunaga, T., et al., 1988. Jpn. J. Cancer Res. 79:682-686; Tokunaga, T., et al, 1984, JNCI 12:955-962; Messina, J.P., et al., 1991, J. Immunol. 147:1759-1764; and reviewed in Krieg, 1998, hi: Applied Oligonucleotide Technology, CA. Stein and A.M. Krieg, (Eds.), John Wiley and Sons, Inc., New York, NY, pp. 431-448) and Krieg. A. M.
  • the immune stimulatory effects of bacterial DNA can be mimicked with synthetic oligodeoxynucleotides (ODN) containing these CpG motifs.
  • Such CpG ODN have highly stimulatory effects on human and murine leukocytes, inducing B cell proliferation; cytokine and immunoglobulin secretion; natural killer (NK) cell lytic activity and IFN- ⁇ secretion; and activation of dendritic cells (DCs) and other antigen presenting cells to express costimulatory molecules and secrete cytokines, especially the Thl-like cytokines that are important in promoting the development of Thl-like T cell responses.
  • DCs dendritic cells
  • CpG motif is generally used to refer to a hexamer motif in which the CpG dinucleotide is located at the center.
  • the invention involves the finding that specific sub-classes of CpG immunostimulatory oligonucleotides having a 5 'CpG are highly effective in mediating immune stimulatory effects.
  • CpG nucleic acids are useful therapeutically and prophylactically for stimulating the immune system to treat cancer, infectious diseases, allergy, asthma and other disorders and to help protect against opportunistic infections following cancer chemotherapy.
  • the strong yet balanced, cellular and humoral immune responses that result from CpG stimulation reflect the body's own natural defense system against invading pathogens and cancerous cells.
  • the invention is a composition comprising an oligonucleotide: 5'TCGX ⁇ X 2 N ⁇ 3', wherein Ni is 2-95 nucleotides and, when Xi is C or A, X 2 is A, T, or C (SEQ. ID NO.: 61); when X] is T, X 2 is A or G (SEQ. ID NO.: 62); and when Xi is G, X 2 is any nucleotide (SEQ. ID NO.: 63).
  • the invention in other aspects, relates to an oligonucleotide comprising
  • 5'TCGTN ⁇ 3' (SEQ. ID NO.: 64).
  • N is 3-96 nucleotides, but when Ni is 16 nucleotides Ni does not include a C, 2 (5'-CCCCCCCCC-3' SEQ. ID NO.: 65), and when Ni is 8 nucleotides N, is at least 50% C or 70% T (SEQ. ID NO.: 66).
  • an oligonucleotide comprising 5'TCGAN ⁇ 3' SEQ.
  • Ni is 3-96 nucleotides, but when Ni is 19 nucleotides Ni is at least 55% pyrimidine (SEQ. ID NO.: 68), and when Ni is 8 nucleotides Ni is at least 50% T or C (SEQ. ID NO.: 69).
  • an oligonucleotide comprising 5'TCGN]3' is provided.
  • Ni is 10-96 nucleotides, and the C content of the oligonucleotide is less than or equal to 60%, and the A content of the oligonucleotide is less than or equal to 30%.
  • an oligonucleotide that comprises 5'TYZN ⁇ 3'.
  • Ni is 4-97 nucleotides, and the oligonucleotide does not include an unmethylated CG motif.
  • Y is a cytosine or modified cystosine.
  • Z is a guanine or modified guanine.
  • Y is 5'methyl cytosine, 5-methyl- deoxycytosine, 5-methyl-deoxyisocytosine, 5-hydroxy-deoxycytosine, deoxyuridine, N4- ethyl-deoxycytosine, 2'-deoxyuridine, 5-fluoro-2'-dU, and dSpacer.
  • Z is 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, hypoxanthine, 2,6-diaminopurine, 2-aminopurine, purine, 8-substituted guanine such as
  • oligonucleotide formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide.
  • the oligonucleotide has one of the following structures: 5' ⁇ *C*G*A*G*G*A*C*T*T*C*T*C*T*C*T*C*A*G*G*T*T 3' (SEQ. ID NO.: 50) or 5' T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T* ⁇ * ⁇ * ⁇ 3.
  • SE Q_ ID N0 . 2 The * refers to a phosphorothioate linkage.
  • the oligonucleotide includes at least 1 modified internucleotide linkage. In other embodiments the oligonucleotide includes at least 50% modified internucleotide linkages. Optionally all internucleotide linkages of the oligonucleotide are modified.
  • the stabilized internucleotide linkage may be a phosphorothioate linkage.
  • the oligonucleotide is 20-100 nucleotides in length. In other embodiments it is 40 or less nucleotides in length.
  • Ni is free of unmethylated CG motifs.
  • Ni may be defined by N 2 N 3 , such that N 2 is 8-94 nucleotides, or in some embodiments 8-40 nucleotides, and N 3 is 2-5 pyrimidines.
  • N 3 is TTTTT, TTTT, TTT, or TT.
  • i may be at least 50% pyrimidine or at least 80% pyrimidine.
  • Ni is free of Poly-A and Poly-G sequences.
  • Ni is TN 2 and N 2 is 8-94 nucleotides.
  • the invention involves, in one aspect, the discovery that the 5' sequence of immunostimulatory nucleotides, their length and internucleotide linkage have specific influences on the cytokine profile of the induced immune response and that these discoveries can be used to design a subset of CpG immunostimulatory oligonucleotides that have improved immune stimulatory properties.
  • the preferred CpG immunostimulatory oligonucleotides fall within one of the following 6 general formulas: 5'-X ⁇ YRM ⁇ -3', 5'-X 2 CGM 2 -3', 5'-X 3 CGM 3 -3', 5'-X 4 CGM 4 -3', 5'-X 5 CGM 5 -3' and 5'- TTGM 6 -3'.
  • the ODN has the general formula 5 ' -Xj YRM . -3 ' , wherein Xi is a single nucleotide; Y is a cytosine or a modified cytosine; R is a guanine or a modified guanine; and Mj is a nucleic acid of 1-3 nucleotides.
  • the internucleotide linkages of the oligonucleotide are all stabilized phosphorothioate internucleotide linkages.
  • the internucleotide linkage between Y and R is a phosphodiester linkage in an Rp configuration.
  • the modified cytosine has a C5 substitution and/or the modified guanine has a C8 or C7 substitution.
  • the substituted or modified C or G is selected from the group consisting of 5-substituted cytosines (e.g.
  • N,N' -propyl ene cytosine or phenoxazine N,N' -propyl ene cytosine or phenoxazine
  • uracil and its derivatives e.g. 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4- thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil), thymine derivatives (e.g.
  • N2-methyl-guanine 5-amino-3-methyl-3H,6H-thiazolo[4,5- d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, adenine, substituted adenines (e.g. N6-methyl-adenine, 8-oxo-adenine) 8-substituted guanine (e.g. 8 -hydroxy guanine and 8-bromoguanine), and 6-thioguanine.
  • the base is substituted by a universal base (e.g.
  • the oligonucleotide is associated with a carrier linked to the 3' end of the oligonucleotide.
  • the carrier is selected from the group consisting of a microparticle, dendrimer, liposome, cationic complex, and antigen.
  • the ODN is administered to the subject along with an antigen.
  • the CpG immunostimulatory oligonucleotides are useful for treating subjects in combination with the administration of a therapeutic protocol to the subject. In some embodiments of the invention, the therapeutic protocol is surgery.
  • the oligonucleotide is not associated with a carrier. In other embodiments the oligonucleotide is in a multimerized complex.
  • the multimerized complex includes the oligonucleotide linked by a multimerization unit to a second oligonucleotide.
  • the second oligonucleotide may have the formula 5'-X ⁇ YRM ⁇ - 3'.
  • the immunostimulatory oligonucleotide of the invention has the general formula 5'-X 2 YRM 2 -3' with a multimerization unit linked to the 3' end of the oligonucleotide.
  • X 2 is a nucleic acid that consists of a single nucleotide, or a dinucleotide or a trinucleotide that does not comprise a CG dinucleotide.
  • Y is a cytosine or a modified cytosine.
  • R is a guanine or a modified guanine.
  • M 2 is a nucleic acid of 0- 27 nucleotides.
  • the immunostimulatory oligonucleotides have the following structures: 5'-TCG-3', 5'-TCGT-3', 5'-UCG-3', 5'-UCGT-3'.
  • M 2 is free of a CG dinucleotide.
  • X 2 is a single nucleotide, and X 2 is a pyrimidine.
  • the internucleotide linkages of the oligonucleotide are all stabilized phosphodiester internucleotide linkages.
  • the multimerization unit is a carrier selected from the group consisting of a microparticle, dendrimer, liposome, cationic complex, cholesterol and antigen. In other embodiments the multimerization unit is a linker between the 3' end of the oligonucleotide and a second oligonucleotide.
  • the ODN is administered to the subject along with an antigen.
  • the CpG immunostimulatory oligonucleotides are useful for treating subjects in combination with the administration of a therapeutic protocol to the subject.
  • the therapeutic protocol is surgery.
  • the immunostimulatory oligonucleotide has the general formula 5'-X CGM 3 -3', wherein X is a single nucleotide that does not comprise a CG dinucleotide; M 3 is a nucleic acid of 3-27 nucleotides that is free of a CG dinucleotide, and M has at least one of the following properties: is free of a TC dinucleotide, is at least 30% T nucleotides, consists of A, T, and G or is free of a CCTTCC hexamer having at least one modified internucleotide linkage.
  • M 3 has at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%>, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
  • the immunostimulatory oligonucleotide has the general formula
  • M 4 is a nucleic acid of 2-26 nucleotides that is free of a CG dinucleotide and it has at least one of the following properties: is free of a TG or a GT dinucleotide, is at least 38% T nucleotides or consists of A and T.
  • M 4 has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% A or T or modified versions thereof.
  • the immunostimulatory oligonucleotide has the general formula 5'-X 5 CGM 5 -3', X 5 is a trinucleotide that does not comprise a CG dinucleotide; M 5 is a nucleic acid of 1-25 nucleotides that is free of a CG dinucleotide, and wherein Ms has at least one of the following properties: is free of a CT dinucleotide and does not include at least one phosphorothioate linkage, is at least 41% T nucleotides, or consists of A and C. In some embodiments M 4 has at least 45%, 50%, 55%, 60%, 65%, 70%, 75%>, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
  • the immunostimulatory oligonucleotide has the general formula 5'-TTGM 6 -3', M 6 is a nucleic acid that consists of 5-21 nucleotides, wherein M 6 does not comprise a CG dinucleotide, wherein M 6 is comprised of at least 30% T nucleotides, and wherein said nucleotide is 10-24 nucleotides in length.
  • M 4 has at least 35%, 40%, 45%, 50%o, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
  • the oligonucleotide has one of the following structures:
  • An oligonucleotide comprising: 5 '-X 6 CGM 7 -3' is provided according to an aspect of the invention.
  • 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide.
  • X 6 is 1-3 nucleotides and does not include a CG dinucleotide.
  • M 7 is a nucleic acid of 6-27 nucleotides and includes at least three CG dinucleotides and is at least 50%) T nucleotides. In one embodiment M 7 is 16-18 nucleotides in length.
  • M includes at least four CG dinucleotides. In other embodiments at least one CG dinucleotide includes a phosphodiester internucleotide linkage. Optionally at least three CG dinucleotides includes a phosphodiester internucleotide linkage.
  • the oligonucleotide may be selected from the group consisting of SEQ ID NO. 33, 34, 35, 36, and 37.
  • the invention is an oligonucleotide comprising: 5'-'TTGM 8 -3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein M 7 is a nucleic acid of 6-18 nucleotides and includes at least one CG dinucleotide and is at least 50% T nucleotides.
  • M 8 is 14 nucleotides in length.
  • the immunostimulatory oligonucleotides generally have a length in the range of between 3 and 35 nucleotides. In some embodiments the length is in the range of 4-6, 3- 32, 6-30, or 10-24 nucleotides or any integer range therebetween.
  • the oligonucleotide includes at least 1 modified internucleotide linkage. In other embodiments the oligonucleotide includes at least 50% modified internucleotide linkages. Optionally all internucleotide linkages of the oligonucleotide are modified.
  • the stabilized internucleotide linkage may be a phosphorothioate linkage.
  • the invention in another aspect, relates to a method for treating allergy or asthma.
  • the method is performed by administering to a subject having or at risk of having allergy or asthma an immunostimulatory CpG oligonucleotide described herein in an effective amount to treat allergy or asthma.
  • the oligonucleotide is administered to a mucosal surface, such as a respiratory tissue.
  • the oligonucleotide is administered in an aerosol formulation.
  • the oligonucleotide is administered intranasally.
  • the subject has or is at risk of developing allergic asthma.
  • a method for inducing cytokine production is provided according to another aspect of the invention.
  • the method is performed by administering to a subject an immunostimulatory CpG oligonucleotide described herein in an effective amount to induce a cytokine selected from the group consisting of IP10, IL6, IL 8, IL12, IL18, TNF, IFN- ⁇ , chemokines, and IFN- ⁇ .
  • the invention is a composition of the CpG immunostimulatory oligonucleotides described herein in combination with an antigen or other therapeutic compound, such as an anti-microbial agent or an anti-cancer agent.
  • an antigen or other therapeutic compound such as an anti-microbial agent or an anti-cancer agent.
  • the anti-microbial agent may be, for instance, an anti-viral agent, an anti-parasitic agent, an anti-bacterial agent or an anti-fungal agent.
  • the composition may optionally include a pharmaceutical carrier and/or be formulated in a delivery device.
  • the delivery device is selected from the group consisting of cationic lipids, cell permeating proteins, and sustained release devices.
  • the sustained release device is a biodegradable polymer or a microparticle.
  • a method of stimulating an immune response involves administering a CpG immunostimulatory oligonucleotide to a subject in an amount effective to induce an immune response in the subject.
  • the CpG immunostimulatory oligonucleotide is administered orally, locally, in a sustained release device, mucosally, systemically, parenterally, or intramuscularly.
  • the CpG immunostimulatory oligonucleotide When the CpG immunostimulatory oligonucleotide is administered to the mucosal surface it may be delivered in an amount effective for inducing a mucosal immune response or a systemic immune response.
  • the mucosal surface is an oral, nasal, rectal, vaginal, or ocular surface.
  • the method includes exposing the subject to an antigen wherein the immune response is an antigen-specific immune response.
  • the antigen is selected from the group consisting of a tumor antigen, a viral antigen, a bacterial antigen, a parasitic antigen and a peptide antigen.
  • CpG immunostimulatory oligonucleotides are capable of provoking a broad spectrum of immune response. For instance these CpG immunostimulatory oligonucleotides can be used to redirect a Th2 to a Thl immune response. CpG immunostimulatory oligonucleotides may also be used to activate an immune cell, such as a lymphocyte (e.g., B and T cells), a dendritic cell, and an NK cell.
  • a lymphocyte e.g., B and T cells
  • a dendritic cell e.g., a dendritic cell
  • NK cell e.g., NK cell
  • the activation can be performed in vivo, in vitro, or ex vivo, i.e., by isolating an immune cell from the subject, contacting the immune cell with an effective amount to activate the immune cell of the CpG immunostimulatory oligonucleotide and re-administering the activated immune cell to the subject, hi some embodiments the dendritic cell presents a cancer antigen.
  • the dendritic cell can be exposed to the cancer antigen ex vivo.
  • the immune response produced by CpG immunostimulatory oligonucleotides may also result in induction of cytokine production, e.g., production of IP 10, IL6, IL 8, IL12, IL18, TNF, IFN- ⁇ , chemokines, and IFN- ⁇ .
  • cytokine production e.g., production of IP 10, IL6, IL 8, IL12, IL18, TNF, IFN- ⁇ , chemokines, and IFN- ⁇ .
  • the CpG immunostimulatory oligonucleotides are useful for treating cancer in a subject having or at risk of developing a cancer.
  • the cancer may be selected from the group consisting of biliary tract cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, gastric cancer, intraepithelial neoplasms, lymphomas, liver cancer, lung cancer (e.g. small cell and non-small cell), melanoma, neuroblastomas, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, sarcomas, thyroid cancer, and renal cancer, as well as other carcinomas and sarcomas.
  • the cancer is selected from the group consisting of bone cancer, brain and CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer.
  • CpG immunostimulatory oligonucleotides may also be used for increasing the responsiveness of a cancer cell to a cancer therapy (i.e., an anti-cancer therapy), optionally when the CpG immunostimulatory oligonucleotide is administered in conjunction with an anti-cancer therapy.
  • the anti-cancer therapy may be, for instance, a chemotherapy, a vaccine (e.g., an in vitro primed dendritic cell vaccine or a cancer antigen vaccine) or an immunotherapeutic agent such as an antibody based therapy.
  • This latter therapy may also involve administering an antibody specific for a cell surface antigen of, for example, a cancer cell, wherein the immune response results in antibody dependent cellular cytotoxicity (ADCC).
  • ADCC antibody dependent cellular cytotoxicity
  • the antibody may be selected from the group consisting of Ributaxin, Herceptin, Quadramet, Panorex, IDEC- Y2B8, BEC2, C225, Oncolym, SMART M195, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-11, MDX-22, OV103, 3622W94, anti-VEGF, Zenapax, MDX- 220, MDX-447, MELIMMUNE-2, MELIMMUNE-1, CEACIDE, Pretarget, NovoMAb- G2, TNT, Gliomab-H, GNI-250, EMD-72000, LymphoCide, CMA 676, Monopharm-C, 4B5, ior egf.r3, ior c5, BABS, anti-FLK-2, MDX-260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab and ImmuRAIT-CEA.
  • a subject having cancer or at risk of having a cancer is administered a CpG immunostimulatory oligonucleotide and an anti-cancer therapy.
  • the anti-cancer therapy is selected from the group consisting of a chemotherapeutic agent, an immunotherapeutic agent and a cancer vaccine.
  • the subject may be further administered interferon- ⁇ .
  • the invention is a method for inducing an innate immune response by administering to the subject a CpG immunostimulatory oligonucleotide in an amount effective for activating an innate immune response.
  • a method for treating a viral or retroviral infection is provided. The method involves administering to a subject having or at risk of having a viral or retroviral infection, an effective amount for treating the viral or retroviral infection of any of the compositions of the invention.
  • the virus is caused by a hepatitis virus e.g., hepatitis B, hepatitis C, HIV, herpes virus, or papillomavirus.
  • a method for treating a bacterial infection involves administering to a subject having or at risk of having a bacterial infection, an effective amount for treating the bacterial infection of any of the compositions of the invention.
  • the bacterial infection is due to an intracellular bacteria.
  • the invention is a method for treating a parasite infection by administering to a subject having or at risk of having a parasite infection, an effective amount for treating the parasite infection of any of the compositions of the invention.
  • the parasite infection is due to an intracellular parasite.
  • the parasite infection is due to a non-helminthic parasite.
  • the subject is a human and in other embodiments the subject is a non-human vertebrate such as a dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, or sheep.
  • the invention relates to a method for inducing a TH1 immune response by administering to a subject any of the compositions of the invention in an effective amount to produce a TH1 immune response.
  • Figure 1 is a bar graph depicting the effect of a 5'-TCG motif on the immunostimulatory activity of non-CpG or CpG ODNs through induction of IL-10.
  • Figure 2 is a bar graph depicting effect of a 5'-TCG motif on the immunostimulatory activity of non-CpG or CpG ODNs through induction of IFN- ⁇ .
  • Figure 3 is a bar graph depicting the effect of a 5'TCG on Poly-A and Poly-T sequences.
  • Figure 4 is a bar graph depicting the effect of shifting the CpG dinucleotide from the 5' to the 3' end of an ODN.
  • Figure 5 is a bar graph demonstrating that the length of an ODN has an effect on stimulatory activity in addition to a 5'-TCG.
  • Figure 6 is a bar graph depicting the effect of other 5' modifications in addition to 5'-TCG.
  • Figure 7 is a set of bar graphs depicting the effect of a 5' -TCG modification on stimulatory capability of CpG ODNs as shown by different cellular effects: 7A (IL-10 induction) 7B (IFN- ⁇ induction) and 7C (IL-6 induction).
  • 7A IL-10 induction
  • 7B IFN- ⁇ induction
  • 7C IL-6 induction
  • Figure 8 is a set of bar graphs that shows IL-10 secretion induced by ODN with 5' -TCG.
  • Figure 9 is a set of bar graphs that shows IL-10 secretion induced by ODN with 5'-TCG and increasing numbers of thymidines.
  • Figure 10 is set of bar graphs that depicts ODN's with a 5'-TCG as the most potent and efficient ODN's to induce a strong Thl-mediated immune response: 10A (IL- 10 induction) and 10B (IFN- ⁇ induction).
  • 10A IL- 10 induction
  • 10B IFN- ⁇ induction
  • Figure 11 is a set of bar graphs that depicts how the position of CpG dinucleotides in immune stimulatory ODN determines the strength of type I IFN secretion.
  • Figure 12 is a set of bar graphs that shows type I IFN secretion induced by short 5'-TCG ODN's.
  • Figure 13 is a set of bar graphs that shows the in vitro immune stimulation by a panel of newly generated CpG ODN's according to the observations described herein: 13A (IL-10 induction) and 13B (IFN- ⁇ induction).
  • Figure 14 is a set of bar graphs depicting B cell stimulation by short CpG ODN's.
  • Figure 15 is a bar graph that shows IL-10 induction by a panel of CpG ODN's and which demonstrates that some ODN having phosphodiester linkage between C and G have increased potency.
  • Figure 16 is a bar graph that shows IL-10 induction by a panel of CpG ODN's and which demonstrates that some ODN having a modified 5 ' TCG induce IL- 10.
  • the invention in one aspect involves the finding that specific sub-classes of CpG immunostimulatory oligonucleotides having a 5'TCG are highly effective in mediating immune stimulatory effects.
  • CpG nucleic acids are useful therapeutically and prophylactically for stimulating the immune system to treat cancer, infectious diseases, allergy, asthma and other disorders and to help protect against opportunistic infections following cancer chemotherapy.
  • the strong yet balanced, cellular and humoral immune responses that result from CpG stimulation reflect the body's own natural defense system against invading pathogens and cancerous cells.
  • the invention involves, in one aspect, the discovery that a subset of CpG immunostimulatory oligonucleotides have improved immune stimulatory properties.
  • the preferred CpG immunostimulatory oligonucleotides fall within one of the following 5 general formulas: 5'TCGX ⁇ X 2 Nj3', 5'TCGTN]3', 5'TCGAN ⁇ 3 ⁇ 5'TCGN ⁇ 3' and 5'TYZN ⁇ 3' (SEQ. ID NO.: 61-69).
  • Xi and X 2 refer to single nucleotides.
  • the formulas define subsets of the class of CpG oligonucleotides which demonstrated excellent immune stimulating properties and yet do not include additional unmethylated CpG motifs.
  • 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide.
  • Ni encompasses a variable set of nucleotide sequences.
  • the nucleotide sequences may range from 2-97 nucleotides in length or any integer range therebetween.
  • the findings of the invention are based in part on the discovery of the importance of the positional effects of CpG or YpZ motif. It has been discovered that oligonucleotides having a 5'TCG or 5' TYZ without any additional unmethylated CpG motifs therein are strong immunostimulatory capability. The remainder of the oligonucleotide may be any combination of nucleotides or modified nucleotides as long as the 5' end of the molecule includes the requisite motif.
  • N ]5 when combined with the 5'TCG or 5' TYZ produce molecules having even greater immunostimulatory activity.
  • N] when N] is at least 50% pyrimidine the oligonucleotide produces enhanced Thl biased immune induction.
  • Nj is at least 55%, 60%, 65%, 70%, 75%>, 80%, 85%), 90%, 95%, or 100 pyrimidine, e.g. C or T.
  • a pyrimidine is T or C or modified versions thereof.
  • the 3' most nucleotides of Ni are pyrimidines.
  • the 3' end may be TTTTT, TTTT, TTT, TT, T, CCCCC, CCCC, CCC, CC, C, CTT, CCTT, or any other possible combination of pyri idines.
  • Ni is free of a C. 2 (5'-CCCCCCCCCC-3 5 (SEQ. ID NO.: 65)).
  • the invention involves, in one aspect, the discovery that the 5' sequence of immunostimulatory nucleotides, their length and internucleotide linkage have specific influences on the cytokine profile of the induced immune response and that these discoveries can be used to design a subset of CpG immunostimulatory oligonucleotides that have improved immune stimulatory properties.
  • the preferred CpG immunostimulatory oligonucleotides fall within one of the following 6 general formulas: 5'-X ⁇ YRM ⁇ -3', 5'-X 2 CGM 2 -3', 5'-X 3 CGM 3 -3', 5'-X 4 CGM 4 -3', 5'-X 5 CGM 5 -3' and 5'- TTGMe-3'.
  • the formulas define subsets of the class of CpG oligonucleotides which demonstrated excellent immune stimulating properties and yet do not include additional unmethylated CpG motifs, hi the formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide.
  • Xi is a single nucleotide
  • Y is a cytosine or a modified cytosine
  • R is a guanine or a modified guanine
  • Mi is a nucleic acid of 1-3 nucleotides.
  • such a oligonucleotide can be
  • X 2 is a nucleic acid that consists of a single nucleotide, or a dinucleotide or a trinucleotide that does not comprise a CG dinucleotide; and M 2 is a nucleic acid of 0-27 nucleotides.
  • the oligonucleotides have the following structures: 5'-TCG-3', 5'- TCGT-3', 5'-UCG-3', 5'-UCGT-3'.
  • M 2 is free of a CG dinucleotide.
  • X 3 is a single nucleotide that does not comprise a CG dinucleotide; M 3 is a nucleic acid of 3-27 nucleotides that is free of a CG dinucleotide, and M 3 has at least one of the following properties: is free of a TC dinucleotide, is at least 30% T nucleotides, consists of A, T, and G or is free of a CCTTCC hexamer having at least one modified internucleotide linkage.
  • M 3 has at least 35%>, 40%, 45%, 50%, 55%, 60%>, 65%, 70%), 75%, 80%, 85%), 90%, 95%> or 100%) T or modified versions thereof.
  • X 4 is a dinucleotide that does not comprise a CG dinucleotide
  • M 4 is a nucleic acid of 2-26 nucleotides that is free of a CG dinucleotide and it has at least one of the following properties: is free of a TG or a GT dinucleotide, is at least 38% T nucleotides or consists of A and T.
  • M 4 has at least 40%, 45%>, 50%, 55%>, 60%, 65%>, 70%, 75%o, 80%, 85%, 90%>, 95% or 100% A or T or modified versions thereof.
  • X 5 is a trinucleotide that does not comprise a CG dinucleotide; Ms is a nucleic acid of 1-25 nucleotides that is free of a CG dinucleotide, and wherein Ms has at least one of the following properties: is free of a CT dinucleotide and does not include at least one phosphorothioate linkage, is at least 41%> T nucleotides, or consists of A and C. In some embodiments M 4 has at least 45%, 50%, 55%>, 60%>, 65%, 70%, 75%, 80%, 85%, 90%), 95% or 100%) T or modified versions thereof.
  • M 6 is a nucleic acid that consists of 5-21 nucleotides, wherein M 6 does not comprise a CG dinucleotide, wherein M 6 is comprised of at least 30%> T nucleotides, and wherein said nucleotide is 10-24 nucleotides in length.
  • M 4 has at least 35%>, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
  • the oligonucleotides may have one or two accessible 5' ends. Since the importance of the 5'TCG and 5'TYZ motif has been discovered, it also possible to create modified oligonucleotides having two such 5' ends. This may be achieved, for instance by attaching two oligonucleotides through a 3'-3' linkage to generate an oligonucleotide having one or two accessible 5' ends. Such a structure might have a formula such as 5 CGN ⁇ -N ⁇ GCT5' (SEQ. ID NO.: 13).
  • the 3'3'-linkage may be a phosphodiester, phosphorothioate or any other modified internucleoside bridge. Methods for accomplishing such linkages are known in the art.
  • 3 '3 '-linked ODNs where the linkage between the 3 '-terminal nucleosides is not a phosphodiester, phosphorothioate or other modified bridge, can be prepared using an additional spacer, such as tri- or tetra-ethylenglycol phosphate moiety (Durand, M. et al, Triple-helix formation by an oligonucleotide containing one (dA)12 and two (dT)12 sequences bridged by two hexaethylene glycol chains, Biochemistry (1992), 31(38), 9197-204, US Patent No. 5658738, and US Patent No. 5668265).
  • an additional spacer such as tri- or tetra-ethylenglycol phosphate moiety (Durand, M. et al, Triple-helix formation by an oligonucleotide containing one (dA)12 and two (dT)12 sequences bridged by two hexaethylene glycol chains
  • the non-nucleotidic linker may be derived from ethanediol, propanediol, or from an abasic deoxyribose (dSpacer) unit (Fontanel, Marie Laurence et al, Sterical recognition by T4 polynucleotide kinase of non-nucleosidic moieties 5'-attached to oligonucleotides; Nucleic Acids Research (1994), 22(11), 2022-7) using standard phosphoramidite chemistry.
  • the non-nucleotidic linkers can be incorporated once or multiple times, or combined with each other allowing for any desirable distance between the 3'-ends of the two ODNs to be linked.
  • the oligonucleotide in some embodiments does not include an unmethylated CG motif, other than the 5 'TCG.
  • the oligonucleotide has one of the following structures: 5' T*C*G*A*G*G*A*C*T*T*C*T*C*T*C*T*C*A*G*G*T*T (SEQ.
  • the immunostimulatory oligonucleotides generally have a length in the range of between 7 and 100 nucleotides. In some embodiments the length is in the range of 7-40, 13-100, 13-40, 13-30, 15-50, or 15- 30 nucleotides or any integer range therebetween.
  • the oligonucleotide is associated with a carrier linked to the 3' end by, but not limited to, the aforementioned linkers and methods.
  • the carrier can be selected from but not limited to the group consisting of microparticles, dendrimers, liposomes, cationic complexes and antigens.
  • nucleic acid and “oligonucleotide” are used interchangeably to mean multiple nucleotides (i.e., molecules comprising a sugar (e.g., ribose or deoxyribose) linked to a phosphate group and to an exchangeable organic base, which is either a substituted pyrimidine (e.g., cytosine (C), thymine (T) or uracil (U)) or a substituted purine (e.g., adenine (A) or guanine (G)).
  • cytosine C
  • T thymine
  • U uracil
  • purine e.g., adenine (A) or guanine (G)
  • nucleic acid and oligonucleotide refer to oligoribonucleotides as well as oligodeoxyribonucleotides.
  • nucleic acid and oligonucleotide shall also include polynucleosides (i.e., a polynucleotide minus the phosphate) and any other organic base containing polymer.
  • Nucleic acid molecules can be obtained from existing nucleic acid sources (e.g., genomic or cDNA), but are preferably synthetic (e.g., produced by nucleic acid synthesis).
  • nucleic acid and oligonucleotide also encompass nucleic acids or oligonucleotides with substitutions or modifications, such as in the bases and/or sugars.
  • nucleic acids having backbone sugars that are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 2' position and other than a phosphate group or hydroxy group at the 5' position.
  • modified nucleic acids may include a 2'-O-alkylated ribose group.
  • modified nucleic acids may include sugars such as arabinose or 2'-fluoiOarabinose instead of ribose.
  • the nucleic acids may be heterogeneous in backbone composition thereby containing any possible combination of polymer units linked together such as peptide- nucleic acids (which have an amino acid backbone with nucleic acid bases). Other examples are described in more detail below.
  • the immunostimulatory oligonucleotides of the instant invention can encompass various chemical modifications and substitutions, in comparison to natural RNA and DNA, involving a phosphodiester internucleoside bridge, a ⁇ -D-ribose unit and/or a natural nucleoside base (adenine, guanine, cytosine, thymine, uracil).
  • Examples of chemical modifications are known to the skilled person and are described, for example, in Uhlmann E et al. (1990) Chem Rev 90:543; "Protocols for Oligonucleotides and Analogs" Synthesis and Properties & Synthesis and Analytical Techniques, S.
  • An oligonucleotide according to the invention may have one or more modifications, wherein each modification is located at a particular phosphodiester internucleoside bridge and/or at a particular ⁇ -D-ribose unit and/or at a particular natural nucleoside base position in comparison to an oligonucleotide of the same sequence which is composed of natural DNA or RNA.
  • the oligonucleotides may comprise one or more modifications and wherein each modification is independently selected from: a) the replacement of a phosphodiester internucleoside bridge located at the 3' and/or the 5' end of a nucleoside by a modified internucleoside bridge, b) the replacement of phosphodiester bridge located at the 3' and/or the 5' end of a nucleoside by a dephospho bridge, c) the replacement of a sugar phosphate unit from the sugar phosphate backbone by another unit, d) the replacement of a ⁇ -D-ribose unit by a modified sugar unit, and e) the replacement of a natural nucleoside base by a modified nucleoside base.
  • each modification is independently selected from: a) the replacement of a phosphodiester internucleoside bridge located at the 3' and/or the 5' end of a nucleoside by a modified internucleoside bridge, b) the replacement of phosphodiester bridge located
  • the oligonucleotides may include modified internucleotide linkages, such as those described in a or b above. These modified linkages may be partially resistant to degradation (e.g., are stabilized).
  • a "stabilized oligonucleotide molecule" shall mean an oligonucleotide that is relatively resistant to in vivo degradation (e.g. via an exo- or endo- nuclease) resulting form such modifications.
  • Oligonucleotides having phosphorothioate linkages may provide maximal activity and protect the oligonucleotide from degradation by intracellular exo- and endo-nucleases.
  • a phosphodiester internucleoside bridge located at the 3' and/or the 5' end of a nucleoside can be replaced by a modified internucleoside bridge, wherein the modified internucleoside bridge is for example selected from phosphorothioate, phosphorodithioate, NR 1 R 2 -phosphoramidate, boranophosphate, ⁇ -hydroxybenzyl phosphonate, phosphate-(C ⁇ -C 2 ⁇ )-O-alkyl ester, phosphate-[(C 6 -C ⁇ )aryl-(C ⁇ -C 2 ⁇ )-O- alkyljester, (C ⁇ -C 8 )alkylphosphonate and/or (C 6 -C] 2 )arylphosphonate bridges, (C 7 -C] 2 )- ⁇ -hydroxymethyl-aryl (e.g., disclosed in WO 95/01363), wherein (C 6 -C ⁇ 2 )aryl, (C 6 - C 20
  • dephospho bridges are described, for example, in Uhlmann E and Peyman A in "Methods in Molecular Biology", Vol. 20, “Protocols for Oligonucleotides and Analogs", S. Agrawal, Ed, Humana Press, Totowa 1993, Chapter 16, pp. 355 ff), wherein a dephospho bridge is for example selected from the dephospho bridges formacetal, 3'-thioformacetal, methylhydroxylamine, oxime, methylenedim ethyl - hydrazo, dimethylenesulfone and/or silyl groups.
  • a sugar phosphate unit i.e., a ⁇ -D-ribose and phosphodiester internucleoside bridge together forming a sugar phosphate unit
  • the sugar phosphate backbone i.e., a sugar phosphate backbone is composed of sugar phosphate units
  • the other unit is for example suitable to build up a "morpholino- derivative" oligomer (as described, for example, in Stirchak EP et al.
  • Nucleic Acids Res 17:6129-41 that is, e.g., the replacement by a morpholino-derivative unit; or to build up a polyamide nucleic acid ("PNA"; as described for example, in Nielsen PE et al. (1994) Bioconjug Chem 5:3-7), that is, e.g., the replacement by a PNA backbone unit, e.g, by 2-aminoethylglycine.
  • PNA polyamide nucleic acid
  • the oligonucleotide may have other carbohydrate backbone modifications and replacements, such as peptide nucleic acids with phosphate groups (PHONA), locked nucleic acids (LNA), and oligonucleotides having backbone sections with alkyl linkers or amino linkers.
  • the alkyl linker may be branched or unbranched, substituted or unsubstituted, and chirally pure or a racemic mixture.
  • a ⁇ -ribose unit or a ⁇ -D-2'-deoxyribose unit can be replaced by a modified sugar unit, wherein the modified sugar unit is for example selected from ⁇ -D-ribose, ⁇ -D-2'- deoxyribose, L-2'-deoxyribose, 2'-F-2'-deoxyribose, 2'-F-arabinose, 2'-O-(C ⁇ -C 6 )alkyl- ribose, preferably 2'-O-(C ⁇ -C6)alkyl-ribose is 2'-O-methylribose, 2'-O-(C 2 -C 6 )alkenyl- ribose, 2'-[O-(C ⁇ -C 6 )alkyl-O-(C C 6 )alkyl]-ribose, 2'-NH 2 -2'-deoxyribose, ⁇ -D-xylo- furanose, ⁇
  • the sugar is 2'-O-methylribose, particularly for one or both nucleotides linked by a phosphodiester or phosphodiester-like internucleoside linkage.
  • Nucleic acids also include substituted purines and pyrimidines such as C-5 propyne pyrimidine and 7-deaza-7-substituted purine modified bases.
  • Purines and pyrimidines include but are not limited to adenine, cytosine, guanine, and thymine, and other naturally and non-naturally occurring nucleobases, substituted and unsubstituted aromatic moieties.
  • a modified base is any base which is chemically distinct from the naturally occurring bases typically found in DNA and RNA such as T, C, G, A, and U, but which share basic chemical structures with these naturally occurring bases.
  • the modified nucleoside base may be, for example, selected from hypoxanthine, uracil, dihydrouracil, pseudouracil, 2-thiouracil, 4-thiouracil, 5-aminouracil, 5-(Ci-C 6 )-alkyluracil, 5-(C 2 -C 6 )- alkenyluracil, 5-(C 2 -C 6 )-alkynyluracil, 5-(hydroxymethyl)uracil, 5-chlorouracil, 5-fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(C ⁇ -C6)-alkylcytosine, 5-(C -C 6 )- alkenylcytosine, 5-(C 2 -Co)-alkynylcytosine, 5-chlorocyto
  • a set of modified bases is defined.
  • the letter Y is used to refer to a nucleotide containing a cytosine or a modified cytosine.
  • a modified cytosine as used herein is a naturally occurring or non-naturally occurring pyrimidine base analog of cytosine which can replace this base without impairing the immunostimulatory activity of the oligonucleotide.
  • Modified cytosines include but are not limited to 5-substituted cytosines (e.g.
  • N,N'-propylene cytosine or phenoxazine N,N'-propylene cytosine or phenoxazine
  • uracil and its derivatives e.g. 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4- thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil.
  • Some of the preferred cytosines include 5-methyl-cytosine, 5-fluoro-cytosine, 5-hydroxy-cytosine, 5-hydroxymethyl- cytosine, and N4-ethyl-cytosine.
  • the cytosine base is substituted by a universal base (e.g. 3-nitropyrrole, P-base), an aromatic ring system (e.g.
  • a modified guanine as used herein is a naturally occurring or non-naturally occurring purine base analog of guanine which can replace this base without impairing the immunostimulatory activity of the oligonucleotide.
  • Modified guanines include but are not limited to 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, hypoxanthine, N2-substituted guanines (e.g. N2-methyl- guanine), 5-amino-3-methyl-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, adenine, substituted adenines (e.g.
  • the guanine base is substituted by a universal base (e.g. 4-methyl-indole, 5-nitro-indole, and K-base), an aromatic ring system (e.g. benzimidazole or dichloro- benzimidazole, 1- methyl- lH-[l,2,4]triazole-3-carboxylic acid amide) or a hydrogen atom (dSpacer).
  • a universal base e.g. 4-methyl-indole, 5-nitro-indole, and K-base
  • an aromatic ring system e.g. benzimidazole or dichloro- benzimidazole, 1- methyl- lH-[l,2,4]triazole-3-carboxylic acid amide
  • dSpacer a hydrogen atom
  • the oligonucleotides of the invention can be synthesized de novo using any of a number of procedures well known in the art.
  • the b-cyanoethyl phosphoramidite method eaucage, S.L, and Caruthers, M.H, Tet. Let. 22:1859, 1981
  • nucleoside H-phosphonate method Gagg et al, Tet. Let. 27:4051-4054, 1986; Froehler et al, Nucl. Acid. Res. 14:5399-5407, 1986, ; Garegg et al, Tet. Let. 27:4055-4058, 1986, Gaffhey et al, Tet. Let.
  • oligonucleotide generally refers to an oligonucleotide which is separated from components which it is normally associated with in nature.
  • an isolated oligonucleotide may be one which is separated from a cell, from a nucleus, from mitochondria or from chromatin.
  • the immunostimulatory nucleic acid molecules of the instant invention can have phosphodiester internuclotide linkages.
  • a phosphodiester internucleotide linkage is the type of linkage characteristic of nucleic acids found in nature. As shown in Figure 20, the phosphodiester internucleotide linkage includes a phosphorus atom flanked by two bridging oxygen atoms and bound also by two additional oxygen atoms, one charged and the other uncharged. Phosphodiester internucleotide linkage is particularly preferred when it is important to reduce the tissue half-life of the oligonucleotide.
  • a phosphodiester-like internucleotide linkage is a phosphorus-containing bridging group that is chemically and/or diastereomerically similar to phosphodiester. Measures of similarity to phosphodiester include susceptibility to nuclease digestion and ability to activate RNAse H. Thus for example phosphodiester, but not phosphorothioate, oligonucleotides are susceptible to nuclease digestion, while both phosphodiester and phosphorothioate oligonucleotides activate RNAse H.
  • the phosphodiester-like internucleotide linkage is boranophosphate (or equivalently, boranophosphonate) linkage.
  • the phosphodiester-like internucleotide linkage is diasteromerically pure Rp phosphorothioate. It is believed that diasteromerically pure Rp phosphorothioate is more susceptible to nuclease digestion and is better at activating RNAse H than mixed or diastereomerically pure Sp phosphorothioate. Stereoisomers of CpG oligonucleotides are the subject of co-pending U.S.
  • the immunostimulatory nucleic acid molecules of the instant invention can have chimeric backbone.
  • a chimeric backbone refers to a partially stabilized backbone, wherein at least one internucleotide linkage is phosphodiester or phosphodiester-like, and wherein at least one other internucleotide linkage is a stabilized internucleotide linkage, wherein the at least one phosphodiester or phosphodiester-like linkage and the at least one stabilized linkage are different.
  • boranophosphonate linkages have been reported to be stabilized relative to phosphodiester linkages, for purposes of the chimeric nature of the backbone, boranophosphonate linkages can be classified either as phosphodiester-like or as stabilized, depending on the context.
  • a chimeric backbone according to the instant invention could in one embodiment include at least one phosphodiester (phosphodiester or phosphodiester-like) linkage and at least one boranophosphonate (stabilized) linkage.
  • a chimeric backbone according to the instant invention could include boranophosphonate (phosphodiester or phosphodiester- like) and phosphorothioate (stabilized) linkages.
  • a “stabilized internucleotide linkage” shall mean an internucleotide linkage that is relatively resistant to in vivo degradation (e.g., via an exo- or endo-nuclease), compared to a phosphodiester internucleotide linkage.
  • Preferred stabilized internucleotide linkages include, without limitation, phosphorothioate, phosphorodithioate, methylphosphonate, and methylphosphorothioate.
  • Other stabilized internucleotide linkages include, without limitation: peptide, alkyl, dephospho, and others as described above.
  • Modified backbones such as phosphorothioates may be synthesized using automated techniques employing either phosphoramidate or H-phosphonate chemistries.
  • Aryl-and alkyl-phosphonates can be made, e.g., as described in U.S. Patent No. 4,469,863; and alkylphosphotriesters (in which the charged oxygen moiety is alkylated as described in U.S. Patent No. 5,023,243 and European Patent No. 092,574) can be prepared by automated solid phase synthesis using commercially available reagents. Methods for making other DNA backbone modifications and substitutions have been described (e.g., Uhlmann, E. and Peyman, A, Chem. Rev.
  • CpG phosphorothioate oligonucleotides with strong stimulatory activity in the mouse system tend to show lower activity on human and other non-rodent immune cells.
  • CpG phosphorothioate oligonucleotides with strong stimulatory activity in the mouse system tend to show lower activity on human and other non-rodent immune cells.
  • DNA containing these 5'TCG or 5 'TYZ CpG motifs strongly stimulated human peripheral blood cells to produce IL-10, IL-6, IP- 10 and IFN- ⁇ .
  • the 5'TCG containing ODN could be further optimized by selecting ODNs of a particular length. For instance, ODNs of 22 nucleotides in length are more stimulatory than shorter ODN.
  • CpG immunostimulatory oligonucleotides have dramatic immune stimulatory effects on human cells such as PBMC, suggesting that these CpG immunostimulatory oligonucleotides are effective therapeutic agents for human vaccination, cancer immunotherapy, asthma immunotherapy, general enhancement of immune function, enhancement of hematopoietic recovery following radiation or chemotherapy, and other immune modulatory applications.
  • the terms treat, treated, or treating when used with respect to a disorder such as an infectious disease, cancer, allergy, or asthma refers to a prophylactic treatment which increases the resistance of a subject to development of the disease (e.g., to infection with a pathogen) or, in other words, decreases the likelihood that the subject will develop the disease (e.g., become infected with the pathogen) as well as a treatment after the subject has developed the disease in order to fight the disease (e.g., reduce or eliminate the infection) or prevent the disease from becoming worse.
  • the CpG immunostimulatory oligonucleotides are useful in some aspects of the invention as a vaccine for the treatment of a subject having or at risk of developing allergy or asthma, an infection with an infectious organism or a cancer in which a specific cancer antigen has been identified.
  • the CpG immunostimulatory oligonucleotides can also be given alone without the antigen or allergen for protection against infection, allergy or cancer or may be administered with other therapeutic agents. Repeated doses may allow longer term protection.
  • a subject at risk as used herein is a subject who has any risk of exposure to an infection causing pathogen or a cancer or an allergen or a risk of developing cancer.
  • a subject at risk may be a subject who is planning to travel to an area where a particular type of infectious agent is found or it may be a subject who through lifestyle or medical procedures is exposed to bodily fluids which may contain infectious organisms or directly to the organism or even any subject living in an area where an infectious organism or an allergen has been identified.
  • Subjects at risk of developing infection also include general populations to which a medical agency recommends vaccination with a particular infectious organism antigen. If the antigen is an allergen and the subject develops allergic responses to that particular antigen and the subject may be exposed to the antigen, i.e., during pollen season, then that subject is at risk of exposure to the antigen.
  • a subject at risk of developing an allergy to asthma includes those subjects that have been identified as having an allergy or asthma but that don't have the active disease during the CpG immunostimulatory oligonucleotide treatment as well as subjects that are considered to be at risk of developing these diseases because of genetic or environmental factors.
  • a subject at risk of developing a cancer is one who has a high probability of developing cancer. These subjects include, for instance, subjects having a genetic abnormality, the presence of which has been demonstrated to have a correlative relation to a higher likelihood of developing a cancer and subjects exposed to cancer causing agents such as tobacco, asbestos, or other chemical toxins, or a subject who has previously been treated for cancer and is in apparent remission.
  • a subject at risk of developing a cancer is treated with a CpG immunostimulatory oligonucleotide and optionally an antigen specific for the type of cancer to which the subject is at risk of developing, the subject may be able to kill the cancer cells as they develop. If a tumor begins to form in the subject, the subject will develop an innate immune response or a i specific immune response against the tumor antigen.
  • the invention also encompasses the use of the CpG immunostimulatory oligonucleotides for the treatment of a subject having an infection, an allergy, asthma, or a cancer.
  • a subject having an infection is a subject that has been exposed to an infectious pathogen and has acute or chronic detectable levels of the pathogen in the body.
  • CpG immunostimulatory oligonucleotides can be used with or without an antigen or other therapeutic to mount an innate or an antigen specific systemic or mucosal immune response that is capable of reducing the level of or eradicating the infectious pathogen.
  • An infectious disease as used herein, is a disease arising from the presence of a foreign microorganism in the body. It is particularly important to develop effective vaccine strategies and treatments to protect the body's mucosal surfaces, which are the primary site of pathogenic entry.
  • a subject having an allergy is a subject that is capable of developing an allergic reaction in response to an allergen.
  • An allergy refers to acquired hypersensitivity to a substance (allergen).
  • Allergic conditions include but are not limited to eczema, allergic rhinitis or coryza, hay fever, conjunctivitis, bronchial asthma, allergic asthma, urticaria (hives) and food allergies, and other atopic conditions.
  • Th2 immune response The other major type of immune response, which is associated with the production of IL-4 and IL-5 cytokines, is termed a Th2 immune response. In general, it appears that allergic diseases are mediated by Th2 type immune responses.
  • an effective dose for inducing an immune response of a CpG immunostimulatory oligonucleotide can be administered to a subject to treat asthma and allergy.
  • the CpG immunostimulatory oligonucleotides have significant therapeutic utility in the treatment of allergic conditions and asthma.
  • Th2 cytokines especially IL-4 and IL-5 are elevated in the airways of asthmatic subjects. These cytokines promote important aspects of the asthmatic inflammatory response, including IgE isotope switching, eosinophil chemotaxis and activation and mast cell growth.
  • Thl cytokines especially IFN- ⁇ and IL-12, can suppress the formation of Th2 clones and production of Th2 cytokines.
  • Asthma refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively associated with atopic or allergic symptoms. Thus, asthma includes allergic asthma and non-allergic asthma.
  • a subject having a cancer is a subject that has detectable cancerous cells.
  • the cancer may be a malignant or non-malignant cancer.
  • Cancers or tumors include but are not limited to biliary tract cancer; brain cancer; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; intraepithelial neoplasms; lymphomas; liver cancer; lung cancer (e.g. small cell and non-small cell); melanoma; neuroblastomas; oral cancer; ovarian cancer; pancreas cancer; prostate cancer; rectal cancer; sarcomas; skin cancer; testicular cancer; thyroid cancer; and renal cancer, as well as other carcinomas and sarcomas.
  • the cancer is hairy cell leukemia, chronic myelogenous leukemia, cutaneous T-cell leukemia, multiple myeloma, follicular lymphoma, malignant melanoma, squamous cell carcinoma, renal cell carcinoma, prostate carcinoma, bladder cell carcinoma, or colon carcinoma, CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer, as well as other carcinomas and sarcomas.
  • a subject shall mean a human or vertebrate animal including but not limited to a dog, cat, horse, cow, pig, sheep, goat, turkey, chicken, primate, e.g., monkey, and fish (aquaculture species), e.g. salmon.
  • the compounds may be used to treat cancer and tumors, infections, and allergy/asthma in human and non human subjects.
  • Cancer is one of the leading causes of death in companion animals (i.e., cats and dogs).
  • the subject may be exposed to the antigen.
  • the term exposed to refers to either the active step of contacting the subject with an antigen or the passive exposure of the subject to the antigen in vivo.
  • an antigen is administered directly to the subject by any means such as intravenous, intramuscular, oral, transdermal, mucosal, intranasal, intratracheal, or subcutaneous administration.
  • the antigen can be administered systemically or locally.
  • Methods for administering the antigen and the CpG immunostimulatory oligonucleotide are described in more detail below.
  • a subject is passively exposed to an antigen if an antigen becomes available for exposure to the immune cells in the body.
  • a subject may be passively exposed to an antigen, for instance, by entry of a foreign pathogen into the body or by the development of a tumor cell expressing a foreign antigen on its surface.
  • the methods in which a subject is passively exposed to an antigen can be particularly dependent on timing of administration of the CpG immunostimulatory oligonucleotide.
  • the subject may be administered the CpG immunostimulatory oligonucleotide on a regular basis when that risk is greatest, i.e., during allergy season or after exposure to a cancer causing agent.
  • the CpG immunostimulatory oligonucleotide may be administered to travelers before they travel to foreign lands where they are at risk of exposure to infectious agents.
  • an antigen as used herein is a molecule capable of provoking an immune response.
  • Antigens include but are not limited to cells, cell extracts, proteins, polypeptides, peptides, polysaccharides, polysaccharide conjugates, peptide and nonpeptide mimics of polysaccharides and other molecules, small molecules, lipids, glycolipids, carbohydrates, viruses and viral extracts and muticellular organisms such as parasites and allergens.
  • antigen broadly includes any type of molecule which is recognized by a host immune system as being foreign.
  • Antigens include but are not limited to cancer antigens, microbial antigens, and allergens.
  • a cancer antigen as used herein is a compound, such as a peptide or protein, associated with a tumor or cancer cell surface and which is capable of provoking an immune response when expressed on the surface of an antigen presenting cell in the context of an MHC molecule.
  • Cancer antigens can be prepared from cancer cells either by preparing crude extracts of cancer cells, for example, as described in Cohen, et al, 1994, Cancer Research, 54:1055, by partially purifying the antigens, by recombinant technology, or by de novo synthesis of known antigens.
  • Cancer antigens include but are not limited to antigens that are recombinantly expressed, an immunogenic portion thereof, or a whole tumor or cancer cell. Such antigens can be isolated or prepared recombinantly or by any other means known in the
  • cancer antigen and “tumor antigen” are used interchangeably to refer to antigens which are differentially expressed by cancer cells and can thereby be exploited in order to target cancer cells.
  • Cancer antigens are antigens which can potentially stimulate apparently tumor-specific immune responses. Some of these antigens are encoded, although not necessarily expressed, by normal cells. These antigens can be characterized as those which are normally silent (i.e., not expressed) in normal cells, those that are expressed only at certain stages of differentiation and those that are temporally expressed such as embryonic and fetal antigens.
  • cancer antigens are encoded by mutant cellular genes, such as oncogenes (e.g., activated ras oncogene), suppressor genes (e.g., mutant p53), fusion proteins resulting from internal deletions or chromosomal translocations. Still other cancer antigens can be encoded by viral genes such as those carried on RNA and DNA tumor viruses.
  • a microbial antigen as used herein is an antigen of a microorganism and includes but is not limited to virus, bacteria, parasites, and fungi.
  • antigens include the intact microorganism as well as natural isolates and fragments or derivatives thereof and also synthetic compounds which are identical to or similar to natural microorganism antigens and induce an immune response specific for that microorganism.
  • a compound is similar to a natural microorganism antigen if it induces an immune response (humoral and/or cellular) to a natural microorganism antigen.
  • Such antigens are used routinely in the art and are well known to those of ordinary skill in the art.
  • Retroviridae e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HTLV-III, LAV or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV-LP; Picornaviridae (e.g. polio viruses, hepatitis A virus; enteroviruses, human Coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (e.g. strains that cause gastroenteritis); Togaviridae (e.g. equine encephalitis viruses, rubella viruses); Flaviridae (e.g.
  • Coronoviridae e.g. coronaviruses
  • Rhabdoviradae e.g. vesicular stomatitis viruses, rabies viruses
  • Coronoviridae e.g. coronaviruses
  • Rhabdoviridae e.g. vesicular stomatitis viruses, rabies viruses
  • Filoviridae e.g. ebola viruses
  • Paramyxoviridae e.g. parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus
  • Orthomyxoviridae e.g. influenza viruses
  • Bungaviridae e.g.
  • African swine fever virus African swine fever virus
  • unclassified viruses e.g. the agent of delta hepatitis (thought to be a defective satellite of hepatitis B virus), Hepatitis C; Norwalk and related viruses, and astroviruses).
  • Both gram negative and gram positive bacteria serve as antigens in vertebrate animals.
  • Such gram positive bacteria include, but are not limited to, Pasteurella species, Staphylococci species, and Streptococcus species.
  • Gram negative bacteria include, but are not limited to, Escherichia coli, Pseudomonas species, and Salmonella species.
  • infectious bacteria include but are not limited to, Helicobacter pylons, Borelia burgdorferi, Legionella pneumophilia, Mycobacteria sps (e.g. M. tuberculosis, M. avium, M. intracellular e, M. kansaii, M.
  • fungi examples include Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, Candida albicans.
  • Plasmodium spp such as Plasmodium falciparum, Plasmodium malariae, Plasmodium ovale, and Plasmodium vivax and Toxoplasma gondii.
  • Blood-borne and/or tissues parasites include Plasmodium spp, Babesia microti, Babesia divergens, Leishmania tropica, Leishmania spp, Leishmania braziliensis, Leishmania donovani, Trypanosoma gambiense and Trypanosoma rhodesiense (African sleeping sickness), Trypanosoma cruzi (Chagas' disease), and Toxoplasma gondii.
  • An allergen refers to a substance (antigen) that can induce an allergic or asthmatic response in a susceptible subject.
  • the list of allergens is enormous and can include pollens, insect venoms, animal dander dust, fungal spores and drugs (e.g. penicillin).
  • Examples of natural, animal and plant allergens include but are not limited to proteins specific to the following genuses: Canine (Ca is familiaris); Dermatophagoides (e.g. Dermatophagoides farinae); Felis (Felis domesticus); Ambrosia (Ambrosia artemiisfolia; Lolium (e.g.
  • Lolium perenne or Lolium multiflorum Lolium perenne or Lolium multiflorum); Cryptomeria (Cryptomeria japonica); Alternaria (Alternaria alternata); Alder; Alnus (Alnus gultinoasa); Betula (Betula verrucosa); Quercus (Quercus alba); Olea (Olea europa); Artemisia (Artemisia vulgaris); Plantago (e.g. Plantago lanceolata); Parietaria (e.g. Parietaria officinalis or Parietaria judaica); Blattella (e.g. Blattella germanica); Apis (e.g. Apis multiflorum); Cupressus (e.g.
  • Cupressus semper ⁇ virens Cupressus arizonica and Cupressus macrocarpd
  • Juniperus e.g. Juniperus sabinoides, Juniperus virginiana, Juniperus communis and Juniperus ashei
  • Thuya e.g. Thuya orientalis
  • Chamaecyparis e.g. Chamaecyparis obtusci
  • Periplaneta e.g. Periplaneta americana
  • Agropyron e.g. Agropyron repens
  • Secale e.g. Secale cereale
  • Triticum e.g. Triticum aestivum
  • Dactylis e.g. Dactylis glomerata
  • Festuca e.g. Festuca elatior
  • Poa e.g. Poapratensis or Poa compressa
  • Avena e.g. Avena sativa
  • Holcus e.g. Holcus lanatus
  • Anthoxanthum e.g.
  • Anthoxanthum odoratum Arrhenatherum (e.g. Arrhenatherum elatius); Agrostis (e.g. Agrostis alba); Phleum (e.g. Phleum pratense); Phalaris (e.g. Phalaris arundinacea); Paspalum (e.g. Paspalum notatum); Sorghum (e.g. Sorghum halepensis); and Bromus (e.g. Bromus inermis).
  • Arrhenatherum e.g. Arrhenatherum elatius
  • Agrostis e.g. Agrostis alba
  • Phleum e.g. Phleum pratense
  • Phalaris e.g. Phalaris arundinacea
  • Paspalum e.g. Paspalum notatum
  • Sorghum e.g. Sorghum halepensis
  • Bromus e.g. Bromus iner
  • the antigen may be substantially purified.
  • substantially purified as used herein refers to an antigen, i.e., a polypeptide which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated.
  • One skilled in the art can purify polypeptide antigens using standard techniques for protein purification.
  • the substantially pure polypeptide will often yield a single major band on a non-reducing polyacrylamide gel. In the case of partially glycosylated polypeptides or those that have several start codons, there may be several bands on a non-reducing polyacrylamide gel, but these will form a distinctive pattern for that polypeptide.
  • the purity of the polypeptide antigen may also be determined by amino-terminal amino acid sequence analysis. Other types of antigens such as polysaccharides, small molecule, mimics etc are included within the invention and may optionally be substantially pure.
  • the oligonucleotides of the invention may be administered to a subject with an anti-microbial agent.
  • An anti-microbial agent refers to a naturally- occurring or synthetic compound which is capable of killing or inhibiting infectious microorganisms.
  • the type of anti-microbial agent useful according to the invention will depend upon the type of microorganism with which the subject is infected or at risk of becoming infected.
  • Anti-microbial agents include but are not limited to anti-bacterial agents, anti-viral agents, anti-fungal agents and anti-parasitic agents.
  • anti-bacterial agents kill or inhibit bacteria, and include antibiotics as well as other synthetic or natural compounds having similar functions.
  • Antibiotics are low molecular weight molecules which are produced as secondary metabolites by cells, such as microorganisms. In general, antibiotics interfere with one or more bacterial functions or structures which are specific for the microorganism and which are not present in host cells.
  • Anti-viral agents can be isolated from natural sources or synthesized and are useful for killing or inhibiting viruses.
  • Anti-fungal agents are used to treat superficial fungal infections as well as opportunistic and primary systemic fungal infections. Anti-parasitic agents kill or inhibit parasites.
  • anti-parasitic agents also referred to as parasiticides useful for human administration
  • examples of anti-parasitic agents include but are not limited to albendazole, amphotericin B, benznidazole, bithionol, chloroquine HCI, chloroquine phosphate, clindamycin, dehydroemetine, diethylcarbamazine, diloxanide furoate, eflornithine, furazolidaone, glucocorticoids, halofantrine, iodoquinol, ivermectin, mebendazole, mefloquine, meglumine antimoniate, melarsoprol, metrifonate, metronidazole, niclosamide, nifurtimox, oxamniquine, paromomycin, pentamidine isethionate, piperazine, praziquantel, primaquine phosphate, proguanil, pyr
  • Antibacterial agents kill or inhibit the growth or function of bacteria.
  • a large class of antibacterial agents is antibiotics.
  • Antibiotics which are effective for killing or inhibiting a wide range of bacteria, are referred to as broad spectrum antibiotics.
  • Other types of antibiotics are predominantly effective against the bacteria of the class gram- positive or gram-negative. These types of antibiotics are referred to as narrow spectrum antibiotics.
  • Other antibiotics which are effective against a single organism or disease and not against other types of bacteria are referred to as limited spectrum antibiotics.
  • Antibacterial agents are sometimes classified based on their primary mode of action. In general, antibacterial agents are cell wall synthesis inhibitors, cell membrane inhibitors, protein synthesis inhibitors, nucleic acid synthesis or functional inhibitors, and competitive inhibitors.
  • Antiviral agents are compounds which prevent infection of cells by viruses or replication of the virus within the cell. There are many fewer antiviral drugs than antibacterial drugs because the process of viral replication is so closely related to DNA replication within the host cell, that non-specific antiviral agents would often be toxic to the host. There are several stages within the process of viral infection which can be blocked or inhibited by antiviral agents. These stages include, attachment of the virus to the host cell (immunoglobulin or binding peptides), uncoating of the virus (e.g. amantadine), synthesis or translation of viral mRNA (e.g. interferon), replication of viral RNA or DNA (e.g. nucleoside analogues), maturation of new virus proteins (e.g. protease inhibitors), and budding and release of the virus.
  • attachment of the virus to the host cell immunoglobulin or binding peptides
  • uncoating of the virus e.g. amantadine
  • synthesis or translation of viral mRNA e
  • Nucleotide analogues are synthetic compounds which are similar to nucleotides, but which have an incomplete or abnormal deoxyribose or ribose group. Once the nucleotide analogues are in the cell, they are phosphorylated, producing the triphosphate form which competes with normal nucleotides for incorporation into the viral DNA or RNA. Once the triphosphate form of the nucleotide analogue is incorporated into the growing nucleic acid chain, it causes irreversible association with the viral polymerase and thus chain termination.
  • Nucleotide analogues include, but are not limited to, acyclovir (used for the treatment of herpes simplex virus and varicella-zoster virus), gancyclovir (useful for the treatment of cytomegalovirus), idoxuridine, ribavirin (useful for the treatment of respiratory syncitial virus), dideoxyinosine, dideoxycytidine, zidovudine (azidothymidine), imiquimod, and resimiquimod.
  • acyclovir used for the treatment of herpes simplex virus and varicella-zoster virus
  • gancyclovir used for the treatment of cytomegalovirus
  • idoxuridine used for the treatment of cytomegalovirus
  • ribavirin used for the treatment of respiratory syncitial virus
  • dideoxyinosine dideoxycytidine
  • zidovudine zidovudine
  • imiquimod imiquimod
  • resimiquimod
  • the interferons are cytokines which are secreted by virus-infected cells as well as immune cells.
  • the interferons function by binding to specific receptors on cells adjacent to the infected cells, causing the change in the cell which protects it from infection by the virus, ⁇ and ⁇ -interferon also induce the expression of Class I and Class II MHC molecules on the surface of infected cells, resulting in increased antigen presentation for host immune cell recognition, ⁇ and ⁇ -interferons are available as recombinant forms and have been used for the treatment of chronic hepatitis B and C infection. At the dosages which are effective for anti-viral therapy, interferons have severe side effects such as fever, malaise and weight loss.
  • Anti-viral agents useful in the invention include but are not limited to immunoglobulins, amantadine, interferons, nucleoside analogues, and protease inhibitors.
  • Specific examples of anti-virals include but are not limited to Acemannan; Acyclovir; Acyclovir Sodium; Adefovir; Alovudine; Alvircept Sudotox; Amantadine Hydrochloride; Aranotin; Arildone; Atevirdine Mesylate; Avridine; Cidofovir; Cipamfylline; Cytarabine Hydrochloride; Delavirdine Mesylate; Desciclovir; Didanosine; Disoxaril; Edoxudine; Enviradene; Enviroxime; Famciclovir; Famotine Hydrochloride; Fiacitabine; Fialuridine; Fosarilate; Foscarnet Sodium; Fosfonet Sodium; Ganciclovir; Ganciclovir
  • Anti-fungal agents are useful for the treatment and prevention of infective fungi. Anti-fungal agents are sometimes classified by their mechanism of action. Some anti- fungal agents function as cell wall inhibitors by inhibiting glucose synthase. These include, but are not limited to, basiungin/ECB. Other anti-fungal agents function by destabilizing membrane integrity. These include, but are not limited to, immidazoles, such as clotrimazole, sertaconzole, fluconazole, itraconazole, ketoconazole, miconazole, and voriconacole, as well as FK 463, amphotericin B, BAY 38-9502, MK 991, pradimicin, UK 292, butenafine, and terbinafine. Other anti-fungal agents function by breaking down chitin (e.g. chitinase) or immunosuppression (501 cream).
  • immidazoles such as clotrimazole, sertaconzole, fluconazole,
  • CpG immunostimulatory oligonucleotides can be combined with other therapeutic agents such as adjuvants to enhance immune responses.
  • the CpG immunostimulatory oligonucleotide and other therapeutic agent may be administered simultaneously or sequentially.
  • the other therapeutic agents When the other therapeutic agents are administered simultaneously they can be administered in the same or separate formulations, but are administered at the same time.
  • the other therapeutic agents are administered sequentially with one another and with CpG immunostimulatory oligonucleotide, when the administration of the other therapeutic agents and the CpG immunostimulatory oligonucleotide is temporally separated. The separation in time between the administration of these compounds may be a matter of minutes or it may be longer.
  • compositions of the invention may also be administered with non-nucleic acid adjuvants.
  • a non-nucleic acid adjuvant is any molecule or compound except for the CpG immunostimulatory oligonucleotides described herein which can stimulate the humoral and/or cellular immune response.
  • Non-nucleic acid adjuvants include, for instance, adjuvants that create a depo effect, immune stimulating adjuvants, and adjuvants that create a depo effect and stimulate the immune system.
  • the CpG immunostimulatory oligonucleotides are also useful as mucosal adjuvants. It has previously been discovered that both systemic and mucosal immunity are induced by mucosal delivery of CpG nucleic acids. Thus, the oligonucleotides may be administered in combination with other mucosal adjuvants.
  • Immune responses can also be induced or augmented by the co-administration or co-linear expression of cytokines (Bueler & Mulligan, 1996; Chow et al, 1997; Geissler et al, 1997; Iwasaki et al, 1997; Kim et al, 1997) or co-stimulatory molecules such as B7 (Iwasaki et al, 1997; Tsuji et al, 1997) with the CpG immunostimulatory oligonucleotides.
  • cytokines Buseler & Mulligan, 1996; Chow et al, 1997; Geissler et al, 1997; Iwasaki et al, 1997; Kim et al, 1997) or co-stimulatory molecules such as B7 (Iwasaki et al, 1997; Tsuji et al, 1997) with the CpG immunostimulatory oligonucleotides.
  • cytokine is used as a generic name for a diverse group of soluble proteins and peptides which act as humoral regulators at nano- to picomolar concentrations and which, either under normal or pathological conditions, modulate the functional activities of individual cells and tissues. These proteins also mediate interactions between cells directly and regulate processes taking place in the extracellular environment.
  • cytokines include, but are not limited to IP- 10, IL-1, IL-2, IL- 4, IL-5, IL-6, IL-7, IL-10, IL-12, IL-15, IL-18, granulocyte-macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), interferon- ⁇ ( ⁇ -IFN), IFN- ⁇ , tumor necrosis factor (TNF), TGF- ⁇ , FLT-3 ligand, and CD40 ligand.
  • the CpG oligonucleotides may be used in combination with antibodies against certain cytokines, such as anti-IL-10 and anti -TGF- ⁇ , as well as Cox inhibitors, i.e. COX-1 and COX-2 inhibitors.
  • the oligonucleotides are also useful for redirecting an immune response from a Th2 immune response to a Thl immune response. This results in the production of a relatively balanced Thl/Th2 environment. Redirection of an immune response from a Th2 to a Thl immune response can be assessed by measuring the levels of cytokines produced in response to the nucleic acid (e.g., by inducing monocytic cells and other cells to produce Thl cytokines, including IL-12, IFN- ⁇ and GM-CSF). The redirection or rebalance of the immune response from a Th2 to a Thl response is particularly useful for the treatment of asthma.
  • an effective amount for treating asthma can be that amount; useful for redirecting a Th2 type of immune response that is associated with asthma to a Thl type of response or a balanced Thl/Th2 environment.
  • Th2 cytokines, especially IL-4 and IL-5 are elevated in the airways of asthmatic subjects.
  • the CpG immunostimulatory oligonucleotides described herein cause an increase in Thl cytokines which helps to rebalance the immune system, preventing or reducing the adverse effects associated with a predominately Th2 immune response.
  • the CpG immunostimulatory oligonucleotides have the unique capability to promote cell survival, differentiation, activation and maturation of dendritic cells, and are useful for in vitro, in vivo, and ex vivo methods involving dendritic cells.
  • CpG immunostimulatory oligonucleotides also increase natural killer cell lytic activity and antibody dependent cellular cytotoxicity (ADCC).
  • ADCC can be performed using a CpG immunostimulatory oligonucleotide in combination with an antibody specific for a cellular target, such as a cancer cell.
  • a cellular target such as a cancer cell.
  • the antibodies useful in the ADCC procedure include antibodies which interact with a cell in the body. Many such antibodies specific for cellular targets have been described in the art and many are commercially available.
  • the CpG immunostimulatory oligonucleotides may also be administered in conjunction with an anti-cancer therapy.
  • Anti-cancer therapies include cancer medicaments, radiation and surgical procedures.
  • a "cancer medicament” refers to an agent which is administered to a subject for the purpose of treating a cancer.
  • treating cancer includes preventing the development of a cancer, reducing the symptoms of cancer, and/or inhibiting the growth of an established cancer.
  • the cancer medicament is administered to a subject at risk of developing a cancer for the purpose of reducing the risk of developing the cancer.
  • Various types of medicaments for the treatment of cancer are described herein.
  • cancer medicaments are classified as chemotherapeutic agents, immunotherapeutic agents, cancer vaccines, hormone therapy, and biological response modifiers. Additionally, the methods of the invention are intended to embrace the use of more than one cancer medicament along with the CpG immunostimulatory oligonucleotides. As an example, where appropriate, the CpG immunostimulatory oligonucleotides may be administered with both a chemotherapeutic agent and an immunotherapeutic agent.
  • the cancer medicament may embrace an immunotherapeutic agent and a cancer vaccine, or a chemotherapeutic agent and a cancer vaccine, or a chemotherapeutic agent, an immunotherapeutic agent and a cancer vaccine all administered to one subject for the purpose of treating a subject having a cancer or at risk of developing a cancer.
  • the chemotherapeutic agent may be selected from the group consisting of methotrexate, vincristine, adriamycin, cisplatin, non-sugar containing chloroethylnitrosoureas, 5-fluorouracil, mitomycin C, bleomycin, doxorubicin, dacarbazine, taxol, fragyline, Meglamine GLA, valrubicin, carmustaine and poliferposan, MMI270, BAY 12-9566, RAS famesyl transferase inhibitor, famesyl transferase inhibitor, MMP, MTA/LY231514, LY264618/Lometexol, Glamolec, CI-994, TNP-470, Hycamtin/Topotecan, PKC412, Valspodar/PSC833, Novantrone/Mitroxantrone, Metaret/Suramin, Batimastat, E7070, BCH-4556, CS-682,
  • the immunotherapeutic agent may be selected from the group consisting of Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncolym, SMART M195, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-11, MDX-22, OV103, 3622 W94, anti-VEGF, Zenapax, MDX-220, MDX-447, MELIMMUNE-2, MELIMMUNE-1, CEACIDE, Pretarget, NovoMAb-G2, TNT,
  • Gliomab-H GNI-250, EMD-72000, LymphoCide, CMA 676, Monopharm-C, 4B5, ior egf.r3, ior c5, BABS, anti-FLK-2, MDX-260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab and ImmuRAIT-CEA, but it is not so limited.
  • the cancer vaccine may be selected from the group consisting of EGF, Anti- idiotypic cancer vaccines, Gp75 antigen, GMK melanoma vaccine, MGV ganglioside conjugate vaccine, Her2/neu, Ovarex, M-Vax, O-Vax, L-Vax, STn-KHL theratope, BLP25 (MUC-1), liposomal idiotypic vaccine, Melacine, peptide antigen vaccines, toxin/antigen vaccines, MVA-based vaccine, PACIS, BCG vacine, TA-HPV, TA-CIN, DISC-virus and ImmuCyst/TheraCys, but it is not so limited.
  • CpG immunostimulatory oligonucleotides in conjunction with immunotherapeutic agents such as monoclonal antibodies is able to increase long-term survival through a number of mechanisms including significant enhancement of ADCC (as discussed above), activation of natural killer (NK) cells and an increase in IFN ⁇ levels.
  • the nucleic acids when used in combination with monoclonal antibodies serve to reduce the dose of the antibody required to achieve a biological result.
  • the invention also includes methods for inducing antigen non-specific innate immune activation and broad spectrum resistance to infectious challenge using the CpG immunostimulatory oligonucleotides.
  • innate immune activation refers to the activation of immune cells other than memory B cells and for instance can include the activation of NK cells, T cells and/or other immune cells that can respond in an antigen independent fashion.
  • a broad spectrum resistance to infectious challenge is induced because the immune cells are in active form and are primed to respond to any invading compound or microorganism. The cells do not have to be specifically primed against a particular antigen. This is particularly useful in biowarfare, and the other circumstances described above such as travelers.
  • the CpG immunostimulatory oligonucleotides may be directly administered to the subject or may be administered in conjunction with a nucleic acid delivery complex.
  • a nucleic acid delivery complex shall mean a nucleic acid molecule associated with (e.g. ionically or covalently bound to; or encapsulated within) a targeting means (e.g. a molecule that results in higher affinity binding to target cell.
  • a targeting means e.g. a molecule that results in higher affinity binding to target cell.
  • nucleic acid delivery complexes include nucleic acids associated with a sterol (e.g. cholesterol), a lipid (e.g. a cationic lipid, virosome or liposome), or a target cell specific binding agent (e.g. a ligand recognized by target cell specific receptor).
  • Preferred complexes may be sufficiently stable in vivo to prevent significant uncoupling prior to internalization by the target cell.
  • the complex can be cleavable under appropriate conditions within the cell so that the oligonucleotide is released in a functional form.
  • the CpG immunostimulatory oligonucleotide and/or the antigen and/or other therapeutics may be administered alone (e.g., in saline or buffer) or using any delivery vehicles known in the art.
  • an effective amount of a CpG immunostimulatory oligonucleotide refers to the amount necessary or sufficient to realize a desired biologic effect.
  • an effective amount of a CpG immunostimulatory oligonucleotide administered with an antigen for inducing mucosal immunity is that amount necessary to cause the development of IgA in response to an antigen upon exposure to the antigen, whereas that amount required for inducing systemic immunity is that amount necessary to cause the development of IgG in response to an antigen upon exposure to the antigen.
  • an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the particular subject.
  • the effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular CpG immunostimulatory oligonucleotide being administered the size of the subject, or the severity of the disease or condition.
  • One of ordinary skill in the art can empirically determine the effective amount of a particular CpG immunostimulatory oligonucleotide and/or antigen and/or other therapeutic agent without necessitating undue experimentation.
  • Subject doses of the compounds described herein for mucosal or local delivery typically range from about 0.1 ⁇ g to 10 mg per administration, which depending on the application could be given daily, weekly, or monthly and any other amount of time therebetween or as otherwise required. More typically mucosal or local doses range from about 10 ⁇ g to 5 mg per administration, and most typically from about 100 ⁇ g to 1 mg, with 2 - 4 administrations being spaced days or weeks apart. More typically, immune stimulant doses range from 1 ⁇ g to 10 mg per administration, and most typically lO ⁇ g to 1 mg, with daily or weekly administrations.
  • Subject doses of the compounds described herein for parenteral delivery for the purpose of inducing an antigen-specific immune response are typically 5 to 10,000 times higher than the effective mucosal dose for vaccine adjuvant or immune stimulant applications, and more typically 10 to 1,000 times higher, and most typically 20 to 100 times higher.
  • Doses of the compounds described herein for parenteral delivery for the purpose of inducing an innate immune response or for increasing ADCC or for inducing an antigen specific immune response when the CpG immunostimulatory oligonucleotides are administered in combination with other therapeutic agents or in specialized delivery vehicles typically range from about 1.0 ⁇ g to 100 mg per administration, which depending on the application could be given daily, weekly, or monthly and any other amount of time therebetween or as otherwise required. More typically parenteral doses for these purposes range from about 100 ⁇ g to 50 mg per administration, and most typically from about 200 ⁇ g to 2 mg, with 2 - 4 administrations being spaced days or weeks apart. In some embodiments, however, parenteral doses for these purposes may be used in a range of 5 to 10,000 times higher than the typical doses described above.
  • the therapeutically effective amount can be initially determined from animal models.
  • a therapeutically effective dose can also be determined from human data for other CpG oligonucleotides which have been tested in humans (human clinical trials are ongoing) and for compounds which are known to exhibit similar pharmacological activities, such as other adjuvants, e.g., LT and other antigens for vaccination purposes. Higher doses may be required for parenteral administration.
  • the applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.
  • compositions of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.
  • an effective amount of the CpG immunostimulatory oligonucleotide an/or other therapeutics can be administered to a subject by any mode that delivers the compound to the desired surface, e.g., local, mucosal, systemic.
  • Administering the pharmaceutical composition of the present invention may be accomplished by any means known to the skilled artisan.
  • Preferred routes of administration include but are not limited to oral, parenteral, intramuscular, intranasal, sublingual, intratracheal, inhalation, ocular, vaginal, and rectal.
  • the compounds i.e., CpG immunostimulatory oligonucleotides, antigens and/or other therapeutic agents
  • the compounds can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated.
  • Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the oral formulations may also be formulated in saline or buffers for neutralizing internal acid conditions or may be administered without any carriers.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds may be administered by inhalation to pulmonary tract, especially the bronchi and more particularly into the alveoli of the deep lung, using standard inhalation devices.
  • the compounds may be delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • An inhalation apparatus may be used to deliver the compounds to a subject.
  • An inhalation apparatus as used herein, is any device for administering an aerosol, such as dry powdered form of the compounds. This type of equipment is well known in the art and has been described in detail, such as that description found in Remington: The Science and Practice of
  • “Powder” as used herein refers to a composition that consists of finely dispersed solid particles. Preferably the compounds are relatively free flowing and capable of being dispersed in an inhalation device and subsequently inhaled by a subject so that the compounds reach the lungs to permit penetration into the alveoli.
  • a “dry powder” refers to a powder composition that has a moisture content such that the particles are readily dispersible in an inhalation device to form an aerosol. The moisture content is generally below about 10%> by weight (% w) water, and in some embodiments is below about 5% w and preferably less than about 3% w.
  • the powder may be formulated with polymers or optionally may be formulated with other materials such as liposomes, albumin and/or other carriers.
  • Aerosol dosage and delivery systems may be selected for a particular therapeutic application by one of skill in the art, such as described, for example in Gonda, I. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract,” in Critical Reviews in Therapeutic Drug Carrier Systems, 6:273-313 (1990), and in Moren, "Aerosol dosage forms and formulations,” in Aerosols in Medicine. Principles, Diagnosis and Therapy, Moren, et al, Eds, Esevier, Amsterdam, 1985.
  • the compounds when it is desirable to deliver them system ically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents .
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • suspensions of the active compounds may be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation.
  • Such long acting formulations may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin.
  • the pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above.
  • the pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see Langer, Science 249:1527-1533, 1990, which is incorporated herein by reference.
  • the CpG immunostimulatory oligonucleotides and optionally other therapeutics and/or antigens may be administered per se (neat) or in the form of a pharmaceutically acceptable salt.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (l-3%> w/v); boric acid and a salt (0.5-2.5%) w/v); and phosphoric acid and a salt (0.8-2%) w/v).
  • Suitable preservatives include benzalkonium chloride (0.003-0.03%) w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004- 0.02% w/v).
  • compositions of the invention contain an effective amount of a CpG immunostimulatory oligonucleotide and optionally antigens and/or other therapeutic agents optionally included in a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
  • ODNs Oligodeoxynucleotides All ODNs were provided by Coley Pharmaceutical GmbH (Langenfeld, Germany). ODNs were diluted in phosphate-buffered saline (Sigma, Germany), and stored at -20° C. All dilutions were carried out using pyrogen-free reagents. The ODNs used in the studies described below are shown in Table 1.
  • PBMC peripheral blood buffy coat preparations from healthy male and female human donors were obtained from the German Red Cross (Rathingen, Germany) or from the Blood Bank of the University of Dusseldorf (Germany) and from these, PBMC were purified by centrifugation over Ficoll-Hypaque (Sigma). The purified PBMC were either used fresh (for most assays) or were suspended in freezing medium and stored at -70°C.
  • Cytokine detection Thawed or fresh PBMC were resuspended at a concentration of 3xl0 6 /ml to 5xl0 6 /ml and added to plates which had previously received nothing or ODN in a variety of concentrations. The cells were cultured in a humidified incubator at 37°C. Culture supernatants were collected after the indicated time points. If not used immediately, supernatants were frozen at -20°C until required. Amounts of cytokines in the supernatants were assessed using commercially available ELISA Kits or in-house ELISA developed using commercially available antibodies (e.g. from Becton Dickinson, Germany).
  • Example 1 5'-TCG enhances immunostimulatory activity of non-CpG or CpG ODNs (IL-10).
  • Example 3 Enhancement of the immune response is dependent on the CpG dinucleotide.
  • Human PBMC of two representative donors were incubated for 48h with the indicated ODNs ( Figure 3).
  • Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. Shown are the effect of a 5' -TCG, SEQ. ID NO.: 2, and a poly T sequence, SEQ. ID NO.: 8.
  • a 5'-TGC, SEQ. ID NO.: 10 may have some minimal effect, the 5' -TCG modification clearly resulted in a much stronger potentiation of cytokine secretion.
  • a 5'-TCG modification of a 17mer poly A ODN did not appear to have an effect, SEQ.
  • Example 4 Shifting the CpG dinucleotide from the 5' to the 3' end of an
  • ODN results in graded loss of immunostimulatory capability.
  • Example 6 A 5'-TCG is the most stimulatory 5' modification, but other modifications also lead to enhanced immunostimulation.
  • Example 7 The 5'-TCG modification enhances the stimulatory capability of CpG ODNs as shown by different cellular effects.
  • Figures 7A to 7C demonstrate that the 5' -TCG is able to enhance the stimulatory capacity of an ODN in a variety of assays.
  • the parent CpG ODN SEQ. ID NO.: 18 does not have a CpG dinucleotide directly at the 5' end. Modifying the sequence with a 5'- TCG, SEQ. ID NO.: 19, also enhanced the activity of the CpG ODN.
  • Example 8 IL-10 secretion induced by ODN with 5'-TCG.
  • Example 9 IL-10 secretion induced by ODN with 5'-TCG and increasing numbers of thymidines.
  • a 5'-TCG was sufficient to enhance immunostimulation by phosphorothioate ODN independent of the nucleotide sequence. Nevertheless, an increasing number of pyrimidines further contributed to this stimulation. Only two to four thymidines 3' of the 5'-TCG (here: about at least 20% thymidines) were sufficient to lead to a significant increase of cytokine secretion (SEQ. ID NO.: 42 and SEQ. ID NO.: 43).
  • Example 10 ODN with a 5'-TCG are the most potent and efficient ODN to induce a strong Thl-mediated immune response.
  • Example 11 The position of CpG dinucleotides in immune stimulatory ODN determines the strength of type I IFN secretion.
  • Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations. Supernatants were harvested and IFN- ⁇ measured by ELISA as described in Materials and Methods. Shown is the Mean. Shifting the CpG dinucleotide (essential for efficient immune stimulation) from the 5' to the 3' end led to a graded loss of immune stimulation (measured as secretion of the B cell related cytokine IL-10).
  • Figure 11 demonstrates that the position of the CpG also strongly influences the strength of type I IFN secretion.
  • Example 12 Type I IFN secretion induced by short 5'-TCG ODN.
  • Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations.
  • Supernatants were harvested and IFN- ⁇ measured by ELISA as described in Materials and Methods. Shown is the Mean ⁇ SEM.
  • Previous findings have shown decrease of immune stimulation (measured as IL-10 secretion) upon shortening of the ODN's length. Nevertheless, when the secretion of IFN- ⁇ by shortened ODN's (e.g. 13mer SEQ. ID NO.: 32 with 5'-TCG) was measured, surprisingly a strongly increased IFN- ⁇ secretion compared to the 17mer SEQ. ID NO.: 2 was observed (Figure 12).
  • Example 13 In vitro immune stimulation by a panel of newly generated
  • a 5' TCG supports efficient and potent IFN- ⁇ (a Thl related cytokine) as well as IL-10 secretion (a B cell related cytokine); b. Shifting the CpG dinucleotide from the 5' to the 3' end led first to an increase of type I IFN secretion and further 3' shifts led to a decrease (B cell activation was only decreased or only slightly changed by CpG shifts); c. Shortening an ODN with a 5'-TCG led to a strong increase in the potential to induce IFN- ⁇ (in contrast to other effects, e.g. secretion of IL-10);
  • Example 14 Short CpG ODN are perfectly able to induce efficient B cell stimulation.
  • Example 15 A phosphodiester linkage between the C and G of the 5' CpG dinucleotide results in enhancement of potency of immune stimulation.
  • Example 16 Modifications of the T preceeding the 5'-CG are allowed.
  • an ODN with a 5'-UCG (SEQ. ID NO. 54) induced similar strong cytokine secretion as an ODN with a 5'-TCG (SEQ. ID NO.: 2). Both ODN were superior to a pure poly T ODN (SEQ. ID NO.: 8). This result suggests that a variety of chemical modified nucleotides 5' to the CpG are allowed to induce an enhanced immune stimulation.
  • ODN with a 5'-TCU (SEQ. ID NO. 55) or 5'-TUG (SEQ. ID NO. 56) also demonstrated enhanced cytokine secretion when compared to a poly T ODN (SEQ. ID NO.: 8).

Abstract

The invention relates to a class of CpG immunostimulatory oligonucleotides containing a 5’TCG motif or a CG at or near the 5’ end that are useful for stimulating an immune response.

Description

5' CPG NUCLEIC ACIDS AND METHODS OF USE
FIELD OF THE INVENTION The present invention relates generally to immunostimulatory nucleic acids, compositions thereof and methods of using the immunostimulatory nucleic acids.
BACKGROUND OF THE INVENTION Bacterial DNA has immune stimulatory effects to activate B cells and natural killer cells, but vertebrate DNA does not (Tokunaga, T., et al., 1988. Jpn. J. Cancer Res. 79:682-686; Tokunaga, T., et al, 1984, JNCI 12:955-962; Messina, J.P., et al., 1991, J. Immunol. 147:1759-1764; and reviewed in Krieg, 1998, hi: Applied Oligonucleotide Technology, CA. Stein and A.M. Krieg, (Eds.), John Wiley and Sons, Inc., New York, NY, pp. 431-448) and Krieg. A. M. CpG motifs in bacterial DNA and their immune effects (2002) Annu. Rev. Immunol. 20: 709-760. It is now understood that these immune stimulatory effects of bacterial DNA are a result of the presence of unmethylated CpG dinucleotides in particular base contexts (CpG motifs), which are common in bacterial DNA, but methylated and underrepresented in vertebrate DNA (Krieg et al, 1995 Nature 374:546-549; Krieg, 1999 Biochim. Biophys. Acta 93321:1- 10). The immune stimulatory effects of bacterial DNA can be mimicked with synthetic oligodeoxynucleotides (ODN) containing these CpG motifs. Such CpG ODN have highly stimulatory effects on human and murine leukocytes, inducing B cell proliferation; cytokine and immunoglobulin secretion; natural killer (NK) cell lytic activity and IFN-γ secretion; and activation of dendritic cells (DCs) and other antigen presenting cells to express costimulatory molecules and secrete cytokines, especially the Thl-like cytokines that are important in promoting the development of Thl-like T cell responses. These immune stimulatory effects of native phosphodiester backbone CpG ODN are highly CpG specific in that the effects are dramatically reduced if the CpG motif is methylated, changed to a GpC, or otherwise eliminated or altered (Krieg et al, 1995 Nature 374:546-549; Hartmann et al, 1999 Proc. Natl. Acad. Sci USA 96:9305-10). In early studies, it was thought that the immune stimulatory CpG motif followed the formula purine-purine-CpG-pyrimidine-pyrimidine (Krieg et al, 1995 Nature 374:546-549; Pisetsky, 1996 J. Immunol. 156:421-423; Hacker et al, 1998 EMBO J. 17:6230-6240; Lipford et al, 1998 Trends in Microbiol. 6:496-500). However, it is now clear that mouse lymphocytes respond quite well to phosphodiester CpG motifs that do not follow this "formula" (Yi et al, 1998 J. Immunol. 160:5898-5906) and the same is true of human B cells and dendritic cells (Hartmann et al, 1999 Proc. Natl. Acad. Sci USA 96:9305-10; Liang, 1996 J. Clin. Invest. 98:1119-1129). Nevertheless, the term "CpG motif is generally used to refer to a hexamer motif in which the CpG dinucleotide is located at the center.
SUMMARY OF THE INVENTION The invention involves the finding that specific sub-classes of CpG immunostimulatory oligonucleotides having a 5 'CpG are highly effective in mediating immune stimulatory effects. These CpG nucleic acids are useful therapeutically and prophylactically for stimulating the immune system to treat cancer, infectious diseases, allergy, asthma and other disorders and to help protect against opportunistic infections following cancer chemotherapy. The strong yet balanced, cellular and humoral immune responses that result from CpG stimulation reflect the body's own natural defense system against invading pathogens and cancerous cells.
In particular, immunostimulatory CpG containing oligonucleotides having a 5'TCG motif, rather than the conventional hexamer motif have important therapeutic properties. It has been discovered that oligonucleotides having a '5TCG motif without any additional unmethylated CpG motifs have strong immunostimulatory capability. In one aspect the invention is a composition comprising an oligonucleotide: 5'TCGXιX2Nι3', wherein Ni is 2-95 nucleotides and, when Xi is C or A, X2 is A, T, or C (SEQ. ID NO.: 61); when X] is T, X2 is A or G (SEQ. ID NO.: 62); and when Xi is G, X2 is any nucleotide (SEQ. ID NO.: 63). The invention, in other aspects, relates to an oligonucleotide comprising
5'TCGTNι3' (SEQ. ID NO.: 64). In the oligonucleotide N, is 3-96 nucleotides, but when Ni is 16 nucleotides Ni does not include a C,2 (5'-CCCCCCCCCCC-3' SEQ. ID NO.: 65), and when Ni is 8 nucleotides N, is at least 50% C or 70% T (SEQ. ID NO.: 66). According to other aspects, an oligonucleotide comprising 5'TCGANι3' (SEQ.
ID NO.: 67) is provided. In the oligonucleotide Ni is 3-96 nucleotides, but when Ni is 19 nucleotides Ni is at least 55% pyrimidine (SEQ. ID NO.: 68), and when Ni is 8 nucleotides Ni is at least 50% T or C (SEQ. ID NO.: 69).
According to other aspects, an oligonucleotide comprising 5'TCGN]3' is provided. In the oligonucleotide Ni is 10-96 nucleotides, and the C content of the oligonucleotide is less than or equal to 60%, and the A content of the oligonucleotide is less than or equal to 30%.
According to other aspects, an oligonucleotide is provided that comprises 5'TYZNι3'. In the oligonucleotide Ni is 4-97 nucleotides, and the oligonucleotide does not include an unmethylated CG motif. Y is a cytosine or modified cystosine. Z is a guanine or modified guanine. In one embodiment Y is 5'methyl cytosine, 5-methyl- deoxycytosine, 5-methyl-deoxyisocytosine, 5-hydroxy-deoxycytosine, deoxyuridine, N4- ethyl-deoxycytosine, 2'-deoxyuridine, 5-fluoro-2'-dU, and dSpacer. In other embodiments Z is 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, hypoxanthine, 2,6-diaminopurine, 2-aminopurine, purine, 8-substituted guanine such as
8-hydroxyguanine, and 6-thioguanine, Inosine, 2-aminopurine, nebularine, and dSpacer. In the oligonucleotide formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide.
In some embodiment the oligonucleotide has one of the following structures: 5' χ*C*G*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T 3' (SEQ. ID NO.: 50) or 5' T*C*G*T*T*T*T*T*T*T*T*T* τ*τ*τ*τ*τ 3. (SEQ_ ID N0 . 2) The * refers to a phosphorothioate linkage.
According to one embodiment the oligonucleotide includes at least 1 modified internucleotide linkage. In other embodiments the oligonucleotide includes at least 50% modified internucleotide linkages. Optionally all internucleotide linkages of the oligonucleotide are modified. The stabilized internucleotide linkage may be a phosphorothioate linkage.
In some embodiments the oligonucleotide is 20-100 nucleotides in length. In other embodiments it is 40 or less nucleotides in length. Ni is free of unmethylated CG motifs. Ni may be defined by N2N3, such that N2 is 8-94 nucleotides, or in some embodiments 8-40 nucleotides, and N3 is 2-5 pyrimidines. In some embodiments N3 is TTTTT, TTTT, TTT, or TT. i, according to other embodiments, may be at least 50% pyrimidine or at least 80% pyrimidine. In yet other embodiments Ni is free of Poly-A and Poly-G sequences. In other embodiments Ni is TN2 and N2 is 8-94 nucleotides.
The invention involves, in one aspect, the discovery that the 5' sequence of immunostimulatory nucleotides, their length and internucleotide linkage have specific influences on the cytokine profile of the induced immune response and that these discoveries can be used to design a subset of CpG immunostimulatory oligonucleotides that have improved immune stimulatory properties. The preferred CpG immunostimulatory oligonucleotides fall within one of the following 6 general formulas: 5'-XιYRMι-3', 5'-X2CGM2-3', 5'-X3CGM3-3', 5'-X4CGM4-3', 5'-X5CGM5-3' and 5'- TTGM6-3'. The formulas define subsets of the class of CpG oligonucleotides which demonstrated excellent immune stimulating properties and yet do not include additional unmethylated CpG motifs. In the formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide. In one aspect of the invention the ODN has the general formula 5 ' -Xj YRM. -3 ' , wherein Xi is a single nucleotide; Y is a cytosine or a modified cytosine; R is a guanine or a modified guanine; and Mj is a nucleic acid of 1-3 nucleotides. According to other embodiments of the invention, the internucleotide linkages of the oligonucleotide are all stabilized phosphorothioate internucleotide linkages. In one embodiment, the internucleotide linkage between Y and R is a phosphodiester linkage in an Rp configuration. In some embodiments of the invention, the modified cytosine has a C5 substitution and/or the modified guanine has a C8 or C7 substitution. In certain embodiments of the invention, the substituted or modified C or G is selected from the group consisting of 5-substituted cytosines (e.g. 5-methyl-cytosine, 5-fluoro-cytosine, 5- chloro-cytosine, 5-bromo-cytosine, 5-iodo-cytosine, 5-hydroxy-cytosine, 5- hydroxymethyl-cytosine, 5-difiuoromethyl-cytosine, and unsubstituted or substituted 5- alkynyl-cytosine), 6-substituted cytosines, N4-substituted cytosines (e.g. N4-ethyl- cytosine), 5-aza-cytosine, 2-mercapto-cytosine, isocytosine, pseudo-isocytosine, cytosine analogs with condensed ring systems (e.g. N,N' -propyl ene cytosine or phenoxazine), and uracil and its derivatives (e.g. 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4- thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil), thymine derivatives (e.g. 2- thiothymine, 4-thiothymine, 6-substituted thymines), 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, 7-deaza-8-aza guanine, hypoxanthine, N2-substituted guanines (e.g. N2-methyl-guanine), 5-amino-3-methyl-3H,6H-thiazolo[4,5- d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, adenine, substituted adenines (e.g. N6-methyl-adenine, 8-oxo-adenine) 8-substituted guanine (e.g. 8 -hydroxy guanine and 8-bromoguanine), and 6-thioguanine. In another embodiment of the invention, the base is substituted by a universal base (e.g. 4-methyl-indole, 5-nitro- indole, 3-nitropyrrole, P-base, and K-base), an aromatic ring system (e.g. benzimidazole or dichloro- benzimidazole, l-methyl-lH-[l,2,4Jtriazole-3-carboxylic acid amide) an aromatic ring system (e.g. fluorobenzene or difluorobenzene) and a hydrogen atom (dSpacer). According to one embodiment of the invention the oligonucleotide is associated with a carrier linked to the 3' end of the oligonucleotide. In some embodiments, the carrier is selected from the group consisting of a microparticle, dendrimer, liposome, cationic complex, and antigen. In yet another embodiment of the invention, the ODN is administered to the subject along with an antigen. In still another embodiment the CpG immunostimulatory oligonucleotides are useful for treating subjects in combination with the administration of a therapeutic protocol to the subject. In some embodiments of the invention, the therapeutic protocol is surgery.
In some embodiments the oligonucleotide is not associated with a carrier. In other embodiments the oligonucleotide is in a multimerized complex. Optionally the multimerized complex includes the oligonucleotide linked by a multimerization unit to a second oligonucleotide. The second oligonucleotide may have the formula 5'-XιYRMι- 3'.
In one aspect the immunostimulatory oligonucleotide of the invention has the general formula 5'-X2YRM2-3' with a multimerization unit linked to the 3' end of the oligonucleotide. X2 is a nucleic acid that consists of a single nucleotide, or a dinucleotide or a trinucleotide that does not comprise a CG dinucleotide. Y is a cytosine or a modified cytosine. R is a guanine or a modified guanine. M2 is a nucleic acid of 0- 27 nucleotides. In some embodiments the immunostimulatory oligonucleotides have the following structures: 5'-TCG-3', 5'-TCGT-3', 5'-UCG-3', 5'-UCGT-3'. In yet another embodiment M2 is free of a CG dinucleotide. According to another embodiment of the invention X2 is a single nucleotide, and X2 is a pyrimidine. According to other embodiments of the invention, the internucleotide linkages of the oligonucleotide are all stabilized phosphodiester internucleotide linkages.
In some embodiments the multimerization unit is a carrier selected from the group consisting of a microparticle, dendrimer, liposome, cationic complex, cholesterol and antigen. In other embodiments the multimerization unit is a linker between the 3' end of the oligonucleotide and a second oligonucleotide.
In yet another embodiment of the invention, the ODN is administered to the subject along with an antigen. In still another embodiment the CpG immunostimulatory oligonucleotides are useful for treating subjects in combination with the administration of a therapeutic protocol to the subject. In some embodiments of the invention, the therapeutic protocol is surgery.
According to another aspect of the invention the immunostimulatory oligonucleotide has the general formula 5'-X CGM3-3', wherein X is a single nucleotide that does not comprise a CG dinucleotide; M3 is a nucleic acid of 3-27 nucleotides that is free of a CG dinucleotide, and M has at least one of the following properties: is free of a TC dinucleotide, is at least 30% T nucleotides, consists of A, T, and G or is free of a CCTTCC hexamer having at least one modified internucleotide linkage. In some embodiments M3 has at least 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%>, 80%, 85%, 90%, 95% or 100% T or modified versions thereof. In another aspect the immunostimulatory oligonucleotide has the general formula
5'-X CGM -3', wherein X4 is a dinucleotide that does not comprise a CG dinucleotide, and M4 is a nucleic acid of 2-26 nucleotides that is free of a CG dinucleotide and it has at least one of the following properties: is free of a TG or a GT dinucleotide, is at least 38% T nucleotides or consists of A and T. In some embodiments M4 has at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% A or T or modified versions thereof.
In yet another aspect the immunostimulatory oligonucleotide has the general formula 5'-X5CGM5-3', X5 is a trinucleotide that does not comprise a CG dinucleotide; M5 is a nucleic acid of 1-25 nucleotides that is free of a CG dinucleotide, and wherein Ms has at least one of the following properties: is free of a CT dinucleotide and does not include at least one phosphorothioate linkage, is at least 41% T nucleotides, or consists of A and C. In some embodiments M4 has at least 45%, 50%, 55%, 60%, 65%, 70%, 75%>, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
According to another aspect of the invention the immunostimulatory oligonucleotide has the general formula 5'-TTGM6-3', M6 is a nucleic acid that consists of 5-21 nucleotides, wherein M6 does not comprise a CG dinucleotide, wherein M6 is comprised of at least 30% T nucleotides, and wherein said nucleotide is 10-24 nucleotides in length. In some embodiments M4 has at least 35%, 40%, 45%, 50%o, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% T or modified versions thereof. In some embodiments the oligonucleotide has one of the following structures:
5'-T*C*G*T*T*T*T*T*T*T*T*T*T-3' (SEQ. ID NO.: 32) e > THST*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T-3' fSEO ID NO 27^ ^-_γΦ *^Φ ΦQ*' *' 'Φ' *' *' *r Φrj,*τ*T*T*T*T-3' fSEO ID NO 28^ The symbol * refers to the presence of a stabilized internucleotide linkage and _: refers to the presence of a phosphodiester linkage.
An oligonucleotide comprising: 5 '-X6CGM7-3' is provided according to an aspect of the invention. 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide. X6 is 1-3 nucleotides and does not include a CG dinucleotide. M7 is a nucleic acid of 6-27 nucleotides and includes at least three CG dinucleotides and is at least 50%) T nucleotides. In one embodiment M7 is 16-18 nucleotides in length.
In some embodiments M includes at least four CG dinucleotides. In other embodiments at least one CG dinucleotide includes a phosphodiester internucleotide linkage. Optionally at least three CG dinucleotides includes a phosphodiester internucleotide linkage. The oligonucleotide may be selected from the group consisting of SEQ ID NO. 33, 34, 35, 36, and 37.
In another aspect the invention is an oligonucleotide comprising: 5'-'TTGM8-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein M7 is a nucleic acid of 6-18 nucleotides and includes at least one CG dinucleotide and is at least 50% T nucleotides. Optionally M8 is 14 nucleotides in length. The immunostimulatory oligonucleotides generally have a length in the range of between 3 and 35 nucleotides. In some embodiments the length is in the range of 4-6, 3- 32, 6-30, or 10-24 nucleotides or any integer range therebetween.
According to one embodiment the oligonucleotide includes at least 1 modified internucleotide linkage. In other embodiments the oligonucleotide includes at least 50% modified internucleotide linkages. Optionally all internucleotide linkages of the oligonucleotide are modified. The stabilized internucleotide linkage may be a phosphorothioate linkage.
In another aspect, the invention relates to a method for treating allergy or asthma. The method is performed by administering to a subject having or at risk of having allergy or asthma an immunostimulatory CpG oligonucleotide described herein in an effective amount to treat allergy or asthma. In one embodiment the oligonucleotide is administered to a mucosal surface, such as a respiratory tissue. In other embodiments the oligonucleotide is administered in an aerosol formulation. Optionally the oligonucleotide is administered intranasally. In other embodiments the subject has or is at risk of developing allergic asthma.
A method for inducing cytokine production is provided according to another aspect of the invention. The method is performed by administering to a subject an immunostimulatory CpG oligonucleotide described herein in an effective amount to induce a cytokine selected from the group consisting of IP10, IL6, IL 8, IL12, IL18, TNF, IFN-α, chemokines, and IFN-γ.
In another aspect the invention is a composition of the CpG immunostimulatory oligonucleotides described herein in combination with an antigen or other therapeutic compound, such as an anti-microbial agent or an anti-cancer agent. The anti-microbial agent may be, for instance, an anti-viral agent, an anti-parasitic agent, an anti-bacterial agent or an anti-fungal agent.
The composition may optionally include a pharmaceutical carrier and/or be formulated in a delivery device. In some embodiments the delivery device is selected from the group consisting of cationic lipids, cell permeating proteins, and sustained release devices. In one embodiment the sustained release device is a biodegradable polymer or a microparticle. According to another aspect of the invention a method of stimulating an immune response is provided. The method involves administering a CpG immunostimulatory oligonucleotide to a subject in an amount effective to induce an immune response in the subject. Preferably the CpG immunostimulatory oligonucleotide is administered orally, locally, in a sustained release device, mucosally, systemically, parenterally, or intramuscularly. When the CpG immunostimulatory oligonucleotide is administered to the mucosal surface it may be delivered in an amount effective for inducing a mucosal immune response or a systemic immune response. In preferred embodiments the mucosal surface is an oral, nasal, rectal, vaginal, or ocular surface. In some embodiments the method includes exposing the subject to an antigen wherein the immune response is an antigen-specific immune response. In some embodiments the antigen is selected from the group consisting of a tumor antigen, a viral antigen, a bacterial antigen, a parasitic antigen and a peptide antigen.
CpG immunostimulatory oligonucleotides are capable of provoking a broad spectrum of immune response. For instance these CpG immunostimulatory oligonucleotides can be used to redirect a Th2 to a Thl immune response. CpG immunostimulatory oligonucleotides may also be used to activate an immune cell, such as a lymphocyte (e.g., B and T cells), a dendritic cell, and an NK cell. The activation can be performed in vivo, in vitro, or ex vivo, i.e., by isolating an immune cell from the subject, contacting the immune cell with an effective amount to activate the immune cell of the CpG immunostimulatory oligonucleotide and re-administering the activated immune cell to the subject, hi some embodiments the dendritic cell presents a cancer antigen. The dendritic cell can be exposed to the cancer antigen ex vivo.
The immune response produced by CpG immunostimulatory oligonucleotides may also result in induction of cytokine production, e.g., production of IP 10, IL6, IL 8, IL12, IL18, TNF, IFN-α, chemokines, and IFN-γ.
In still another embodiment, the CpG immunostimulatory oligonucleotides are useful for treating cancer in a subject having or at risk of developing a cancer. The cancer may be selected from the group consisting of biliary tract cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, gastric cancer, intraepithelial neoplasms, lymphomas, liver cancer, lung cancer (e.g. small cell and non-small cell), melanoma, neuroblastomas, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, sarcomas, thyroid cancer, and renal cancer, as well as other carcinomas and sarcomas. In some important embodiments, the cancer is selected from the group consisting of bone cancer, brain and CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer.
CpG immunostimulatory oligonucleotides may also be used for increasing the responsiveness of a cancer cell to a cancer therapy (i.e., an anti-cancer therapy), optionally when the CpG immunostimulatory oligonucleotide is administered in conjunction with an anti-cancer therapy. The anti-cancer therapy may be, for instance, a chemotherapy, a vaccine (e.g., an in vitro primed dendritic cell vaccine or a cancer antigen vaccine) or an immunotherapeutic agent such as an antibody based therapy. This latter therapy may also involve administering an antibody specific for a cell surface antigen of, for example, a cancer cell, wherein the immune response results in antibody dependent cellular cytotoxicity (ADCC). In one embodiment, the antibody may be selected from the group consisting of Ributaxin, Herceptin, Quadramet, Panorex, IDEC- Y2B8, BEC2, C225, Oncolym, SMART M195, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-11, MDX-22, OV103, 3622W94, anti-VEGF, Zenapax, MDX- 220, MDX-447, MELIMMUNE-2, MELIMMUNE-1, CEACIDE, Pretarget, NovoMAb- G2, TNT, Gliomab-H, GNI-250, EMD-72000, LymphoCide, CMA 676, Monopharm-C, 4B5, ior egf.r3, ior c5, BABS, anti-FLK-2, MDX-260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab and ImmuRAIT-CEA.
Thus, according to some aspects of the invention, a subject having cancer or at risk of having a cancer is administered a CpG immunostimulatory oligonucleotide and an anti-cancer therapy. In some embodiments, the anti-cancer therapy is selected from the group consisting of a chemotherapeutic agent, an immunotherapeutic agent and a cancer vaccine.
In still another embodiment of the methods directed to treating cancer, the subject may be further administered interferon-α.
In other aspects, the invention is a method for inducing an innate immune response by administering to the subject a CpG immunostimulatory oligonucleotide in an amount effective for activating an innate immune response. According to another aspect of the invention a method for treating a viral or retroviral infection is provided. The method involves administering to a subject having or at risk of having a viral or retroviral infection, an effective amount for treating the viral or retroviral infection of any of the compositions of the invention. In some embodiments the virus is caused by a hepatitis virus e.g., hepatitis B, hepatitis C, HIV, herpes virus, or papillomavirus.
A method for treating a bacterial infection is provided according to another aspect of the invention. The method involves administering to a subject having or at risk of having a bacterial infection, an effective amount for treating the bacterial infection of any of the compositions of the invention. In one embodiment the bacterial infection is due to an intracellular bacteria.
In another aspect the invention is a method for treating a parasite infection by administering to a subject having or at risk of having a parasite infection, an effective amount for treating the parasite infection of any of the compositions of the invention. In one embodiment the parasite infection is due to an intracellular parasite. In another embodiment the parasite infection is due to a non-helminthic parasite.
In some embodiments the subject is a human and in other embodiments the subject is a non-human vertebrate such as a dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, or sheep. In another aspect the invention relates to a method for inducing a TH1 immune response by administering to a subject any of the compositions of the invention in an effective amount to produce a TH1 immune response.
Each of the limitations of the invention can encompass various embodiments of the invention. It is, therefore, anticipated that each of the limitations of the invention involving any one element or combinations of elements can be included in each aspect of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS The present invention may be more easily and completely understood when taken in conjunction with the accompanying figures. Figure 1 is a bar graph depicting the effect of a 5'-TCG motif on the immunostimulatory activity of non-CpG or CpG ODNs through induction of IL-10. . Figure 2 is a bar graph depicting effect of a 5'-TCG motif on the immunostimulatory activity of non-CpG or CpG ODNs through induction of IFN-α.
Figure 3 is a bar graph depicting the effect of a 5'TCG on Poly-A and Poly-T sequences.
Figure 4 is a bar graph depicting the effect of shifting the CpG dinucleotide from the 5' to the 3' end of an ODN.
Figure 5 is a bar graph demonstrating that the length of an ODN has an effect on stimulatory activity in addition to a 5'-TCG.
Figure 6 is a bar graph depicting the effect of other 5' modifications in addition to 5'-TCG.
Figure 7 is a set of bar graphs depicting the effect of a 5' -TCG modification on stimulatory capability of CpG ODNs as shown by different cellular effects: 7A (IL-10 induction) 7B (IFN-α induction) and 7C (IL-6 induction).
Figure 8 is a set of bar graphs that shows IL-10 secretion induced by ODN with 5' -TCG.
Figure 9 is a set of bar graphs that shows IL-10 secretion induced by ODN with 5'-TCG and increasing numbers of thymidines.
Figure 10 is set of bar graphs that depicts ODN's with a 5'-TCG as the most potent and efficient ODN's to induce a strong Thl-mediated immune response: 10A (IL- 10 induction) and 10B (IFN-α induction).
Figure 11 is a set of bar graphs that depicts how the position of CpG dinucleotides in immune stimulatory ODN determines the strength of type I IFN secretion.
Figure 12 is a set of bar graphs that shows type I IFN secretion induced by short 5'-TCG ODN's.
Figure 13 is a set of bar graphs that shows the in vitro immune stimulation by a panel of newly generated CpG ODN's according to the observations described herein: 13A (IL-10 induction) and 13B (IFN-α induction).
Figure 14 is a set of bar graphs depicting B cell stimulation by short CpG ODN's. Figure 15 is a bar graph that shows IL-10 induction by a panel of CpG ODN's and which demonstrates that some ODN having phosphodiester linkage between C and G have increased potency.
Figure 16 is a bar graph that shows IL-10 induction by a panel of CpG ODN's and which demonstrates that some ODN having a modified 5 ' TCG induce IL- 10.
DETAILED DESCRIPTION The invention in one aspect involves the finding that specific sub-classes of CpG immunostimulatory oligonucleotides having a 5'TCG are highly effective in mediating immune stimulatory effects. These CpG nucleic acids are useful therapeutically and prophylactically for stimulating the immune system to treat cancer, infectious diseases, allergy, asthma and other disorders and to help protect against opportunistic infections following cancer chemotherapy. The strong yet balanced, cellular and humoral immune responses that result from CpG stimulation reflect the body's own natural defense system against invading pathogens and cancerous cells.
The invention involves, in one aspect, the discovery that a subset of CpG immunostimulatory oligonucleotides have improved immune stimulatory properties. The preferred CpG immunostimulatory oligonucleotides fall within one of the following 5 general formulas: 5'TCGXιX2Nj3', 5'TCGTN]3', 5'TCGANι3\ 5'TCGNι3' and 5'TYZNι3' (SEQ. ID NO.: 61-69). Xi and X2 refer to single nucleotides.
The formulas define subsets of the class of CpG oligonucleotides which demonstrated excellent immune stimulating properties and yet do not include additional unmethylated CpG motifs. In the formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide.
Ni encompasses a variable set of nucleotide sequences. The nucleotide sequences may range from 2-97 nucleotides in length or any integer range therebetween. The findings of the invention are based in part on the discovery of the importance of the positional effects of CpG or YpZ motif. It has been discovered that oligonucleotides having a 5'TCG or 5' TYZ without any additional unmethylated CpG motifs therein are strong immunostimulatory capability. The remainder of the oligonucleotide may be any combination of nucleotides or modified nucleotides as long as the 5' end of the molecule includes the requisite motif.
It has also been discovered that some sequences of N]5 when combined with the 5'TCG or 5' TYZ produce molecules having even greater immunostimulatory activity. For instance, when N] is at least 50% pyrimidine the oligonucleotide produces enhanced Thl biased immune induction. In some embodiments Nj is at least 55%, 60%, 65%, 70%, 75%>, 80%, 85%), 90%, 95%, or 100 pyrimidine, e.g. C or T. A pyrimidine is T or C or modified versions thereof. In some embodiments the 3' most nucleotides of Ni are pyrimidines. For instance the 3' end may be TTTTT, TTTT, TTT, TT, T, CCCCC, CCCC, CCC, CC, C, CTT, CCTT, or any other possible combination of pyri idines. In some limited embodiments Ni is free of a C.2 (5'-CCCCCCCCCCCC-35 (SEQ. ID NO.: 65)).
The invention involves, in one aspect, the discovery that the 5' sequence of immunostimulatory nucleotides, their length and internucleotide linkage have specific influences on the cytokine profile of the induced immune response and that these discoveries can be used to design a subset of CpG immunostimulatory oligonucleotides that have improved immune stimulatory properties. The preferred CpG immunostimulatory oligonucleotides fall within one of the following 6 general formulas: 5'-XιYRMι-3', 5'-X2CGM2-3', 5'-X3CGM3-3', 5'-X4CGM4-3', 5'-X5CGM5-3' and 5'- TTGMe-3'.
The formulas define subsets of the class of CpG oligonucleotides which demonstrated excellent immune stimulating properties and yet do not include additional unmethylated CpG motifs, hi the formulas 5' refers to the free 5' end of the oligonucleotide and 3' refers to the free 3' end of the oligonucleotide. In the preferred embodiment with the general formula 5'-XιYRMι-3', Xi is a single nucleotide; Y is a cytosine or a modified cytosine; R is a guanine or a modified guanine; and Mi is a nucleic acid of 1-3 nucleotides. For example, such a oligonucleotide can be
In the preferred embodiment with the general formula 5'-X2CGM2-3', X2 is a nucleic acid that consists of a single nucleotide, or a dinucleotide or a trinucleotide that does not comprise a CG dinucleotide; and M2 is a nucleic acid of 0-27 nucleotides. In some embodiments the oligonucleotides have the following structures: 5'-TCG-3', 5'- TCGT-3', 5'-UCG-3', 5'-UCGT-3'. In other preferred embodiments M2 is free of a CG dinucleotide.
In the preferred embodiment with the general formula 5'-X3CGM3-3', X3 is a single nucleotide that does not comprise a CG dinucleotide; M3 is a nucleic acid of 3-27 nucleotides that is free of a CG dinucleotide, and M3 has at least one of the following properties: is free of a TC dinucleotide, is at least 30% T nucleotides, consists of A, T, and G or is free of a CCTTCC hexamer having at least one modified internucleotide linkage. In some embodiments M3 has at least 35%>, 40%, 45%, 50%, 55%, 60%>, 65%, 70%), 75%, 80%, 85%), 90%, 95%> or 100%) T or modified versions thereof. In the preferred embodiment with the general formula 5 ' -X4CGM4-3 ' , X4 is a dinucleotide that does not comprise a CG dinucleotide, and M4 is a nucleic acid of 2-26 nucleotides that is free of a CG dinucleotide and it has at least one of the following properties: is free of a TG or a GT dinucleotide, is at least 38% T nucleotides or consists of A and T. In some embodiments M4 has at least 40%, 45%>, 50%, 55%>, 60%, 65%>, 70%, 75%o, 80%, 85%, 90%>, 95% or 100% A or T or modified versions thereof.
In the preferred embodiment with the general formula 5'-X5CGM5-3', X5 is a trinucleotide that does not comprise a CG dinucleotide; Ms is a nucleic acid of 1-25 nucleotides that is free of a CG dinucleotide, and wherein Ms has at least one of the following properties: is free of a CT dinucleotide and does not include at least one phosphorothioate linkage, is at least 41%> T nucleotides, or consists of A and C. In some embodiments M4 has at least 45%, 50%, 55%>, 60%>, 65%, 70%, 75%, 80%, 85%, 90%), 95% or 100%) T or modified versions thereof.
In the preferred embodiment with the general formula 5'-TTGM6-3', M6 is a nucleic acid that consists of 5-21 nucleotides, wherein M6 does not comprise a CG dinucleotide, wherein M6 is comprised of at least 30%> T nucleotides, and wherein said nucleotide is 10-24 nucleotides in length. In some embodiments M4 has at least 35%>, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% T or modified versions thereof.
In some embodiment the oligonucleotide has one of the following structures: 5>.T=. c*G*T*T*T*T*T*T*T*T*T:t:T-3' (SEQ. ID O.: 32)
5>_' #' *C* *'P*T*T*T*T*T*T*T*T*T*T*T*T-3' ($EO ID NO - 27) ;_ >i;^Φτ*(^* *τ*τ*χ*χ*τ*τ* * *τ*τ* *, -.3' (SEO ID NO ' 28^ The symbol * refers to the presence of a stabilized internucleotide linkage and __: refers to the presence of a phosphodiester linkage.
The oligonucleotides may have one or two accessible 5' ends. Since the importance of the 5'TCG and 5'TYZ motif has been discovered, it also possible to create modified oligonucleotides having two such 5' ends. This may be achieved, for instance by attaching two oligonucleotides through a 3'-3' linkage to generate an oligonucleotide having one or two accessible 5' ends. Such a structure might have a formula such as 5 CGNι-NιGCT5' (SEQ. ID NO.: 13). The 3'3'-linkage may be a phosphodiester, phosphorothioate or any other modified internucleoside bridge. Methods for accomplishing such linkages are known in the art. For instance, such linkages have been described in Seliger, H.; et al, Oligonucleotide analogs with terminal 3'-3'- and 5'-5'- internucleotidic linkages as antisense inhibitors of viral gene expression, Nucleosides & Nucleotides (1991), 10(1-3), 469-77 and Jiang, et al. Pseudo-cyclic oligonucleotides: in vitro and in vivo properties, Bioorganic & Medicinal Chemistry (1999), 7(12), 2727- 2735.
Additionally, 3 '3 '-linked ODNs where the linkage between the 3 '-terminal nucleosides is not a phosphodiester, phosphorothioate or other modified bridge, can be prepared using an additional spacer, such as tri- or tetra-ethylenglycol phosphate moiety (Durand, M. et al, Triple-helix formation by an oligonucleotide containing one (dA)12 and two (dT)12 sequences bridged by two hexaethylene glycol chains, Biochemistry (1992), 31(38), 9197-204, US Patent No. 5658738, and US Patent No. 5668265). Alternatively, the non-nucleotidic linker may be derived from ethanediol, propanediol, or from an abasic deoxyribose (dSpacer) unit (Fontanel, Marie Laurence et al, Sterical recognition by T4 polynucleotide kinase of non-nucleosidic moieties 5'-attached to oligonucleotides; Nucleic Acids Research (1994), 22(11), 2022-7) using standard phosphoramidite chemistry. The non-nucleotidic linkers can be incorporated once or multiple times, or combined with each other allowing for any desirable distance between the 3'-ends of the two ODNs to be linked.
The oligonucleotide in some embodiments does not include an unmethylated CG motif, other than the 5 'TCG.
In some embodiment the oligonucleotide has one of the following structures: 5' T*C*G*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T (SEQ. ID NO.: 50), χ*C*G*C*C*C*C*C*C*C*C*C*C*C*C*C*C (SEQ jo N0.: 51), T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T:1:T*T*T*T fSEO ID NO " 13^ T*C*G*U*U*U*U*U*U*U*U*U*U*U*U*U*U (SEQ. ID NO.: 48), T*C Q* *χ*χ*τ*τ*χ* *τ*T*T*T*T*T*T fSEO ID NO 25^ ^*G*Q*χ*γ* *χ*χ* * * *r ,*r *' *'τ*' *rj' ' SEO ID NO - 14)
The symbol * refers to the presence of a stabilized internucleotide linkage and _: refers to the presence of a phosphodiester linkage.
The immunostimulatory oligonucleotides generally have a length in the range of between 7 and 100 nucleotides. In some embodiments the length is in the range of 7-40, 13-100, 13-40, 13-30, 15-50, or 15- 30 nucleotides or any integer range therebetween. In some preferred embodiments the oligonucleotide is associated with a carrier linked to the 3' end by, but not limited to, the aforementioned linkers and methods. The carrier can be selected from but not limited to the group consisting of microparticles, dendrimers, liposomes, cationic complexes and antigens. The terms "nucleic acid" and "oligonucleotide" are used interchangeably to mean multiple nucleotides (i.e., molecules comprising a sugar (e.g., ribose or deoxyribose) linked to a phosphate group and to an exchangeable organic base, which is either a substituted pyrimidine (e.g., cytosine (C), thymine (T) or uracil (U)) or a substituted purine (e.g., adenine (A) or guanine (G)). As used herein, the terms "nucleic acid" and "oligonucleotide" refer to oligoribonucleotides as well as oligodeoxyribonucleotides.
The terms "nucleic acid" and "oligonucleotide" shall also include polynucleosides (i.e., a polynucleotide minus the phosphate) and any other organic base containing polymer. Nucleic acid molecules can be obtained from existing nucleic acid sources (e.g., genomic or cDNA), but are preferably synthetic (e.g., produced by nucleic acid synthesis). The terms "nucleic acid" and "oligonucleotide" also encompass nucleic acids or oligonucleotides with substitutions or modifications, such as in the bases and/or sugars. For example, they include nucleic acids having backbone sugars that are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 2' position and other than a phosphate group or hydroxy group at the 5' position. Thus modified nucleic acids may include a 2'-O-alkylated ribose group. In addition, modified nucleic acids may include sugars such as arabinose or 2'-fluoiOarabinose instead of ribose. Thus the nucleic acids may be heterogeneous in backbone composition thereby containing any possible combination of polymer units linked together such as peptide- nucleic acids (which have an amino acid backbone with nucleic acid bases). Other examples are described in more detail below.
The immunostimulatory oligonucleotides of the instant invention can encompass various chemical modifications and substitutions, in comparison to natural RNA and DNA, involving a phosphodiester internucleoside bridge, a β-D-ribose unit and/or a natural nucleoside base (adenine, guanine, cytosine, thymine, uracil). Examples of chemical modifications are known to the skilled person and are described, for example, in Uhlmann E et al. (1990) Chem Rev 90:543; "Protocols for Oligonucleotides and Analogs" Synthesis and Properties & Synthesis and Analytical Techniques, S. Agrawal, Ed, Humana Press, Totowa, USA 1993; Crooke ST et al. (1996) Annu Rev Pharmacol Toxicol 36: 107-129; and Hunziker J et al. (1995) Mod Synth Methods 7:331-417. An oligonucleotide according to the invention may have one or more modifications, wherein each modification is located at a particular phosphodiester internucleoside bridge and/or at a particular β-D-ribose unit and/or at a particular natural nucleoside base position in comparison to an oligonucleotide of the same sequence which is composed of natural DNA or RNA.
For example, the oligonucleotides may comprise one or more modifications and wherein each modification is independently selected from: a) the replacement of a phosphodiester internucleoside bridge located at the 3' and/or the 5' end of a nucleoside by a modified internucleoside bridge, b) the replacement of phosphodiester bridge located at the 3' and/or the 5' end of a nucleoside by a dephospho bridge, c) the replacement of a sugar phosphate unit from the sugar phosphate backbone by another unit, d) the replacement of a β-D-ribose unit by a modified sugar unit, and e) the replacement of a natural nucleoside base by a modified nucleoside base. More detailed examples for the chemical modification of an oligonucleotide are as follows. The oligonucleotides may include modified internucleotide linkages, such as those described in a or b above. These modified linkages may be partially resistant to degradation (e.g., are stabilized). A "stabilized oligonucleotide molecule" shall mean an oligonucleotide that is relatively resistant to in vivo degradation (e.g. via an exo- or endo- nuclease) resulting form such modifications. Oligonucleotides having phosphorothioate linkages, in some embodiments, may provide maximal activity and protect the oligonucleotide from degradation by intracellular exo- and endo-nucleases. A phosphodiester internucleoside bridge located at the 3' and/or the 5' end of a nucleoside can be replaced by a modified internucleoside bridge, wherein the modified internucleoside bridge is for example selected from phosphorothioate, phosphorodithioate, NR1R2-phosphoramidate, boranophosphate, α-hydroxybenzyl phosphonate, phosphate-(Cι-C2ι)-O-alkyl ester, phosphate-[(C6-Cι )aryl-(Cι-C2ι)-O- alkyljester, (Cι-C8)alkylphosphonate and/or (C6-C]2)arylphosphonate bridges, (C7-C]2)- α-hydroxymethyl-aryl (e.g., disclosed in WO 95/01363), wherein (C6-Cι2)aryl, (C6- C20)aryl and (C6-Ci4)aryl are optionally substituted by halogen, alkyl, alkoxy, nitro, cyano, and where R1 and R2 are, independently of each other, hydrogen, (Cι-C)8)-alkyl, (C6-C2o)-aryl, (C6-Cι )-aryl-(Cι-C8)-alkyl, preferably hydrogen, (C]-C8)-alkyl, preferably (Cι-C4)-alkyl and/or methoxyethyl, or R1 and R2 form, together with the nitrogen atom carrying them, a 5-6-membered heterocyclic ring which can additionally contain a further heteroatom from the group O, S and N.
The replacement of a phosphodiester bridge located at the 3' and/or the 5' end of a nucleoside by a dephospho bridge (dephospho bridges are described, for example, in Uhlmann E and Peyman A in "Methods in Molecular Biology", Vol. 20, "Protocols for Oligonucleotides and Analogs", S. Agrawal, Ed, Humana Press, Totowa 1993, Chapter 16, pp. 355 ff), wherein a dephospho bridge is for example selected from the dephospho bridges formacetal, 3'-thioformacetal, methylhydroxylamine, oxime, methylenedim ethyl - hydrazo, dimethylenesulfone and/or silyl groups. A sugar phosphate unit (i.e., a β-D-ribose and phosphodiester internucleoside bridge together forming a sugar phosphate unit) from the sugar phosphate backbone (i.e., a sugar phosphate backbone is composed of sugar phosphate units) can be replaced by another unit, wherein the other unit is for example suitable to build up a "morpholino- derivative" oligomer (as described, for example, in Stirchak EP et al. (1989) Nucleic Acids Res 17:6129-41), that is, e.g., the replacement by a morpholino-derivative unit; or to build up a polyamide nucleic acid ("PNA"; as described for example, in Nielsen PE et al. (1994) Bioconjug Chem 5:3-7), that is, e.g., the replacement by a PNA backbone unit, e.g, by 2-aminoethylglycine. The oligonucleotide may have other carbohydrate backbone modifications and replacements, such as peptide nucleic acids with phosphate groups (PHONA), locked nucleic acids (LNA), and oligonucleotides having backbone sections with alkyl linkers or amino linkers. The alkyl linker may be branched or unbranched, substituted or unsubstituted, and chirally pure or a racemic mixture.
A β-ribose unit or a β-D-2'-deoxyribose unit can be replaced by a modified sugar unit, wherein the modified sugar unit is for example selected from β-D-ribose, α-D-2'- deoxyribose, L-2'-deoxyribose, 2'-F-2'-deoxyribose, 2'-F-arabinose, 2'-O-(Cι-C6)alkyl- ribose, preferably 2'-O-(Cι-C6)alkyl-ribose is 2'-O-methylribose, 2'-O-(C2-C6)alkenyl- ribose, 2'-[O-(Cι-C6)alkyl-O-(C C6)alkyl]-ribose, 2'-NH2-2'-deoxyribose, β-D-xylo- furanose, α-arabinofuranose, 2,4-dideoxy-β-D-erythro-hexo-pyranose, and carbocyclic (described, for example, in Froehler J (1992) Am Chem Soc 114:8320) and/or open-chain sugar analogs (described, for example, in Vandendriessche et al. (1993) Tetrahedron 49:7223) and/or bicyclosugar analogs (described, for example, in Tarkov M et al. (1993) Helv Chim Acta 16:481).
In some embodiments the sugar is 2'-O-methylribose, particularly for one or both nucleotides linked by a phosphodiester or phosphodiester-like internucleoside linkage.
Nucleic acids also include substituted purines and pyrimidines such as C-5 propyne pyrimidine and 7-deaza-7-substituted purine modified bases. Wagner RW et al. (1996) Nat Biotechnol 14:840-4. Purines and pyrimidines include but are not limited to adenine, cytosine, guanine, and thymine, and other naturally and non-naturally occurring nucleobases, substituted and unsubstituted aromatic moieties.
A modified base is any base which is chemically distinct from the naturally occurring bases typically found in DNA and RNA such as T, C, G, A, and U, but which share basic chemical structures with these naturally occurring bases. The modified nucleoside base may be, for example, selected from hypoxanthine, uracil, dihydrouracil, pseudouracil, 2-thiouracil, 4-thiouracil, 5-aminouracil, 5-(Ci-C6)-alkyluracil, 5-(C2-C6)- alkenyluracil, 5-(C2-C6)-alkynyluracil, 5-(hydroxymethyl)uracil, 5-chlorouracil, 5-fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(Cι-C6)-alkylcytosine, 5-(C -C6)- alkenylcytosine, 5-(C2-Co)-alkynylcytosine, 5-chlorocytosine, 5-fluorocytosine,
5-bromocytosine, N -dimethylguanine, 2,4-diamino-purine, 8-azapuπne, a substituted 7-deazapurine, preferably 7-deaza-7-substituted and/or 7-deaza-8-substituted purine, 5- hydroxymethylcytosine, N4-alkylcytosine, e.g., N4-ethylcytosine, 5- hydroxydeoxycytidine, 5-hydroxymethyldeoxycytidine, N4-alkyldeoxycytidine, e.g., N4-ethyldeoxycytidine, 6-thiodeoxyguanosine, and deoxyribonucleosides of nitropyrrole, C5-propynylpyrimidine, and diaminopurine e.g., 2,6-diaminopurine, inosine, 5-methylcytosine, 2-aminopurine, 2-amino-6-chloropurine, hypoxanthine or other modifications of a natural nucleoside bases. This list is meant to be exemplary and is not to be interpreted to be limiting.
In particular formulas described herein a set of modified bases is defined. For instance the letter Y is used to refer to a nucleotide containing a cytosine or a modified cytosine. A modified cytosine as used herein is a naturally occurring or non-naturally occurring pyrimidine base analog of cytosine which can replace this base without impairing the immunostimulatory activity of the oligonucleotide. Modified cytosines include but are not limited to 5-substituted cytosines (e.g. 5-metlιyl-cytosine, 5-fluoro- cytosine, 5-chloro-cytosine, 5-bromo-cytosine, 5-iodo-cytosine, 5-hydroxy-cytosine, 5- hydroxymethyl-cytosine, 5-difluoromethyl-cytosine, and unsubstituted or substituted 5- alkynyl-cytosine), 6-substituted cytosines, N4-substituted cytosines (e.g. N4-ethyl- cytosine), 5-aza-cytosine, 2-mercapto-cytosine, isocytosine, pseudo-isocytosine, cytosine analogs with condensed ring systems (e.g. N,N'-propylene cytosine or phenoxazine), and uracil and its derivatives (e.g. 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4- thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil). Some of the preferred cytosines include 5-methyl-cytosine, 5-fluoro-cytosine, 5-hydroxy-cytosine, 5-hydroxymethyl- cytosine, and N4-ethyl-cytosine. In another embodiment of the invention, the cytosine base is substituted by a universal base (e.g. 3-nitropyrrole, P-base), an aromatic ring system (e.g. fluorobenzene or difluorobenzene) or a hydrogen atom (dSpacer). The letter Z is used to refer to guanine or a modified guanine base. A modified guanine as used herein is a naturally occurring or non-naturally occurring purine base analog of guanine which can replace this base without impairing the immunostimulatory activity of the oligonucleotide. Modified guanines include but are not limited to 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, hypoxanthine, N2-substituted guanines (e.g. N2-methyl- guanine), 5-amino-3-methyl-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, adenine, substituted adenines (e.g. N6-methyl-adenine, 8-oxo-adenine) 8-substituted guanine (e.g. 8 -hydroxy guanine and 8-bromoguanine), and 6-thioguanine. In another embodiment of the invention, the guanine base is substituted by a universal base (e.g. 4-methyl-indole, 5-nitro-indole, and K-base), an aromatic ring system (e.g. benzimidazole or dichloro- benzimidazole, 1- methyl- lH-[l,2,4]triazole-3-carboxylic acid amide) or a hydrogen atom (dSpacer). For use in the instant invention, the oligonucleotides of the invention can be synthesized de novo using any of a number of procedures well known in the art. For example, the b-cyanoethyl phosphoramidite method (Beaucage, S.L, and Caruthers, M.H, Tet. Let. 22:1859, 1981); nucleoside H-phosphonate method (Garegg et al, Tet. Let. 27:4051-4054, 1986; Froehler et al, Nucl. Acid. Res. 14:5399-5407, 1986, ; Garegg et al, Tet. Let. 27:4055-4058, 1986, Gaffhey et al, Tet. Let. 29:2619-2622, 1988). These chemistries can be performed by a variety of automated nucleic acid synthesizers available in the market. These oligonucleotides are referred to as synthetic oligonucleotides. An isolated oligonucleotide generally refers to an oligonucleotide which is separated from components which it is normally associated with in nature. As an example, an isolated oligonucleotide may be one which is separated from a cell, from a nucleus, from mitochondria or from chromatin.
The immunostimulatory nucleic acid molecules of the instant invention can have phosphodiester internuclotide linkages. A phosphodiester internucleotide linkage is the type of linkage characteristic of nucleic acids found in nature. As shown in Figure 20, the phosphodiester internucleotide linkage includes a phosphorus atom flanked by two bridging oxygen atoms and bound also by two additional oxygen atoms, one charged and the other uncharged. Phosphodiester internucleotide linkage is particularly preferred when it is important to reduce the tissue half-life of the oligonucleotide. A phosphodiester-like internucleotide linkage is a phosphorus-containing bridging group that is chemically and/or diastereomerically similar to phosphodiester. Measures of similarity to phosphodiester include susceptibility to nuclease digestion and ability to activate RNAse H. Thus for example phosphodiester, but not phosphorothioate, oligonucleotides are susceptible to nuclease digestion, while both phosphodiester and phosphorothioate oligonucleotides activate RNAse H. In a preferred embodiment the phosphodiester-like internucleotide linkage is boranophosphate (or equivalently, boranophosphonate) linkage. U.S. Patent No. 5,177,198; U.S. Patent No. 5,859,231; U.S. Patent No. 6,160,109; U.S. Patent No. 6,207,819; Sergueev et al, (1998) J Am Chem Soc 120:9417-27. In another preferred embodiment the phosphodiester-like internucleotide linkage is diasteromerically pure Rp phosphorothioate. It is believed that diasteromerically pure Rp phosphorothioate is more susceptible to nuclease digestion and is better at activating RNAse H than mixed or diastereomerically pure Sp phosphorothioate. Stereoisomers of CpG oligonucleotides are the subject of co-pending U.S. patent application 09/361,575 filed July 27, 1999, and published PCT application PCT/US99/17100 (WO 00/06588). It is to be noted that for purposes of the instant invention, the term "phosphodiester-like internucleotide linkage" specifically excludes phosphorodithioate and methylphosphonate internucleotide linkages.
The immunostimulatory nucleic acid molecules of the instant invention can have chimeric backbone. For purposes of the instant invention, a chimeric backbone refers to a partially stabilized backbone, wherein at least one internucleotide linkage is phosphodiester or phosphodiester-like, and wherein at least one other internucleotide linkage is a stabilized internucleotide linkage, wherein the at least one phosphodiester or phosphodiester-like linkage and the at least one stabilized linkage are different. Since boranophosphonate linkages have been reported to be stabilized relative to phosphodiester linkages, for purposes of the chimeric nature of the backbone, boranophosphonate linkages can be classified either as phosphodiester-like or as stabilized, depending on the context. For example, a chimeric backbone according to the instant invention could in one embodiment include at least one phosphodiester (phosphodiester or phosphodiester-like) linkage and at least one boranophosphonate (stabilized) linkage. In another embodiment a chimeric backbone according to the instant invention could include boranophosphonate (phosphodiester or phosphodiester- like) and phosphorothioate (stabilized) linkages. A "stabilized internucleotide linkage" shall mean an internucleotide linkage that is relatively resistant to in vivo degradation (e.g., via an exo- or endo-nuclease), compared to a phosphodiester internucleotide linkage. Preferred stabilized internucleotide linkages include, without limitation, phosphorothioate, phosphorodithioate, methylphosphonate, and methylphosphorothioate. Other stabilized internucleotide linkages include, without limitation: peptide, alkyl, dephospho, and others as described above. Modified backbones such as phosphorothioates may be synthesized using automated techniques employing either phosphoramidate or H-phosphonate chemistries. Aryl-and alkyl-phosphonates can be made, e.g., as described in U.S. Patent No. 4,469,863; and alkylphosphotriesters (in which the charged oxygen moiety is alkylated as described in U.S. Patent No. 5,023,243 and European Patent No. 092,574) can be prepared by automated solid phase synthesis using commercially available reagents. Methods for making other DNA backbone modifications and substitutions have been described (e.g., Uhlmann, E. and Peyman, A, Chem. Rev. 90:544, 1990; Goodchild, J, Bioconjugate Chem. 1:165, 1990). CpG phosphorothioate oligonucleotides with strong stimulatory activity in the mouse system tend to show lower activity on human and other non-rodent immune cells. In the examples the development of a potent human CpG motif and the characterization of its effects and mechanisms of action on human PBMC, e.g., B-cells, and plasmacytoid dendritic cells is described. DNA containing these 5'TCG or 5 'TYZ CpG motifs strongly stimulated human peripheral blood cells to produce IL-10, IL-6, IP- 10 and IFN- α. The 5'TCG containing ODN could be further optimized by selecting ODNs of a particular length. For instance, ODNs of 22 nucleotides in length are more stimulatory than shorter ODN.
It has been discovered according to the invention that the subsets of CpG immunostimulatory oligonucleotides have dramatic immune stimulatory effects on human cells such as PBMC, suggesting that these CpG immunostimulatory oligonucleotides are effective therapeutic agents for human vaccination, cancer immunotherapy, asthma immunotherapy, general enhancement of immune function, enhancement of hematopoietic recovery following radiation or chemotherapy, and other immune modulatory applications.
As used herein, the terms treat, treated, or treating when used with respect to a disorder such as an infectious disease, cancer, allergy, or asthma refers to a prophylactic treatment which increases the resistance of a subject to development of the disease (e.g., to infection with a pathogen) or, in other words, decreases the likelihood that the subject will develop the disease (e.g., become infected with the pathogen) as well as a treatment after the subject has developed the disease in order to fight the disease (e.g., reduce or eliminate the infection) or prevent the disease from becoming worse. Thus the CpG immunostimulatory oligonucleotides are useful in some aspects of the invention as a vaccine for the treatment of a subject having or at risk of developing allergy or asthma, an infection with an infectious organism or a cancer in which a specific cancer antigen has been identified. The CpG immunostimulatory oligonucleotides can also be given alone without the antigen or allergen for protection against infection, allergy or cancer or may be administered with other therapeutic agents. Repeated doses may allow longer term protection. A subject at risk as used herein is a subject who has any risk of exposure to an infection causing pathogen or a cancer or an allergen or a risk of developing cancer. For instance, a subject at risk may be a subject who is planning to travel to an area where a particular type of infectious agent is found or it may be a subject who through lifestyle or medical procedures is exposed to bodily fluids which may contain infectious organisms or directly to the organism or even any subject living in an area where an infectious organism or an allergen has been identified. Subjects at risk of developing infection also include general populations to which a medical agency recommends vaccination with a particular infectious organism antigen. If the antigen is an allergen and the subject develops allergic responses to that particular antigen and the subject may be exposed to the antigen, i.e., during pollen season, then that subject is at risk of exposure to the antigen. A subject at risk of developing an allergy to asthma includes those subjects that have been identified as having an allergy or asthma but that don't have the active disease during the CpG immunostimulatory oligonucleotide treatment as well as subjects that are considered to be at risk of developing these diseases because of genetic or environmental factors.
A subject at risk of developing a cancer is one who has a high probability of developing cancer. These subjects include, for instance, subjects having a genetic abnormality, the presence of which has been demonstrated to have a correlative relation to a higher likelihood of developing a cancer and subjects exposed to cancer causing agents such as tobacco, asbestos, or other chemical toxins, or a subject who has previously been treated for cancer and is in apparent remission. When a subject at risk of developing a cancer is treated with a CpG immunostimulatory oligonucleotide and optionally an antigen specific for the type of cancer to which the subject is at risk of developing, the subject may be able to kill the cancer cells as they develop. If a tumor begins to form in the subject, the subject will develop an innate immune response or a i specific immune response against the tumor antigen.
In addition to the use of the CpG immunostimulatory oligonucleotides for prophylactic treatment, the invention also encompasses the use of the CpG immunostimulatory oligonucleotides for the treatment of a subject having an infection, an allergy, asthma, or a cancer.
A subject having an infection is a subject that has been exposed to an infectious pathogen and has acute or chronic detectable levels of the pathogen in the body. The
CpG immunostimulatory oligonucleotides can be used with or without an antigen or other therapeutic to mount an innate or an antigen specific systemic or mucosal immune response that is capable of reducing the level of or eradicating the infectious pathogen. An infectious disease, as used herein, is a disease arising from the presence of a foreign microorganism in the body. It is particularly important to develop effective vaccine strategies and treatments to protect the body's mucosal surfaces, which are the primary site of pathogenic entry.
A subject having an allergy is a subject that is capable of developing an allergic reaction in response to an allergen. An allergy refers to acquired hypersensitivity to a substance (allergen). Allergic conditions include but are not limited to eczema, allergic rhinitis or coryza, hay fever, conjunctivitis, bronchial asthma, allergic asthma, urticaria (hives) and food allergies, and other atopic conditions.
Allergies are generally caused by IgE antibody generation against harmless allergens. The cytokines that are induced by systemic or mucosal administration of CpG immunostimulatory oligonucleotides are predominantly of a class called Thl (examples are IL-12, IP-10, IFN-α and IFN-γ) and these induce both humoral and cellular immune responses. The other major type of immune response, which is associated with the production of IL-4 and IL-5 cytokines, is termed a Th2 immune response. In general, it appears that allergic diseases are mediated by Th2 type immune responses. Based on the ability of the CpG immunostimulatory oligonucleotides described herein to shift the immune response in a subject from a predominant Th2 (which is associated with production of IgE antibodies and allergy) to a balanced Th2/Thl response (which is protective against allergic reactions), an effective dose for inducing an immune response of a CpG immunostimulatory oligonucleotide can be administered to a subject to treat asthma and allergy.
Thus, the CpG immunostimulatory oligonucleotides have significant therapeutic utility in the treatment of allergic conditions and asthma. Th2 cytokines, especially IL-4 and IL-5 are elevated in the airways of asthmatic subjects. These cytokines promote important aspects of the asthmatic inflammatory response, including IgE isotope switching, eosinophil chemotaxis and activation and mast cell growth. Thl cytokines, especially IFN-γ and IL-12, can suppress the formation of Th2 clones and production of Th2 cytokines. Asthma refers to a disorder of the respiratory system characterized by inflammation, narrowing of the airways and increased reactivity of the airways to inhaled agents. Asthma is frequently, although not exclusively associated with atopic or allergic symptoms. Thus, asthma includes allergic asthma and non-allergic asthma.
A subject having a cancer is a subject that has detectable cancerous cells. The cancer may be a malignant or non-malignant cancer. Cancers or tumors include but are not limited to biliary tract cancer; brain cancer; breast cancer; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; intraepithelial neoplasms; lymphomas; liver cancer; lung cancer (e.g. small cell and non-small cell); melanoma; neuroblastomas; oral cancer; ovarian cancer; pancreas cancer; prostate cancer; rectal cancer; sarcomas; skin cancer; testicular cancer; thyroid cancer; and renal cancer, as well as other carcinomas and sarcomas. In one embodiment the cancer is hairy cell leukemia, chronic myelogenous leukemia, cutaneous T-cell leukemia, multiple myeloma, follicular lymphoma, malignant melanoma, squamous cell carcinoma, renal cell carcinoma, prostate carcinoma, bladder cell carcinoma, or colon carcinoma, CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer, as well as other carcinomas and sarcomas.
A subject shall mean a human or vertebrate animal including but not limited to a dog, cat, horse, cow, pig, sheep, goat, turkey, chicken, primate, e.g., monkey, and fish (aquaculture species), e.g. salmon. Thus, the compounds may be used to treat cancer and tumors, infections, and allergy/asthma in human and non human subjects. Cancer is one of the leading causes of death in companion animals (i.e., cats and dogs). In the instances when the CpG oligonucleotide is administered with an antigen, the subject may be exposed to the antigen. As used herein, the term exposed to refers to either the active step of contacting the subject with an antigen or the passive exposure of the subject to the antigen in vivo. Methods for the active exposure of a subject to an antigen are well-lcnown in the art. In general, an antigen is administered directly to the subject by any means such as intravenous, intramuscular, oral, transdermal, mucosal, intranasal, intratracheal, or subcutaneous administration. The antigen can be administered systemically or locally. Methods for administering the antigen and the CpG immunostimulatory oligonucleotide are described in more detail below. A subject is passively exposed to an antigen if an antigen becomes available for exposure to the immune cells in the body. A subject may be passively exposed to an antigen, for instance, by entry of a foreign pathogen into the body or by the development of a tumor cell expressing a foreign antigen on its surface.
The methods in which a subject is passively exposed to an antigen can be particularly dependent on timing of administration of the CpG immunostimulatory oligonucleotide. For instance, in a subject at risk of developing a cancer or an infectious disease or an allergic or asthmatic response, the subject may be administered the CpG immunostimulatory oligonucleotide on a regular basis when that risk is greatest, i.e., during allergy season or after exposure to a cancer causing agent. Additionally the CpG immunostimulatory oligonucleotide may be administered to travelers before they travel to foreign lands where they are at risk of exposure to infectious agents. Likewise the CpG immunostimulatory oligonucleotide may be administered to soldiers or civilians at risk of exposure to biowarfare to induce a systemic or mucosal immune response to the antigen when and if the subject is exposed to it. An antigen as used herein is a molecule capable of provoking an immune response. Antigens include but are not limited to cells, cell extracts, proteins, polypeptides, peptides, polysaccharides, polysaccharide conjugates, peptide and nonpeptide mimics of polysaccharides and other molecules, small molecules, lipids, glycolipids, carbohydrates, viruses and viral extracts and muticellular organisms such as parasites and allergens. The term antigen broadly includes any type of molecule which is recognized by a host immune system as being foreign. Antigens include but are not limited to cancer antigens, microbial antigens, and allergens. A cancer antigen as used herein is a compound, such as a peptide or protein, associated with a tumor or cancer cell surface and which is capable of provoking an immune response when expressed on the surface of an antigen presenting cell in the context of an MHC molecule. Cancer antigens can be prepared from cancer cells either by preparing crude extracts of cancer cells, for example, as described in Cohen, et al, 1994, Cancer Research, 54:1055, by partially purifying the antigens, by recombinant technology, or by de novo synthesis of known antigens. Cancer antigens include but are not limited to antigens that are recombinantly expressed, an immunogenic portion thereof, or a whole tumor or cancer cell. Such antigens can be isolated or prepared recombinantly or by any other means known in the art.
As used herein, the terms "cancer antigen" and "tumor antigen" are used interchangeably to refer to antigens which are differentially expressed by cancer cells and can thereby be exploited in order to target cancer cells. Cancer antigens are antigens which can potentially stimulate apparently tumor-specific immune responses. Some of these antigens are encoded, although not necessarily expressed, by normal cells. These antigens can be characterized as those which are normally silent (i.e., not expressed) in normal cells, those that are expressed only at certain stages of differentiation and those that are temporally expressed such as embryonic and fetal antigens. Other cancer antigens are encoded by mutant cellular genes, such as oncogenes (e.g., activated ras oncogene), suppressor genes (e.g., mutant p53), fusion proteins resulting from internal deletions or chromosomal translocations. Still other cancer antigens can be encoded by viral genes such as those carried on RNA and DNA tumor viruses.
A microbial antigen as used herein is an antigen of a microorganism and includes but is not limited to virus, bacteria, parasites, and fungi. Such antigens include the intact microorganism as well as natural isolates and fragments or derivatives thereof and also synthetic compounds which are identical to or similar to natural microorganism antigens and induce an immune response specific for that microorganism. A compound is similar to a natural microorganism antigen if it induces an immune response (humoral and/or cellular) to a natural microorganism antigen. Such antigens are used routinely in the art and are well known to those of ordinary skill in the art.
Examples of viruses that have been found in humans include but are not limited to: Retroviridae (e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HTLV-III, LAV or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV-LP; Picornaviridae (e.g. polio viruses, hepatitis A virus; enteroviruses, human Coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (e.g. strains that cause gastroenteritis); Togaviridae (e.g. equine encephalitis viruses, rubella viruses); Flaviridae (e.g. dengue viruses, encephalitis viruses, yellow fever viruses); Coronoviridae (e.g. coronaviruses); Rhabdoviradae (e.g. vesicular stomatitis viruses, rabies viruses); Coronoviridae (e.g. coronaviruses); Rhabdoviridae (e.g. vesicular stomatitis viruses, rabies viruses); Filoviridae (e.g. ebola viruses); Paramyxoviridae (e.g. parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); Orthomyxoviridae (e.g. influenza viruses); Bungaviridae (e.g. Hantaan viruses, bunga viruses, phleboviruses and Nairo viruses); Arena viridae (hemorrhagic fever viruses); Reoviridae (e.g. reoviruses, orbiviurses and rotaviruses); Birnaviridae; Hepadnaviridae (Hepatitis B virus); Parvovirida (parvoviruses); Papovaviridae (papilloma viruses, polyoma viruses); Adenoviridae (most adenoviruses); Herpesviridae (herpes simplex virus (HSV) 1 and 2, varicella zoster virus, cytomegalovirus (CMV), herpes virus; Poxviridae (variola viruses, vaccinia viruses, pox viruses); and Iridoviridae (e.g. African swine fever virus); and unclassified viruses (e.g. the agent of delta hepatitis (thought to be a defective satellite of hepatitis B virus), Hepatitis C; Norwalk and related viruses, and astroviruses).
Both gram negative and gram positive bacteria serve as antigens in vertebrate animals. Such gram positive bacteria include, but are not limited to, Pasteurella species, Staphylococci species, and Streptococcus species. Gram negative bacteria include, but are not limited to, Escherichia coli, Pseudomonas species, and Salmonella species. Specific examples of infectious bacteria include but are not limited to, Helicobacter pylons, Borelia burgdorferi, Legionella pneumophilia, Mycobacteria sps (e.g. M. tuberculosis, M. avium, M. intracellular e, M. kansaii, M. gordonae), Staphylococcus aureus, Neisseria gonorrhoeae, Neisseria meningitidis, Listeria monocytogenes, Streptococcus pyogenes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecalis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogenic Campylobacter sp., Enterococcus sp., Haemophilus influenzae, Bacillus antracis, cory b acterium diphtheriae, corynebacterium sp., Erysipelothrix rhusiopathiae, Clostridium perfringers, Clostridium tetani, Enterobacter aerogenes, Klebsiella pneumoniae, Pasturella multocida, Bacteroides sp., Fusobacterium nucleatum, Streptobacillus moniliformis, Treponema pallidium, Treponema pertenue, Leptospira, Rickettsia, and Actinomyces israelli.
Examples of fungi include Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, Candida albicans.
Other infectious organisms (i.e., protists) include Plasmodium spp. such as Plasmodium falciparum, Plasmodium malariae, Plasmodium ovale, and Plasmodium vivax and Toxoplasma gondii. Blood-borne and/or tissues parasites include Plasmodium spp, Babesia microti, Babesia divergens, Leishmania tropica, Leishmania spp, Leishmania braziliensis, Leishmania donovani, Trypanosoma gambiense and Trypanosoma rhodesiense (African sleeping sickness), Trypanosoma cruzi (Chagas' disease), and Toxoplasma gondii.
Other medically relevant microorganisms have been described extensively in the literature, e.g., see C.G.A Thomas, Medical Microbiology, Bailliere Tindall, Great Britain 1983, the entire contents of which is hereby incorporated by reference.
An allergen refers to a substance (antigen) that can induce an allergic or asthmatic response in a susceptible subject. The list of allergens is enormous and can include pollens, insect venoms, animal dander dust, fungal spores and drugs (e.g. penicillin). Examples of natural, animal and plant allergens include but are not limited to proteins specific to the following genuses: Canine (Ca is familiaris); Dermatophagoides (e.g. Dermatophagoides farinae); Felis (Felis domesticus); Ambrosia (Ambrosia artemiisfolia; Lolium (e.g. Lolium perenne or Lolium multiflorum); Cryptomeria (Cryptomeria japonica); Alternaria (Alternaria alternata); Alder; Alnus (Alnus gultinoasa); Betula (Betula verrucosa); Quercus (Quercus alba); Olea (Olea europa); Artemisia (Artemisia vulgaris); Plantago (e.g. Plantago lanceolata); Parietaria (e.g. Parietaria officinalis or Parietaria judaica); Blattella (e.g. Blattella germanica); Apis (e.g. Apis multiflorum); Cupressus (e.g. Cupressus semper~virens, Cupressus arizonica and Cupressus macrocarpd); Juniperus (e.g. Juniperus sabinoides, Juniperus virginiana, Juniperus communis and Juniperus ashei); Thuya (e.g. Thuya orientalis);
Chamaecyparis (e.g. Chamaecyparis obtusci); Periplaneta (e.g. Periplaneta americana); Agropyron (e.g. Agropyron repens); Secale (e.g. Secale cereale); Triticum (e.g. Triticum aestivum); Dactylis (e.g. Dactylis glomerata); Festuca (e.g. Festuca elatior); Poa (e.g. Poapratensis or Poa compressa); Avena (e.g. Avena sativa); Holcus (e.g. Holcus lanatus); Anthoxanthum (e.g. Anthoxanthum odoratum); Arrhenatherum (e.g. Arrhenatherum elatius); Agrostis (e.g. Agrostis alba); Phleum (e.g. Phleum pratense); Phalaris (e.g. Phalaris arundinacea); Paspalum (e.g. Paspalum notatum); Sorghum (e.g. Sorghum halepensis); and Bromus (e.g. Bromus inermis).
The antigen may be substantially purified. The term substantially purified as used herein refers to an antigen, i.e., a polypeptide which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. One skilled in the art can purify polypeptide antigens using standard techniques for protein purification. The substantially pure polypeptide will often yield a single major band on a non-reducing polyacrylamide gel. In the case of partially glycosylated polypeptides or those that have several start codons, there may be several bands on a non-reducing polyacrylamide gel, but these will form a distinctive pattern for that polypeptide. The purity of the polypeptide antigen may also be determined by amino-terminal amino acid sequence analysis. Other types of antigens such as polysaccharides, small molecule, mimics etc are included within the invention and may optionally be substantially pure.
The oligonucleotides of the invention may be administered to a subject with an anti-microbial agent. An anti-microbial agent, as used herein, refers to a naturally- occurring or synthetic compound which is capable of killing or inhibiting infectious microorganisms. The type of anti-microbial agent useful according to the invention will depend upon the type of microorganism with which the subject is infected or at risk of becoming infected. Anti-microbial agents include but are not limited to anti-bacterial agents, anti-viral agents, anti-fungal agents and anti-parasitic agents. Phrases such as "anti-infective agent", "anti-bacterial agent", "anti-viral agent", "anti-fungal agent", "anti-parasitic agent" and "parasiticide" have well-established meanings to those of ordinary skill in the art and are defined in standard medical texts. Briefly, anti-bacterial agents kill or inhibit bacteria, and include antibiotics as well as other synthetic or natural compounds having similar functions. Antibiotics are low molecular weight molecules which are produced as secondary metabolites by cells, such as microorganisms. In general, antibiotics interfere with one or more bacterial functions or structures which are specific for the microorganism and which are not present in host cells. Anti-viral agents can be isolated from natural sources or synthesized and are useful for killing or inhibiting viruses. Anti-fungal agents are used to treat superficial fungal infections as well as opportunistic and primary systemic fungal infections. Anti-parasitic agents kill or inhibit parasites.
Examples of anti-parasitic agents, also referred to as parasiticides useful for human administration include but are not limited to albendazole, amphotericin B, benznidazole, bithionol, chloroquine HCI, chloroquine phosphate, clindamycin, dehydroemetine, diethylcarbamazine, diloxanide furoate, eflornithine, furazolidaone, glucocorticoids, halofantrine, iodoquinol, ivermectin, mebendazole, mefloquine, meglumine antimoniate, melarsoprol, metrifonate, metronidazole, niclosamide, nifurtimox, oxamniquine, paromomycin, pentamidine isethionate, piperazine, praziquantel, primaquine phosphate, proguanil, pyrantel pamoate, pyrimethanmine- sulfonamides, pyrimethanmine-sulfadoxine, quinacrine HCI, quinine sulfate, quinidine gluconate, spiramycin, stibogluconate sodium (sodium antimony gluconate), suramin, tetracycline, doxycycline, thiabendazole, tinidazole, trimethroprim-sulfamethoxazole, and tryparsamide some of which are used alone or in combination with others.
Antibacterial agents kill or inhibit the growth or function of bacteria. A large class of antibacterial agents is antibiotics. Antibiotics, which are effective for killing or inhibiting a wide range of bacteria, are referred to as broad spectrum antibiotics. Other types of antibiotics are predominantly effective against the bacteria of the class gram- positive or gram-negative. These types of antibiotics are referred to as narrow spectrum antibiotics. Other antibiotics which are effective against a single organism or disease and not against other types of bacteria, are referred to as limited spectrum antibiotics. Antibacterial agents are sometimes classified based on their primary mode of action. In general, antibacterial agents are cell wall synthesis inhibitors, cell membrane inhibitors, protein synthesis inhibitors, nucleic acid synthesis or functional inhibitors, and competitive inhibitors.
Antiviral agents are compounds which prevent infection of cells by viruses or replication of the virus within the cell. There are many fewer antiviral drugs than antibacterial drugs because the process of viral replication is so closely related to DNA replication within the host cell, that non-specific antiviral agents would often be toxic to the host. There are several stages within the process of viral infection which can be blocked or inhibited by antiviral agents. These stages include, attachment of the virus to the host cell (immunoglobulin or binding peptides), uncoating of the virus (e.g. amantadine), synthesis or translation of viral mRNA (e.g. interferon), replication of viral RNA or DNA (e.g. nucleoside analogues), maturation of new virus proteins (e.g. protease inhibitors), and budding and release of the virus.
Nucleotide analogues are synthetic compounds which are similar to nucleotides, but which have an incomplete or abnormal deoxyribose or ribose group. Once the nucleotide analogues are in the cell, they are phosphorylated, producing the triphosphate form which competes with normal nucleotides for incorporation into the viral DNA or RNA. Once the triphosphate form of the nucleotide analogue is incorporated into the growing nucleic acid chain, it causes irreversible association with the viral polymerase and thus chain termination. Nucleotide analogues include, but are not limited to, acyclovir (used for the treatment of herpes simplex virus and varicella-zoster virus), gancyclovir (useful for the treatment of cytomegalovirus), idoxuridine, ribavirin (useful for the treatment of respiratory syncitial virus), dideoxyinosine, dideoxycytidine, zidovudine (azidothymidine), imiquimod, and resimiquimod.
The interferons are cytokines which are secreted by virus-infected cells as well as immune cells. The interferons function by binding to specific receptors on cells adjacent to the infected cells, causing the change in the cell which protects it from infection by the virus, α and β-interferon also induce the expression of Class I and Class II MHC molecules on the surface of infected cells, resulting in increased antigen presentation for host immune cell recognition, α and β-interferons are available as recombinant forms and have been used for the treatment of chronic hepatitis B and C infection. At the dosages which are effective for anti-viral therapy, interferons have severe side effects such as fever, malaise and weight loss.
Anti-viral agents useful in the invention include but are not limited to immunoglobulins, amantadine, interferons, nucleoside analogues, and protease inhibitors. Specific examples of anti-virals include but are not limited to Acemannan; Acyclovir; Acyclovir Sodium; Adefovir; Alovudine; Alvircept Sudotox; Amantadine Hydrochloride; Aranotin; Arildone; Atevirdine Mesylate; Avridine; Cidofovir; Cipamfylline; Cytarabine Hydrochloride; Delavirdine Mesylate; Desciclovir; Didanosine; Disoxaril; Edoxudine; Enviradene; Enviroxime; Famciclovir; Famotine Hydrochloride; Fiacitabine; Fialuridine; Fosarilate; Foscarnet Sodium; Fosfonet Sodium; Ganciclovir; Ganciclovir Sodium; Idoxuridine; Kethoxal; Lamivudine; Lobucavir; Memotine Hydrochloride; Methisazone; Nevirapine; Penciclovir; Pirodavir; Ribavirin; Rimantadine Hydrochloride; Saquinavir Mesylate; Somantadine Hydrochloride; Sorivudine; Statolon; Stavudine; Tilorone Hydrochloride; Trifluridine; Valacyclovir Hydrochloride; Vidarabine; Vidarabine Phosphate; Vidarabine Sodium Phosphate; Viroxime; Zalcitabine; Zidovudine; and Zinviroxime.
Anti-fungal agents are useful for the treatment and prevention of infective fungi. Anti-fungal agents are sometimes classified by their mechanism of action. Some anti- fungal agents function as cell wall inhibitors by inhibiting glucose synthase. These include, but are not limited to, basiungin/ECB. Other anti-fungal agents function by destabilizing membrane integrity. These include, but are not limited to, immidazoles, such as clotrimazole, sertaconzole, fluconazole, itraconazole, ketoconazole, miconazole, and voriconacole, as well as FK 463, amphotericin B, BAY 38-9502, MK 991, pradimicin, UK 292, butenafine, and terbinafine. Other anti-fungal agents function by breaking down chitin (e.g. chitinase) or immunosuppression (501 cream).
CpG immunostimulatory oligonucleotides can be combined with other therapeutic agents such as adjuvants to enhance immune responses. The CpG immunostimulatory oligonucleotide and other therapeutic agent may be administered simultaneously or sequentially. When the other therapeutic agents are administered simultaneously they can be administered in the same or separate formulations, but are administered at the same time. The other therapeutic agents are administered sequentially with one another and with CpG immunostimulatory oligonucleotide, when the administration of the other therapeutic agents and the CpG immunostimulatory oligonucleotide is temporally separated. The separation in time between the administration of these compounds may be a matter of minutes or it may be longer. Other therapeutic agents include but are not limited to adjuvants, cytokines, antibodies, antigens, etc. The compositions of the invention may also be administered with non-nucleic acid adjuvants. A non-nucleic acid adjuvant is any molecule or compound except for the CpG immunostimulatory oligonucleotides described herein which can stimulate the humoral and/or cellular immune response. Non-nucleic acid adjuvants include, for instance, adjuvants that create a depo effect, immune stimulating adjuvants, and adjuvants that create a depo effect and stimulate the immune system.
The CpG immunostimulatory oligonucleotides are also useful as mucosal adjuvants. It has previously been discovered that both systemic and mucosal immunity are induced by mucosal delivery of CpG nucleic acids. Thus, the oligonucleotides may be administered in combination with other mucosal adjuvants.
Immune responses can also be induced or augmented by the co-administration or co-linear expression of cytokines (Bueler & Mulligan, 1996; Chow et al, 1997; Geissler et al, 1997; Iwasaki et al, 1997; Kim et al, 1997) or co-stimulatory molecules such as B7 (Iwasaki et al, 1997; Tsuji et al, 1997) with the CpG immunostimulatory oligonucleotides. The term cytokine is used as a generic name for a diverse group of soluble proteins and peptides which act as humoral regulators at nano- to picomolar concentrations and which, either under normal or pathological conditions, modulate the functional activities of individual cells and tissues. These proteins also mediate interactions between cells directly and regulate processes taking place in the extracellular environment. Examples of cytokines include, but are not limited to IP- 10, IL-1, IL-2, IL- 4, IL-5, IL-6, IL-7, IL-10, IL-12, IL-15, IL-18, granulocyte-macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), interferon- γ (γ-IFN), IFN-α, tumor necrosis factor (TNF), TGF-β, FLT-3 ligand, and CD40 ligand. In addition to cytokines the CpG oligonucleotides may be used in combination with antibodies against certain cytokines, such as anti-IL-10 and anti -TGF-β, as well as Cox inhibitors, i.e. COX-1 and COX-2 inhibitors.
The oligonucleotides are also useful for redirecting an immune response from a Th2 immune response to a Thl immune response. This results in the production of a relatively balanced Thl/Th2 environment. Redirection of an immune response from a Th2 to a Thl immune response can be assessed by measuring the levels of cytokines produced in response to the nucleic acid (e.g., by inducing monocytic cells and other cells to produce Thl cytokines, including IL-12, IFN-γ and GM-CSF). The redirection or rebalance of the immune response from a Th2 to a Thl response is particularly useful for the treatment of asthma. For instance, an effective amount for treating asthma can be that amount; useful for redirecting a Th2 type of immune response that is associated with asthma to a Thl type of response or a balanced Thl/Th2 environment. Th2 cytokines, especially IL-4 and IL-5 are elevated in the airways of asthmatic subjects. The CpG immunostimulatory oligonucleotides described herein cause an increase in Thl cytokines which helps to rebalance the immune system, preventing or reducing the adverse effects associated with a predominately Th2 immune response.
The CpG immunostimulatory oligonucleotides have the unique capability to promote cell survival, differentiation, activation and maturation of dendritic cells, and are useful for in vitro, in vivo, and ex vivo methods involving dendritic cells.
CpG immunostimulatory oligonucleotides also increase natural killer cell lytic activity and antibody dependent cellular cytotoxicity (ADCC). ADCC can be performed using a CpG immunostimulatory oligonucleotide in combination with an antibody specific for a cellular target, such as a cancer cell. When the CpG immunostimulatory oligonucleotide is administered to a subject in conjunction with the antibody the subject's immune system is induced to kill the tumor cell. The antibodies useful in the ADCC procedure include antibodies which interact with a cell in the body. Many such antibodies specific for cellular targets have been described in the art and many are commercially available.
The CpG immunostimulatory oligonucleotides may also be administered in conjunction with an anti-cancer therapy. Anti-cancer therapies include cancer medicaments, radiation and surgical procedures. As used herein, a "cancer medicament" refers to an agent which is administered to a subject for the purpose of treating a cancer. As used herein, "treating cancer" includes preventing the development of a cancer, reducing the symptoms of cancer, and/or inhibiting the growth of an established cancer. In other aspects, the cancer medicament is administered to a subject at risk of developing a cancer for the purpose of reducing the risk of developing the cancer. Various types of medicaments for the treatment of cancer are described herein. For the purpose of this specification, cancer medicaments are classified as chemotherapeutic agents, immunotherapeutic agents, cancer vaccines, hormone therapy, and biological response modifiers. Additionally, the methods of the invention are intended to embrace the use of more than one cancer medicament along with the CpG immunostimulatory oligonucleotides. As an example, where appropriate, the CpG immunostimulatory oligonucleotides may be administered with both a chemotherapeutic agent and an immunotherapeutic agent. Alternatively, the cancer medicament may embrace an immunotherapeutic agent and a cancer vaccine, or a chemotherapeutic agent and a cancer vaccine, or a chemotherapeutic agent, an immunotherapeutic agent and a cancer vaccine all administered to one subject for the purpose of treating a subject having a cancer or at risk of developing a cancer.
The chemotherapeutic agent may be selected from the group consisting of methotrexate, vincristine, adriamycin, cisplatin, non-sugar containing chloroethylnitrosoureas, 5-fluorouracil, mitomycin C, bleomycin, doxorubicin, dacarbazine, taxol, fragyline, Meglamine GLA, valrubicin, carmustaine and poliferposan, MMI270, BAY 12-9566, RAS famesyl transferase inhibitor, famesyl transferase inhibitor, MMP, MTA/LY231514, LY264618/Lometexol, Glamolec, CI-994, TNP-470, Hycamtin/Topotecan, PKC412, Valspodar/PSC833, Novantrone/Mitroxantrone, Metaret/Suramin, Batimastat, E7070, BCH-4556, CS-682, 9-AC, AG3340, AG3433, Incel/VX-710, VX-853, ZD0101, ISI641, ODN 698, TA 2516/Marmistat,
BB2516/Marmistat, CDP 845, D2163, PD183805, DX8951f, Lemonal DP 2202, FK 317, Picibanil/OK-432, AD 32/Valrubicin, Metastron/strontium derivative, Temodal/Temozolomide, Evacet/liposomal doxorubicin, Yewtaxan/Paclitaxel, Taxol Paclitaxel, Xeload/Capecitabine, Furtulon/Doxifluridine, Cyclopax/oral paclitaxel, Oral Taxoid, SPU-077/Cisplatin, HMR 1275/Flavopiridol, CP-358 (774)/EGFR, CP-609 (754)/RAS oncogene inhibitor, BMS-182751/oral platinum, UFT(Tegafur/Uracil), Ergamisol/Levamisole, Eniluracil/776C85/5FU enhancer, Campto/Levamisole, Camptosar/Irinotecan, Tumodex/Ralitrexed, Leustatin/Cladribine, Paxex/Paclitaxel, Doxil/liposomal doxorubicin, Caelyx/liposomal doxorubicin, Fludara/Fludarabine, Pharmarubicin Epirubicin, DepoCyt, ZD1839, LU 79553/Bis-Naphtalimide, LU 103793 Dolastain, Caetyx/liposomal doxorubicin, Gemzar/Gemcitabine, ZD 0473/Anormed, YM 116, Iodine seeds, CDK4 and CDK2 inhibitors, PARP inhibitors, D4809/Dexifosamide, Ifes/Mesnex/Ifosamide, Vumon/Teniposide, Paraplatin/Carbop latin, Plantinol/cisplatin, Vepeside/Etoposide, ZD 9331, Taxotere/Docetaxel, prodrug of guanine arabinoside, Taxane Analog, nitrosoureas, alkylating agents such as melphelan and cyclophosphamide, Aminoglutethimide, Asparaginase, Busulfan, Carboplatin, Chlorombucil, Cytarabine HCI, Dactinomycin, Daunorubicin HCI, Estramustine phosphate sodium, Etoposide (VP16-213), Floxuridine, Fluorouracil (5-FU), Flutamide, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alfa-2a, Alfa-2b, Leuprolide acetate (LHRH-releasing factor analogue), Lomustine (CCNU), Mechlorethamine HCI (nitrogen mustard), Mercaptopurine, Mesna, Mitotane (o.p'-DDD), Mitoxantrone HCI, Octreotide, Plicamycin, Procarbazine HCI, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Amsacrine (m-AMSA), Azacitidine, Erthropoietin, Hexamethylmelamine (HMM), Interleukin 2, Mitoguazone (methyl-GAG; methyl glyoxal bis-guanylhydrazone; MGBG), Pentostatin (2'deoxycoformycin), Semustine (methyl-CCNU), Teniposide (VM-26) and Vindesine sulfate, but it is not so limited.
The immunotherapeutic agent may be selected from the group consisting of Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncolym, SMART M195, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-11, MDX-22, OV103, 3622 W94, anti-VEGF, Zenapax, MDX-220, MDX-447, MELIMMUNE-2, MELIMMUNE-1, CEACIDE, Pretarget, NovoMAb-G2, TNT,
Gliomab-H, GNI-250, EMD-72000, LymphoCide, CMA 676, Monopharm-C, 4B5, ior egf.r3, ior c5, BABS, anti-FLK-2, MDX-260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab and ImmuRAIT-CEA, but it is not so limited.
The cancer vaccine may be selected from the group consisting of EGF, Anti- idiotypic cancer vaccines, Gp75 antigen, GMK melanoma vaccine, MGV ganglioside conjugate vaccine, Her2/neu, Ovarex, M-Vax, O-Vax, L-Vax, STn-KHL theratope, BLP25 (MUC-1), liposomal idiotypic vaccine, Melacine, peptide antigen vaccines, toxin/antigen vaccines, MVA-based vaccine, PACIS, BCG vacine, TA-HPV, TA-CIN, DISC-virus and ImmuCyst/TheraCys, but it is not so limited. The use of CpG immunostimulatory oligonucleotides in conjunction with immunotherapeutic agents such as monoclonal antibodies is able to increase long-term survival through a number of mechanisms including significant enhancement of ADCC (as discussed above), activation of natural killer (NK) cells and an increase in IFNα levels. The nucleic acids when used in combination with monoclonal antibodies serve to reduce the dose of the antibody required to achieve a biological result.
The invention also includes methods for inducing antigen non-specific innate immune activation and broad spectrum resistance to infectious challenge using the CpG immunostimulatory oligonucleotides. The term innate immune activation as used herein refers to the activation of immune cells other than memory B cells and for instance can include the activation of NK cells, T cells and/or other immune cells that can respond in an antigen independent fashion. A broad spectrum resistance to infectious challenge is induced because the immune cells are in active form and are primed to respond to any invading compound or microorganism. The cells do not have to be specifically primed against a particular antigen. This is particularly useful in biowarfare, and the other circumstances described above such as travelers.
The CpG immunostimulatory oligonucleotides may be directly administered to the subject or may be administered in conjunction with a nucleic acid delivery complex. A nucleic acid delivery complex shall mean a nucleic acid molecule associated with (e.g. ionically or covalently bound to; or encapsulated within) a targeting means (e.g. a molecule that results in higher affinity binding to target cell. Examples of nucleic acid delivery complexes include nucleic acids associated with a sterol (e.g. cholesterol), a lipid (e.g. a cationic lipid, virosome or liposome), or a target cell specific binding agent (e.g. a ligand recognized by target cell specific receptor). Preferred complexes may be sufficiently stable in vivo to prevent significant uncoupling prior to internalization by the target cell. However, the complex can be cleavable under appropriate conditions within the cell so that the oligonucleotide is released in a functional form. The CpG immunostimulatory oligonucleotide and/or the antigen and/or other therapeutics may be administered alone (e.g., in saline or buffer) or using any delivery vehicles known in the art. For instance the following delivery vehicles have been described: Cochleates (Gould-Fogerite et al, 1994, 1996); Emulsomes (Vancott et al, 1998, Lowell et al, 1997); ISCOMs (Mowat et al, 1993, Carlsson et al, 1991, Hu et, 1998, Morein et al, 1999); Liposomes (Childers et al, 1999, Michalek et al, 1989, 1992, de Haan 1995a, 1995b); Live bacterial vectors (e.g., Salmonella, Escherichia coli, Bacillus calmatte-guerin, Shigella, Lactobacillus) (Hone et al, 1996, Pouwels et al, 1998, Chatfield et al, 1993, Stover et al, 1991, Nugent et al, 1998); Live viral vectors (e.g., Vaccinia, adenovirus, Herpes Simplex) (Gallichan et al, 1993, 1995, Moss et al, 1996, Nugent et al, 1998, Flexner et al, 1988, Morrow et al, 1999); Microspheres
(Gupta et al, 1998, Jones et al, 1996, Maloy et al, 1994, Moore et al, 1995, O'Hagan et al, 1994, Eldridge et al, 1989); Nucleic acid vaccines (Fynan et al, 1993, Kuklin et al. 1997, Sasaki et al, 1998, Okada et al, 1997, Ishii et al, 1997); Polymers (e.g. carboxymethylcellulose, chitosan) (Hamajima et al, 1998, Jabbal-Gill et al, 1998); Polymer rings (Wyatt et al, 1998); Proteosomes (Vancott et al, 1998, Lowell et al, 1988, 1996, 1997); Sodium Fluoride (Hashi et al, 1998); Transgenic plants (Tacket et al, 1998, Mason et al, 1998, Haq et al, 1995); Virosomes (Gluck et al, 1992,
Mengiardi et al, 1995, Cryz et al, 1998); Virus-like particles (Jiang et al, 1999, Leibl et al, 1998). Other delivery vehicles are known in the art.
The term effective amount of a CpG immunostimulatory oligonucleotide refers to the amount necessary or sufficient to realize a desired biologic effect. For example, an effective amount of a CpG immunostimulatory oligonucleotide administered with an antigen for inducing mucosal immunity is that amount necessary to cause the development of IgA in response to an antigen upon exposure to the antigen, whereas that amount required for inducing systemic immunity is that amount necessary to cause the development of IgG in response to an antigen upon exposure to the antigen. Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the particular subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular CpG immunostimulatory oligonucleotide being administered the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular CpG immunostimulatory oligonucleotide and/or antigen and/or other therapeutic agent without necessitating undue experimentation.
Subject doses of the compounds described herein for mucosal or local delivery typically range from about 0.1 μg to 10 mg per administration, which depending on the application could be given daily, weekly, or monthly and any other amount of time therebetween or as otherwise required. More typically mucosal or local doses range from about 10 μg to 5 mg per administration, and most typically from about 100 μg to 1 mg, with 2 - 4 administrations being spaced days or weeks apart. More typically, immune stimulant doses range from 1 μg to 10 mg per administration, and most typically lOμg to 1 mg, with daily or weekly administrations. Subject doses of the compounds described herein for parenteral delivery for the purpose of inducing an antigen-specific immune response, wherein the compounds are delivered with an antigen but not another therapeutic agent are typically 5 to 10,000 times higher than the effective mucosal dose for vaccine adjuvant or immune stimulant applications, and more typically 10 to 1,000 times higher, and most typically 20 to 100 times higher. Doses of the compounds described herein for parenteral delivery for the purpose of inducing an innate immune response or for increasing ADCC or for inducing an antigen specific immune response when the CpG immunostimulatory oligonucleotides are administered in combination with other therapeutic agents or in specialized delivery vehicles typically range from about 1.0 μg to 100 mg per administration, which depending on the application could be given daily, weekly, or monthly and any other amount of time therebetween or as otherwise required. More typically parenteral doses for these purposes range from about 100 μg to 50 mg per administration, and most typically from about 200 μg to 2 mg, with 2 - 4 administrations being spaced days or weeks apart. In some embodiments, however, parenteral doses for these purposes may be used in a range of 5 to 10,000 times higher than the typical doses described above.
For any compound described herein the therapeutically effective amount can be initially determined from animal models. A therapeutically effective dose can also be determined from human data for other CpG oligonucleotides which have been tested in humans (human clinical trials are ongoing) and for compounds which are known to exhibit similar pharmacological activities, such as other adjuvants, e.g., LT and other antigens for vaccination purposes. Higher doses may be required for parenteral administration. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.
The formulations of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. For use in therapy, an effective amount of the CpG immunostimulatory oligonucleotide an/or other therapeutics can be administered to a subject by any mode that delivers the compound to the desired surface, e.g., local, mucosal, systemic. Administering the pharmaceutical composition of the present invention may be accomplished by any means known to the skilled artisan. Preferred routes of administration include but are not limited to oral, parenteral, intramuscular, intranasal, sublingual, intratracheal, inhalation, ocular, vaginal, and rectal.
For oral administration, the compounds (i.e., CpG immunostimulatory oligonucleotides, antigens and/or other therapeutic agents) can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers for neutralizing internal acid conditions or may be administered without any carriers.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. The compounds may be administered by inhalation to pulmonary tract, especially the bronchi and more particularly into the alveoli of the deep lung, using standard inhalation devices. The compounds may be delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. An inhalation apparatus may be used to deliver the compounds to a subject. An inhalation apparatus, as used herein, is any device for administering an aerosol, such as dry powdered form of the compounds. This type of equipment is well known in the art and has been described in detail, such as that description found in Remington: The Science and Practice of
Pharmacy, 19th Edition, 1995, Mac Publishing Company, Easton, Pennsylvania, pages 1676-1692. Many U.S. patents also describe inhalation devices, such as U.S. Patent No. 6,116,237.
"Powder" as used herein refers to a composition that consists of finely dispersed solid particles. Preferably the compounds are relatively free flowing and capable of being dispersed in an inhalation device and subsequently inhaled by a subject so that the compounds reach the lungs to permit penetration into the alveoli. A "dry powder" refers to a powder composition that has a moisture content such that the particles are readily dispersible in an inhalation device to form an aerosol. The moisture content is generally below about 10%> by weight (% w) water, and in some embodiments is below about 5% w and preferably less than about 3% w. The powder may be formulated with polymers or optionally may be formulated with other materials such as liposomes, albumin and/or other carriers.
Aerosol dosage and delivery systems may be selected for a particular therapeutic application by one of skill in the art, such as described, for example in Gonda, I. "Aerosols for delivery of therapeutic and diagnostic agents to the respiratory tract," in Critical Reviews in Therapeutic Drug Carrier Systems, 6:273-313 (1990), and in Moren, "Aerosol dosage forms and formulations," in Aerosols in Medicine. Principles, Diagnosis and Therapy, Moren, et al, Eds, Esevier, Amsterdam, 1985.
The compounds, when it is desirable to deliver them system ically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents .
Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides. In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin. The pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above. The pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see Langer, Science 249:1527-1533, 1990, which is incorporated herein by reference.
The CpG immunostimulatory oligonucleotides and optionally other therapeutics and/or antigens may be administered per se (neat) or in the form of a pharmaceutically acceptable salt. When used in medicine the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (l-3%> w/v); boric acid and a salt (0.5-2.5%) w/v); and phosphoric acid and a salt (0.8-2%) w/v). Suitable preservatives include benzalkonium chloride (0.003-0.03%) w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004- 0.02% w/v).
The pharmaceutical compositions of the invention contain an effective amount of a CpG immunostimulatory oligonucleotide and optionally antigens and/or other therapeutic agents optionally included in a pharmaceutically-acceptable carrier. The term pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference.
Examples Materials and Methods:
Oligodeoxynucleotides All ODNs were provided by Coley Pharmaceutical GmbH (Langenfeld, Germany). ODNs were diluted in phosphate-buffered saline (Sigma, Germany), and stored at -20° C. All dilutions were carried out using pyrogen-free reagents. The ODNs used in the studies described below are shown in Table 1.
Table 1: Sequences of ODNs shown in the drawings.
^*γ* *^*^* *r *T*T*T*T*T*T*T*T*T*T*T fSEO ID NO ' 1) T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T (SEO ID NO 2) T*G*A*C*T*G*T*G*A*A*C*G*T*T*C*G*A*G*A*T*G*A (SEQ. ID NO.: 3)
T*C*G*T*G*A*C*T*G*T*G*A*A*C*G*T*T*C*G*A*G*A*T*G*A (SEQ. ID NO.: 4) χ*C*G*C*T*G*T*G*A*A*C*G*T*T*C*G*A*G*A*T*G*A (SEQ. ID NO.:
5)
T*C*C*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T (SEQ. ID NO.: 6) T*C*G*T*C*C*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T SEQ IDN0.: 5 7)
T* ΦT*T*T*T* *T* *T*T* *T.T* *T*T tSEO ID NO ' 8)
T*C*G*A*A*A*A*A*A*A*A*A*A*A*A*A*A (SEQ. ID NO.: 9) T*G*C*T*T*T*T*T*T*T*T*T*T*T*T*T*T fSEO IDNO' 10")
Figure imgf000050_0001
,Q φQ ' ' φ'j' 'j' ' T'φ' ' ' φ πφ' φ' Φ'TΦ' Φ' CSgQ IT) MQ • 1 ".
T*C*G*T*T*T*T*T*T*T*T*T*T*T:!:T*T*T*T*T*T*T fSEO IDNO' 13") A*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T fSEO ID NO 14"! C*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T SEO IDNO- 15) G*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T fSEO IDNO' 16)
J5 'Φ^ΦQ ' φ'J'Φ' φ''Φ' Φ' Φ'pΦ' φ' ' φ' φ' ' Φ' (SEO IDNO" 17)
^*G ^ GΦGΦ^*γ*GΦγ G*G ,φγ GG G q, GΦ^*G G 'rΦτ ('ggQ in
NO.: 18)
T*C*G*T*C*G*T*T*G*T*C*G*T*T*G*T*C*G*T*T*G*T*C*G*T*T fSEO IDNO.: 19) 20 TΦCΦCΦAΦTΦGΦAΦCΦGΦTΦTΦCΦCΦTΦGΦAΦCΦGΦT*T (SEQ ID N0.: 20)
T*C*G*A*T*G*A*C*G*T*T*C*C*T*G*A*C*G*T*T (SEQ. ID NO.: 21) T*C*G*T*C*G*T*C*C*A*G*G*A*C*T*T*C*T*C*T:1:C*A*G*G*T*T tSEO
IDNO.: 22)
T*C*G*T*C*G*C*T*G*T*G*A*A*C*G*T*T*C*G*A*G*A*T*G*A(SEQ. 25 IDNO.: 23)
T*C*G*T*G*A*C*T*G*T*G*A*A*C*G*T*T*C*G*A*G*A*T*G*A(SEQ. IDNO.: 24)
T*C_G*T*T*T*T*T*T*T*T*T*T*T*T*T*T (SEQ. ID NO.: 25) τ*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T*T fSEO ID 30 NO.: 26)
T*T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T fSEO ID NO ' 27)
' ' φ' Q ' ' φ' ' φ' ' Φ' φ' φ' ' Φ' ' (gβQ JJ) O ' 28) ^Φ' ' Φ' ^ QΦ' Φ' Φ' Φ' Φ' ' ' Φ' ' ' ' C§£Q U) sQ • 29)
' φ' φ' ' ' φ φQ ' Φ' φ' ' 'J' ' φ'pφ' ' SEQ ID NO ' 30)
T*C*C*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T (SEQ. ID NO.: 31) T*C*G*T*T*T*T*T*T*T*T*T*T (SEO ID NO 32) T*C_G*T*C_G*T*T*T*T_G*T*C_G*T*T (SEQ. ID NO.: 33)
T*T*C_G*T*C_G*T*T*T*C_G*T*C_G*T*T (SEQ. ID NO.: 34) T*T*C_G*T*C_G*T*T*T*T_G*T*C_G*T*T (SEQ. ID NO.: 35) T*T*C*G*T*C*G*T*T*T*C*G*T*C*G*T*T (SEQ. ID NO.: 36) T*T*T*C_G*T*C_G*T*T*T*C_G*T*C_G*T*T (SEQ. ID NO.: 37) T*T*G_C*T*C_G*T*T*T*C_G*T*C_G*T*T (SEQ. IDNO.: 38)
T*T*G_C*T*G_C*T*T*T*C_G*T*C_G*T*T (SEQ. ID NO.: 39)
T*T*G_C*T*G_C*T*T*T*G_C*T*G_C*T*T (SEQ. ID NO.: 40)
T*C*G*A*A*A*A*A*A*A*A*A*A*T*A*A*A(8EQ. IDNO.: 41)
T*C*G*A*A*A*A*A*A*A*A*A*T*T*A*A*A(SEQ. IDNO.: 42) TΦCΦGΦAΦAΦAΦAΦAΦAΦAΦTΦTΦT*TΦAΦAΦA (SEQ. ID N0.: 43)
T*C*G*A*A*A*A*A*T*T*T*T*T*T*A*A*A (SEQ .ID NO .: 44)
T*C*G*A*A*A*T*T*T*T*T*T*T*T*T*T*A (SEQ. ID NO.: 45)
T*C*G*T*A*A*A*A*A*A*A*A*A*A*A*A*A (SEQ. ID NO.: 46) T*C*GΦTΦTΦTΦAΦAΦAΦAΦAΦAΦAΦAΦAΦAΦA (SEQ. ID N0.47) T*C*G*U*U*U*U*U*U*U*U*U*U*U*l)*U*U (SEQ. ID NO.: 48)
U*U*U*U*U*U*U*U*U*U*U*U*U*U*U*U*U(SEQ. IDNO.: 49) T*C*G*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T (SEQ. ID NO.: 50) T*C*G*C*C*C*C*C*C*C*C*C*C*C*C*C*C (SEQ. IDNO.: 51) T*C*G*T*C*G*A*G*C*G*T*G*C*G*C*C*A*T (SEQ IDNO.52) T*C*G*C*C*C*A*G*C*G*T*G*C*G*C*C*A*T (SEQ ID NO.53)
U*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T (SEQ. ID NO.: 54) T*C*U*T*T*T*T*T*T*T*T*T*T*T*T*T*T (SEQ ID NO 55) T*U*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T (SEQ ID NO 56) T*C*G*T*T*T*T*T*T*T*T*T*T*T*T (SEQ ID NO.57) γΦ^ O G ,p r,Φr Φ' Φ' r r *r r ' Φrp ' Φ' S Q ID NO 58)
^ΦγΦ ,*, Φ' Φrj, ' *' , G GΦ' Φ' ' ' rp*' (SEQ ID NO 59) T*C*T*C*C*C*A*G*C*G*T*G*C*G*C*C*A*T (SEQ ID NO.60) *: Phosphorothioate linkage; _: Phosphodiester linkage.
Cell purification Peripheral blood buffy coat preparations from healthy male and female human donors were obtained from the German Red Cross (Rathingen, Germany) or from the Blood Bank of the University of Dusseldorf (Germany) and from these, PBMC were purified by centrifugation over Ficoll-Hypaque (Sigma). The purified PBMC were either used fresh (for most assays) or were suspended in freezing medium and stored at -70°C. When required, aliquots of these cells were thawed, washed and resuspended in RPMI 1640 culture medium supplemented with 5% (v/v) heat inactivated human AB serum (BioWhittaker, Belgium) or 10%> (v/v) heat inactivated FCS, 1.5mM L-glutamine, lOOU/ml penicillin and lOOμg/ml streptomycin (all from Sigma).
Cytokine detection Thawed or fresh PBMC were resuspended at a concentration of 3xl06/ml to 5xl06/ml and added to plates which had previously received nothing or ODN in a variety of concentrations. The cells were cultured in a humidified incubator at 37°C. Culture supernatants were collected after the indicated time points. If not used immediately, supernatants were frozen at -20°C until required. Amounts of cytokines in the supernatants were assessed using commercially available ELISA Kits or in-house ELISA developed using commercially available antibodies (e.g. from Becton Dickinson, Germany).
Example 1: 5'-TCG enhances immunostimulatory activity of non-CpG or CpG ODNs (IL-10).
Human PBMC of two representative donors were incubated for 48h with the indicated ODNs (Figure 1). Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. The activity of non-CpG ODNs such as poly T ODNs or another non-CpG ODN, SEQ. ID NO.: 6, were strongly enhanced by adding a TCG trinucleotide to the 5' end. CpG ODNs lacking a 5'-TCG such as SEQ. ID NO.: 3, (Magone et al, Eur. J. Immunol. 2000; 30: 1841 - 1850) could also be modified to exhibit higher potency and/or efficacy with the addition of a 5'TCG. Exam le 2: TCG enhances immunostimulatory activity of non-CpG or CpG ODNs (IFN-α).
Human PBMC of two representative donors were incubated for 48h with the indicated ODNs (Figure 2). Supernatants were harvested and IFN-α measured by ELISA as described in Materials and Methods. The effects of a 5'-TCG modification for the same ODNs as shown in Figure 1 are demonstrated in this IFN-α assay.
Example 3: Enhancement of the immune response is dependent on the CpG dinucleotide. Human PBMC of two representative donors were incubated for 48h with the indicated ODNs (Figure 3). Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. Shown are the effect of a 5' -TCG, SEQ. ID NO.: 2, and a poly T sequence, SEQ. ID NO.: 8. Although a 5'-TGC, SEQ. ID NO.: 10, may have some minimal effect, the 5' -TCG modification clearly resulted in a much stronger potentiation of cytokine secretion. A 5'-TCG modification of a 17mer poly A ODN did not appear to have an effect, SEQ. ID NO.: 9. Similar results were obtained for interferon secretion. In contrast to the 5'-TCG plus poly A ODN a 5'-TCG ODN in a poly uracil context lead to enhancement of IL-10 secretion (Shown in Figure 6).
Example 4: Shifting the CpG dinucleotide from the 5' to the 3' end of an
ODN results in graded loss of immunostimulatory capability.
Human PBMC of two representative donors were incubated for 48h with the indicated ODNs (Figure 4). Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. The CpG dinucleotide at the 5' end of an ODN resulted in enhanced IL-10 secretion, SEQ. ID NO.: 2. Shifting the CpG to the 3' end, SEQ. ID NO.: 11, of a poly T ODN resulted in strongly reduced cytokine secretion. A 5'-CG, SEQ. ID NO.: 12, had also a potentiating effect although a 5'-TCG was more efficient in enhancing the cytokine response. Similar results were obtained for interferon secretion. Example 5: The length of an ODN has an effect on stimulatory activity in addition to a 5'-TCG.
Human PBMC of two representative donors were incubated for 48h with the indicated ODNs (Figure 5). Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. The data demonstrate that the length of a CpG ODN plays a role in the stimulatory activity in addition to the 5' -TCG ODNs. A 21mer is more potent and efficient than a 17mer which is more potent than a 15mer or a 13mer.
Example 6; A 5'-TCG is the most stimulatory 5' modification, but other modifications also lead to enhanced immunostimulation.
Human PBMC of three representative donors were incubated for 48h with the indicated ODNs (Figure 6). Supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods. A 5'-TCG is clearly the most potent 5' modification as demonstrated in the above experiment. Nevertheless, other 5' modifications were also able to enhance the stimulatory capability of poly T ODNs on human cells. Surprisingly, a 5'-TC alone was able to enhance cytokine secretion. Other 5' trinucleotides such as ACG, CCG and GCG lead also to enhanced IL-10 secretion although the 5'-TCG showed the strongest effects. Even a 5'-TTG was shown be more stimulatory than a purely poly T ODN. In addition, specific modifications of the sequence 3' to the 5'-TCG retained immunostimulation. In contrast to the 5'-TCG plus poly A ODN (Figure 3) a 5' -TCG ODN in a poly uracil context lead to enhancement of IL-10 secretion.
Example 7: The 5'-TCG modification enhances the stimulatory capability of CpG ODNs as shown by different cellular effects. a. Human PBMC of three representative donors were incubated for 48h with the indicated ODNs ands supernatants were harvested and IL-10 measured by ELISA as described in Materials and Methods (Figure 7A). b. Human PBMC of three representative donors were incubated for 48h with the indicated ODNs and supernatants were harvested and IFN-α measured by ELISA as described in Materials and Methods (Figure 7B).
c. Human PBMC of two representative donors were incubated for 20h with the indicated ODNs and supernatants were harvested and IL-6 measured by ELISA as described in Materials and Methods (Figure 7C).
Figures 7A to 7C demonstrate that the 5' -TCG is able to enhance the stimulatory capacity of an ODN in a variety of assays. The parent CpG ODN SEQ. ID NO.: 18 does not have a CpG dinucleotide directly at the 5' end. Modifying the sequence with a 5'- TCG, SEQ. ID NO.: 19, also enhanced the activity of the CpG ODN.
Example 8: IL-10 secretion induced by ODN with 5'-TCG.
Human PBMC were incubated with increasing concentrations of the indicated ODN's for 48h. Supernatant was harvested and IL-10 measured by ELISA as described in Materials and Methods. Shown is the result for three individual donors. This experiment was a dose response study that investigated the contribution of thymidines 3' of the 5'-TCG trinucleotide with ODN concentrations up to lμM. As shown in Figure 8, at lμM low stimulation of cytokine secretion (IL-10) can be observed with SEQ. ID NO.: 9 (5'-TCG plus poly A).
Example 9: IL-10 secretion induced by ODN with 5'-TCG and increasing numbers of thymidines.
Human PBMC were incubated with the indicated ODN for 48h. Supernatant was harvested and IL-10 measured by ELISA as described in Materials and Methods. Shown is the result for three individual donors. To investigate whether the addition of thymidines to the 3' tail of ODN's would increase in vitro cytokine production, the ODN's were modified by exchanging increasing numbers of adenosines to thymidines. The addition of only one thymidine to the poly A tail led to an increase of immunostimulation as can be observed with SEQ. ID NO.: 41 (Figure 9). Adding more thymidines led to further increase of IL-10 production, dependent on the number of thymidines. A 5'-TCG was sufficient to enhance immunostimulation by phosphorothioate ODN independent of the nucleotide sequence. Nevertheless, an increasing number of pyrimidines further contributed to this stimulation. Only two to four thymidines 3' of the 5'-TCG (here: about at least 20% thymidines) were sufficient to lead to a significant increase of cytokine secretion (SEQ. ID NO.: 42 and SEQ. ID NO.: 43).
Example 10: ODN with a 5'-TCG are the most potent and efficient ODN to induce a strong Thl-mediated immune response.
Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations (Figure 10A and 10B). Supernatants were harvested and IL-10 and IFN-α measured by ELISA as described in Materials and Methods. Shown is the Mean±SEM. The 5' -TCG led to the most potent and efficient immune stimulation of all ODN tested (in terms of the B cell related cytokine IL-10). Nevertheless, when the potential of ODN's with different 5' ends to induce the Thl related cytokine IFN-α was measured, it was observed that only the 5' -TCG supported strong secretion of this cytokine (Figure 10B).
Example 11: The position of CpG dinucleotides in immune stimulatory ODN determines the strength of type I IFN secretion. Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations. Supernatants were harvested and IFN-α measured by ELISA as described in Materials and Methods. Shown is the Mean. Shifting the CpG dinucleotide (essential for efficient immune stimulation) from the 5' to the 3' end led to a graded loss of immune stimulation (measured as secretion of the B cell related cytokine IL-10). Figure 11 demonstrates that the position of the CpG also strongly influences the strength of type I IFN secretion. Surprisingly, shifting the CpG only one to three positions to the 3' end led to strongly enhanced IFN-α secretion especially with ODN SEQ. ID NO.: 27 and SEQ. ID NO.: 28. Shifting further to the 3' end led to strong decrease of IFN-α secretion below the level of SEQ. ID NO.: 2 (5'-TCG).
Example 12: Type I IFN secretion induced by short 5'-TCG ODN. Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations. Supernatants were harvested and IFN-α measured by ELISA as described in Materials and Methods. Shown is the Mean±SEM. Previous findings have shown decrease of immune stimulation (measured as IL-10 secretion) upon shortening of the ODN's length. Nevertheless, when the secretion of IFN-α by shortened ODN's (e.g. 13mer SEQ. ID NO.: 32 with 5'-TCG) was measured, surprisingly a strongly increased IFN-α secretion compared to the 17mer SEQ. ID NO.: 2 was observed (Figure 12).
Example 13: In vitro immune stimulation by a panel of newly generated
CpG ODN according to the observations described herein.
The important observations that were described in the previous examples were: a. A 5' TCG supports efficient and potent IFN-α (a Thl related cytokine) as well as IL-10 secretion (a B cell related cytokine); b. Shifting the CpG dinucleotide from the 5' to the 3' end led first to an increase of type I IFN secretion and further 3' shifts led to a decrease (B cell activation was only decreased or only slightly changed by CpG shifts); c. Shortening an ODN with a 5'-TCG led to a strong increase in the potential to induce IFN-α (in contrast to other effects, e.g. secretion of IL-10);
These observations were combined and a panel of short CpG ODN's was created that were tested for their potential to induce the secretion of IFN-α as well as to activate B cells. Human PBMC of three representative donors were incubated for 48h with the indicated ODN concentrations. Supernatants were harvested and IL-10 and IFN-α measured by ELISA as described in Materials and Methods. Shown is the Mean±SEM. As demonstrated in Figurel3A ODN's were generated with lengths below 20 nucleotides that induced more efficient IFN-α secretion than a typical 24mer B-Class ODN, SEQ. ID NO.: 26. The difference between ODN SEQ. ID NO.: 36 (PS) and SEQ. ID NO.: 35 (semi-soft) in Figure 13A indicated a shift of the bell-shaped curve to lower ODN concentrations (down-turn of the activation curve can be observed at lower ODN concentrations with SEQ. ID NO.: 35). In addition, loss of single to all CpG dinucleotides as in SEQ. ID NO.: 38 to SEQ. ID NO.: 40 led to a decrease of cytokine secretion, confirming that the observed effects were CpG-dependent. Figures 13B (and Figure 14) demonstrate that such short ODN were perfectly able to induce the activation of B cells (measured as CD80 up-regulation on CD19-positive B cells as well as secretion of the cytokine IL-10 produced by B cells).
Example 14: Short CpG ODN are perfectly able to induce efficient B cell stimulation.
Human PBMC of three representative donors were incubated for 24h with the indicated ODN concentrations and cells harvested and stained for CD 19, CD 14 and CD80. Expression of CD80 on CD19-positive B cells was measured by flow cytometry as described. Figure 14 demonstrates that such short ODN were perfectly able to induce the activation of B cells (measured as CD80 up-regulation on CD19-positive B cells as well as secretion of the cytokine IL-10 produced by B cells).
Example 15: A phosphodiester linkage between the C and G of the 5' CpG dinucleotide results in enhancement of potency of immune stimulation.
Human PBMC of three representative donors were incubated with the indicated ODN concentrations for 48h. Supernatants were harvested and IL-10 measured by ELISA as described above. The introduction of a phosphodiester linkage between the CpG dinucleotide in an ODN with a 5'-TCG, SEQ. ID NO.: 25, led to a shift of the IL- 10 secretion to lower ODN concentrations compared to an ODN with an unmodified 5'- TCG (SEQ. ID NO.: 2). The data is shown in Figure 15. A similar result was obtained for IFN-alpha secretion.
Example 16: Modifications of the T preceeding the 5'-CG are allowed.
Human PBMC of three representative donors were incubated with the indicated ODN concentrations for 48h. Supernatants were harvested and IL-10 measured by ELISA as described above. The result shown in Figure 16 demonstrate that:
1. an ODN with a 5'-UCG (SEQ. ID NO. 54) induced similar strong cytokine secretion as an ODN with a 5'-TCG (SEQ. ID NO.: 2). Both ODN were superior to a pure poly T ODN (SEQ. ID NO.: 8). This result suggests that a variety of chemical modified nucleotides 5' to the CpG are allowed to induce an enhanced immune stimulation. 2. ODN with a 5'-TCU (SEQ. ID NO. 55) or 5'-TUG (SEQ. ID NO. 56)also demonstrated enhanced cytokine secretion when compared to a poly T ODN (SEQ. ID NO.: 8). Nevertheless, a 5'-TCG was superior to these two modifications and the 5'-TCU induced more efficient IL-10 secretion than the 5'- TUG. These results suggest that a variety of chemical modifications at the CpG dinucleotide are allowed to induce enhanced immune stimulation. The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. Various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention.
We claim:

Claims

1. An oligonucleotide comprising: 5'TCGXιX2Nι3' wherein Xi is any nucleotide, X2 is A, T, or C when Xi is C or A, X2 is A or G when Xi is T, X2 is any nucleotide when X] is G, Ni is 2-95 nucleotides, wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, and wherein Ni does not include an unmethylated CG motif.
2. An oligonucleotide comprising: 5'TCGTN,3' wherein Nj is 3-96 nucleotides, wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein N] does not include an unmethylated CG motif and when Ni is 16 nucleotides Ni does not include a Cι2 and when JvTi is 8 nucleotides Ni is at least 50% C or 70% T.
3. An oligonucleotide comprising: 5'TCGANι3' wherein Ni is 3-96 nucleotides, wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein Nj does not include an unmethylated CG motif and when i is 19 nucleotides Ni is at least 55%> pyrimidine, and when Ni is 8 nucleotides Ni is at least 50%> T or C.
4. An oligonucleotide comprising: 5'TCGN)3' wherein Ni is 10-96 nucleotides, wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein the C content of the oligonucleotide is less than or equal to 60%>, and the A content is less than or equal to 30%>, and wherein Ni does not include an unmethylated CG motif.
5. An oligonucleotide comprising:
5'TYZN,35 wherein Y is a cytosine or modified cystosine, wherein Z is a guanine or modified guanine, Ni is 4-97 nucleotides, wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, and wherein the oligonucleotide does not include an unmethylated CG motif.
6. The oligonucleotide of any one of claims 1-5, wherein the oligonucleotide includes at least 1 modified internucleotide linkage.
7. The oligonucleotide of any one of claims 1-5, wherein the oligonucleotide includes at least 50%> modified internucleotide linkage.
8. The oligonucleotide of any one of claims 1-5, wherein all internucleotide linkages of the oligonucleotide are modified.
9. The oligonucleotide of any one of claims 1-5, wherein the oligonucleotide is
20-100 nucleotides in length.
10. The oligonucleotide of claim 6, wherein the stabilized internucleotide linkage is a phosphorothioate linkage.
11. The oligonucleotide of any one of claims 3 or 4, wherein the oligonucleotide has the following structure: 5' T*C*G*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T 3' (SEQ. ID NO.: 50) and wherein * refers to a phosphorothioate linkage.
12. The oligonucleotide of any one of claims 2 or 4, wherein the oligonucleotide has the following structure: 5' τ*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T 3' (SEQ. ID NO.: 2) and wherein * refers to a phosphorothioate linkage.
13. The oligonucleotide of any one of claims 1-5, wherein Nj is N2N3 and wherein N is 8-94 nucleotides and N3 is 2-5 pyrimidines.
14. The oligonucleotide of claim 13, wherein N3 is TTTTT.
15. The oligonucleotide of claim 13, wherein N3 is TT.
16. The oligonucleotide of claim 13, wherein N2 is 8-40 nucleotides.
17. The oligonucleotide of any one of claims 1-5, wherein i is at least 50%) pyrimidine.
18. The oligonucleotide of any one of claims 1-5, wherein Ni is at least 80%> pyrimidine.
19. The oligonucleotide of any one of claims 1-5, wherein Ni is free of Poly-A and Poly-G sequences.
20. The oligonucleotide of any one of claims 1-5, wherein Ni is TN2 and wherein
N2 is 8-94 nucleotides.
21. The oligonucleotide of any one of claims 1-5, wherein Y is selected from the group of modified cystosine bases consisting of 5-methyl cytosine, , 5-methyl- isocytosine, 5-hydroxy-cytosine, 5-halogeno cytosine, uracil, N4-ethyl-cytosine, , 5- fluoro-uracil, and hydrogen.
22. The oligonucleotide of any one of claims 1-5, wherein Z is selected from the ' group of modified guanine bases consisting of 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine), 7-deaza-8-substituted guanine, hypoxanthine, 2,6-diaminopurine, 2-aminopurine, purine, 8-substituted guanine such as 8-hydroxyguanine, and 6-thioguanine, , 2-aminopurine, , and hydogen
23. The oligonucleotide of any one of claims 1-5, wherein the oligonucleotide has a 3'-3' linkage with one or two accessible 5' ends.
24. The oligonucleotide of claim 23, wherein the oligonucleotide has two accessible 5' ends, each of which are 5'TCG.
25. A method for treating allergy or asthma, comprising: administering to a subject having or at risk of having allergy or asthma an oligonucleotide of any one of claims 1-5 in an effective amount to treat allergy or asthma.
26. The method of claim 25, wherein the oligonucleotide is administered to a respiratory tissue.
27. The method of claim 25, wherein the subject has or is at risk of developing allergic asthma.
28. A method for inducing cytokine production, comprising: administering to a subject an oligonucleotide of any one of claims 1-5 in an effective amount to induce a cytokine selected from the group consisting of IP 10, IL6, IL12, IL18, TNF, chemokines, IFN-α and IFN-γ.
29. A method for treating infectious disease, comprising: administering to a subject having or at risk of having an infectious disease an oligonucleotide of any one of claims 1-5 in an effective amount to treat the infectious disease.
30. The method of claim 29 wherein the subject has or is at risk of having a bacterial infection.
31. The method of claim 29 wherein the subject has or is at risk of having a viral infection.
32. A method for treating cancer, comprising: administering to a subject having or at risk of having cancer an oligonucleotide of any one of claims 1-5 in an effective amount to treat cancer.
33. The method of claim 32, wherein the cancer is selected from the group consisting of biliary tract cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, gastric cancer, intraepithelial neoplasms, lymphomas, liver cancer, lung cancer (e.g. small cell and non-small cell), melanoma, neuroblastomas, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, sarcomas, thyroid cancer, renal cancer, bone cancer, brain and CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer, as well as other carcinomas and sarcomas.
34. The method of claim 32, further comprising administering an anti-cancer agent.
35. A method for inducing innate immunity in a subject, comprising: administering to a subject an oligonucleotide of any one of claims 1-5 in an effective amount to induce innate immunity.
36. A method for inducing a Thl immune response, comprising: administering to a subject an oligonucleotide of any one of claims 1-5 in an effective amount to induce a Thl immune response.
37. A method of modulating an immune response in a subject, comprising administering to the subject an effective amount for modulating an immune response of an oligonucleotide comprising: 5'-XιYRMj-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein Xi is a nucleotide, wherein Y is a cytosine or a modified cytosine, wherein R is a guanine or a modified guanine, and wherein Mi is a nucleic acid of 1-3 nucleotides.
38. The method of claim 37, wherein the internucleotide linkages of the oligonucleotide are stabilized phosphorothioate internucleotide linkages.
39. The method of claim 38, wherein the internucleotide linkage between Y and R is a phosphodiester linkage in an Rp configuration.
40. The method of claim 37, wherein the modified cytosine has a C5 substitution.
41. The method of claim 37, wherein the modified guanine has a C8 or C7 substitution.
42. The method of claim 37, wherein the modified or modified cytosine or guanine is selected from the group consisting of 5 -substituted cytosines (e.g. 5-methyl- cytosine, 5-fluoro-cytosine, 5-chloro-cytosine, 5-bromo-cytosine, 5-iodo-cytosine, 5- hydroxy-cytosine, 5-hydroxymethyl-cytosine, 5-difluoromethyl-cytosine, and unsubstituted or substituted 5-alkynyl-cytosine), 6-substituted cytosines, N4-substituted cytosines (e.g. N4-ethyl-cytosine), 5-aza-cytosine, 2-mercapto-cytosine, isocytosine, pseudo-isocytosine, cytosine analogs with condensed ring systems (e.g. N,N'-propylene cytosine or phenoxazine), and uracil and its derivatives (e.g. 5-fluoro-uracil, 5-bromo- uracil, 5-bromovinyl-uracil, 4-thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil), thymine derivatives (e.g. 2-thiothymine, 4-thiothymine, 6-substituted thymines), 7-deazaguanine, 7-deaza-7-substituted guanine (such as 7-deaza-7-(C2-C6)alkynylguanine),
7-deaza-8-substituted guanine, 7-deaza-8-aza guanine, hypoxanthine, N2-substituted guanines (e.g. N2-methyl-guanine), 5-amino-3-methyl-3H,6H-thiazolo[4,5- d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, adenine, substituted adenines (e.g. N6-methyl-adenine, 8-oxo-adenine) 8-substituted guanine (e.g. 8 -hydroxy guanine and 8-bromoguanine), and 6-thioguanine. In another embodiment of the invention, the base is substituted by a universal base (e.g. 4-methyl- indole, 5-nitro-indole, 3-nitropyrrole, P-base, and K-base), an aromatic ring system (e.g. benzimidazole or dichloro- benzimidazole, l-methyl-lH-[l,2,4]triazole-3-carboxylic acid amide) an aromatic ring system (e.g. fluorobenzene or difluorobenzene) and a hydrogen atom (dSpacer).
43. The method of claim 37 wherein the oligonucleotide is associated with a carrier linked to the 3' end of the oligonucleotide.
44. The method of claim 43, wherein the carrier is selected from the group consisting of a microparticle, dendrimer, cholesterol, liposome, cationic complex, and antigen.
45. The method of claim 37, further comprising administering an antigen to the subject.
46. The method of claim 37, further comprising administering a therapeutic protocol to the subject.
47. The method of claim 46, wherein the therapeutic protocol is surgery.
48. The method of claim 37 wherein the oligonucleotide is not associated with a carrier.
49. The method of claim 37 wherein the oligonucleotide is in a multimerized complex..
50. The method of claim 49, wherein the multimerized complex includes the oligonucleotide linked by a multimerization unit to a second oligonucleotide.
51. The method of claim 49, wherein the second oligonucleotide has the formula 5'-XιYRM,-3'.
52. A composition, comprising a multimerized complex of an oligonucleotide comprising:
5'-X2YRM2-3' wherein X2 is a nucleic acid that consists of a single nucleotide, or a dinucleotide or a trinucleotide that does not comprise a CG dinucleotide, wherein Y is a cytosine or a modified cytosine, wherein R is a guanine or a modified guanine, wherein M2 is a nucleic acid of 0-27 nucleotides, and a multimerization unit linked to the 3' end of the oligonucleotide.
53. The composition of claim 52, wherein the multimerization unit is a carrier selected from the group consisting of a microparticle, dendrimer, liposome, cationic complex, cholesterol and antigen.
54. The composition of claim 52, wherein the oligonucleotide is 5'TCG3', 5 CGT3', 5'UCG3', or 5'UCGT3'.
55. The composition of claim 52, wherein X2 is a single nucleotide.
56. The composition of claim 52, wherein X is a pyrimidine.
57. The composition of claim 52, wherein the oligonucleotide has phosphodiester internucleotide linkages.
58. The composition of claim 52, wherein M is free of a CG dinucleotide.
59. The composition of claim 52, further comprising administering an antigen to the subject.
60. The composition of claim 52, further comprising administering a therapeutic protocol to the subject.
61. The composition of claim 60, wherein the therapeutic protocol is surgery.
62. The composition of claim 52, wherein the multimerization unit is a linker between the,3' end of the oligonucleotide and a second oligonucleotide.
63. An oligonucleotide comprising: 5'-X3CGM3-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X3 is a single nucleotide that does not comprise a CG dinucleotide, wherein M3 is a nucleic acid of 3-27 nucleotides that is free of a CG dinucleotide, and wherein M has at least one of the following properties: is free of a TC dinucleotide, is at least 30%> T nucleotides, consists of A, T, and G or is free of a CCTTCC hexamer having at least one modified internucleotide linkage.
64. An oligonucleotide comprising: 5'-X4CGM4-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X4 is a dinucleotide that does not comprise a CG dinucleotide, wherein M is a nucleic acid of 2-26 nucleotides that is free of a CG dinucleotide, and wherein M has at least one of the following properties: is free of a TG or a GT dinucleotide, is at least 38%> T nucleotides or consists of A and T.
65. An oligonucleotide comprising: 5'-XsCGM5-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X5 is a trinucleotide that does not comprise a CG dinucleotide, wherein M5 is a nucleic acid of 1-25 nucleotides that is free of a CG dinucleotide, and wherein Ms has at least one of the following properties: is free of a CT dinucleotide and does not include at least one phosphorothioate linkage, is at least 41%) T nucleotides, or consists of A and C.
66. The oligonucleotide of claim 65, wherein the internucleotide linkage between the C and G nucleotides is a phosphodiester linkage.
67. The oligonucleotide of claim 65 wherein the oligonucleotide includes at least two modified internucleotide linkages.
68. An oligonucleotide comprising: 5'-TTGM6-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein M6 is a nucleic acid that consists of 5-21 nucleotides, wherein M does not comprise a CG dinucleotide, wherein M6 is comprised of at least 30% T nucleotides, and wherein said nucleotide is 10-24 nucleotides in length.
69. An oligonucleotide comprising: 5'-X6CGM7-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X6 is 1-3 nucleotides and does not include a CG dinucleotide, wherein M7 is a nucleic acid of 6-27 nucleotides and includes at least three CG dinucleotides and is at least 50% T nucleotides.
70. The oligonucleotide of claim 69, wherein M7 includes at least four CG dinucleotides.
71. The oligonucleotide of claim 69, wherein at least one CG dinucleotide includes a phosphodiester internucleotide linkage.
72. The oligonucleotide of claim 69, wherein at least three CG dinucleotides includes a phosphodiester internucleotide linkage.
73. The oligonucleotide of claim 69, wherein M7 is 16-18 nucleotides in length.
74. The oligonucleotide of claim 69, wherein the oligonucleotide is selected from the group consisting of SEQ ID NO. 33, 34, 35, 36, and 37.
75. An oligonucleotide comprising: 5'-'TTGM8-3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein M7 is a nucleic acid of 6-18 nucleotides and includes at least one CG dinucleotide and is at least 50% T nucleotides.
76. The oligonucleotide of claim 75, wherein M8 is 14 nucleotides in length.
77. The oligonucleotide of claim 75, wherein the oligonucleotide is selected from the group consisting of SEQ ID NO. 38, 39, and 40.
78. A method for inducing an immune response, comprising: administering to a subject an oligonucleotide of any one of claims 63 to 77 or a composition of claim 52 in an effective amount to induce an immune response.
79. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to induce a cytokine selected from the group consisting of Type I and Type II IFN.
80. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to treat the infectious disease.
81. The method of claim 78 wherein the subject has or is at risk of having a bacterial infection.
82. The method of claim 78 wherein the subject has or is at risk of having a viral infection.
83. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to treat cancer.
84. The method of claim 78, wherein the cancer is selected from the group consisting of biliary tract cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, endometrial cancer, gastric cancer, intraepithelial neoplasms, lymphomas, liver cancer, lung cancer (e.g. small cell and non-small cell), melanoma, neuroblastomas, ovarian cancer, pancreatic cancer, prostate cancer, rectal cancer, sarcomas, thyroid cancer, renal cancer, bone cancer, brain and CNS cancer, connective tissue cancer, esophageal cancer, eye cancer, Hodgkin's lymphoma, larynx cancer, oral cavity cancer, skin cancer, and testicular cancer, as well as other carcinomas and sarcomas.
85. The method of claim 78, further comprising administering an anti-cancer agent.
86. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to induce innate immunity.
87. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to induce a Thl immune response.
88. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount treat allergy.
89. The method of claim 78, wherein the oligonucleotide is administered to the subject in an effective amount to treat asthma.
PCT/US2003/039775 2002-12-11 2003-12-11 5’ cpg nucleic acids and methods of use WO2004053104A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2003300919A AU2003300919A1 (en) 2002-12-11 2003-12-11 5' cpg nucleic acids and methods of use
CA002502015A CA2502015A1 (en) 2002-12-11 2003-12-11 5' cpg nucleic acids and methods of use
JP2005511961A JP2006512927A (en) 2002-12-11 2003-12-11 5 'CPG nucleic acids and methods of use thereof
EP03813010A EP1578954A4 (en) 2002-12-11 2003-12-11 5' cpg nucleic acids and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US43240902P 2002-12-11 2002-12-11
US60/432,409 2002-12-11
US50610803P 2003-09-25 2003-09-25
US60/506,108 2003-09-25

Publications (2)

Publication Number Publication Date
WO2004053104A2 true WO2004053104A2 (en) 2004-06-24
WO2004053104A3 WO2004053104A3 (en) 2005-12-15

Family

ID=32511644

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/039775 WO2004053104A2 (en) 2002-12-11 2003-12-11 5’ cpg nucleic acids and methods of use

Country Status (6)

Country Link
US (1) US7956043B2 (en)
EP (1) EP1578954A4 (en)
JP (2) JP2006512927A (en)
AU (1) AU2003300919A1 (en)
CA (1) CA2502015A1 (en)
WO (1) WO2004053104A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1538904A2 (en) * 2002-08-19 2005-06-15 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
WO2006135434A3 (en) * 2004-10-20 2007-05-10 Coley Pharm Group Inc Semi-soft c-class immunostimulatory oligonucleotides
US8128944B2 (en) 2007-08-13 2012-03-06 Coley Pharmaceutical Gmbh RNA sequence motifs in the context of defined internucleotide linkages inducing specific immune modulatory profiles
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses

Families Citing this family (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US7935675B1 (en) 1994-07-15 2011-05-03 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
EP0855184A1 (en) * 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
WO1999051259A2 (en) * 1998-04-03 1999-10-14 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
DE69932717T2 (en) 1998-05-22 2007-08-09 Ottawa Health Research Institute, Ottawa METHODS AND PRODUCTS FOR INDUCING MUCOSAL IMMUNITY
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
ATE464907T1 (en) 1999-02-17 2010-05-15 Csl Ltd IMMUNOGENIC COMPLEXES AND METHODS RELATING THEM
EP1176966B1 (en) * 1999-04-12 2013-04-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Oligodeoxynucleotide and its use to induce an immune response
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
AP2006003503A0 (en) * 1999-09-25 2006-02-28 Univ Iowa Res Found Immunostimulatory nucleic acids.
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US20030129251A1 (en) 2000-03-10 2003-07-10 Gary Van Nest Biodegradable immunomodulatory formulations and methods for use thereof
AU7013401A (en) * 2000-06-22 2002-01-02 Univ Iowa Res Found Methods for enhancing antibody-induced cell lysis and treating cancer
WO2002022809A2 (en) * 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
CN100334228C (en) * 2001-06-21 2007-08-29 戴纳瓦克斯技术公司 Cimeric immunomodulatory compounds and methods of using the same
SI1446162T1 (en) 2001-08-17 2009-04-30 Coley Pharm Gmbh Combination motif immune stimulatory oligonucleotides with improved activity
AU2002360278A1 (en) * 2001-10-12 2003-11-11 Coley Pharmaceutical Gmbh Methods and products for enhancing immune responses using imidazoquinoline compounds
US7276489B2 (en) * 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
US8153141B2 (en) 2002-04-04 2012-04-10 Coley Pharmaceutical Gmbh Immunostimulatory G, U-containing oligoribonucleotides
KR100456681B1 (en) 2002-05-22 2004-11-10 주식회사 대웅 Immnune-stimulating and controlling Composition comprising bacterial chromosomal DNA fragments and detoxified lipopolysaccharides
US7569553B2 (en) * 2002-07-03 2009-08-04 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7807803B2 (en) 2002-07-03 2010-10-05 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US20040053880A1 (en) * 2002-07-03 2004-03-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
MXPA05004588A (en) 2002-10-29 2005-12-14 Coley Pharmaceutical Group Ltd Use of cpg oligonucleotides in the treatment of hepatitis c virus infection.
AU2003300919A1 (en) 2002-12-11 2004-06-30 Coley Pharmaceutical Gmbh 5' cpg nucleic acids and methods of use
JP2007524615A (en) 2003-06-20 2007-08-30 コーリー ファーマシューティカル ゲーエムベーハー Low molecular weight Toll-like receptor (TLR) antagonist
CA2536139A1 (en) * 2003-09-25 2005-04-07 Coley Pharmaceutical Group, Inc. Nucleic acid-lipophilic conjugates
CA2540949A1 (en) 2003-10-30 2005-05-12 Coley Pharmaceutical Gmbh C-class oligonucleotide analogs with enhanced immunostimulatory potency
US20050100983A1 (en) * 2003-11-06 2005-05-12 Coley Pharmaceutical Gmbh Cell-free methods for identifying compounds that affect toll-like receptor 9 (TLR9) signaling
EP1730281A2 (en) * 2004-04-02 2006-12-13 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for inducing il-10 responses
US20060213010A1 (en) * 2005-03-22 2006-09-28 Davis David T Mattress sled
US20060241076A1 (en) * 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
US7754679B2 (en) * 2005-11-16 2010-07-13 Idexx Laboratories, Inc. Pharmaceutical compositions for the administration of aptamers
US8114440B2 (en) * 2005-11-16 2012-02-14 Idexx Laboratories Inc. Pharmaceutical compositions for the administration of aptamers
DK1957647T3 (en) 2005-11-25 2015-04-07 Zoetis Belgium S A Immunostimulatory oligoribonucleotides
US20080026986A1 (en) * 2006-06-05 2008-01-31 Rong-Fu Wang Reversal of the suppressive function of specific t cells via toll-like receptor 8 signaling
MX2009003398A (en) 2006-09-27 2009-08-12 Coley Pharm Gmbh Cpg oligonucleotide analogs containing hydrophobic t analogs with enhanced immunostimulatory activity.
US8628927B2 (en) 2008-11-07 2014-01-14 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
WO2010053587A2 (en) 2008-11-07 2010-05-14 Mlc Dx Incorporated Methods of monitoring conditions by sequence analysis
US9506119B2 (en) 2008-11-07 2016-11-29 Adaptive Biotechnologies Corp. Method of sequence determination using sequence tags
US9365901B2 (en) 2008-11-07 2016-06-14 Adaptive Biotechnologies Corp. Monitoring immunoglobulin heavy chain evolution in B-cell acute lymphoblastic leukemia
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
PT2376107E (en) 2008-12-09 2014-07-25 Coley Pharm Group Inc Immunostimulatory oligonucleotides
US8552165B2 (en) * 2008-12-09 2013-10-08 Heather Davis Immunostimulatory oligonucleotides
EP2387627B1 (en) 2009-01-15 2016-03-30 Adaptive Biotechnologies Corporation Adaptive immunity profiling and methods for generation of monoclonal antibodies
AU2010229835B2 (en) 2009-03-25 2015-01-15 The Board Of Regents Of The University Of Texas System Compositions for stimulation of mammalian innate immune resistance to pathogens
RU2539032C2 (en) 2009-06-25 2015-01-10 Фред Хатчинсон Кансэр Рисёч Сентер Method for measuring artificial immunity
US10385475B2 (en) 2011-09-12 2019-08-20 Adaptive Biotechnologies Corp. Random array sequencing of low-complexity libraries
EP2768982A4 (en) 2011-10-21 2015-06-03 Adaptive Biotechnologies Corp Quantification of adaptive immune cell genomes in a complex mixture of cells
AU2012347460B2 (en) 2011-12-09 2017-05-25 Adaptive Biotechnologies Corporation Diagnosis of lymphoid malignancies and minimal residual disease detection
US9499865B2 (en) 2011-12-13 2016-11-22 Adaptive Biotechnologies Corp. Detection and measurement of tissue-infiltrating lymphocytes
WO2013134162A2 (en) 2012-03-05 2013-09-12 Sequenta, Inc. Determining paired immune receptor chains from frequency matched subunits
SG10201507700VA (en) 2012-05-08 2015-10-29 Adaptive Biotechnologies Corp Compositions and method for measuring and calibrating amplification bias in multiplexed pcr reactions
CN105189779B (en) 2012-10-01 2018-05-11 适应生物技术公司 The immunocompetence carried out by adaptive immunity receptor diversity and Clonal characterization is assessed
US10150996B2 (en) 2012-10-19 2018-12-11 Adaptive Biotechnologies Corp. Quantification of adaptive immune cell genomes in a complex mixture of cells
US9708657B2 (en) 2013-07-01 2017-07-18 Adaptive Biotechnologies Corp. Method for generating clonotype profiles using sequence tags
AU2015227054A1 (en) 2014-03-05 2016-09-22 Adaptive Biotechnologies Corporation Methods using randomer-containing synthetic molecules
US10066265B2 (en) 2014-04-01 2018-09-04 Adaptive Biotechnologies Corp. Determining antigen-specific t-cells
US10286065B2 (en) 2014-09-19 2019-05-14 Board Of Regents, The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds
AU2015339191A1 (en) 2014-10-29 2017-05-18 Adaptive Biotechnologies Corp. Highly-multiplexed simultaneous detection of nucleic acids encoding paired adaptive immune receptor heterodimers from many samples
US10246701B2 (en) 2014-11-14 2019-04-02 Adaptive Biotechnologies Corp. Multiplexed digital quantitation of rearranged lymphoid receptors in a complex mixture
EP3498866A1 (en) 2014-11-25 2019-06-19 Adaptive Biotechnologies Corp. Characterization of adaptive immune response to vaccination or infection using immune repertoire sequencing
US11047008B2 (en) 2015-02-24 2021-06-29 Adaptive Biotechnologies Corporation Methods for diagnosing infectious disease and determining HLA status using immune repertoire sequencing
EP3277294A4 (en) 2015-04-01 2018-11-14 Adaptive Biotechnologies Corp. Method of identifying human compatible t cell receptors specific for an antigenic target
JP2019513692A (en) * 2016-02-19 2019-05-30 ジェニスフィア・エルエルシー Nucleic acid carriers and therapeutic uses
US10428325B1 (en) 2016-09-21 2019-10-01 Adaptive Biotechnologies Corporation Identification of antigen-specific B cell receptors
US11254980B1 (en) 2017-11-29 2022-02-22 Adaptive Biotechnologies Corporation Methods of profiling targeted polynucleotides while mitigating sequencing depth requirements

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002555A1 (en) * 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6426336B1 (en) * 1996-10-04 2002-07-30 The Regents Of The University Of California Method for treating allergic lung disease
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6476000B1 (en) * 1999-08-13 2002-11-05 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US6498148B1 (en) * 1997-09-05 2002-12-24 The Regents Of The University Of California Immunization-free methods for treating antigen-stimulated inflammation in a mammalian host and shifting the host's antigen immune responsiveness to a Th1 phenotype
US6562798B1 (en) * 1998-06-05 2003-05-13 Dynavax Technologies Corp. Immunostimulatory oligonucleotides with modified bases and methods of use thereof

Family Cites Families (289)

* 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
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US5766920A (en) 1982-08-11 1998-06-16 Cellcor, Inc. Ex vivo activation of immune cells
US5308626A (en) 1985-06-28 1994-05-03 Toni N. Mariani Lymphokine activated effector cells for antibody-dependent cellular cytotoxicity (ADCC) treatment of cancer and other diseases
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US5276019A (en) * 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
CA1339596C (en) 1987-08-07 1997-12-23 New England Medical Center Hospitals, Inc. Viral expression inhibitors
US5268365A (en) 1988-03-11 1993-12-07 Rudolph Frederick B Nucleotides, nucleosides, and nucleobases in immune function restoration enhancement or maintenance
US5004810A (en) * 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
US5087617A (en) * 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5786189A (en) 1989-11-29 1998-07-28 Smithkline Beecham Biologicals (S.A.) Vaccine
US5457189A (en) 1989-12-04 1995-10-10 Isis Pharmaceuticals Antisense oligonucleotide inhibition of papillomavirus
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5514788A (en) * 1993-05-17 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5514577A (en) * 1990-02-26 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide therapies for modulating the effects of herpes viruses
US5248670A (en) 1990-02-26 1993-09-28 Isis Pharmaceuticals, Inc. Antisense oligonucleotides for inhibiting herpesviruses
US5166195A (en) 1990-05-11 1992-11-24 Isis Pharmaceuticals, Inc. Antisense inhibitors of the human immunodeficiency virus phosphorothioate oligonucleotides
WO1991019813A1 (en) 1990-06-11 1991-12-26 The University Of Colorado Foundation, Inc. Nucleic acid ligands
EP0468520A3 (en) 1990-07-27 1992-07-01 Mitsui Toatsu Chemicals, Inc. Immunostimulatory remedies containing palindromic dna sequences
WO1994008003A1 (en) 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
US5582986A (en) 1991-06-14 1996-12-10 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of the ras gene
US6030954A (en) * 1991-09-05 2000-02-29 University Of Connecticut Targeted delivery of poly- or oligonucleotides to cells
US5576302A (en) 1991-10-15 1996-11-19 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating hepatitis C virus having phosphorothioate linkages of high chiral purity
US6498147B2 (en) 1992-05-22 2002-12-24 The Scripps Research Institute Suppression of nuclear factor-κb dependent processes using oligonucleotides
AU678769B2 (en) 1992-07-27 1997-06-12 Hybridon, Inc. Oligonucleotide alkylphosphonothioates
US5523389A (en) 1992-09-29 1996-06-04 Isis Pharmaceuticals, Inc. Inhibitors of human immunodeficiency virus
AU678415B2 (en) 1992-10-05 1997-05-29 Hybridon, Inc. Therapeutic anti-HIV oligonucleotide and pharmaceutical
US5567604A (en) * 1993-04-23 1996-10-22 Aronex Pharmaceuticals, Inc. Anti-viral guanosine-rich oligonucleotides
KR960703170A (en) * 1993-06-23 1996-06-19 알버트 디. 프리센. ANTISENSE OLIGONUCLEOTIDES AND THERAPEUTIC USE THEREOF IN HUMAN IMMUNODEFICIENCY VIRUS INFECTION
US6605708B1 (en) 1993-07-28 2003-08-12 Hybridon, Inc. Building blocks with carbamate internucleoside linkages and oligonucleotides derived therefrom
US5804566A (en) 1993-08-26 1998-09-08 The Regents Of The University Of California Methods and devices for immunizing a host through administration of naked polynucleotides with encode allergenic peptides
US5679647A (en) 1993-08-26 1997-10-21 The Regents Of The University Of California Methods and devices for immunizing a host against tumor-associated antigens through administration of naked polynucleotides which encode tumor-associated antigenic peptides
DE4338704A1 (en) * 1993-11-12 1995-05-18 Hoechst Ag Stabilized oligonucleotides and their use
US5595756A (en) 1993-12-22 1997-01-21 Inex Pharmaceuticals Corporation Liposomal compositions for enhanced retention of bioactive agents
DE69432315T2 (en) 1993-12-23 2004-02-12 Biognostik Gesellschaft für Biomolekulare Diagnostik mbH ANTISENSE NUCLEIC ACIDS FOR PREVENTING AND TREATING COMPLAINTS IN WHICH THE EXPRESSION OF C-ERBB-2 PLAYS A ROLE
US5646126A (en) 1994-02-28 1997-07-08 Epoch Pharmaceuticals Sterol modified oligonucleotide duplexes having anticancer activity
US5596091A (en) * 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US6727230B1 (en) 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US5696248A (en) 1994-06-15 1997-12-09 Hoechst Aktiengesellschaft 3'-modified oligonucleotide derivatives
US5741516A (en) 1994-06-20 1998-04-21 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5543152A (en) 1994-06-20 1996-08-06 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US7935675B1 (en) * 1994-07-15 2011-05-03 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US20030050263A1 (en) * 1994-07-15 2003-03-13 The University Of Iowa Research Foundation Methods and products for treating HIV infection
JPH10502820A (en) 1994-07-18 1998-03-17 ユニバーシティ・オブ・ノース・カロライナ・アット・チャペル・ヒル Oligonucleoside compounds and methods for inhibiting tumor growth, invasion and metastasis
US5646262A (en) * 1994-07-28 1997-07-08 Georgetown University Antisense oligonucleotides against hepatitis B viral replication
AU3559695A (en) 1994-09-30 1996-04-26 Inex Pharmaceuticals Corp. Glycosylated protein-liposome conjugates and methods for their preparation
US5785992A (en) 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5753613A (en) 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
DE19502912A1 (en) 1995-01-31 1996-08-01 Hoechst Ag G-Cap Stabilized Oligonucleotides
US5932556A (en) 1995-09-17 1999-08-03 Tam; Robert C Methods and compositions for regulation of CD28 expression
GB9505438D0 (en) 1995-03-17 1995-05-03 Sod Conseils Rech Applic Antisense oligonucleotides
AU716486B2 (en) * 1995-04-13 2000-02-24 Milkhaus Laboratory, Inc. Methods for treating respiratory disease
US5955059A (en) 1995-06-06 1999-09-21 Trustees Of Boston University Use of locally applied DNA fragments
US6025339A (en) * 1995-06-07 2000-02-15 East Carolina University Composition, kit and method for treatment of disorders associated with bronchoconstriction and lung inflammation
AU723163B2 (en) 1995-06-07 2000-08-17 Tekmira Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US6410690B1 (en) 1995-06-07 2002-06-25 Medarex, Inc. Therapeutic compounds comprised of anti-Fc receptor antibodies
US5994315A (en) 1995-06-07 1999-11-30 East Carolina University Low adenosine agent, composition, kit and method for treatment of airway disease
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US6040296A (en) * 1995-06-07 2000-03-21 East Carolina University Specific antisense oligonucleotide composition & method for treatment of disorders associated with bronchoconstriction and lung inflammation
US5705385A (en) 1995-06-07 1998-01-06 Inex Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5968909A (en) 1995-08-04 1999-10-19 Hybridon, Inc. Method of modulating gene expression with reduced immunostimulatory response
US5780448A (en) 1995-11-07 1998-07-14 Ottawa Civic Hospital Loeb Research DNA-based vaccination of fish
JP4359654B2 (en) 1996-01-30 2009-11-04 ザ リージェンツ オブ ザ ユニバーシティー オブ カリフォルニア Gene expression vector for generating antigen-specific immune response and method of use thereof
US20030078223A1 (en) * 1996-01-30 2003-04-24 Eyal Raz Compositions and methods for modulating an immune response
US6620805B1 (en) 1996-03-14 2003-09-16 Yale University Delivery of nucleic acids by porphyrins
US6610661B1 (en) 1996-10-11 2003-08-26 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates
EP0855184A1 (en) * 1997-01-23 1998-07-29 Grayson B. Dr. Lipford Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
US20030064945A1 (en) * 1997-01-31 2003-04-03 Saghir Akhtar Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
AU738513B2 (en) 1997-02-28 2001-09-20 University Of Iowa Research Foundation, The Use of nucleic acids containing unmethylated CpG dinucleotide in the treatment of LPS-associated disorders
EP1005368B1 (en) 1997-03-10 2009-09-02 Ottawa Hospital Research Institute Use of nucleic acids containing unmethylated CpG dinucleotide in combination with alum as adjuvants
US5965542A (en) 1997-03-18 1999-10-12 Inex Pharmaceuticals Corp. Use of temperature to control the size of cationic liposome/plasmid DNA complexes
US6426334B1 (en) 1997-04-30 2002-07-30 Hybridon, Inc. Oligonucleotide mediated specific cytokine induction and reduction of tumor growth in a mammal
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
US20030104044A1 (en) 1997-05-14 2003-06-05 Semple Sean C. Compositions for stimulating cytokine secretion and inducing an immune response
US6835395B1 (en) 1997-05-14 2004-12-28 The University Of British Columbia Composition containing small multilamellar oligodeoxynucleotide-containing lipid vesicles
EP0983289A4 (en) 1997-05-19 2001-04-25 Merck & Co Inc Oligonucleotide adjuvant
EP1003531B1 (en) * 1997-05-20 2007-08-22 Ottawa Health Research Institute Processes for preparing nucleic acid constructs
US6589940B1 (en) 1997-06-06 2003-07-08 Dynavax Technologies Corporation Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
US20040006034A1 (en) * 1998-06-05 2004-01-08 Eyal Raz Immunostimulatory oligonucleotides, compositions thereof and methods of use thereof
ATE432348T1 (en) * 1997-06-06 2009-06-15 Univ California INHIBITORS OF IMMUNO-STIMULATIVE DNA SEQUENCE ACTIVITY
US6221882B1 (en) 1997-07-03 2001-04-24 University Of Iowa Research Foundation Methods for inhibiting immunostimulatory DNA associated responses
AU8428998A (en) 1997-07-24 1999-02-16 Inex Pharmaceuticals Corporation Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
US5877309A (en) * 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
EP1007657B1 (en) 1997-08-19 2006-02-08 Idera Pharmaceuticals, Inc. Hiv-specific oligonucleotides and methods of their use
US6749856B1 (en) 1997-09-11 2004-06-15 The United States Of America, As Represented By The Department Of Health And Human Services Mucosal cytotoxic T lymphocyte responses
WO1999033493A1 (en) 1997-12-23 1999-07-08 Inex Pharmaceuticals Corporation Polyamide oligomers
GB9727262D0 (en) 1997-12-24 1998-02-25 Smithkline Beecham Biolog Vaccine
US20050031638A1 (en) * 1997-12-24 2005-02-10 Smithkline Beecham Biologicals S.A. Vaccine
JPH11209289A (en) 1998-01-22 1999-08-03 Taisho Pharmaceut Co Ltd Mucosal immunity inducer
WO1999051259A2 (en) 1998-04-03 1999-10-14 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
ATE331739T1 (en) 1998-04-28 2006-07-15 Inex Pharmaceuticals Corp POLYANIONIC POLYMERS WITH IMPROVED FUSOGEN CAPACITY
AU3884199A (en) 1998-05-06 1999-11-23 Ottawa Health Research Institute Methods for the prevention and treatment of parasitic infections and related diseases using cpg oligonucleotides
NZ508650A (en) * 1998-05-14 2003-05-30 Coley Pharm Gmbh Regulating hematopoiesis using unmethylated C and G CpG-oligonucleotides with a phosphorothioate modification
DE69932717T2 (en) 1998-05-22 2007-08-09 Ottawa Health Research Institute, Ottawa METHODS AND PRODUCTS FOR INDUCING MUCOSAL IMMUNITY
US6881561B1 (en) 1998-05-27 2005-04-19 Cheil Jedang Corporation Endonuclease of immune cell, process for producing the same and immune adjuvant using the same
EP1089764B1 (en) 1998-06-10 2004-09-01 Biognostik Gesellschaft für biomolekulare Diagnostik mbH Stimulating the immune system
US20040247662A1 (en) 1998-06-25 2004-12-09 Dow Steven W. Systemic immune activation method using nucleic acid-lipid complexes
US6693086B1 (en) * 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
CA2335393C (en) 1998-07-20 2008-09-23 Inex Pharmaceuticals Corporation Liposomal encapsulated nucleic acid-complexes
EP1100807A1 (en) 1998-07-27 2001-05-23 University Of Iowa Research Foundation STEREOISOMERS OF CpG OLIGONUCLEOTIDES AND RELATED METHODS
GB9817052D0 (en) * 1998-08-05 1998-09-30 Smithkline Beecham Biolog Vaccine
EP0979869A1 (en) 1998-08-07 2000-02-16 Hoechst Marion Roussel Deutschland GmbH Short oligonucleotides for the inhibition of VEGF expression
US20010034330A1 (en) 1998-08-10 2001-10-25 Charlotte Kensil Innate immunity-stimulating compositions of CpG and saponin and methods thereof
US7049302B1 (en) 1998-08-10 2006-05-23 Antigenics Inc. Compositions of CPG and saponin adjuvants and uses thereof
EP1108017A2 (en) 1998-09-03 2001-06-20 Coley Pharmaceutical GmbH G-motif oligonucleotides and uses thereof
FR2783170B1 (en) 1998-09-11 2004-07-16 Pasteur Merieux Serums Vacc IMMUNOSTIMULATING EMULSION
CA2343052A1 (en) 1998-09-18 2000-03-30 Dynavax Technologies Corporation Methods of treating ige-associated disorders and compositions for use therein
WO2000020039A1 (en) 1998-10-05 2000-04-13 The Regents Of The University Of California Methods and adjuvants for stimulating mucosal immunity
AU6425999A (en) 1998-10-09 2000-05-01 Dynavax Technologies Corporation Anti hiv compositions comprising immunostimulatory polynucleotides and hiv antigens
PL201482B1 (en) 1998-10-16 2009-04-30 Smithkline Beecham Biolog Adjuvant systems and vaccines
AUPP807399A0 (en) 1999-01-08 1999-02-04 Csl Limited Improved immunogenic lhrh composition and methods relating thereto
JP2002534438A (en) 1999-01-12 2002-10-15 スミスクライン ビーチャム バイオロジカルズ ソシエテ アノニム New treatment
US6887464B1 (en) 1999-02-02 2005-05-03 Biocache Pharmaceuticals, Inc. Advanced antigen presentation platform
EP1165773A4 (en) 1999-02-02 2005-01-05 Biocache Pharmaceuticals Inc Advanced antigen presentation platform
JP2002536344A (en) 1999-02-05 2002-10-29 ジエンザイム コーポレイション Use of cationic lipids to generate anti-tumor immunity
WO2000054803A2 (en) 1999-03-16 2000-09-21 Panacea Pharmaceuticals, Llc Immunostimulatory nucleic acids and antigens
FR2790955B1 (en) 1999-03-19 2003-01-17 Assist Publ Hopitaux De Paris USE OF STABILIZED OLIGONUCLEOTIDES AS ANTI-TUMOR ACTIVE INGREDIENT
SI1163000T1 (en) 1999-03-19 2008-06-30 Glaxosmithkline Biolog Sa Vaccine against antigens from bacteriae
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
EP1176966B1 (en) 1999-04-12 2013-04-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Oligodeoxynucleotide and its use to induce an immune response
AU4642600A (en) 1999-04-15 2000-11-02 Regents Of The University Of California, The Methods and compositions for use in potentiating antigen presentation by antigenpresenting cells
PL203951B1 (en) 1999-04-19 2009-11-30 Smithkline Beecham Biolog Vaccines
US6558670B1 (en) 1999-04-19 2003-05-06 Smithkline Beechman Biologicals S.A. Vaccine adjuvants
AU4978100A (en) 1999-04-29 2000-11-17 Coley Pharmaceutical Gmbh Screening for immunostimulatory dna functional modifyers
US6737066B1 (en) 1999-05-06 2004-05-18 The Immune Response Corporation HIV immunogenic compositions and methods
AP1891A (en) 1999-05-06 2008-09-23 The Immune Response Corp HIV immunogenic compositions and methods.
AU776268B2 (en) 1999-06-08 2004-09-02 Aventis Pasteur Immunostimulant oligonucleotide
US6479504B1 (en) 1999-06-16 2002-11-12 The University Of Iowa Research Foundation Antagonism of immunostimulatory CpG-oligonucleotides by 4-aminoquinolines and other weak bases
US6514948B1 (en) 1999-07-02 2003-02-04 The Regents Of The University Of California Method for enhancing an immune response
DE19935756A1 (en) 1999-07-27 2001-02-08 Mologen Forschungs Entwicklung Covalently closed nucleic acid molecule for immune stimulation
EP1204425B1 (en) 1999-08-19 2009-01-07 Dynavax Technologies Corporation Methods of modulating an immune response using immunostimulatory sequences and compositions for use therein
GB9921147D0 (en) 1999-09-07 1999-11-10 Smithkline Beecham Biolog Novel composition
GB9921146D0 (en) 1999-09-07 1999-11-10 Smithkline Beecham Biolog Novel composition
AP2006003503A0 (en) * 1999-09-25 2006-02-28 Univ Iowa Res Found Immunostimulatory nucleic acids.
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US20070031446A1 (en) 2003-07-11 2007-02-08 Intercell Ag Hcv vaccines
US7223398B1 (en) 1999-11-15 2007-05-29 Dynavax Technologies Corporation Immunomodulatory compositions containing an immunostimulatory sequence linked to antigen and methods of use thereof
AU781812B2 (en) 2000-01-13 2005-06-16 Antigenics, Inc. Innate immunity-stimulating compositions of CPG and saponin and methods thereof
EP1322655B1 (en) * 2000-01-14 2007-11-14 The Government of the United States of America, as represented by the Secretary of the Department of Health and Human Services Oligodeoxynucleotide and its use to induce an immune response
AU3108001A (en) 2000-01-20 2001-12-24 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for inducing a th2 immune response
US6852705B2 (en) * 2000-01-21 2005-02-08 Merial DNA vaccines for farm animals, in particular bovines and porcines
US6815429B2 (en) 2000-01-26 2004-11-09 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
AT409085B (en) 2000-01-28 2002-05-27 Cistem Biotechnologies Gmbh PHARMACEUTICAL COMPOSITION FOR IMMUNULATING AND PRODUCING VACCINES
MXPA02007413A (en) 2000-01-31 2004-07-30 Smithkline Beecham Biolog Novel use.
US6552006B2 (en) 2000-01-31 2003-04-22 The Regents Of The University Of California Immunomodulatory polynucleotides in treatment of an infection by an intracellular pathogen
US7585847B2 (en) * 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
FR2805265B1 (en) 2000-02-18 2002-04-12 Aventis Pasteur IMMUNOSTIMULATING OLIGONUCLEOTIDES
US20030130217A1 (en) 2000-02-23 2003-07-10 Eyal Raz Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
US6613751B2 (en) * 2000-02-23 2003-09-02 The Regents Of The University Of California Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
US6423539B2 (en) 2000-02-24 2002-07-23 The Board Of Trustees Of The Leland Stanford Junior University Adjuvant treatment by in vivo activation of dendritic cells
US20020156033A1 (en) 2000-03-03 2002-10-24 Bratzler Robert L. Immunostimulatory nucleic acids and cancer medicament combination therapy for the treatment of cancer
US20040131628A1 (en) 2000-03-08 2004-07-08 Bratzler Robert L. Nucleic acids for the treatment of disorders associated with microorganisms
US20020098199A1 (en) 2000-03-10 2002-07-25 Gary Van Nest Methods of suppressing hepatitis virus infection using immunomodulatory polynucleotide sequences
US20020028784A1 (en) * 2000-03-10 2002-03-07 Nest Gary Van Methods of preventing and treating viral infections using immunomodulatory polynucleotide sequences
US20020107212A1 (en) 2000-03-10 2002-08-08 Nest Gary Van Methods of reducing papillomavirus infection using immunomodulatory polynucleotide sequences
US7129222B2 (en) 2000-03-10 2006-10-31 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US20030129251A1 (en) 2000-03-10 2003-07-10 Gary Van Nest Biodegradable immunomodulatory formulations and methods for use thereof
US20010046967A1 (en) 2000-03-10 2001-11-29 Gary Van Nest Methods of preventing and treating respiratory viral infection using immunomodulatory polynucleotide
US7157437B2 (en) 2000-03-10 2007-01-02 Dynavax Technologies Corporation Methods of ameliorating symptoms of herpes infection using immunomodulatory polynucleotide sequences
AU2001249609A1 (en) 2000-03-28 2001-10-08 Department Of Veterans Affairs Methods for increasing a cytotoxic T lymphocyte response in vivo
AU2001251407A1 (en) 2000-04-07 2001-10-23 The Regents Of The University Of California Synergistic improvements to polynucleotide vaccines
WO2001085910A2 (en) 2000-05-05 2001-11-15 The Regents Of The University Of California Agents that modulate dna-pk activity and methods of use thereof
US6339630B1 (en) * 2000-05-18 2002-01-15 The United States Of America As Represented By The United States Department Of Energy Sealed drive screw operator
AU2001275294A1 (en) 2000-06-07 2001-12-17 Biosynexus Incorporated. Immunostimulatory RNA/DNA hybrid molecules
AU7013401A (en) 2000-06-22 2002-01-02 Univ Iowa Res Found Methods for enhancing antibody-induced cell lysis and treating cancer
US20020165178A1 (en) 2000-06-28 2002-11-07 Christian Schetter Immunostimulatory nucleic acids for the treatment of anemia, thrombocytopenia, and neutropenia
WO2002009748A1 (en) 2000-07-31 2002-02-07 Yale University Innate immune system-directed vaccines
US20020198165A1 (en) 2000-08-01 2002-12-26 Bratzler Robert L. Nucleic acids for the prevention and treatment of gastric ulcers
WO2002018631A2 (en) * 2000-09-01 2002-03-07 Epigenomics Ag Diagnosis of illnesses or predisposition to certain illnesses
US20020091097A1 (en) 2000-09-07 2002-07-11 Bratzler Robert L. Nucleic acids for the prevention and treatment of sexually transmitted diseases
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
US6787524B2 (en) 2000-09-22 2004-09-07 Tanox, Inc. CpG oligonucleotides and related compounds for enhancing ADCC induced by anti-IgE antibodies
CA2423487C (en) 2000-09-26 2015-12-15 Hybridon, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
FR2814958B1 (en) 2000-10-06 2003-03-07 Aventis Pasteur VACCINE COMPOSITION
KR100831139B1 (en) * 2000-10-18 2008-05-20 글락소스미스클라인 바이오로지칼즈 에스.에이. Vaccines
EP1330520A2 (en) 2000-11-02 2003-07-30 Inex Pharmaceuticals Corp. Therapeutic oligonucleotides of reduced toxicity
ES2307568T3 (en) 2000-12-08 2008-12-01 Coley Pharmaceutical Gmbh CPG TYPE NUCLEIC ACIDS AND SAME USE METHODS.
US20030055014A1 (en) 2000-12-14 2003-03-20 Bratzler Robert L. Inhibition of angiogenesis by nucleic acids
DE60142410D1 (en) * 2000-12-27 2010-07-29 Dynavax Tech Corp IMMUNOMODULATORY POLYNUCLEOTIDES AND METHOD FOR THE USE THEREOF
US20030050268A1 (en) 2001-03-29 2003-03-13 Krieg Arthur M. Immunostimulatory nucleic acid for treatment of non-allergic inflammatory diseases
US7105495B2 (en) 2001-04-30 2006-09-12 Idera Pharmaceuticals, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US7176296B2 (en) 2001-04-30 2007-02-13 Idera Pharmaceuticals, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides
US20030129605A1 (en) 2001-05-04 2003-07-10 Dong Yu Immunostimulatory activity of CpG oligonucleotides containing non-ionic methylphosophonate linkages
EP1395262A4 (en) 2001-06-15 2006-04-12 Ribapharm Corp Nucleoside vaccine adjuvants
CN100334228C (en) 2001-06-21 2007-08-29 戴纳瓦克斯技术公司 Cimeric immunomodulatory compounds and methods of using the same
US20040132677A1 (en) 2001-06-21 2004-07-08 Fearon Karen L. Chimeric immunomodulatory compounds and methods of using the same-IV
US7785610B2 (en) 2001-06-21 2010-08-31 Dynavax Technologies Corporation Chimeric immunomodulatory compounds and methods of using the same—III
WO2003000232A2 (en) 2001-06-25 2003-01-03 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method for preparation of vesicles loaded with immunostimulator y oligodeoxynucleotides
CN1931365A (en) 2001-06-29 2007-03-21 希龙公司 Hcv e1e2 vaccine compositions
WO2003040308A2 (en) 2001-07-27 2003-05-15 The Government Of 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
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
WO2003012061A2 (en) 2001-08-01 2003-02-13 Coley Pharmaceutical Gmbh Methods and compositions relating to plasmacytoid dendritic cells
JP2005518343A (en) 2001-08-03 2005-06-23 メダレックス, インク. Novel PGC-1 isoforms and their use to improve use-mediated immunotherapy
WO2003014316A2 (en) 2001-08-07 2003-02-20 Dynavax Technologies Corporation Immunomodulatory compositions, formulations, and methods for use thereof
US7354909B2 (en) * 2001-08-14 2008-04-08 The United States Of America As Represented By Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
SI1446162T1 (en) 2001-08-17 2009-04-30 Coley Pharm Gmbh Combination motif immune stimulatory oligonucleotides with improved activity
US20030133913A1 (en) 2001-08-30 2003-07-17 3M Innovative Properties Company Methods of maturing plasmacytoid dendritic cells using immune response modifier molecules
EP2196217A1 (en) 2001-09-14 2010-06-16 Cytos Biotechnology AG Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
US7514415B2 (en) 2002-08-01 2009-04-07 The United States Of America As Represented By The Department Of Health And Human Services Method of treating inflammatory arthropathies with suppressors of CpG oligonucleotides
US20030119774A1 (en) 2001-09-25 2003-06-26 Marianna Foldvari Compositions and methods for stimulating an immune response
CA2461315A1 (en) 2001-10-05 2003-04-17 Coley Pharmaceutical Gmbh Toll-like receptor 3 signaling agonists and antagonists
CN100438908C (en) 2001-10-06 2008-12-03 梅瑞尔有限公司 CpG formulations and related methods
AU2002360278A1 (en) 2001-10-12 2003-11-11 Coley Pharmaceutical Gmbh Methods and products for enhancing immune responses using imidazoquinoline compounds
US7276489B2 (en) 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
WO2003035836A2 (en) 2001-10-24 2003-05-01 Hybridon Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5' ends
US20030125292A1 (en) 2001-11-07 2003-07-03 Sean Semple Mucoscal vaccine and methods for using the same
TW200303759A (en) 2001-11-27 2003-09-16 Schering Corp Methods for treating cancer
AU2002356763A1 (en) 2001-11-30 2003-06-10 Medigene Aktiengesellschaft Use of a technically modified cell as a vaccine for treating tumoral disease
CA2474709A1 (en) 2002-02-04 2003-08-14 Biomira, Inc. Immunostimulatory, covalently lipidated oligonucleotides
US8088388B2 (en) 2002-02-14 2012-01-03 United Biomedical, Inc. Stabilized synthetic immunogen delivery system
US20030232443A1 (en) 2002-06-18 2003-12-18 Isis Pharmaceuticals Inc. Antisense modulation of centromere protein B expression
US8153141B2 (en) 2002-04-04 2012-04-10 Coley Pharmaceutical Gmbh Immunostimulatory G, U-containing oligoribonucleotides
US7737264B2 (en) 2002-04-05 2010-06-15 Enzon Pharmaceuticals, Inc. Oligomeric compounds for the modulation HIF-1α expression
CA2483012C (en) * 2002-04-22 2011-05-24 Bioniche Life Sciences Inc. Oligonucleotide compositions and their use for the modulation of immune responses
CA2487452A1 (en) 2002-05-28 2003-12-04 Robinson Ramirez-Pineda A method for generating antigen-presenting cells
US20040009949A1 (en) 2002-06-05 2004-01-15 Coley Pharmaceutical Group, Inc. Method for treating autoimmune or inflammatory diseases with combinations of inhibitory oligonucleotides and small molecule antagonists of immunostimulatory CpG nucleic acids
US7605138B2 (en) 2002-07-03 2009-10-20 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US20040013688A1 (en) * 2002-07-03 2004-01-22 Cambridge Scientific, Inc. Vaccines to induce mucosal immunity
DE10229872A1 (en) 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
US7807803B2 (en) 2002-07-03 2010-10-05 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US20040053880A1 (en) * 2002-07-03 2004-03-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7569553B2 (en) * 2002-07-03 2009-08-04 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
US7576066B2 (en) 2002-07-03 2009-08-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
AU2003255969A1 (en) 2002-07-17 2004-02-02 Coley Pharmaceutical Gmbh Use of cpg nucleic acids in prion-disease
US20050209183A1 (en) 2002-07-25 2005-09-22 Phenion Gmbh & Co. Kg Cosmetic or pharmaceutical preparations comprising nucleic acids based on non-methylated CPG motifs
AR040996A1 (en) 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
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
AU2003278845A1 (en) 2002-09-19 2004-04-08 Coley Pharmaceutical Gmbh Toll-like receptor 9 (tlr9) from various mammalian species
MXPA05004588A (en) 2002-10-29 2005-12-14 Coley Pharmaceutical Group Ltd Use of cpg oligonucleotides in the treatment of hepatitis c virus infection.
WO2004041183A2 (en) 2002-11-01 2004-05-21 The Regents Of The University Of California Methods of treating pulmonary fibrotic disorders
US8039443B2 (en) 2002-11-21 2011-10-18 Archemix Corporation Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
AU2003300919A1 (en) 2002-12-11 2004-06-30 Coley Pharmaceutical Gmbh 5' cpg nucleic acids and methods of use
KR100525321B1 (en) 2002-12-13 2005-11-02 안웅식 Pharmaceutical composition for prophylaxis and treatment of papillomavirus-derived diseases comprising papillomavirus antigen protein and CpG-oligodeoxynucleotide
JP2006516099A (en) 2002-12-23 2006-06-22 ダイナバックス テクノロジーズ コーポレイション Branched immunomodulatory compounds and methods of using the compounds
CN100546998C (en) * 2002-12-23 2009-10-07 戴纳伐克斯技术股份有限公司 Immunostimulatory sequence oligonucleotides and using method
US7851453B2 (en) * 2003-01-16 2010-12-14 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by utilizing modified immunostimulatory dinucleotides
US20040235770A1 (en) 2003-04-02 2004-11-25 Coley Pharmaceutical Group, Ltd. Immunostimulatory nucleic acid oil-in-water formulations and related methods of use
US20050004144A1 (en) * 2003-04-14 2005-01-06 Regents Of The University Of California Combined use of IMPDH inhibitors with toll-like receptor agonists
EP1631687A2 (en) 2003-04-22 2006-03-08 Coley Pharmaceutical GmbH Methods and products for identification and assessment of tlr ligands
EP2371834B1 (en) 2003-06-11 2016-02-17 Idera Pharmaceuticals, Inc. Stabilized immunomodulatory oligonucleotides
JP2007524615A (en) 2003-06-20 2007-08-30 コーリー ファーマシューティカル ゲーエムベーハー Low molecular weight Toll-like receptor (TLR) antagonist
KR20060031607A (en) 2003-07-10 2006-04-12 사이토스 바이오테크놀로지 아게 Packaged virus-like particles
US20050013812A1 (en) * 2003-07-14 2005-01-20 Dow Steven W. Vaccines using pattern recognition receptor-ligand:lipid complexes
AU2004259204B2 (en) 2003-07-15 2010-08-19 Idera Pharmaceuticals, Inc. Synergistic stimulation of the immune system using immunostimulatory oligonucleotides and/or immunomer compounds in conjunction with cytokines and/or chemotherapeutic agents or radiation therapy
CA2544240A1 (en) * 2003-07-22 2005-02-17 Cytos Biotechnology Ag Cpg-packaged liposomes
WO2005021726A2 (en) 2003-08-28 2005-03-10 The Immune Response Corporation Immunogenic hiv compositions and related methods
EP1671643A1 (en) 2003-09-08 2006-06-21 Intellectual Property Consulting Inc. Medicinal composition for treating chronic hepatitis c
CA2536139A1 (en) 2003-09-25 2005-04-07 Coley Pharmaceutical Group, Inc. Nucleic acid-lipophilic conjugates
WO2005034979A2 (en) 2003-10-11 2005-04-21 Inex Pharmaceuticals Corporation Methods and compositions for enhancing innate immunity and antibody dependent cellular cytotoxicity
US20050215501A1 (en) 2003-10-24 2005-09-29 Coley Pharmaceutical Group, Inc. Methods and products for enhancing epitope spreading
CA2540949A1 (en) * 2003-10-30 2005-05-12 Coley Pharmaceutical Gmbh C-class oligonucleotide analogs with enhanced immunostimulatory potency
US20050239733A1 (en) 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
US20050100983A1 (en) 2003-11-06 2005-05-12 Coley Pharmaceutical Gmbh Cell-free methods for identifying compounds that affect toll-like receptor 9 (TLR9) signaling
US7846436B2 (en) 2003-11-28 2010-12-07 Chemgenes Corporation Oligonucleotides and related compounds
WO2005060377A2 (en) 2003-12-08 2005-07-07 Hybridon, Inc. Modulation of immunostimulatory properties by small oligonucleotide-based compounds
US9090673B2 (en) 2003-12-12 2015-07-28 City Of Hope Synthetic conjugate of CpG DNA and T-help/CTL peptide
WO2005072097A2 (en) 2003-12-16 2005-08-11 University Of Massachusetts Toll-like receptor 9 modulators
EP1550458A1 (en) 2003-12-23 2005-07-06 Vectron Therapeutics AG Synergistic liposomal adjuvants
KR100558851B1 (en) 2004-01-08 2006-03-10 학교법인연세대학교 Modified CpG oligodeoxynucleotide with improved immunoregulatory function
WO2005072290A2 (en) 2004-01-23 2005-08-11 Joslin Diabetes Center Methods of treating, reducing, or preventing autoimmune conditions
WO2005079419A2 (en) 2004-02-17 2005-09-01 The Regents Of The University Of California Methods of treating immunopathological disorders
EP1720568A2 (en) 2004-02-19 2006-11-15 Coley Pharmaceutical Group, Inc. Immunostimulatory viral rna oligonucleotides
EP1730281A2 (en) * 2004-04-02 2006-12-13 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for inducing il-10 responses
AU2005326144A1 (en) 2004-06-08 2006-08-03 Coley Pharmaceutical Gmbh Abasic oligonucleotide as carrier platform for antigen and immunostimulatory agonist and antagonist
CA2574090A1 (en) * 2004-07-18 2006-12-21 Coley Pharmaceutical Group, Ltd. Methods and compositions for inducing innate immune responses
EP1781325A2 (en) 2004-07-18 2007-05-09 CSL Limited Immuno stimulating complex and oligonucleotide formulations for inducing enhanced interferon-gamma responses
MY159370A (en) 2004-10-20 2016-12-30 Coley Pharm Group Inc Semi-soft-class immunostimulatory oligonucleotides
US20060105979A1 (en) 2004-11-08 2006-05-18 Hybridon, Inc. Synergistic inhibition of VEGF and modulation of the immune response
CN101160401A (en) * 2005-02-24 2008-04-09 科勒制药集团公司 Immunostimulatory oligonucleotides
EP1874325A2 (en) 2005-04-08 2008-01-09 Coley Pharmaceutical Group, Inc. Methods for treating infectious disease exacerbated asthma
US20060241076A1 (en) 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
WO2007088423A2 (en) 2005-09-16 2007-08-09 Coley Pharmaceutical Gmbh Immunostimulatory single-stranded ribonucleic acid with phosphodiester backbone
EA013375B1 (en) 2005-09-16 2010-04-30 Коли Фармасьютикал Гмбх MODULATION OF IMMUNOSTIMULATORY PROPERTIES OF SHORT INTERFERING RIBONUCLEIC ACID (siRNA) BY NUCLEOTIDE MODIFICATION
BRPI0616770A2 (en) 2005-09-27 2011-06-28 Coley Pharm Gmbh modulation of tlr-mediated immune responses employing adapter oligonucleotides
DK1957647T3 (en) 2005-11-25 2015-04-07 Zoetis Belgium S A Immunostimulatory oligoribonucleotides
EP2405002B1 (en) * 2006-02-15 2014-09-24 AdiuTide Pharmaceuticals GmbH Compositions and methods for oligonucleotide formulations
DE102006007433A1 (en) 2006-02-17 2007-08-23 Curevac Gmbh Immunostimulant adjuvant useful in vaccines against cancer or infectious diseases comprises a lipid-modified nucleic acid
US8027888B2 (en) 2006-08-31 2011-09-27 Experian Interactive Innovation Center, Llc Online credit card prescreen systems and methods
WO2008033432A2 (en) 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immune modulation by chemically modified ribonucleosides and oligoribonucleotides
MX2009003398A (en) 2006-09-27 2009-08-12 Coley Pharm Gmbh Cpg oligonucleotide analogs containing hydrophobic t analogs with enhanced immunostimulatory activity.
MX2009003403A (en) 2006-09-27 2009-04-09 Coley Pharm Group Inc Compositions of tlr ligands and antivirals.
RU2009115687A (en) 2006-10-26 2010-11-10 Коли Фармасьютикал Гмбх (De) OLIGORIBONUCLEOTIDES AND THEIR APPLICATION

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002555A1 (en) * 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6194388B1 (en) * 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6426336B1 (en) * 1996-10-04 2002-07-30 The Regents Of The University Of California Method for treating allergic lung disease
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6498148B1 (en) * 1997-09-05 2002-12-24 The Regents Of The University Of California Immunization-free methods for treating antigen-stimulated inflammation in a mammalian host and shifting the host's antigen immune responsiveness to a Th1 phenotype
US6562798B1 (en) * 1998-06-05 2003-05-13 Dynavax Technologies Corp. Immunostimulatory oligonucleotides with modified bases and methods of use thereof
US6476000B1 (en) * 1999-08-13 2002-11-05 Hybridon, Inc. Modulation of oligonucleotide CpG-mediated immune stimulation by positional modification of nucleosides

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
AGRAWAL D.K.: 'Novel immunomodulatory oligonucleotides prevent development of allergic airway inflammation and airway hyperresponsiveness in asthma' INTERNATIONAL IMMUNOPHARMACOLOGY vol. 4, 2004, pages 127 - 138, XP002992302 *
BOCHNER B.S.: 'Advances in mechanisms of allergy' J. ALLERGY CLIN. IMMUNOL. vol. 113, 2004, pages 868 - 875, XP002992303 *
BROIDE D.: 'DNA-based immunization for asthma' INT. ARCH. ALLERGY IMMNOL. vol. 118, 1999, pages 453 - 456, XP002267485 *
JAIN V.V.: 'CpG DNA and immunotherapy of allergic airway diseases' CLIN. EXP. ALLERGY vol. 33, 2003, pages 1330 - 1335, XP002992306 *
JAIN V.V.: 'CpG DNA: immunomodulation and remodelling of the asthmatic airway' EXPERT OPIN. BIOL. THER. vol. 4, no. 9, 2004, pages 1533 - 1540, XP008054965 *
JAIN V.V.: 'CpG-oligodeoxynucleotides inhibit airway remodeling in a murine model of chronic asthma' J. ALLERGY CLIN. IMMUNOL. vol. 110, 2002, pages 867 - 872, XP002976934 *
KITAGAKI K.: 'Immunomodulary effects of CpG oligodeoxynucleotides on established Th2 response' CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY vol. 9, no. 6, November 2002, pages 1260 - 1269, XP002992305 *
KLINE J.N.: 'CpG oligodeoxynucleotides do not require Th1 cytokines to prevent eosinophilic airway inflammation in a murine model of asthma' J. ALLERGY CLIN. IMMUNOL. vol. 104, no. 6, 1999, pages 1258 - 1264, XP008054966 *
KLINE J.N.: 'Cutting Edge: Modulation of airway inflammation by CpG oligodeoxynucleotides in a murine model for asthma' J. IMMUNOLOGY vol. 160, 1998, pages 2555 - 2559, XP002319386 *
KLINE J.N.: 'Effects of CpG DNA on Th1/Th2 balance in asthma' CURRENT TOPICS MICROBIOL. IMMUNOL. vol. 247, 2000, pages 211 - 225, XP008054963 *
KLINE J.N.: 'Treatment of established asthma in a murine model using CpG oligodeoxynucleotides' AM. J. PHYSIOL. LUNG CELL MOL. PHYSIOL. vol. 283, July 2002, pages L170 - L179, XP002992307 *
REDECKE V.: 'Cutting Edge: Activation of Toll-Like receptor 2 induces a Th2 immune response and promotes experimental asthma' J. IMMUNOLOGY vol. 172, 2004, pages 2739 - 2743, XP002992304 *
See also references of EP1578954A2 *
SUR S.: 'Long term prevention of allergic lung inflammation in a mouse model of asthma by CpG oligodeoxynucleotides' J. IMMUNOLOGY vol. 162, 1999, pages 6284 - 6293, XP001113080 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8283328B2 (en) 2002-08-19 2012-10-09 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
EP2290078A3 (en) * 2002-08-19 2013-02-13 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
EP1538904A4 (en) * 2002-08-19 2008-03-05 Coley Pharm Group Inc Immunostimulatory nucleic acids
EP1538904A2 (en) * 2002-08-19 2005-06-15 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
US8304396B2 (en) 2002-08-19 2012-11-06 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids
US7795235B2 (en) 2004-10-20 2010-09-14 Coley Pharmaceutical Gmbh Semi-soft c-class immunostimulatory oligonucleotides
WO2006135434A3 (en) * 2004-10-20 2007-05-10 Coley Pharm Group Inc Semi-soft c-class immunostimulatory oligonucleotides
US8227447B2 (en) 2007-08-13 2012-07-24 Coley Pharmaceutical Gmbh RNA sequence motifs in the context of defined internucleotide linkages inducing specific immune modulatory profiles
US8128944B2 (en) 2007-08-13 2012-03-06 Coley Pharmaceutical Gmbh RNA sequence motifs in the context of defined internucleotide linkages inducing specific immune modulatory profiles
US8466124B2 (en) 2007-08-13 2013-06-18 Coley Pharmaceutical Gmbh RNA sequence motifs in the context of defined internucleotide linkages inducing specific immune modulatory profiles
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9764031B2 (en) 2010-05-26 2017-09-19 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses

Also Published As

Publication number Publication date
US7956043B2 (en) 2011-06-07
AU2003300919A1 (en) 2004-06-30
EP1578954A2 (en) 2005-09-28
JP2006512927A (en) 2006-04-20
WO2004053104A3 (en) 2005-12-15
JP2010148524A (en) 2010-07-08
CA2502015A1 (en) 2004-06-24
US20040171571A1 (en) 2004-09-02
EP1578954A4 (en) 2010-08-11

Similar Documents

Publication Publication Date Title
US7956043B2 (en) 5′ CpG nucleic acids and methods of use
US8188254B2 (en) C-class oligonucleotide analogs with enhanced immunostimulatory potency
AU2004275876B2 (en) Nucleic acid-lipophilic conjugates
US20180171338A1 (en) Compositions and methods for oligonucleotide formulations
US9382545B2 (en) CpG oligonucleotide analogs containing hydrophobic T analogs with enhanced immunostimulatory activity
EP2290078B1 (en) Immunostimulatory nucleic acids
US20060211644A1 (en) Immunostimulatory oligonucleotides
EP2197488A1 (en) Immune stimulatory oligonucleotide analogs containing modified sugar moieties

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE 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 NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG UZ VC VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2502015

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 168451

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2003300919

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005511961

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003813010

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003813010

Country of ref document: EP