CA2560108A1 - Immunostimulatory nucleic acids for inducing il-10 responses - Google Patents

Immunostimulatory nucleic acids for inducing il-10 responses Download PDF

Info

Publication number
CA2560108A1
CA2560108A1 CA002560108A CA2560108A CA2560108A1 CA 2560108 A1 CA2560108 A1 CA 2560108A1 CA 002560108 A CA002560108 A CA 002560108A CA 2560108 A CA2560108 A CA 2560108A CA 2560108 A1 CA2560108 A1 CA 2560108A1
Authority
CA
Canada
Prior art keywords
oligonucleotide
subject
cytosine
modified
nucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002560108A
Other languages
French (fr)
Inventor
Arthur M. Krieg
Joerg Vollmer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Coley Pharmaceutical GmbH
Coley Pharmaceutical Group Inc
Original Assignee
Coley Pharmaceutical Group, Inc.
Coley Pharmaceutical Gmbh
Arthur M. Krieg
Joerg Vollmer
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, Arthur M. Krieg, Joerg Vollmer filed Critical Coley Pharmaceutical Group, Inc.
Publication of CA2560108A1 publication Critical patent/CA2560108A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/17Immunomodulatory nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention relates to methods and products for inducing IL-10 expression using immunostimulatory nucleic acids. In particular, the invention relates to methods and products for inducing IL-10 expression without inducing high levels of IFN-.alpha. expression. IL-10-inducing immunostimulatory nucleic acids preferably include a TC dinucleotide at the 5' end and a CG dinucleotide towards the 3' end, but not near the 5' end. The invention is useful for treating and preventing disorders associated with a Thl or Th2 immune response or for promoting a T regulatory cell environment suitable for suppressing inappropriate immune responses (e.g., for controlling or suppressing excessive immune responses).

Description

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE I)E CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST ~.E TOME 1 DE 2 NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional vohxmes please contact the Canadian Patent Oi~ice.

RESPONSES
RELATED APPLICATIONS
This application claims priority to U.S. Provisional Patent Application filed April 2, 2004, entitled "IMMUNOSTIMULATORY NUCLEIC ACIDS FOR
INDUCING IL-10 RESPONSES", Serial No. 60/558,951, the contents of which are incorporated by reference herein in their entirety.
1o FIELD OF THE INVENTION
The present invention relates generally to immunostimulatory nucleic acids, and particularly to CpG containing immunostimulatory nucleic acids and their therapeutic uses.
BACKGROUND OF INVENTION
is The existence of functionally polarized T cell responses based on the profile of cytokines secreted by CD4+ T helper (Th) cells 'has been well established. In general, Thl cells secrete interferon-gamma (IFN-y), interleukin (IL)-2, and tumor necrosis factor-beta (TNF(3), and axe important in macrophage activation, the generation of both humoral and cell-mediated immune responses and phagocyte-dependent 20 protective responses. Th2 cells secrete IL-4, IL-5, IL-10, and IL-13 and are more important in the generation of humoral immunity, eosinophil activation, regulation of cell-mediated immune responses, control of macrophage function and the stimulation of particular Ig isotypes (Morel et al., 1998, Rornagnani, 1999). Thl cells generally develop following infections by intracellular pathogens, whereas Th2 cells 25 predominate in response to intestinal nematodes. In addition to their roles in protective immunity, Thl and Th2 cells are responsible for different types of immunopathological disorders. Fox example, Thl cells tend to predominate in organ-specific autoimmune disorders, Crohn's disease, Helicobaeter pylori-induced peptic ulcer, acute solid organ allogxaft rejection, and unexplained recurrent abortion, whereas Th2 cells tend to predominate in Omenn's syndrome, systemic lupus erythematosus, transplantation tolerance, chronic graft versus host disease, idiopathic pulmonary fibrosis, and progressive systemic sclerosis, and are involved in triggering of allergic reactions including most asthma (Romagnani 1999, Singh et al., 1999). In many diseases, such as lupus, there is evidence for both a Thl and Th2 component contributing to pathogenesis either at the same or different times during disease development.
An additional type of T cell response was observed when T cells were activated in the presence of interleukin 10 (IL-10). IL-10 activation results in the generation of a T cell subset known as regulatory T cells. Regulatory T cells have a cytokine profile that differs from both the Thl and Th2 cytokine profiles.
Regulatory T cells were also observed to have inhibitory effects on Ag-specific or Ag-nonspecific T cell activation, including both Thl and Th2 responses.
In recent years, a number of studies have demonstrated the ability of unmethylated CpG dinucleotides (i.e., the cytosine is unmethylated) within the context of certain flanking sequences (CpG motifs) to stimulate both innate and specific immune responses. Such sequences are commonly found in bacterial DNA
which is immunostimulatory. Similar immunostimulation is also possible with synthetic oligodeoxynucleotides (ODN) containing CpG motifs (CpG ODN). It has been demonstrated that CpG DNA can induce stimulation of B cells to proliferate and secrete immunoglobulin (Ig), IL-6 and IL-12, and to be protected from apoptosis (Krieg et al., 1995, Yi et al., 1996, Klinxnan et al., 1996). These effects contribute to the ability of CpG DNA to have adjuvant activity. In addition, CpG DNA
enhances expression of class II MHC and B7 co-stimulatory molecules (Davis et al., 1998, Sparwasser et al., 1998), that leads to improved antigen presentation.
Furthermore, CpG DNA also directly activates dendritic cells in mice to secrete various cytokines and chemokines (LThlmann and Vollmer, 2003) that can provide T-helper functions.
These ira vitYO effects were believed to be specific to the unmethylated CpG
motifs since they were not induced by methylated bacterial DNA or in general by ODN
that do not contain unmethylated CpG motifs.
Immunization of animals against a variety of antigens delivered both parenterally and mucosally demonstrate that addition of CpG ODN induces more Thl-dominated responses as indicated by strong cytotoxic T lymphocytes (CTL) stimulation, high levels of IgG2a antibodies, and predominantly Thl cytokines (e.g., IL-I2 and IFN-y but not IL-4 or IL-5) (Klinman et al., 1996, Davis et al., 1998, Roman et al., 1997, Chu et al., 1997, Lipford et al., 1997 Weiner et al., 1997, McCluskie and Davis, 1998, 1999).
In contrast, immunization experiments using nucleic acids lacking a CpG
demonstrate that mucosal administration of these nucleic acids can induce a Th2-1o dominated response.
SUMMARY OF THE INVENTION
The invention provides a subset of CpG containing nucleic acids that induce high levels of interleukin 10 (IL-10) expression without significant induction of 15 interferon alpha (IFN-cc) expression and type I interferon-mediated effects.
In one aspect, the invention provides CpG containing immunostimulatory nucleic acids that include a 5' TC dinucleotide separated from one or more CpG
dinucleotides located towards the 3' end of the nucleic acid. In preferred embodiments, the nucleic acid contains only one GpG dinucleotide.
20 In one aspect, the CpG immunostimulatory nucleic acids of the invention are useful for stimulating IL-10 expression without stimulating IFN-a expression and type I interferon-mediated effects.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful for obtaining a regulatory T cell response. In particular, the CpG
25 immunostimulatory nucleic acids are useful for treating diseases or conditions where a regulatory T cell response is favorable.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful for obtaining a regulatory B cell response. In particular, the CpG
immunostimulatory nucleic acids are useful for treating diseases or conditions where a regulatory B cell response is favorable.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful for stimulating B cells. In particular, the CpG immunostimulatory nucleic acids are useful for treating diseases or conditions where B cell stimulation is favorable.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful for obtaining a regulatory B cell response. In particular, the CpG
immunostimulatory nucleic acids are useful for treating diseases or conditions where a regulatory B cell response is favorable.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful to reduce or minimize a host subject's rejection of an organ transplant or tissue graft.
In another aspect, the CpG immunostimulatory nucleic acids of the invention 15 are useful to treat asthma, allergy, autoimmune diseases, and other inflammatory disorders.
In another aspect, the CpG immunostimulatory nucleic acids of the invention are useful for antigen-specific vaccinations in patients with an autoimmune disease.
In another aspect, the invention is an oligonucleotide chosen from: a) 5' 2o XYNIYZN2 3', wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X is a T or modified T
nucleotide, wherein Y is a C or modified C nucleotide, wherein Z is a G or modified G nucleotide, wherein Nl and N2 are polynucleotides that do not include a CG
dinucleotide, wherein Nl does not include 5' Z nucleotide, and wherein a 3' 25 polynucleotide consisting of the YZ dinucleotide and the N2 polynucleotide contains a number of nucleotides that is at most 45% of the number of nucleotides in the oligonucleotide; and b) 5' XY N1YZ N2 3', wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X
is a T
or modified T nucleotide, wherein Y is a C or modified C nucleotide, wherein Z
is a 3o G or modified G nucleotide, wherein Nl is a polynucleotide of 5 to 10 nucleotides, wherein N1 does not include a CG dinucleotide, wherein NI does not include 5' Z
nucleotide, and wherein NZ is a polynucleotide of 5 to 30 nucleotides.
In some embodiments, the oligonucleotide includes at least 1 modified internucleotide linkage. In othex embodiments, the oligonucleotide includes at least 50% modified internucleotide linkages. In other embodiments, all internucleotide linkages of the oligonucleotide are modified. In yet other embodiments, between 0%
and 10%, between 10% and 20%, between 20% and 30%, between 30% and 40%, between 40% and 50%, between 50% and 60%, between 60% and 70%, between 70%
and 80%, between 80% and 90%, or between 90% and 100% modified internucleotide linkages. In other embodiments, the oligonucleotide consists of 10 to 100 nucleotides. Tn some embodiments, the modified internucleotide linkage is a phosphorothioate linkage. In some embodiments, the oligonucleotide comprises a phosphodiester linkage between a 5' C nucleotide and a 3' G nucleotide. In other embodiments, the oligonucleotide comprises a R-phosphorothioate linkage between a , 5' C nucleotide and a 3' G nucleotide.
In some embodiments, Y is a modified C nucleotide comprising a modified cytosine base selected from the group consisting of 5-substituted cytosines, 6-substituted cytosines, N4-substituted cytosines, cytosine analogs With condensed ring systems, uxacil, uracil derivatives, a universal base, an aromatic ring system, and a 2o hydrogen atom. In other embodiments, Y is a modified C nucleotide comprising a modified cytosine base selected from the group consisting of 5-methyl-cytosine, 5-fluoro-cytosine, 5-chloro-cytosine, 5-bromo-cytosine, 5-iodo-cytosine, 5-hydroxy-cytosine, 5-hydroxymethyl-cytosine, 5-difluorornethyl-cytosine, unsubstituted or substituted 5-alkynyl-cytosine, N4-ethyl-cytosine, 5-aza-cytosine, 2-mercapto-cytosine, isocytosine, pseudo-isocytosine, N,N'-propylene cytosine or phenoxazine, 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4-thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil, 3-nitropyrrole, P-base, fluorobenzene, and difluorobenzene.
In some embodiments, Z is a modified G nucleotide comprising a modified guanine base selected from the group consisting of 7-deazaguanine, 7-deaza-7-substituted guanine, 7-deaza-7-(C2-C6)alkynylguanine, 7-deaza-8-substituted guanine, hypoxanthine, N2-substituted guanines, N2-methyl-guanine, 5-amino-3-methyl-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, inosine, adenine, substituted adenines, N6-methyl-adenine, 8-oxo-adenine, 8-substituted guanine, 8-hydroxyguanine, 8-bromoguanine, 6-thioguanine, a universal base, 4-methyl-indole, 5-nitro-indole, K-base, an aromatic ring system, benzimidazole, dichloxo-benzimidazole, 1-methyl-1H-[1,2,4]triazole-3-carboxylic acid amide, arid a hydrogen atom.
In some embodiments, the oligonucleotide comprises a 3'-3' linkage with one or two accessible 5' ends.
In some embodiments, the oligonucleotide comprises a nucleotide sequence l0 that does not contain an optimal CpG hexameric sequence. In other embodiments, the oligonucleotide comprises a nucleotide sequence that does not contain a palindromic sequence. In other embodiments, the oligonucleotide does not form a stable secondary structure.
In some embodiments, the oligonucleotide is conjugated to a moiety selected from the group consisting of antigens and cytokines. In some embodiments, the antigen can be selected from the group consisting of infectious disease antigens. In some embodiments, the cytokine can be selected from the group consisting of IL-4, IL-lO,IL-12.
In one embodiment, the oligonucleotide has the following structure: 5' 2o T*C*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T 3' (SEQ ID N0:4) and wherein refexs to a phosphoxothioate linkage. In another embodiment, the oligonucleotide has the following structure: 5' T*T*G*C*G*T*G*C*G*T*T*T*T*G*A*C*G*T*T*T*T*T*T*T3'(SEQID
N0:62) and wherein * refers to a phosphorothioate linkage. In another embodiment, the oligonucleotide has the following structure: 5' T*C*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T 3' (SEQ ID N0:2) and wherein refers to a phosphorothioate linkage.
In some embodiments, NI is a poly-T polynucleotide. In other embodiments, N2 is a poly-T polynucleotide. Both Nl and N2 can also be poly-T
polynucleotides.
3o The poly-T polynucleotide can contain one or more modified T nucleotides.
In preferred embodiments, the poly-T polynucleotide contains between 5 and 20 T
nucleotides, between 5 and 10 T nucleotides, more than 20 T nucleotides, or at least 55% T nucleotides.
In another aspect, the invention is a pharmaceutical composition including an oligonucleotide described herein in combination with a therapeutic agent selected from the group consisting of chemotherapeutic agents, radiotherapeutic agents, monoclonal antibodies, and anticancer agents. In some embodiments, the pharmaceutical composition comprises an oligonucleotide in combination with a polycation Garner.
In another aspect, the invention is a method of specifically increasing IL-10 expression relative to IFN-oc expression in a subject, including the step of administering an oligonucleotide or a pharmaceutical composition of the invention to a subject in whom inducing a T regulatory response may be beneftcial. In preferred embodiments, the step of administering is selected from the group consisting of respiratory, oral, topical, subcutaneous, and infra-venous administrations.
In another aspect, the invention is a method of inducing an antigen-specific regulatory T or B cell response in a subject, including the step of:
administering an immunostimulatory nucleic acid or composition of the invention to a subject exposed to an antigen. In some embodiments, the antigen is administered to the subject along with the immunostimulatory nucleic acid or composition. In other embodiments, the 2o antigen is administered to the subject after the immunostimulatory nucleic acid or composition. In other embodiments, the antigen is present in a food and the subject is exposed to the antigen by ingesting the food. In yet other embodiments, the antigen is inhaled by the subject.
In another aspect, the invention is a method of treating an allergy ox asthma, including the steps of exposing a subject to an allergen and administering an immunostimulatory nucleic acid or composition of the invention to the subject, wherein the immunostirnulatory nucleic acid or composition is administered in an amount sufficient to prevent or alleviate an allergic response to the allergen in the subject. In some embodiments, the method also includes administering IL-10 to the subject. In some embodiments, the subject has or is at risk of developing allergic asthma.

_g_ In another aspect, the invention is a method of treating an autoimmune disease in a subject, including the steps of exposing a subject to a self antigen and administering an immunostimulatory nucleic acid or composition of the invention to the subject, wherein the immunostimulatory nucleic acid or composition is administered in an amount sufficient to prevent or treat an autoimmune disease in the subject. In some embodiments, the method also includes administering IL-10 to the subject. In some embodiments, the autoimmune disease is arthritis, multiple sclerosis, Type 1 diabetes mellitus, Multiple sclerosis, Myasthenia gravis, Autoimmune neuropathies, such as Guillain-Barre, Autoimmune uveitis, Autoimmune hemolytic l0 anemia, Pernicious anemia, Autoimmune thrombocytopenia, Temporal arteritis, Anti-phospholipid syndrome, Psoriasis, Pemphigus vulgaris, Vasculitides such as Wegener's granulomatosis, Vitiligo, Crohn's Disease, Ulcexative colitis, Primary biliary cirrhosis, Autoirnmune hepatitis, Type 1 or immune-mediated, diabetes mellitus, Grave's Disease, Hashimoto's thyroiditis, Autoimmune oophoritis and orchitis, Autoimmune disease of the adrenal gland, Rheumatoid arthritis, Systemic lupus erythematosus, Scleroderma, Polymyositis, dermatomyositis, Spondyloarthropathies, such as ankylosing spondylitis, or Sjogren's syndrome.
In some embodiments, the autoimmune disease is caused by an infection, for example Lyme disease.
2o In another aspect, the invention is a method of reducing an antigen-specific response to an implant in a subject, including the steps of exposing a subject to an implant antigen and administering an immunostimulatory nucleic acid or composition of the invention to the subject, wherein the immunostimulatory nucleic acid or composition is administered in an amount sufficient to prevent or reduce an antigen-specific response to the implant in the subject. In some embodiments, the method also includes administering IL-10 to the subject. In some embodiments, the implant is an autologous tissue implant. In other embodiments, the implant is a non-autologous tissue implant. In other embodiments, the implant is a recombinant cellular implant.
In other embodiments, the implant is a synthetic implant.
In some embodiments, the invention does not include one or more nucleic acids, or use thereof, having one or more of the following sequences (shown 5' to 3'):

TCAACGCT; TCAACGTT; TCAACGTT; TCAAGCTT; TCAAGCTT;
TCACATGTGGAGCCGCGT (SEQ ID N0:63); TCACGGTT; TCAGCGCT;
TCAGCGCT;TCATCGAT;TCATCGAT;TCCAAGACGTTCCTGATGCT(SEQ
ID N0:64); TCCATAACGTTCCTGATGCT (SEQ ID N0:65);
TCCATAACGTTCCTGATGCT (SEQ ID NO:66); TCCATATTGCACCTGATGCT
(SEQ ID N0:67); TCCATCACGTGCCTGATGCT (SEQ TD N0:68);
TCCATCACGTGCCTGATGCT (SEQ ID N0:69);
TCCATCGCCAAGGAGATCGAGCTGGAGGATCCGTACGAGAAGATC(SEQ
ID N0:70); TCCATGACGGTCCTGATGCT (SEQ TD N0:71).
1o TCCATGACGGTCCTGATGCT (SEQ ID N0:72); TCCATGACGTCCCTGATGCT
(SEQ ID N0:73); TCCATGACGTCCCTGATGCT (SEQ ID NO:74);
TCCATGACGTTCCTGATGCT (SEQ ID N0:75); TCCATGACGTTCCTGATGCT
(SEQ ID N0:76); TCCATGACGTTCCTGATGCT (SEQ ID NO:77);
TCCATGACGTTCCTGATGCT (SEQ ID N0:78); TCCATGACGTTCCTGATGCT
is (SEQ ID N0:79); TCCATGACGTTCCTGATGCT (SEQ ID N0:80);
TCCATGACGTTCCTGATGCT (SEQ ID NO:81); TCCATGAGCTTCCTGAGTCT
(SEQ ID N0:82); TCCATGAGCTTCCTGATGCT (SEQ ID N0:83);
TCCATGAGCTTCCTGATGCT (SEQ ID N0:84); TCCATGCCGGTCCTGATGCT
(SEQ ID N0:85); TCCATGCCGGTCCTGATGCT (SEQ ID N0:86);
2o TCCATGCTGGTCCTGATGCT (SEQ ID N0:87); TCCATGCTGGTCCTGATGCT
(SEQ ID N0:88); TCCATGGCGGTCCTGATGCT (SEQ ID NO:89);
TCCATGGCGGTCCTGATGCT (SEQ TD NO:90); TCCATGTCGATCCTGATGCT
(SEQ ID N0:91); TCCATGTCGATCCTGATGCT (SEQ ID N0:92);
TCCATGTCGGTCCTGATGCT (SEQ ID N0:93); TCCATGTCGCTCCTGATGCT
25 (SEQ ID NO:94); TCCATGTCGGTCCTGATGCT (SEQ ID N0:95);
TCCATGTCGGTCCTGATGCT (SEQ ID N0:96); TCCATGTCGGTCCTGATGCT
(SEQ ID N0:97); TCCATGTCGGTCCTGATGCT (SEQ ID N0:98);
TCCATGTCGGTCCTGCTGAT (SEQ ID N0:99); TCCATGTCGGTZCTGATGCT
(SEQ ID NO:100); TCCATGTCGTTCCTGATGCT (SEQ ID NO:101);
30 TCCATGTCGTTCCTGATGCT (SEQ ID N0:102); TCCATGTCGTTCCTGATGCT
(SEQ ID NO:103); TCCATGTZGGTCCTGATGCT(SEQ ID N0:104);
TCCATGTZGTTCCTGATGCT(SEQ ID NO:I05);

TCCCCCATGCCGCCCTCCGGG (SEQ ID N0:106); TCCGCGTT;
TCCGCTGACGTCGCCGCCCAGATGGCCTCC (SEQ ID N0:107);
TCCTCCTCCTCCTCC (SEQ ID N0:108); TCGACGTC;
TCGGCGGTGAAGAAGACT (SEQ ID N0:109); TCGGTCAACGTTGAGATGCT
s (SEQ ID NO:110); TCGGTGAACGTTATGTCGCAGGACCCGGTC (SEQ ID
NO:I 11); TCGGTGACCGGTATGTCGCAGGACCCGGTC (SEQ ID N0:112);
TCGGTGAGCGCTATGTCGCAGGACCCGGTC (SEQ ID N0:113);
TCGGTGCAGGGAATGTCGCAGGACCCGGTC (SEQ ID N0:114);
TCGGTGCAGGGAATGTCGCAGGACCCGGTCGCGGTGGCGGCCTCG(SEQ
1o ID NO:115); TCGGTGCAGGGAATGTCGCAGGACGACGTC (SEQ ID NO:l 16);
TCGGTGGACGTCATGTCGCAGGACCCGGTC (SEQ ID NO:l 17);
TCGGTGGACGTCATGTCGCAGGACCCGGTC (SEQ ID NO:l 18);
TCGGTGGACTGCATGTCGCAGGACCCGGTC (SEQ ID N0:119);
TCGGTGGACTGCATGTCGCAGGACCCGGTC (SEQ ID N0:120); TCGTCG;
15 TCGTCGCTGTCTCCG (SEQ ID N0:121); TCGTCGCTGTCTCCGCTTCTT (SEQ
ID N0:122); TCGTCGCTGTCTCCGCTTCTTCTTGCC (SEQ ID N0:123);
TCGTCGCTGTCTCCGCTTCTTCTTGCC (SEQ ID N0:124);
TCGTCGCTGTCTCCGCTTCTTCTTGCC (SEQ ID N0:125);
TCGTCGCTGTCTCCGCTTCTTCTTGCCA (SEQ ID N0:126);
2o TCGTCGGGGGGGGGGG (SEQ ID N0:127); TCGTCGTCG; TCGTCGTCGTCG
(SEQ ID N0:128); TCGTCGTCGTCGTCG (SEQ ID NO:129);
TCTCCATGATGGTTTTATCG (SEQ ID N0:130); TCTCCCAGCGTGCGCCAT
(SEQ ID N0:131); TCTCCCAGCGTGCGCCAT (SEQ ID N0:132);
TCTCCCAGZGTGZGCCAT (SEQ ID N0:133); TCTTCGAA; TCTTCGAA;
2s TCTTCTGCCCCCTGTGCA (SEQ ID N0:134); TGACGTTTGACGTTTGACGTT
(SEQ ID N0:135); TGACTGTGAACGTTCGAGATGA (SEQ ID N0:136);
TGATCTTCCATCTATTAG (SEQ ID N0:137); TGCACAGGGGGCAGAAGA
(SEQ ID N0:138); TGGTGGTGGTGGTGG (SEQ ID N0:139);
TTGCTTCCATCTTCCTCGTC (SEQ ID NO:140);
3o TTGGTGAAGCTAACGTTGAGGGGCAT (SEQ ID N0:141).

This invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description ox illustrated in the drawings. The invention is capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the puxpose of description and should not be regarded as limiting. The use of "including," "comprising," or "having," "containing", "involving", and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
1o BRIEF DESCRIPTION OF DRAWINGS
The accompanying drawings, are not intended to be drawn to scale. In the drawings, each identical or nearly identical component that is illustrated in various figuxes is represented by a like numeral. For purposes of clarity, not every component may be labeled in every drawing. In the drawings:
Figure 1 shows that shifting a CpG dinucleotide from a 5' end to a 3' end of an oligonucleotide xesults in decreased IFN-a production and a constant IL-10 stimulation: Figure lA shows IFN-a production in response to different oligonucleotides; Figure 1B shows IL-10 production in response to different oligonucleotides;
Figure 2 shows that oligonucleotides with strongly reduced IFN-a production result in optimal IL-10 stimulation when they contain an unmodified C in the CpG
dinucleotide;
Figure 3 shows that oligonucleotides with a higher T content result in higher IL-10 stimulation;
Figure 4 shows that a 5'-TCG is required for efficient IFN-a production, whereas a 5'-TC is sufficient fox potent IL-10 secretion;
Figure 5 shows that IL-10 stimulation is maintained when the thymidine of the 5'-TC is chemically modified;

Figure 6 shows that oligonucleotides with a 5'-TC or a 3' shifted CpG
dinucleotide induce stronger IL-10 production than oligonucleotides lacking a 5'-TC
or a CpG;
Figure 7 shows that oligonucleotides with a 5'-TC and a 3' shifted CpG
dinucleotide induce strong secretion of IL-6 or IL-10 but result in inefficient stimulation of cytokines or chemokines such as IFN-a, or IP-10;
Figure 8 shows that oligonucleotides with a 5'-TC and a 3' shifted CpG
efficiently induce the production of IL-6 and IL-10 from highly purified human B
cells;
l0 Figure 9 shows that cells expressing the human TLR9 and an NFxB-Luciferase reporter are stimulated by oligonucleotides with a 5'-TC and a 3' shifted CpG; and Figure 10 shows TLR9-mediated NFkB responses to oligonucleotides with CpG dinucleotides at different 3' positions: Figure l0A shows human cell responses;
15 Figure l OB shows murine cell responses.
DETAILED DESCRIPTION
The invention provides CpG dinucleotide containing immunostimulatory nucleic acids that increase IL-10 expression without significantly increasing IFN-a 20 expression. The nucleic acids of the invention are useful for treating diseases and disorders including autoimmune disorders.
In one aspect, the invention provides a nucleic acid, preferably an oligonucleotide, that includes a TC dinucleotide at its 5' end and a CpG
dinucleotide separated from the TC dinucleotide by at least two nucleotides.
25 In one embodiment, the CpG dinucleotide is separated from the TC
dinucleotide by at least 2 nucleotides, and more preferably by 3, 4, 5, 6, 7, 8, ,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 25, 26, 27, 28, 29, or 30 or more nucleotides. In another embodiment, the CpG dinucleotide is included in the 3' 80%, 7S%, 70%, 6S% 60%, SS%, SO%, 4S%, 40%, 3S%, 30%, 2S%, 20%, 15%, 10%, S%, or 2.S% of the length of the nucleic acid molecule.
In some embodiments, the nucleic acid has two or more TC dinucleotides, two or more CpG dinucleotides, or combinations thereof. The S'-most CpG
dinucleotide is preferably separated from the 3' most TC dinucleotide (which is S' to the S' most CpG dinucleotide) by 1, 2, 3, 4, S, 6, 7, 8, ,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 2S, 26, 27, 28, 29, or 30 ox more nucleotides. The TC
dinucleotides are preferably in the S' 10%, 20%, 30%, 40%, or SO% of the length of the nucleic acid.
The CpG dinucleotides are in the 3' SO%, 40%, 30%, 20%, or 10% of the length of to the nucleic acid. However, the TC and CpG dinucleotides can be interspersed provided that there is a TC dinucleotide at the S' end of the molecule and that the S' most CpG is separated from the TC dinucleotide by 2, 3, 4, S, 6, 7, 8, 9, 10, 1 l, 12, 13, 14, 1 S, 16, 17, 18, 19, 20, 21, 22, 23, 2S, 26, 27, 28, 29, or 30 or more nucleotides, and the optimal distance between the S' TC and the CpG dinucleotide can depend on the length of the nucleic acid molecule. In preferred embodiments, the 3' dinucleotide is preferably not a CpG dinucleotide.
In some embodiments, the 5' dinucleotide is AC, GC, CC, TA, TG, or TT.
However, a nucleic acid with a S' TC, stimulates IL-10 production more effectively.
In some embodiments, the nucleic acid has a modified C in the CpG
dinucleotide.
However, in other embodiments a nucleic acid with an unmodified C in the CpG
dinucleotide can be used for ease of synthesis or to reduce potential in vivo toxicity.
Nucleic acids of the invention preferably have one or more stretches of poly T
(e.g. 3T, 4T, ST, 6T, 7T, 8T, 9T, l OT, or longer stretches of poly T). A
preferred nucleic acid includes between 2S% and 99%, preferably between 30% and 90%, preferably more than 3S%, more than 40%, more than 45%, more than SO%, more than SS%, more than 60%, more than 65%, more than 70%, more than 7S%. more than 80%, more than 8S%, more than 90%, or more than 9S% T nucleotides.
Preferred nucleic acids are between S and 100 nucleotides long, and preferably longer than about 10, 1S, 20, 25, 30, 3S, or 40 nucleotides long. However, longer 3o nucleic acids are also embraced by the invention. A preferred nucleic acid is between about 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 nucleotides long.
Preferred nucleic acids do not have a 5' TCG trinucleotide. Nucleic acids can be provided as double-stranded molecules. Nucleic acids are preferably single-stranded molecules, and more preferably DNA molecules. However, one or more of the nucleotides and/or the internucleotide linkages can be modified as described herein.
In one embodiment, a nucleic acid of the invention has the following general formula:
5' XYN1YZN2 3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X is a T or modified T nucleotide, wherein Y is a C or modified C nucleotide, wherein Z is a G or modified G nucleotide, wherein Nl and N2 are polynucleotides that do not include a CG dinucleotide, wherein Nl does not include 5' Z nucleotide, and wherein a 3' polynucleotide consisting of the YZ
dinucleotide and the NZ polynucleotide contains a number of nucleotides that is at most 45% of the number of nucleotides in the oligonucleotide.
In another embodiment, a nucleic acid of the invention has the following general formula:
5' XY N1YZ N2 3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X is a T or modified T nucleotide, wherein Y is a C or modified C nucleotide, wherein Z is a G or modified G nucleotide, wherein Nl is a polynucleotide of 5 to 10 nucleotides, wherein Nl does not include a CG
dinucleotide, wherein Nl does not include 5' Z nucleotide, and wherein N2 is a polynucleotide of 5 to 30 nucleotides;
Nucleic acids of the invention stimulate the production of IL-10 relative to that of IFN-a. The ratio of IL-10 induction relative to IFN-a induction is preferably between 1.5 and 10, and can be higher. In some embodiments, the ratio of induction 3o is more than about 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, or 10Ø

Immunostimulatory CpG nucleic acids of the invention form a subset of CpG
nucleic acids that have distinct properties from immunostimulatory CpG nucleic acids previously studied. Three classes of CpG ODN have been described so far: the A-, B-and C-Classes. The most striking attribute of these described CpG ODN classes is their ability to stimulate the secretion of IFN-a from pDC and, therefore, of other effects that are mediated by type I interferons such as IP-10 production from monocytes (Blackwell (2003), J. Immunol. 170: 4061). Nevertheless, differences appear to exist between the stimulation of the two TLR9 expressing cells described to date: pDC and B cells (LThlmann (2003), Current Drugs 6: 204). B cells are to stimulated by immune modulatory ODN to secrete cytokines such as IL-6 or IL-(Krieg (2002), Annu. Rev. Immunol. 20:709). PDCs are, in contrast, stimulated to produce type I interferons. The CpG ODN classes described to date stimulate both PDC activation and cytokine production as well as B cell activation (LThlmann (2003), Current Drugs 6: 204). However, the invention provides ODN sequences that i5 stimulate few to no IFN-a secretion or related effects (such as IP-10 production from monocytes) but stimulate strong cytokine secretion from B cells in a TLR9-dependent way. The CpG immunostimulatory nucleic acids of the invention, termed T-Class ODN, lack a 5'-CG that is mainly responsible for the strong stimulatory effects mediated by CpG on human cells. In preferred embodiments, they contain a 5'TC
2o that was shown to still retain potent and efficient cytokine production from B cells. In addition, such preferred ODN still bear a CpG dinucleotide, although in a more 3' position. This CpG shift towards the 3' end results in a strong decrease of pDC IFN-a production but not B cell IL-10 secretion. The CpG immunostimulatory nucleic acids of the invention induce efficient IL-10 production but don't induce efficient IFN-a 25 production.
Although IL-10 is often considered to be a Th2-inducing cytokine, it can be a "suppressive" cytokine under certain conditions, for example when IL-10 production is out of proportion relative to other Th2 cytokines such as IL-4, IL-5, and IL-13.
Studies demonstrated that IL-10 is involved in the reduction of inflammatory 30 responses and autoimmune diseases (Mocellin (2003), TRENDS 24: 36). This effect involves regulatory lymphocytes, T cells as well as B cells (Shevach (2002), Nature Reviews Immunol. 2: 389; Sakaguchi (2003), Nature Immunol. 4: 10; Fillatreau (2002), Nature Immunol. 10: 944; Mauri (2003), J. Exp. Med. 197: 489;
Mizoguchi (2002), Immunity 16: 219). IL-10 was demonstrated in vitro to be responsible for the generation of IL-I O producing regulatory T cells (Shevach (2002), Nature Reviews Ixnmunol. 2: 389). These T cells appear to influence the immune response of the host to e.g. bacterial infections. These T cells were also demonstrated to help to protect from autoimmune disease development (Shevach (2002), Nature Reviews Immunol.
2: 389). The same effect was observed with regulatory B cells (Fillatreau (2002), Nature Immunol. 10: 944; Mauri (2003), J. Exp. Med. I97: 489; Mizoguchi (2002), to Immunity 16: 219). In one embodiment of the invention, T-class CpG ODN are used to mediate strong stimulation of B cells that produce high levels of IL-10, and are useful as therapy for autoimmune diseases.
In one aspect, CpG stimulatory nucleic acids of the invention are useful to induce increased IL-10 levels in relation to IFN-a levels. In one embodiment, the ratio of IL-I0/IFN-a expression induced by an oligonucleotide of the invention is at least SO% higher than the ratio of IL-10/IFN-a expression induced by a reference oligonucleotide, for example: S' T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G*T*T3'(SEQID
N0:54), 5' T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*T*T*T*C*G*A 3' (SEQ ID N0:142), or 5' T*C*G*T*C*G*T*T*T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T3'(SEQID NO:
143).
The ratio may be even higher, e.g., 2 fold, 3 fold, 4 fold, 5 fold, 10 fold, fold, 100 fold, ox more. The ratio of IL-10/IFN-a induced by an oligonucleotide may be calculated by dividing the induced amount or percent of IL-10 increase by the induced amount or percent of IFN-a increase. The induced amount or percent increase of expression of a molecule may be calculated by comparing the expxession levels of the molecule before and aftex treatment with the oligonucleotide.
The expression levels may be RNA or protein expression levels.
3o In one embodiment, an oligonucleotide of the invention induces an increase in IL-10 expression that is similar to that of a reference oligonucleotide (e.g., one of the reference oligonucleotides described above). However, the induced increase in IFN-a expression may be significantly lower (e.g., 2 fold, 3, fold, 4 fold, 5 fold, 10 fold, or 50 fold lower, etc.) than that obtained with the reference oligonucleotide.
This results in a higher ratio of IL-10/IFN-a induction using an oligonucleotide of the invention.
In one embodiment, only background levels of IFN-a are obtained with an immunostimulatory nucleic acid of the invention.
However, in other embodiments, the absolute level of IL-10 induction obtained with an oligonucleotide of the invention is higher than that obtained with a reference oligonucleotide (e.g., 50% more, 2 fold, 3 fold, 4 fold, 5 fold, 10 fold, or l0 50 fold higher, etc.).
Accordingly, in one aspect of the invention, T-class CpG stimulatory nucleic acids are used to stimulate IL-10 production. In some embodiments, the CpG
stimulatory nucleic acids indirectly stimulate IL-10 production from macrophages. In other embodiments, the CpG stimulatory nucleic acids stimulate IL-10 production 15 from B cells. In yet further embodiments, the CpG stimulatory nucleic acids stimulate IL-10 production from one or more cell types. IL-10 production in the absence of IFN-a production is useful to treat diseases and conditions such as autoimmune diseases or disorders. In some embodiments, IL-10 production is useful to activate T regulatory cells. In other embodiments, IL-10 production is useful to 2o activate B regulatory cells. In yet further embodiments, IL-10 production is useful to suppress Thl cytokines. IL-10 production can be particularly useful to treat a subject with, or at risk of developing, one or more Th2-mediated allergic diseases or disorders. IL-10 can also be used to control autoimmune diseases such as autoimmune encephalomyelitis. Autoimmune diseases include, but are not limited to, 25 rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, pemphigus (e.g., pemphigus vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, 3o polymyositis, pernicious anemia, idiopathic Addison's disease, autoimmune-associated infertility, glomerulonephritis (e.g., crescentic glomerulonephritis, proliferative glomerulonephritis), bullous pemphigoid, Sjogren's syndrome, insulin resistance, and autoimmune diabetes mellitus.
In another aspect, CpG stimulatory nucleic acids of the invention are useful to stimulate a regulatory T cell response. Regulatory T cells can control diseases such as inflammatory bowel disease and are involved in the control of other immune responses including autoimmune responses.
Regulatory T cell activation can be used to regulate antibody specific responses, particularly in the context of allergies and autoimmune diseases.
In some embodiments, the CpG immunostimulatory nucleic acids are used for treating and to preventing antibody-mediated autoimmune diseases. In some autoimmune diseases, a subject's own antibodies react with host tissue or in which immune effector T
cells are autoreactive to endogenous self peptides and cause destruction of tissue.
Thus an immune response is mounted against a subject's own antigens, referred to as self antigens. Autoimmune diseases include but are not limited to rheumatoid arthritis, 15 Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, pemphigus (e.g., pemphigus vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious 2o anemia, idiopathic Addison's disease, autoimmune-associated infertility, glomerulonephritis (e.g., crescentic glomerulonephritis, proliferative glomerulonephritis), bullous pemphigoid, Sjogren's syndrome, insulin resistance, and autoimmune diabetes mellitus. Some of these autoimmune diseases can also associated with organ-specific autoimmune disorders involving a Th2 response.
25 In some embodiments, antigen-speciftc regulatory T cell responses can be stimulated by administering a speciftc antigen, preferably a self antigen, along with (not long before, simultaneously, or not long after) an immunostimulatory CpG
nucleic acid of the invention. In some instances, the CpG immunostimulatory nucleic acids are delivered with low doses of self antigens.
3o A "self antigen" as used herein refers to an antigen of a normal host tissue.
Normal host tissue does not include cancer cells. Thus an immune response mounted against a self antigen, in the context of an autoimmune disease, is an undesirable immune response and contributes to destruction and damage of normal tissue, whereas an immune response mounted against a cancer antigen is a desirable immune response and contributes to the destruction of the tumor or cancer.
In yet another aspect, CpG immunostimulatory nucleic acids of the invention are used to stimulate a regulatory B cell response. The stimulation of regulatory B
cells can be used to control diseases such as autoimmune disorders. In some embodiments, antigen-specific regulatory B cell responses can be stimulated by administering a specific antigen before, with, or after an immunostimulatory CpG
nucleic acid of the invention. In some embodiments, Th2-mediated diseases such as asthma and allergy can be treated by administering one or more CpG
immunostimulatory nucleic acids of the invention with one or more allergens.
In another embodiment, SLE can be treated by administering one or more CpG
stimulatory nucleic acids of the invention with one or more antigens such as purified components of nucleosomes or ribonucleoproteins. In a further embodiment, rheumatoid arthritis can be treated by administering one or more CpG
stimulatory nucleic acids of the invention with one or more antigens such as an immunoglobulin.
In a further aspect, CpG stimulatory nucleic acids of the invention are used to stimulate a T regulatory response. These nucleic acids can be administered (e.g. as an 2o adjuvant for vaccines or as a monotherapy) in a number of diseases for which strong T
regulatory responses might be more important such as Crohn's disease, allograft rejection or spontaneous abortion (McCluskie (2001), Vaccine 19: 413). In some embodiments, the CpG stimulatory nucleic acids of the invention are administered mucosally, Examples of mucosal administration methods and formulations are disclosed in (US Patent Publication 20010044416), the entire disclosure of which is incorporated herein by reference.
Stimulation of a T regulatory response can be useful to treat certain autoimmune diseases and conditions such as organ specific autoimmune disorders (e.g. Crohn's disease, peptic ulcer, acute solid organ allograft rejection, and unexplained recurrent abortion). Stimulation of a T regulatory response can also be useful to induce an antigen-specific response by administering an antigen to a subject along with a nucleic acid of the invention in an amount effective to produce an antigen-specific immune response.
According to the invention, the terms "nucleic acid" and "oligonucleotide"
also encompass nucleic acids or oligonucleotides with substitutions or modifications, such as in the bases and/or sugars. Fox 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 ox 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 1o as arabinose or 2'-fluoroarabinose 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).
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 Riotech~aol 14:840-4. Purines and pyrimidines include but are not limited to adenine, cytosine, guanine, thymine, 5-methylcytosine, 5-hydroxycytosine, 5-fluorocytosine, 2-aminopurine, 2-amino-6-chloropurine, 2,6-diaminopurine, hypoxanthine, and other naturally and non-naturally occurring nucleobases, 2o substituted and unsubstituted aromatic moieties. Other such modifications are well known to those of skill in the art.
The immunostimulatory oligonucleotides of the instant invention can encompass various chemical modifications and substitutions, in comparison to natural RNA and DNA, involving a phosphodiester internucleot'ide bridge, a ~i-D-ribose unit andlor a natural nucleotide base (adenine, guanine, cytosine, thymine, uracil).
Examples of chemical modifications are known to the skilled person and are described, fox example, in IJhimann E et al. (1990) Chem Rev 90:543;
"Protocols for Oligonucleotides and Analogs" Synthesis and Properties & Synthesis and Analytical Techniques, S. Agrawal, Ed, Humane Press, Totowa, USA 1993; Crooke ST et al.
(1996) Ararau 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 internucleotide bridge and/or at a particular (3 -D-ribose unit and/or at a particular natural nucleotide base position in comparison to an oligonucleotide of the same sequence which is composed of natural DNA or RNA.
For example, the invention relates to an oligonucleotide which may comprise one or more modifications and wherein each modification is independently selected from:
a) the replacement of a phosphodiester internucleotide bridge located at the 3' and/or the 5' end of a nucleotide by a modified internucleotide bridge, l0 b) the replacement of phosphodiester bridge located at the 3' and/or the 5' end of a nucleotide 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 (3 -D-ribose unit by a modified sugar unit, and 15 e) the replacement of a natural nucleotide base by a modified nucleotide base.
More detailed examples for the chemical modification of an oligonucleotide are as follows.
A phosphodiester internucleotide bridge located at the 3' and/or the 5' end of a 2o nucleotide can be replaced by a modified internucleotide bridge, wherein the modified internucleotide bridge is for example selected from phosphorothioate, phosphorodithioate, NR1R2-phosphoramidate, boranophosphate, a-hydroxybenzyl phosphonate, phosphate-(C1-CZl)-O-alkyl ester, phosphate-[(C6-C12)aryl-(C1-C21)-O-alkyl]ester, (C1-C8)alkylphosphonate and/or (C6-C12)arylphosphonate bridges, (C~-25 C12)-a-hydroxymethyl-aryl (e.g., disclosed in WO 95/01363), wherein (C6-C1z)aryl, (C6-C2o)aryl and (C6-C14)aryl are optionally substituted by halogen, alkyl, alkoxy, nitro, cyano, and where Rl and RZ are, independently of each other, hydrogen, (C1-Cis)-alkyl, (C6-CZO)-aryl, (C6-C14)-aryl-(C~-C$)-alkyl, preferably hydrogen, (C1-Cg)-alkyl, preferably (C1-Cq.)-alkyl and/or methoxyethyl, or Rl 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 S' end of a nucleotide 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, methylenedimethyl-hydrazo, dimethylenesulfone and/or silyl groups.
A sugar phosphate unit (i.e., a (3 -D-ribose and phosphodiester internucleotide 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 Chena 5:3-7), that is, e.g., the replacement by a PNA backbone unit, e.g., by 2-aminoethylglycine.
A (3 -ribose unit or a ~i -D-2'-deoxyribose unit can be replaced by a modified 2o sugar unit, wherein the modified sugar unit is for example selected from (3-D-ribose, a-D-2'-deoxyribose, L-2'-deoxyribose, 2'-F-2'-deoxyribose, 2'-F-arabinose, 2'-O-(C1-C6)alkyl-ribose, preferably 2'-O-(C1-C6)alkyl-ribose is 2'-O-methylribose, 2'-O-(C2-C6)alkenyl-ribose, 2'-[O-(C1-C6)alkyl-O-(C1-C6)alkyl]-ribose, 2'-NH2-2'-deoxyribose, [3-D-xylo-fuxanose, a-arabinofuranose, 2,4-dideoxy-(3-D-erythro-hexo-pyranose, and carbocyclic (described, for example, in Froehler J (1992) Am Claem S'oc 114:8320) and/or open-chain sugar analogs (described, for example, in Vandendriessche et al. (1993) Tetrahedron 49:7223) and/or bicyclosugar analogs (described, fox example, in Tarkov M et al. (1993) Helv Chirn Acta 76:481).

In some preferred embodiments the sugar is 2'-O-methylribose, particularly for one or both nucleotides linked by a phosphodiester or phosphodiester-like internucleotide 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 occurnng nucleobases, substituted and unsubstituted aromatic moieties.
A modified base is any base which is chemically distinct from the naturally to 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 nucleotide 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, 15 5-(hydroxymethyl)uracil, 5-chlorouracil, 5-fluorouracil, 5-bromouracil, 5-hydroxycytosine, 5-(Cl-C6)-alkylcytosine, 5-(C2-C6)-alkenylcytosine, S-(C2-C6)-alkynylcytosine, 5-chlorocytosine, 5-fluorocytosine, 5-bromocytosine, NZ-dimethylguanine, 2,4-diamino-purine, 8-azapurine, a substituted 7-deazapurine, preferably 7-deaza-7-substituted andlor 7-deaza-8-substituted purine, 5-20 hydroxymethylcytosine, N4-alkylcytosine, e.g., N4-ethylcytosine, 5-hydroxydeoxycytidine, 5-hydroxymethyldeoxycytidine, N4-alkyldeoxycytidine, e.g., N4-ethyldeoxycytidine, 6-thiodeoxyguanosine, and deoxyribonucleotides of nitropyrrole, C5-propynylpyrimidine, and diaminopurine e.g., 2,6-diaminopurine, inosine, 5-methylcytosine, 2-aminopurine, 2-amino-6-chloropurine, hypoxanthine or 25 other modifications of a natural nucleotide 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 3o non-naturally occurnng 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-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, 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, 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, 2o 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-hydroxyguanine 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-[1,2,4]triazole-3-carboxylic acid amide) or a hydrogen atom (dSpacer).
The oligonucleotides may have one or more accessible 5' ends. It is 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. The 3'3'-linkage may be a phosphodiester, phosphorothioate or any other modified internucleotide 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, Nucleotides & 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 nucleic acids where the linkage between the 3'-terminal nucleotides 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.
It recently has been reported that CpG oligonucleotides appear to exert their immunostimulatory effect through interaction with Toll-like receptor 9 (TLR9).
Hemmi H et al. (2000) Nature 408: 740-5. TLR9 signaling activity thus can be measured in response to CpG oligonucleotide or other immunostimulatory nucleic acid by measuring NF-xB, NF-xB-related signals, and suitable events and intermediates upstream of NF-~cB.
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, 3o M.H., Tet. Let. 22:1859, 1981); nucleotide 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, Gaffney 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 oligonucleotides are ,partially resistant to degradation (e.g., are stabilized).
A "stabilized oligonucleotide molecule" shall mean an oligonucleotide that is to relatively resistant to in vivo degradation (e.g. via an exo- or endo-nuclease). Nucleic acid stabilization can be accomplished via backbone modifications.
Oligonucleotides having phosphorothioate linkages provide maximal activity and protect the oligonucleotide from degradation by intracellular exo- and endo-nucleases.
Other modified oligonucleotides include phosphodiester modified nucleic acids, 15 combinations of phosphodiester and phosphorothioate nucleic acid, methylphosphonate, methylphosphorothioate, phosphorodithioate, p-ethoxy, and combinations thereof.
Modified backbones such as phosphorothioates may be synthesized using automated techniques employing either phosphoramidate or H-phosphonate 2o 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 25 substitutions have been described (e.g., Uhlmann, E. and Peyman, A., Claena. Rev.
90:544, 1990; Goodchild, J., Bioconjasgate Chena. 1:165, 1990).
Other stabilized oligonucleotides include: nonionic DNA analogs, such as alkyl- and aryl-phosphates (in which the charged phosphonate oxygen is replaced by an alkyl or aryl group), phosphodiester and alkylphosphotriesters, in which the 3o charged oxygen moiety is alkylated. Nucleic acids which contain diol, such as tetraethyleneglycol or hexaethyleneglycol, at either or both termini have also been shown to be substantially resistant to nuclease degradation.
As described herein, the oligonucleotides of the invention rnay have phosphodiester or phosphodiester like linkages between C and G. One example of a phosphodiester-like linkage is a phosphorothioate linkage in an Rp conformation.
Oligonucleotide p-chirality can have apparently opposite effects on the immune activity of a CpG oligonucleotide, depending upon the time point at which activity is measured. At an early time point of 40 minutes, the Rp but not the SP
stereoisomer of phosphorothioate CpG oligonucleotide induces JNK phosphorylation in mouse spleen cells. In contrast, when assayed at a late time point of 44 hr, the SP but not the Rp stereoisomer is active in stimulating spleen cell proliferation. This difference in the kinetics and bioactivity of the RP and SP stereoisomers does not result from any difference in cell uptake, but rather most likely is due to two opposing biologic roles of the p-chirality. First, the enhanced activity of the Rp stereoisomer compared to the Sp for stimulating immune cells at early time points indicates that the Rp may be more effective at interacting with the CpG receptor, TLR9, or inducing the downstream signaling pathways. On the other hand, the faster degradation of the Rp PS-oligonucleotides compared to the Sp results in a much shorter duration of signaling, so that the Sp PS-oligonucleotides appear to be more biologically active when tested at later time points.
A surprisingly strong effect is achieved by the p-chirality at the CpG
dinucleotide itself. In comparison to a stereo-random CpG oligonucleotide the congener in which the single CpG dinucleotide was linked in Rp was slightly more active, while the congener containing an Sp linkage was nearly inactive for inducing spleen cell proliferation.
According to the invention, 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 ash (aquaculture species), e.g. salmon. Thus, the invention can also be used to treat cancer and tumors, infections, and allergylasthma 3o in non human subjects. Cancer is one of the leading causes of death in companion animals (i.e., cats and dogs).

_~8_ As used herein, the term treat, treated, or treating when used with respect to an 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.
In the instances when the CpG oligonucleotide is administered with an to 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 iTa vivo. Methods for the active exposure of a subject to an antigen are well-known in the art. In general, an antigen is administered directly to the subject by any means such as intravenous, intramuscular, 15 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 nucleic acid 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 20 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 nucleic acid. For instance, in a subject at risk of developing a cancer or an infectious 25 disease or an allergic or asthmatic response, the subject may be administered the CpG
immunostimulatory nucleic acid 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 nucleic acid may be administered to travelers before they travel to foreign lands where they are at risk of exposure to infectious agents.
30 Likewise the CpG immunostimulatory nucleic acid 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 non-peptide 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 to are not limited to cancer antigens, microbial antigens, and allergens.
In methods of the invention, the CpG immunostimulatory nucleic acids 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 2o 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.
Delivery vehicles or delivery devices for delivering antigen and oligonucleotides to surfaces have been described. The CpG immunostimulatory nucleic acid 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; Emulsomes;
ISCOMs; Liposomes; Live bacterial vectors (e.g., Salrrzonella, Esclaerichia coli, Bacillus calrnatte-guerin, Shigella, Lactobacillus); Live viral vectors (e.g., Vaccinia, adenovirus, Herpes Simplex); Microspheres; Nucleic acid vaccines; Polymers (e.g.
carboxymethylcellulose, chitosan); Polymer rings; Proteosomes; Sodium Fluoride;

Transgenic plants; Virosomes; Virus-like particles. Other delivery vehicles are known in the art and some additional examples are provided herein.
The term effective amount of a CpG immunostimulatory nucleic acid refers to the amount necessary or sufficient to realize a desired biologic effect. For example, an effective amount of a CpG immunostimulatory nucleic acid 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.
to 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.
15 The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular CpG
immunostimulatory nucleic acid 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 nucleic acid 20 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 qg to 10 mg per administration, which depending on the application could be given daily, weekly, or monthly and any other amount of 25 time therebetween. More typically mucosal or local doses range from about 10 ~,g to mg per administration, and most typically from about 100 ~g to 1 mg, with 2 -administrations being spaced days or weeks apart. More typically, immune stimulant doses range from 1 ~.g to 10 mg per administration, and most typically 10~.g to 1 mg, with daily or weekly administrations. Subject doses of the compounds described 3o 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 nucleic acids are administered in combination with other therapeutic agents or in specialized delivery vehicles 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. More typically parenteral doses for these purposes range from about 10 p,g to 5 mg per administration, and most typically from about 100 p,g to 1 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 CpG oligonucleotides which have been tested in humans (human clinical trials have been initiated) 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 nucleic acid can be administered to a subject by any mode that delivers the oligonucleotide to the desired surface, e.g., 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 nucleic acids, antigens and other therapeutic agents) can be formulated readily by combining the active compounds) 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 2o sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers, i.e. EDTA for neutralizing internal acid conditions or may be administered without any carriers.
Also specifically contemplated are oral dosage forms of the above component or components. The component or components may be chemically modiEed so that oral delivery of the derivative is efftcacious. Generally, the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of proteolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the component or components and increase in circulation time in the body.
Examples of 3o such moieties include: polyethylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. Abuchowski and Davis, 1981, "Soluble Polymer-Enzyme Adducts" In: En~rnes as Drugs, Hocenberg and Roberts, eds., Wiley-Interscience, New York, NY, pp. 367-383; Newmark, et al., 1982, J. Appl. Biochem. 4:185-189.
Other polymers that could be used are poly-1,3-dioxolane and poly-1,3,6-tioxocane.
Preferred for pharmaceutical usage, as indicated above, are polyethylene glycol moieties.
For the component (or derivative) the location of release may be the stomach, the small intestine (the duodenum, the jejunum, or the ileum), or the large intestine. One skilled in the art has available formulations which will not dissolve in the stomach, yet will release the material in the duodenum or elsewhere in the intestine.
Preferably, the release will avoid the deleterious effects of the stomach environment, either by protection of the oligonucleotide (or derivative) or by release of the biologically active material beyond the. stomach environment, such as in the intestine.
To ensure full gastric resistance a coating impermeable to at least pH 5.0 is essential. Examples of the more common inert ingredients that are used as enteric coatings are cellulose acetate trimellitate (CAT), hydroxypropylmethylcellulose phthalate (HPMCP), HPMCP 50, HPMCP 55, polyvinyl acetate phthalate (PVAP), Eudragit L30D, Aquateric, cellulose acetate phthalate (CAP), Eudragit L, Eudragit S, and Shellac. These coatings may be used as mixed films.
A coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach. This can include sugar coatings, or coatings which make the tablet easier to swallow. Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic i.e. powder; for liquid forms, a soft gelatin shell may be used. The shell material of cachets could be thick starch or other edible paper.
For pills, lozenges, molded tablets or tablet triturates, moist massing techniques can be used.
The therapeutic can be included in the formulation as fine multi-particulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets. The therapeutic could be prepared by compression.

Colorants and flavoring agents may all be included. For example, the oligonucleotide (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.
One may dilute or increase the volume of the therapeutic with an inert material.
These diluents could include carbohydrates, especially mannitol, a-lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch. Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride. Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell.
Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used. Another form of the disintegrants are the insoluble cationic exchange resins.
Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants.
Binders may be used to hold the therapeutic agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxyrnethyl cellulose (CMC). Polyvinyl pyrrolidone (PVP) and hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.
~ An anti-frictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to;
stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes. Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.
To aid dissolution of the therapeutic into the aqueous environment a surfactant might be added as a wetting agent. Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
Cationic detergents might be used and could include benzalkonium chloride or to benzethomium chloride. The list of potential non-ionic detergents that could be included in the formulation as surfactants are lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. These surfactants could be present in the formulation of the 15 oligonucleotide or derivative either alone or as a mixture in different ratios.
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-ftt capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or 20 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 25 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.
For administration by inhalation, the compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray 3o 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. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
Also contemplated herein is pulmonary delivery of the oligonucleotides (or derivatives thereof). The oligonucleotide (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood to stream. Other reports of inhaled molecules include Adjei et al., 1990, Pharmaceutical Research, 7:565-569; Adjei et al., 1990, International Journal of Pharmaceutics, 63:135-144 (leuprolide acetate); Braquet et al., 1989, Journal of Cardiovascular Pharmacology, 13(suppl. 5):143-146 (endothelin-1); Hubbard et al., 1989, Annals of Internal Medicine, Vol. III, pp. 206-212 (al-antitrypsin); Smith et al., 1989, J. Clin.
15 Invest. 84:1145-1146 (a-1-proteinase); Oswein et al., 1990, "Aerosolization of Proteins", Proceedings of Symposium on Respiratory Drug Delivery II, Keystone, Colorado, March, (recombinant human growth hormone); Debs et al., 1988, J. Immunol.
140:3482-3488 (interferon-g and tumor necrosis factor alpha) and Platz et al., U.S.
Patent No. 5,284,656 (granulocyte colony stimulating factor). A method and 2o composition for pulmonary delivery of drugs for systemic effect is described in U.S.
Patent No. 5,451,569, issued September 19, 1995 to Wong et al.
Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which 25 are familiar to those skilled in the art.
Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent0 nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Missouri; the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colorado; the Ventolin~ metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, North Carolina; and the Spinhaler~ powder inhaler, manufactured by Fisons Corp., Bedford, Massachusetts.
All such devices require the use of formulations suitable for the dispensing of oligonucleotide (or derivative). Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy.
Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of .
carriers is contemplated. Chemically modified oligonucleotide may also be prepared in different formulations depending on the type of chemical modification or the type of to device employed.
Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise oligonucleotide (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active oligonucleotide per mL of solution. The formulation may also include a buffer and a simple sugar (e.g., for oligonucleotide 15 stabilization and regulation of osmotic pressure). The nebulizer formulation may also contain a surfactant, to .reduce or prevent surface induced aggregation of the oligonucleotide caused by atomization of the solution in forming the aerosol.
Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the oligonucleotide (or derivative) suspended in a 20 propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin.
25 Oleic acid may also be useful as a surfactant.
Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing oligonucleotide (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.
3o The oligonucleotide (or derivative) should most advantageously be prepared in particulate form with an average particle size of less than 10 mm (or microns), most preferably 0.5 to 5 mm, for most effective delivery to the distal lung.
Nasal delivery of a pharmaceutical composition of the present invention is also contemplated. Nasal delivery allows the passage of a pharmaceutical composition of the present invention to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung. Formulations for nasal delivery include those with dextran or cyclodextran.
For nasal administration, a useful device is a small, hard bottle to which a metered dose sprayer is attached. In one embodiment, the metered dose is delivered by drawing the pharmaceutical composition of the present invention solution into a chamber of defined volume, which chamber has an aperture dimensioned to aerosolize and aerosol formulation by forming a spray when a liquid in the chamber is compressed. The chamber is compressed to administer the pharmaceutical composition of the present invention. In a specific embodiment, the chamber is a piston arrangement. Such devices are commercially available.
Alternatively, a plastic squeeze bottle with an aperture or opening dimensioned to aerosolize an aerosol formulation by forming a spray when squeezed is used. The opening is usually found in the top of the bottle, and the top is generally 2o tapered to partially fit in the nasal passages for efficient administration of the aerosol formulation. Preferably, the nasal inhaler will provide a metered amount of the aerosol formulation, for administration of a measured dose of the drug.
The compounds, when it is desirable to deliver them systemically, 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 mufti-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 to 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 15 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 20 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.
25 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, 30 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 nucleic acids and optionally other therapeutics and/or antigens may be administered per se (neat) or in the form of a to 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, malefic, acetic, salicylic, p-toluene 15 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 2o and a salt (1-3% 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 25 of a CpG immunostimulatory nucleic acid and optionally antigens and/or other therapeutic agents optionally included in a pharmaceutically-acceptable Garner. 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 Garner denotes an organic or 3o 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.
In some embodiments, an immunostimulatory oligonucleotide of the invention can be linked to one or more lipophilic groups (L).
A lipophilic group L is preferably a cholesteryl or modified cholesteryl residue. The cholesterol moiety may be reduced (e.g. as in cholestan) or may be substituted (e.g. by halogen). A combination of different lipophilic groups in one molecule is also possible. Other lipophilic groups include but are not limited to bile acids, cholic acid or taurocholic acid, deoxycholate, oleyl litocholic acid, oleoyl cholenic acid, glycolipids, phospholipids, sphingolipids, isoprenoids, such as steroids, vitamins, such as vitamin E, fatty acids either saturated or unsaturated, fatty acid esters, such as triglycerides, pyrenes, porphyrines, Texaphyrine, adamantine, acridines, biotin, coumarin, fluorescein, rhodamine, Texas-Red, digoxygenin, dimethoxytrityl, t-butyldimethylsilyl, t-butyldiphenylsilyl, cyanine dyes (e.g. Cy3 or Cy5), Hoechst 33258 dye, psoralen, or ibuprofen.
In some embodiments, L is preferably at or near the 3' end of an oligonucleotide. L may be connected to the oligonucleotide by a linker moiety.
Optionally the linker moiety is a non-nucleotidic linker moiety. Non-nucleotidic linkers are e.g. abasic residues (dSpacer), oligoethyleneglycol, such as triethyleneglycol (spacer 9) or hexaethylenegylcol (spacer 18), or alkane-diol, such as butanediol. The spacer units are preferably linked by phosphodiester or phosphorothioate bonds. The linker units may appear just once in the molecule or may be incorporated several times, e.g. via phosphodiester, phosphorothioate, methylphosphonate, or amide linkages.
The lipophilic group L may be attached at various positions of an oligonucleotide.
In some embodiments, the lipophilic group L is linked to the 3'-end of the oligonucleotide, where it also serves the purpose to enhance the stability of the oligomer against 3'-exonucleases. Alternatively, it rnay be linked to an internal 3o nucleotide or a nucleotide on a branch. The lipophilic group L may be attached to a 2'-position of the nucleotide. The lipophilic group L may also be linked to the heterocyclic base of the nucleotide.
. 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 ODN were purchased from Biospring (Frankfurt, Germany), controlled for identity and purity by Coley Pharmaceutical Group (Langenfeld, Germany) and had undetectable endotoxin levels (<O.lEU/ml) measured by the Limulus assay (BioWhittaker, Verviers, Belgium). ODN were suspended in sterile, endotoxin-free Tris-EDTA (Sigma, Deisenhofen, Germany), and stored and handled under aseptic conditions to prevent both microbial and endotoxin contamination. All dilutions were carried out using pyrogen-free phosphate-buffered saline (Life Technologies, Eggenstein, Germany).
The following table shows the sequences of the oligonucleotides (shown 5' to 3') used in the following experiments (* is a phosphorothioate, and - is a phosphodiester or phosphodiester like).
SEQID N0:1 T*C*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQ ID NO: 2 T*C*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T5'-TC + CpG
3~

SEQ ID NO: 3 T*C*T*T*T*T*T*T*T*T*T*T*T*C*G*T*T5'-TC + CpG
3~

SEQ ID NO: 4 T*C*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T5'-TC + CpG
3' SEQ ID NO: 5 T*C*T*T*T*T*T*T*T*T*T*G*T*C*G*T*T5~-TC + CpG
3' SEQ ID NO: 6 T*C*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T*T*T*T*T*T 5'-TC + CpG 3' ' SEQ (D NO: 7 T*C*T*T*T*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T*T*T 5'-TC + CpG 3' SEQ ID NO: 8 T*C*T*T*T*T*T*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T 5'-TC + CpG 3' SEQ ID NO: 9 T*C*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T*T*T*T*T*T 5'-TC + CpG 3' SEQ ID NO: 10 T*C*T*T*T*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T*T*T 5'-TC + CpG 3' SEQ ID NO: 11 T*C*T*T*T*T*T*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T 5'-TC + CpG 3' SEQ ID NO: 12 T*C*T*T*T*T*T*T T*T*C*G*T*T*T*T*T 5'-TC + CpG 3' TTCG w/ PO bond SEQ ID NO: 13 T*C*T*T*T*T*T*T T*T*C*G*T*T*T*T*T*T*T*T*T*T 5'-TC + CpG 3' TTCG
w/ PO bond SEQ ID NO: 14 T*C*T*T*T*T T*T*C*G*T*T*T*T*T*T*T*T*T*T*T*T 5'-TC + CpG 3' TTCG
w/ PO bond SEQID N0:15 T*C*C*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T
SEQID N0:16G*C*C*A*G*G*A*C*T*T*C*T*C*T*C*A*G*G*T*T5'-GC

SEQID NO:17T*C*C*A*T*T*A*C*T*T*C*T*C*T*C*A*T*T*T*TGGtO TT

SEQID NO:18T*C*C*A*G*G*A*T*C*T*C*T*C*T*C*A*G*G*T*TCTtO TC

SEQID N0:19T*C*C*A*G*G*A*C*T*T*G*T*G*T*G*A*G*G*T*TTCtO TG

SEQ ID G*C*C*A*G*G*A*C*A*C*C*T*C*A*C*A*G*G*A*T5'-GC and NO: 20 T to A

SEQ ID T*C*T*T*T*T*T*T*C*T*T*T*C*T*T*T*TTC ODN
NO: 21 SEQID NO:22T*C*T*T*C*T*T*T*T*T*T*T*T*T*T*T*TTC ODN

SEQID NO:23T*C*T*T*T*T*T*C*T*T*C*T*C*T*C*T*T*T*T*T

SEQID NO:24 T*C*T*T*T*T*T*T*G*T*C G*T*T*T*T*T*T*T*T*T*T
SEQID NO:25 T*C T*T*T*T*T*T*G*T*C*G*T*T*T*T*T*T*T*T*T*T
SEQID NO:26 T*C T*T*T*T*T*T*G*T*C G*T*T*T*T*T*T*T*T*T*T
T*C*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQ ID NO: 27 *T*T 24mer SEQID NO:28 T*A*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T 5'-TA

SEQ NO: T*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T5'-TG

SEQIDN0:30T*Z*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T5'-TZ

SEQ NO: U*C*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T5'- UC

SEQ NO: 5T*C*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T5T: 5-Methoxythymidine SEQ NO: T*5H*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T5H: 5-Hydroxy-deoxycytidine SEQ ID T*C*G*A*A*A*A*A*A*A*A*A*A*T*A*A*Apoly A + 5' TCG increasing NO: 34 T amount SEQ ID T*C*G*A*A*A*A*A*A*A*A*A*T*T*A*A*Apoly A + 5' TCG increasing NO: 35 T amount SEQ ID T*C*G*A*A*A*A*A*A*A*T*T*T*T*A*A*Apoly A + 5' TCG increasing NO: 36 T amount SEQ ID T*C*G*A*A*A*A*A*T*T*T*T*T*T*A*A*Apoly A + 5' TCG increasing NO: 37 T amount SEQ ID T*C*G*A*A*A*T*T*T*T*T*T*T*T*T*T*Apoly A + 5' TCG increasing NO: 38 T amount SEQ ID T*C*G*T*A*A*A*A*A*A*A*A*A*A*A*A*Apoly A + 5' TCG increasing NO: 39 T amount SEQ ID T*C*G*T*T*T*A*A*A*A*A*A*A*A*A*A*Apoly A + 5' TCG increasing NO: 40 T amount SEQ ID T*C*G*A*A*A*A*A*A*A*A*A*A*A*A*A*Apoly A + TCG 5' NO: 41 SEQ NO: T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T 1X 5' + poly SEQIDN0:43T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*T

SEQ NO: T*T*T*T*T*T*T*T*T*T*T*T*T*T*T*C*G poly TCG 3' +

SEQ NO: T*T*T*C*G*T*T*T*T*T*T*T*T*T*T*T*T poly CG VarIOUS
ID 45 T pOSItIOrIS
+

SEQ NO: T*T*T*T*T*T*C*G*T*T*T*T*T*T*T*T*T poly CG V8rI0US
ID 46 T pOSitIOflS
+

SEQ NO: T*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T*T poly CG VarIOUS
ID 47 T pOSItIOrIS
+

SEQ NO: T*T*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T CG
ID 48 Shift SEQ NO: T*T*T*T*C*G*T*T*T*T*T*T*T*T*T*T*T CG
ID 49 shift SEQ NO: T*T*T*T*T*C*G*T*T*T*T*T*T*T*T*T*T poly CG ODN 5XT
ID 50 T 5' +

T*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T*T*T*T*T*T*T

SEQ NO: *T 24mer SEQIDNO:52T*T*T*T*T*T*T*T*T*Z*G*T*T*T*T*T*T ZpG

SEQID N0:53 A*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T*T 5~A
T*C*G*T*C*G*T*T*T*T*G*T*C*G*T*T*T*T*G*T*C*G
SEQ ID NO: 54 *T*T
SEQID N0:55 T*C*G*C*C*C*C*C*C*C*C*C*C*C*C*C*C
SEQID NO:56 A*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQID NO:57 C*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQID NO:58 G*C*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQID N0:59 T*T*G*T*T*T*T*T*T*T*T*T*T*T*T*T*T
SEQID N0:60 T*C*G*T*C*G*T*T*T*T*C*G*G*C*G*C*G*C*G*C*C*G
SEQID NO:61 T*C*C*A*T*G*A*C*G*T*T*C*C*T*G*A*C*G*T*T
TLR9 assays:
Stably transfected HEK293 cells expressing the human or mouse TLR9 were described before. Briefly, HEI~293 cells were transfected by electroporation with vectors expressing the human or mouse TLR9 and a 6xNFxB-luciferase reporter plasmid. Stable transfectants (3x104 cells/well) were incubated with ODN for 16h at 37°C in a humidified incubator. Each data point was done in triplicate.
Cells were lysed and assayed for luciferase gene activity (using the BriteLite kit from Perkin-Elmer, Zaventem, Belgium). Stimulation indices were calculated in reference to to reporter gene activity of medium without addition of ODN.
Cell purification:
Peripheral blood buffy coat preparations from healthy human donors were obtained from the Blood Bank of the University of Dusseldorf (Germany) and PBMC
were purified by centrifugation over Ficoll-Hypaque (Sigma). Cells were cultured in a humidified incubator at 37°C in RPMI 1640 medium supplemented with 5%
(v/v) heat inactivated human AB serum (BioWhittaker) or 10% (v/v) heat inactivated FCS, 2mM L-glutamine, 100U/ml penicillin and 100~.glml streptomycin (all from Sigma).

Cytokine detection:
PBMC were resuspended at a concentration of 3x106 cells/ml and added to 48 well flat-bottomed plates (lml/well) or 96 well round-bottomed plates (250~.1/well).
PBMC were incubated with various ODN concentrations and culture supernatants (SN) were collected after the indicated time points. If not used immediately, SN were frozen at -20°C until required.
Amounts of cytokines in the SN were assessed using commercially available ELISA
Kits (IL-6, IP-10, IFN-y or IL-10; from Diaclone, Besan~on, France) or an in-house ELISA for IFN-a developed using commercially available antibodies (from PBL, to New Brunswick, NJ, USA for detection of multiple IFN-a species).
Isolation of human B cells:
Human B cells were isolated from whole PBMC with the CD19 B cell isolation kit as described by the manufacturer (Miltenyi, Bergisch-Gladbach, Germany). To determine purity cells were stained with mAb to CD20 and CD14 and 15 cells identified by flow cytometry. In all experiments B cells were more than 95%
pure. Purified B cells (2x105 to 5x105 cells/ml) were incubated with increasing concentrations of ODN for 24h and IL-6 or IL-10 measured as described above.
Example 1:
20 By shifting the immunostimulatory CpG dinucleotide in a phosphorothioate ODN from the 5' end to the 3' end, a graded decrease of IFN-a production was observed while retaining IL-10 stimulation. Human PBMC were incubated with increasing concentrations of the indicated ODN for 48h. SN were harvested and IFN-a (A) and IL-10 (B) measured by ELISA. Figure 1 shows the Mean~SEM of three 25 donors for each experimental condition.
The data demonstrate that although the production of IFN-a decreases with ODNs containing a CpG dinucleotide shifted toward the 3' end, the level of IL-secretion remains relatively constant. Therefore, a 5' CpG location causes IFN-a production. Shifting the CpG dinucleotide to the 3' end does not result in loss of immune stimulation, only in loss of efficient IFN-a secretion.
Example 2:
Human PBMC were incubated with increasing concentrations of the indicated ODN for 48h. SN were harvested and IL-10 measured by ELISA. Figure 2 shows the Mean~SEM of three donors for each experimental condition.
The data demonstrate that for ODNs with a 3' shifted CpG dinucleotide, the cytosine has to be 5-unmethylated for efficient IL-10 induction. In addition, increasing the length of the ODN appears to result in enhanced IL-10 stimulation (SEQ ID NO: 51).
Example 3:
The T content of an ODN determines its immune stimulatory activity. Human PBMC were incubated with the indicated concentrations of ODN with decreasing T
content for 48h. SN were 'harvested and IL-10 measured by ELISA. Figure 3 shows the Mean~SEM of three donors for each experimental condition.
The data demonstrate that the content of thymidine nucleobases in a phosphorothioate ODN determines its capacity to induce IL-10 production. An ODN
2o with a 5'-TCG and an increasing number of adenosine nucleotides looses its capacity to efficiently stimulate IL-10 production. Therefore, a certain thymidine content is required for efficient IL-10 production.
Example 4:
A 5'-TCG is required for efficient IFN-a production, whereas a 5'-TC is sufficient for potent IL-10 secretion. Human PBMC were incubated with increasing concentrations of the indicated ODN for 48h. SN were harvested and IFN-a (A) and IL-10 (B) measured by ELISA. Figure 4 shows the Mean~SEM of three donors for each experimental condition.
The data demonstrate that a 5'-TCG in a phosphorothioate ODN is required to induce efficient IFN-a secretion. All other 5' trinucleotides (5'-ACG, CCG or GCG) do not appear to have an effect on type I interferon secretion. In addition, exchange of the 5'-CG to 5'-TG or 5'-CT (from 5'-TCG to 5'-TTG or 5'-TCT) also results in a strong decrease of IFN-a production (shown in A). In contrast to IFN-a production, the secretion of IL-10 is efficiently induced by ODN with a 5'-TC lacking a 5'-CG (as shown by SEQ ID NO: 1) (shown in B). This ODN appears to be more potent for l0 inducing IL-10 secretion than an ODN with a 5'-TTG (as shown by SEQ ID NO:
59).
Therefore, ODNs that do not contain a 5'-TCG, but contain a 5'TC, efficiently induce IL-10 production from human PBMC.
Example 5:
The thymidine of the 5'-TC can be chemically modified. No nucleobases other than cytosine or modifications thereof are effective in the 5'-TC
dinucleotide.
Human PBMC were incubated with increasing concentrations of the indicated ODN
for 48h. SN were harvested and IL-10 measured by ELISA. Figure 5 Shows the Mean~SEM of three donors for each experimental condition.
The data demonstrate that introducing a cytosine (as in SEQ ID NO: 1) or a modified cytosine (as in SEQ ID NO: 30: S-methyl-cytosine, and SEQ ID NO: 33:

hydroxy-deoxycytidine) in a thymidine-rich ODN (poly-T SEQ TD NO: 43) results in increased IL-10 amounts. This result cannot be reproduced using other bases such as guanosine or adenosine (as in SEQ ID NO: 29 or SEQ ID NO: 28). ODN with a 5'-TC, 5'-UC (U: uracile), 5'-5TC (ST: 5-methoxythymidine) all appear to have similar activities. Therefore, a cytosine or cytosine analogue is required for efEcient IL-10 production.

Example 6:
ODN with a 5'-TC as well as a 3' shifted CpG both induce stronger IL-10 production relative to their respective ODN sequences lacking a 5'-TC or CpG.
Human PBMC were incubated with increasing concentrations of the indicated ODN
for 48h. SN were harvested and IL-10 measured by ELISA. Figure 6 shows the Mean~SEM of three donors for each experimental condition.
Example 7:
ODN with a 5'-TC as well as a 3' shifted CpG dinucleotide induce strong secretion of IL-6 or IL-10 but show inefficient stimulation of Thl cytokines or chemokines such as IFN-a or IP-10. Human PBMC were incubated with increasing concentrations of the indicated ODN for 48h. SN were harvested and IL-10 (A), IFN-a (B), IP-10 (C) or IL-6 (D) measured by ELISA. Figure 7 shows the Mean~SEM of two (B) or three donors (A, C and D).
The data demonstrate that combining a 5'-TC with a central CpG dinucleotide results in ODN with potent and efficient stimulation of a variety of cytokines such as IL-6 or IL-10. In contrast, these ODNs result in weak IFN-a and IP-10 secretion compared to the B-Class ODN SEQ ID NO: 54 and C-Class ODN SEQ ID NO: 60.
These ODNs are referred to as T-Class ODNs.
Example 8:
T-Class ODNs efficiently induce the production of IL-6 and IL-10 from highly purifted human B cells. B cells were isolated from human PBMC and cultured with the indicated ODN for 24h. SN were harvested and IL-6 (A) or IL-10 (B) measured by ELISA. Figure 8 shows the Mean~SEM of two donors for each experimental condition.
The data demonstrate that the source of IL-10 or IL-6 produced upon culture of human PBMC with T-Class ODNs are B cells. Therefore, this appears to be a direct effect. Indeed, IL-10 secreting B cells were previously demonstrated to play an important role as IL-10 producers and, therefore, in Th2-biased immune responses or the induction of regulatory T or B cells.
Examule 9:
Cells expressing the human TLR9 and an NF~cB-Luciferase reporter are stimulated by T-Class ODN. Transfectants expressing the human TLR9 are incubated for 16h with the indicated ODN concentrations. Cells were lysed and NFxB
stimulation was measured through luciferase activity. The results are shown in Figure 9. Stimulation indices were calculated in reference to luciferase activity of medium without addition of ODN (fold induction of luciferase activity).
The data demonstrate that reconstitution of TLR9 expression in a non-expressing cell leads to the ability to mediate NFxB stimulation upon incubation with T-Class ODN. Therefore, the data strongly suggest that T-Class ODN stimulate the immune system via TLR9.
Example 10:
TLR9-mediated NFkB activation was measured in cells transfected with murine or human TLR9. Figure 10 shows the results for human cells in panel A
and murine cells in panel B. A surprisingly strong dependency on the position of the CpG
dinucleotide was observed in the murine TLR9 transfectants relative to the human TLR9 transfectants with this class of ODN (T-Class). In these experiments, murine TLR9 did not show a significant NFkB signaling response to ODN with CpG at positions 14 (cytosine) and 15 (guanosine) or further to the 3' end (B). In contrast, human TLR9 transfectants responded strongly to ODN with CpG at positions 14 and 15 (A). In addition, in these experiments, the T-Class ODN resulted in a more powerful stimulation in human than in murine TLR9 transfectants.
Having thus described several aspects of at least one embodiment of this invention, it is to be appreciated various alterations, modifications, and improvements will readily occur to those skilled in the art. Such alterations, modifications, and improvements are intended to be part of this disclosure, and are intended to be within the spirit and scope of the invention. Accordingly, the foregoing description and drawings are by way of example only.
The disclosures of all of the patents, patent applications, scientific publications, and other references are incorporated herein by reference in their entirety.

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPRI~:ND PLUS D'UN TOME.
CECI EST L,E TOME 1 DE 2 NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional valumes please contact the Canadian Patent Office.

Claims (53)

1. An oligonucleotide chosen from:

a) 5' XYN1Y2N2 3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X is a T or modified T nucleotide, wherein Y is a C or modified C nucleotide, wherein Z is a G or modified G nucleotide, wherein N1 and N2 are polynucleotides that do not include a CG dinucleotide, wherein N1 does not include 5' Z nucleotide, and wherein a 3' polynucleotide consisting of the YZ
dinucleotide and the N2 polynucleotide contains a number of nucleotides that is at most 45% of the number of nucleotides in the oligonucleotide; and b) 5' XY N1YZ N2 3' wherein 5' designates the 5' end of the oligonucleotide and 3' designates the 3' end of the oligonucleotide, wherein X is a T or modified T nucleotide, wherein Y is a C or modified C nucleotide, wherein Z is a G or modified G nucleotide, wherein N1 is a polynucleotide of 5 to 10 nucleotides, wherein N1 does not include a CG
dinucleotide, wherein N1 does not include 5' Z nucleotide, and wherein N2 is a polynucleotide of 5 to 30 nucleotides.
2. The oligonucleotide of claim 1, wherein the oligonucleotide includes at least 1 modified internucleotide linkage.
3. The oligonucleotide of claim 1, wherein the oligonucleotide includes at least 50% modified internucleotide linkage.
4. The oligonucleotide of claim 1, wherein all internucleotide linkages of the oligonucleotide are modified.
5. The oligonucleotide of claim 1, wherein the oligonucleotide consists of 10 to 100 nucleotides.
6. The oligonucleotide of claim 2, wherein the modified internucleotide linkage is a phosphorothioate linkage.
7. The oligonucleotide of claim 2, comprising a phosphodiester linkage between a 5' C nucleotide and a 3' G nucleotide.
8. The oligonucleotide of claim 2, comprising a R-phosphorothioate linkage between a 5' C nucleotide and a 3' G nucleotide.
9. The oligonucleotide of claim 1, wherein Y is a modified C nucleotide comprising a modified cytosine base selected from the group consisting of 5-substituted cytosines, 6-substituted cytosines, N4-substituted cytosines, cytosine analogs with condensed ring systems, uracil, uracil derivatives, a universal base, an aromatic ring system, and a hydrogen atom.
10. The oligonucleotide of claim 9, wherein Y is a modified C nucleotide comprising a modified cytosine base selected from the group consisting of 5-methyl-cytosine, 5-fluoro-cytosine, 5-chloro-cytosine, 5-bromo-cytosine, 5-iodo-cytosine, 5-hydroxy-cytosine, 5-hydroxymethyl-cytosine, 5-difluoromethyl-cytosine, unsubstituted or substituted 5-alkynyl-cytosine, N4-ethyl-cytosine, 5-aza-cytosine, 2-mercapto-cytosine, isocytosine, pseudo-isocytosine, N,N'-propylene cytosine or phenoxazine, 5-fluoro-uracil, 5-bromo-uracil, 5-bromovinyl-uracil, 4-thio-uracil, 5-hydroxy-uracil, 5-propynyl-uracil, 3-nitropyrrole, P-base, fluorobenzene, and difluorobenzene.
11. The oligonucleotide of claim 1, wherein Z is a modified G nucleotide comprising a modified guanine base selected from the group consisting of 7-deazaguanine, 7-deaza-7-substituted guanine, 7-deaza-7-(C2-C6)alkynylguanine, 7-deaza-8-substituted guanine, hypoxanthine, N2-substituted guanines, N2-methyl-guanine, 5-amino-3-methyl-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione, 2,6-diaminopurine, 2-aminopurine, purine, indole, inosine, adenine, substituted adenines, N6-methyl-adenine, 8-oxo-adenine, 8-substituted guanine, 8-hydroxyguanine, 8-bromoguanine, 6-thioguanine, a universal base, 4-methyl-indole, 5-nitro-indole, K-base, an aromatic ring system, benzimidazole, dichloro-benzimidazole, 1-methyl-1H-[1,2,4]triazole-3-carboxylic acid amide, and a hydrogen atom.
12. The oligonucleotide of claim 1, wherein the oligonucleotide comprises a 3'-3' linkage with one or two accessible 5' ends.
13. The oligonucleotide of claim 1, comprising a nucleotide sequence that does not contain an optimal CpG hexameric sequence.
14. The oligonucleotide of claim 1, comprising a nucleotide sequence that does not contain a palindromic sequence.
15. The oligonucleotide of claim 1, wherein the oligonucleotide does not form a stable secondary structure.
16. The oligonucleotide of claim 1, wherein the oligonucleotide is conjugated to a moiety selected from the group consisting of antigens and cytokines.
17. The oligonucleotide of claim 16, wherein the antigen is selected from the group consisting of infectious disease antigens.
18. The oligonucleotide of claim 16, wherein the cytokine is IL-10.
19. The oligonucleotide of claim l, wherein the oligonucleotide has the following structure: 5' T*C*T*T*T*T*T*T*G*T*C*G*T*T*T*T*T 3' (SEQ ID
NO:4) and wherein * refers to a phosphorothioate linkage.
20. The oligonucleotide of claim 1, wherein the oligonucleotide has the following structure: 5' T*T*G*C*G*T*G*C*G*T*T*T*T*G*A*C*G*T*T*T*T*T*T*T3'(SEQ ID
NO:62) and wherein * refers to a phosphorothioate linkage.
21. The oligonucleotide of claim 1, wherein the oligonucleotide has the following structure: 5' T*C*T*T*T*T*T*T*T*T*C*G*T*T*T*T*T 3' (SEQ ID
NO:2) and wherein * refers to a phosphorothioate linkage.
22. The oligonucleotide of claim 1 a) or 1 b), wherein N1 is a poly-T
polynucleotide.
23. The oligonucleotide of claim 1 a) or 1 b), wherein N2 is a poly-T
polynucleotide.
24. The oligonucleotide of claim 1 a) or 1 b), wherein both N1 and N2 are poly-T polynucleotides.
25. The oligonucleotide of any one of claims 22-24, wherein the poly-T
polynucleotide contains one or more modified T nucleotides.
26. The oligonucleotide of any one of claims 22-24, wherein the poly-T
polynucleotide contains between 5 and 20 T nucleotides.
27. The oligonucleotide of any one of claims 22-24, wherein the poly-T
polynucleotide contains between 5 and 10 T nucleotides.
28. The oligonucleotide of any one of claims 22-24, wherein the poly-T
polynucleotide contains more than 20 T nucleotides.
29. The oligonucleotide of claim 1 consisting of at least 55% T nucleotides.
30. A pharmaceutical composition comprising an oligonucleotide of any one of claims 1-32 in combination with a therapeutic agent selected from the group consisting of chemotherapeutic agents, radiotherapeutic agents, monoclonal antibodies, and anticancer agents.
31. A method of specifically increasing IL-10 expression relative to IFN-.alpha.
expression in a subject, the method comprising the step of administering an oligonucleotide or a pharmaceutical composition of any of claims 1-32 to a subject an need of increased IL-10 expression relative to IFN-.alpha. expression.
32. The method of claim 31, wherein the ratio of induced IL-10 to IFN-.alpha.
is higher than a reference ratio of IL-10 to IFN-.alpha..
33. The method of claim 32, wherein the step of administering is selected from the group consisting of respiratory, oral, topical, subcutaneous, and infra-venous administrations.
34. A method of inducing an antigen-specific regulatory T cell response in a subject, the method comprising the step of:
administering an immunostimulatory nucleic acid or composition of any of claims 1-31 to a subject exposed to an antigen.
35. A method of inducing an antigen-specific regulatory B cell response in a subject, the method comprising the step of:
administering an immunostimulatory nucleic acid or composition of any of claims 1-31 to a subject exposed to an antigen.
36. The method of claim 34 or 35, wherein the antigen is administered to the subject along with the immunostimulatory nucleic acid or composition.
37. The method of claim 34 or 35, wherein the antigen is administered to the subject after the immunostimulatory nucleic acid or composition.
38. The method of claim 34 or 35, wherein the antigen is present in a food and the subject is exposed to the antigen by ingesting the food.
39. The method of claim 34 or 35, wherein the antigen is inhaled by the subject.
40. A method of treating an allergy or asthma, the method comprising the steps of:
exposing a subject to an allergen; and administering an immunostimulatory nucleic acid or composition of any one of claims 1-31 to the subject, wherein the immunostimulatory nucleic acid or composition is administered in an amount sufficient to prevent or alleviate an allergic response to the allergen in the subject.
41. The method of claim 40, further comprising the step of administering IL-to the subject.
42. A method of treating an autoimmune disease in a subject, the method comprising the steps of:
exposing a subject to a self antigen; and administering an immunostimulatory nucleic acid or composition of any one of claims 1-31 to the subject, wherein the immunostimulatory nucleic acid or composition is administered in an amount sufficient to prevent or treat an autoimmune disease in the subject.
43. The method of claim 42, further comprising the step of administering IL-10 to the subject.
44. A method of reducing an antigen-specific response to an implant in a subject, the method comprising the steps of:
exposing a subject to an implant antigen; and administering an immunostimulatory nucleic acid or composition of any one of claims 1-31 to the subject, wherein the immunostimulatory nucleic acid or composition is administered in an amount sufficient to prevent or reduce an antigen-specific response to the implant in the subject.
45. The method of claim 44, further comprising the step of administering IL-to the subject.
46. The method of claim 40, wherein the subject has or is at risk of developing allergic asthma.
47. The method of claim 42, wherein the autoimmune disease is chosen from arthritis, systemic lupus erythematosus, multiple sclerosis, Crohn's disease, Type 1 diabetes mellitus, Multiple sclerosis, Myasthenia gravis, Autoimmune neuropathies, such as Guillain-Barré, Autoimmune uveitis, Autoimmune hemolytic anemia, Pernicious anemia, Autoimmune thrombocytopenia, Temporal arteritis, Anti-phospholipid syndrome, Psoriasis, Pemphigus vulgaris, Vasculitides such as Wegener's granulomatosis, Vitiligo, Crohn's Disease, Ulcerative colitis, Primary biliary cirrhosis, Autoimmune hepatitis, Type 1 or immune-mediated, diabetes mellitus, Grave's Disease, Hashimoto's thyroiditis, Autoimmune oophoritis and orchitis, Autoimmune disease of the adrenal gland, Rheumatoid arthritis, Systemic lupus, erythematosus, Scleroderma, Polymyositis, dermatomyositis, Spondyloarthropathies, such as ankylosing spondylitis, and Sjogren's syndrome.
48. The method of claim 42, wherein the autoimmune disease is caused by an infection.
49. The method of claim 48, wherein the infection is Lyme disease.
50. The method of claim 44, wherein the implant is an autologous tissue implant.
51. The method of claim 44, wherein the implant is a non-autologous tissue implant.
52. The method of claim 44, wherein the implant is a recombinant cellular implant.
53. The method of claim 44, wherein the implant is a synthetic implant.
CA002560108A 2004-04-02 2005-04-04 Immunostimulatory nucleic acids for inducing il-10 responses Abandoned CA2560108A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US55895104P 2004-04-02 2004-04-02
US60/558,951 2004-04-02
PCT/US2005/011827 WO2005111057A2 (en) 2004-04-02 2005-04-04 Immunostimulatory nucleic acids for inducing il-10 responses

Publications (1)

Publication Number Publication Date
CA2560108A1 true CA2560108A1 (en) 2005-11-24

Family

ID=35394719

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002560108A Abandoned CA2560108A1 (en) 2004-04-02 2005-04-04 Immunostimulatory nucleic acids for inducing il-10 responses

Country Status (6)

Country Link
US (1) US20060019916A1 (en)
EP (1) EP1730281A2 (en)
JP (1) JP2007531746A (en)
AU (2) AU2005243250A1 (en)
CA (1) CA2560108A1 (en)
WO (1) WO2005111057A2 (en)

Families Citing this family (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6727230B1 (en) * 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
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
US6239116B1 (en) * 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
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
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
AU760549B2 (en) * 1998-04-03 2003-05-15 University Of Iowa Research Foundation, The Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
WO1999058118A2 (en) * 1998-05-14 1999-11-18 Cpg Immunopharmaceuticals Gmbh METHODS FOR REGULATING HEMATOPOIESIS USING CpG-OLIGONUCLEOTIDES
ES2628744T3 (en) 1998-05-22 2017-08-03 Ottawa Hospital Research Institute Methods and products to induce immunity in mucous membranes
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines 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
EP2204186B1 (en) * 1999-02-17 2016-04-06 CSL Limited Immunogenic complexes and methods relating thereto
US6949520B1 (en) * 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US7585847B2 (en) * 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
US20040131628A1 (en) * 2000-03-08 2004-07-08 Bratzler Robert L. Nucleic acids for the treatment of disorders associated with microorganisms
ES2332444T3 (en) * 2000-06-22 2010-02-05 University Of Iowa Research Foundation COMBINATION OF CPG AND ANTIBODIES DIRECTED AGAINST CD19, CD20, CD22 OR CD40 FOR THE TREATMENT OR PREVENTION OF CANCER.
KR100917101B1 (en) * 2000-08-04 2009-09-15 도요 보세키 가부시키가이샤 Flexible metal laminate and production method thereof
WO2002022809A2 (en) * 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
ATE398175T1 (en) * 2000-12-08 2008-07-15 Coley Pharmaceuticals Gmbh CPG-TYPE NUCLEIC ACIDS AND METHODS OF USE THEREOF
ATE411054T1 (en) * 2001-08-17 2008-10-15 Coley Pharm Gmbh COMBINATION MOTIF-IMMUNO-STIMULATING OLIGONUCLEOTIDES WITH IMPROVED EFFECT
EP1499187B1 (en) * 2002-04-04 2015-06-17 Zoetis Belgium S.A. Immunostimulatory g,u-containing oligoribonucleotides
US7605138B2 (en) * 2002-07-03 2009-10-20 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
US7576066B2 (en) * 2002-07-03 2009-08-18 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
US20040053880A1 (en) * 2002-07-03 2004-03-18 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
AR040996A1 (en) * 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
BR0315810A (en) * 2002-10-29 2005-09-13 Coley Pharmaceutical Group Ltd Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
CA2502015A1 (en) * 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 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
US20050013812A1 (en) * 2003-07-14 2005-01-20 Dow Steven W. Vaccines using pattern recognition receptor-ligand:lipid complexes
UA88457C2 (en) * 2003-10-30 2009-10-26 Коли Фармасьютикал Гмбх Immunostimulatory nucleic acid with enhanced immunostimulatory potency
US20050239733A1 (en) * 2003-10-31 2005-10-27 Coley Pharmaceutical Gmbh Sequence requirements for inhibitory oligonucleotides
AU2005230938A1 (en) * 2004-02-19 2005-10-20 Coley Pharmaceutical Gmbh Immunostimulatory viral RNA oligonucleotides
EP2484374A1 (en) * 2004-07-18 2012-08-08 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
WO2006053430A1 (en) * 2004-11-17 2006-05-26 Protiva Biotherapeutics, Inc. Sirna silencing of apolipoprotein b
CA2598992A1 (en) * 2005-02-24 2006-08-31 Coley Pharmaceutical Group, Inc. Immunostimulatory oligonucleotides
KR20080008350A (en) * 2005-04-08 2008-01-23 콜레이 파마시티컬 그룹, 인코포레이티드 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
EA200800268A1 (en) * 2005-07-07 2008-06-30 Коли Фармасьютикал Груп, Инк. COMBINED THERAPY WITH ANTIBODY AGAINST CTLA-4 AND CONTAINING A CpG-MOTIVE SYNTHETIC OLYGESOXYNECLETOSIDE FOR THE TREATMENT OF MALIGNANT TUMOR
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
WO2007088423A2 (en) * 2005-09-16 2007-08-09 Coley Pharmaceutical Gmbh Immunostimulatory single-stranded ribonucleic acid with phosphodiester backbone
BRPI0616069A2 (en) * 2005-09-16 2011-06-07 Coley Pharm Gmbh modulation of immunomodulatory properties of small interfering ribonucleic acid (sirna) by nucleotide modification
EP1948674A4 (en) 2005-11-02 2009-02-04 Protiva Biotherapeutics Inc Modified sirna molecules and uses thereof
BRPI0618857B1 (en) 2005-11-25 2022-07-19 Zoetis Belgium S.A ISOLATED RNA OLIGONUCLEOTIDE, AND METHOD TO NEGATIVELY REGULATE IMMUNOSUPPRESSANT CD4+ REGULATORY CELLS
ES2526879T3 (en) * 2006-02-15 2015-01-16 Adiutide Pharmaceuticals Gmbh Compositions and procedures for oligonucleotide formulations
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
EP2056880A4 (en) * 2006-08-16 2010-10-13 Protiva Biotherapeutics Inc Nucleic acid modulation of toll-like receptor-mediated immune stimulation
KR101251707B1 (en) 2006-09-27 2013-04-11 콜리 파마슈티칼 게엠베하 CpG oligonucleotide analogs containing hydrophobic T analogs with enhanced immunostimulatory activity
WO2008057529A2 (en) * 2006-11-06 2008-05-15 Coley Pharmaceutical Group, Inc. Peptide-based vaccine compositions to endogenous cholesteryl ester transfer protein (cetp)
PL2170353T3 (en) * 2007-05-18 2015-10-30 Adiutide Pharmaceuticals Gmbh Phosphate-modified oligonucleotide analogs with immunostimulatory activity
RU2010112771A (en) * 2007-10-09 2011-11-20 Коули Фармасьютикал ГмбХ (DE) IMMUNITY MULATING ANALOGUES OF OLIGONUCLEOTIDES CONTAINING MODIFIED SUGAR GROUPS
EP2288702A4 (en) * 2008-06-18 2011-06-29 Index Pharmaceuticals Ab Combination therapies against cancer
EP2350282A1 (en) * 2008-11-04 2011-08-03 Index Pharmaceuticals AB Increased expression of specific antigens
KR101856462B1 (en) 2009-03-25 2018-05-10 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Compositions for stimulation of mammalian innate immune resistance to pathogens
WO2010129351A1 (en) * 2009-04-28 2010-11-11 Schepens Eye Research Institute Method to identify and treat age-related macular degeneration
WO2014175333A1 (en) * 2013-04-23 2014-10-30 株式会社プリベンテック External skin preparation for treating and/or preventing psoriasis vulgaris
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
JP6581604B2 (en) 2014-06-04 2019-09-25 イグジキュア, インコーポレーテッドExicure, Inc. Multivalent delivery of immunomodulators with liposomal globular nucleic acids for prophylactic or therapeutic applications
WO2016044839A2 (en) 2014-09-19 2016-03-24 The Board Of Regents Of The University Of Texas System Compositions and methods for treating viral infections through stimulated innate immunity in combination with antiviral compounds
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
TWI739740B (en) * 2015-04-22 2021-09-21 德商奎爾法克股份有限公司 Rna containing composition for treatment of tumor diseases
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5464943A (en) * 1989-04-26 1995-11-07 Takeda Chemical Industries, Ltd. DNA encoding glycosylated FGF and production thereof
US5695926A (en) * 1990-06-11 1997-12-09 Bio Merieux Sandwich hybridization assays using very short capture probes noncovalently bound to a hydrophobic support
AU2738392A (en) * 1991-11-11 1993-05-13 Ciba-Geigy Ag Novel hybrid transforming growth factors
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
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030050263A1 (en) * 1994-07-15 2003-03-13 The University Of Iowa Research Foundation Methods and products for treating HIV infection
AU760549B2 (en) * 1998-04-03 2003-05-15 University Of Iowa Research Foundation, The Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
US6537751B1 (en) * 1998-04-21 2003-03-25 Genset S.A. Biallelic markers for use in constructing a high density disequilibrium map of the human genome
EP1100807A1 (en) * 1998-07-27 2001-05-23 University Of Iowa Research Foundation STEREOISOMERS OF CpG OLIGONUCLEOTIDES AND RELATED METHODS
WO2000021539A1 (en) * 1998-10-09 2000-04-20 Anadys Pharmaceuticals, Inc. Aryldiamine derivatives useful as antibacterial agents
SK287400B6 (en) * 1999-09-25 2010-08-09 University Of Iowa Research Foundation Immunostimulatory nucleic acid composition and the use thereof for stimulating an immune response
US6949520B1 (en) * 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
ES2332444T3 (en) * 2000-06-22 2010-02-05 University Of Iowa Research Foundation COMBINATION OF CPG AND ANTIBODIES DIRECTED AGAINST CD19, CD20, CD22 OR CD40 FOR THE TREATMENT OR PREVENTION OF CANCER.
ATE398175T1 (en) * 2000-12-08 2008-07-15 Coley Pharmaceuticals Gmbh CPG-TYPE NUCLEIC ACIDS AND METHODS OF USE THEREOF
US20030050268A1 (en) * 2001-03-29 2003-03-13 Krieg Arthur M. Immunostimulatory nucleic acid for treatment of non-allergic inflammatory diseases
EP1505942B1 (en) * 2002-05-10 2008-08-13 Tekmira Pharmaceuticals Corporation Pathogen vaccines and methods for using the same
DE10222632B4 (en) * 2002-05-17 2006-03-09 Con Cipio Gmbh Microsatellite markers for genetic analysis and for the differentiation of roses
US7576066B2 (en) * 2002-07-03 2009-08-18 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
US7605138B2 (en) * 2002-07-03 2009-10-20 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
EP1393745A1 (en) * 2002-07-29 2004-03-03 Hybridon, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5'ends
AR040996A1 (en) * 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
BR0315810A (en) * 2002-10-29 2005-09-13 Coley Pharmaceutical Group Ltd Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
CA2502015A1 (en) * 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5' cpg nucleic acids and methods of use
EP1663316A2 (en) * 2003-09-25 2006-06-07 Coley Pharmaceutical Group, Inc. Nucleic acid lipophilic conjugates
MY159370A (en) * 2004-10-20 2016-12-30 Coley Pharm Group Inc Semi-soft-class immunostimulatory oligonucleotides

Also Published As

Publication number Publication date
AU2010202893A1 (en) 2010-07-29
EP1730281A2 (en) 2006-12-13
JP2007531746A (en) 2007-11-08
WO2005111057A2 (en) 2005-11-24
WO2005111057A3 (en) 2006-07-27
US20060019916A1 (en) 2006-01-26
AU2005243250A1 (en) 2005-11-24

Similar Documents

Publication Publication Date Title
US20060019916A1 (en) Immunostimulatory nucleic acids for inducing IL-10 responses
DK1538904T3 (en) IMMUNSTIMULATING NUCLEIC ACIDS
US9382545B2 (en) CpG oligonucleotide analogs containing hydrophobic T analogs with enhanced immunostimulatory activity
US7956043B2 (en) 5′ CpG nucleic acids and methods of use
AU2008309264B2 (en) Immune stimulatory oligonucleotide analogs containing modified sugar moieties
US20060211644A1 (en) Immunostimulatory oligonucleotides
US20060229271A1 (en) Methods for treating infectious disease exacerbated asthma
CA2536139A1 (en) Nucleic acid-lipophilic conjugates

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued

Effective date: 20130404