Immunization-free methods for treating antigen-stimulated inflammation in a mammalian host and shifting the host's antigen immune responsiveness to a Th1 phenotype

Raz, Eyal

Patent Application Summary

U.S. patent application number 10/229208 was filed with the patent office on 2003-05-15 for immunization-free methods for treating antigen-stimulated inflammation in a mammalian host and shifting the host's antigen immune responsiveness to a th1 phenotype. Invention is credited to Raz, Eyal.

Application Number20030092663 10/229208
Document ID /
Family ID25454211
Filed Date2003-05-15

United States Patent Application 20030092663
Kind Code A1
Raz, Eyal May 15, 2003

Immunization-free methods for treating antigen-stimulated inflammation in a mammalian host and shifting the host's antigen immune responsiveness to a Th1 phenotype

Abstract

The invention relates to methods for preventing or reducing antigen-stimulated, granulocytemediated inflammation in tissue of an antigen-sensitized mammal host by delivering an immunostimulatory oligonucleotide to the host. In addition, methods for using the immunostimulatory oligonucleotides to boost a mammal host's immune responsiveness to a sensitizing antigen (without immunization of the host by the antigen) and shifting the host's immune responsiveness to a Th1 phenotype to achieve various therapeutic ends are provided. Kits for practicing the methods of the invention are also provided.


Inventors: Raz, Eyal; (Del Mar, CA)
Correspondence Address:
    BOZICEVIC, FIELD & FRANCIS LLP
    200 MIDDLEFIELD RD
    SUITE 200
    MENLO PARK
    CA
    94025
    US
Family ID: 25454211
Appl. No.: 10/229208
Filed: August 26, 2002

Related U.S. Patent Documents

Application Number Filing Date Patent Number
10229208 Aug 26, 2002
09235742 Jan 21, 1999
6498148
09235742 Jan 21, 1999
08927120 Sep 5, 1997

Current U.S. Class: 514/44R ; 424/178.1; 514/1.7; 514/12.2; 514/18.7; 514/20.9; 514/26
Current CPC Class: A61P 37/02 20180101; A61K 39/35 20130101; A61K 2039/53 20130101; A61P 21/00 20180101; A61K 2039/55561 20130101; A61P 37/08 20180101; A61K 2039/543 20130101; A61P 27/16 20180101; A61P 11/06 20180101; A61P 29/00 20180101; A61P 27/02 20180101
Class at Publication: 514/44 ; 514/8; 424/178.1; 514/26
International Class: A61K 048/00

Goverment Interests



[0001] This invention was made with Government support under Grant No. AI37305, awarded by the National Institutes of Health. The Government may have certain rights in this invention.
Claims



The invention claimed is:

1. A method for preventing or reducing antigen-stimulated, granulocyte-mediated inflammation in a tissue of an antigen-sensitized mammalian host comprising delivering an immunostimulatory oligonucleotide (ISS-ODN) to the host; wherein a reduction in, or the absence of, a Th2 type immune response measured in the host; or a reduction in, or the absence of other clinical signs of inflammation in the host after antigen challenge, indicates that the desired prevention or reduction in granulocyte-mediated inflammation has been achieved.

2. The method according to claim 1 wherein the ISS-ODN includes a hexameric nucleotide sequence consisting of 5'-Purine-Purine-[C]-[G]-Pyri- midine-Pyrimidine-3'.

3. The method according to claim 2 wherein the hexameric nucleotide sequence consists of AACGTT.

4. The method according to claim 2 wherein the hexameric nucleotide sequence is selected from the group of sequences consisting of AGCGTC, GACGTT, GGCGTT, AACGTC, AGCGTC, GACGTC, GGCGTC, AACGCC, AGCGCC, GACGCC, GGCGCC, AGCGCT, GACGCT, GGCGCT, TTCGAA, GGCGTT and AACGCC.

5. The method according to claim 1 wherein the ISS-ODN is conjugated to an immunostimulatory or anti-inflammatory partner selected from the group consisting of non-antigenic polypeptides, antigenic polypeptides, polysaccharides, antibodies, glycoproteins, lipids and steroids.

6. The method according to claim 1 wherein the host is suffering from an inflammatory condition induced by the sensitizing antigen selected from the group of inflammatory conditions consisting of asthma, nasal polyposis, allergic rhinitis, atopic dermatitis, allergic conjunctivitis, eosinophilic fasciitis, ideopathic hypereosinophilic syndrome and cutaneous basophil hypersensitivity.

7. The method according to claim 1 wherein the inflamed tissue is skin or mucosa.

8. The method according to claim 7 wherein the inflamed tissue is respiratory tissue.

9. The method according to claim 8 wherein the host is suffering from asthma.

10. A method for boosting the immune responsiveness of a mammalian host to a sensitizing antigen without immunization of the host by the sensitizing antigen comprising delivering an immunostimulatory oligonucleotide (ISS-ODN) to the host to the host, wherein an increase in the magnitude of the host immune response to the sensitizing antigen indicates that the desired boost to the host immune responsiveness has been achieved.

11. The method according to claim 10 wherein the IS S-ODN includes a hexameric nucleotide sequence consisting of 5'-Purine-Purine-[C]-[G]-Pyri- midine-Pyrimidine-3'.

12. The method according to claim 11 wherein the hexameric nucleotide sequence consists of AACGTT.

13. The method according to claim 1 wherein the hexameric nucleotide sequence is selected from the group of sequences consisting of AGCGTC, GACGTT, GGCGTT, AACGTC, AGCGTC, GACGTC, GGCGTC, AACGCC, AGCGCC, GACGCC, GGCGCC, AGCGCT, GACGCT, GGCGCT, TTCGAA, GGCGTT and AACGCC.

14. The method according to claim 10 wherein the ISS-ODN is conjugated to an immunostimulatory or anti-inflammatory partner selected from the group consisting of non-antigenic polypeptides, antigenic polypeptides, polysaccharides, antibodies, glycoproteins, lipids and steroids.

15. The method according to claim 10 wherein the host is suffering from an inflammatory condition induced by the sensitizing antigen selected from the group of inflammatory conditions consisting of asthma, nasal polyposis, allergic rhinitis, atopic dermatitis, allergic conjunctivitis, eosinophilic fasciitis, ideopathic hypereosinophilic syndrome and cutaneous basophil hypersensitivity.

16. The method according to claim 10 wherein the inflamed tissue is skin or mucosa.

17. The method according to claim 16 wherein the inflamed tissue is respiratory tissue.

18. The method according to claim 17 wherein the host is suffering from asthma and the host's immune responsiveness to a respiratory allergen is boosted.

19. The method according to claim 10 wherein the antigen is presented by a pathogen and the host's immune responsiveness to an intracellular infection by the pathogen is boosted.

20. The method according to claim 19 wherein the pathogen is a virus.

21. A method for shifting the immune response of a mammal host to a sensitizing antigen toward a Th1 phenotype comprising delivering an immunostimulatory oligonucleotide (ISS-ODN) to the host, wherein detection of a Th1 type immune response by the host indicates that the desired shift to the Th1 phenotype has been achieved.

22. The method according to claim 21 wherein the ISS-ODN includes a hexameric nucleotide sequence consisting of 5'-Purine-Purine-[C]-[G]-Pyri- midine-Pyrimidine-3'.

23. The method according to claim 22 wherein the hexameric nucleotide sequence consists of AACGTT.

24. The method according to claim 22 wherein the hexameric nucleotide sequence is selected from the group of sequences consisting of AACGTT, AGCGTC, GACGTT, GGCGTT, AACGTC, AGCGTC, GACGTC, GGCGTC, AACGCC, AGCGCC, GACGCC, GGCGCC, AGCGCT, GACGCT, GGCGCT, TTCGAA, AACGTT, GGCGTT and AACGCC.

25. The method according to claim 21 wherein the ISS-ODN is conjugated to an immunostimulatory or anti-inflammatory partner selected from the group consisting of non-antigenic polypeptides, antigenic polypeptides, polysaccharides, antibodies, glycoproteins, lipids and steroids.

26. The method according to claim 21 wherein the host is suffering from an inflammatory condition induced by the sensitizing antigen selected from the group of inflammatory conditions consisting of asthma, nasal polyposis, allergic rhinitis, atopic dermatitis, allergic conjunctivitis, eosinophilic fasciitis, ideopathic hypereosinophilic syndrome and cutaneous basophil hypersensitivity, and the shift to the Th1 phenotype reduces granulocyte-mediated inflammation in the affected host tissue.

27. The method according to claim 26 wherein the inflamed tissue is skin or mucosa.

28. The method according to claim 27 wherein the inflamed tissue is respiratory tissue.

29. The method according to claim 28 wherein the host is suffering from asthma and the shift to the Th1 phenotype reduces eosinophil infiltration of the host lung tissue.

30. The method according to claim 21 wherein the host is suffering from an intracellular infection by a pathogen and the shift to the Th1 phenotype strengthens the host immune response to the pathogen.

31. The method according to claim 30 wherein the pathogen is a virus.

32. The method according to claim 21 wherein the host is suffering from reduced blood flow to a tissue and the shift to the Th1 phenotype stimulates angiogenesis in the treated tissue.

33. The method according to claim 32 wherein the host is suffering from diabetic retinopathy.

34. The method according to claims 1, 10 or 21 wherein the desired result is measured by determining any of the following values in a sample containing lymphocytes obtained from the ISS-ODN treated host: (1) a reduction in levels of IL-4, IL-5 and/or IL-10 measured before and after antigen challenge or detection of lower levels of IL-4, IL-5 and/or IL-10 in the IS S-ODN treated host as compared to an antigen-challenged control; (2) an increase in levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) before and after antigen challenge or detection of higher levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) in the ISS ODN treated host as compared to an antigen-challenged control; (3) IgG2a antibody production in the ISS-ODN treated host; or (4) a reduction in levels of antigen-specific IgE as measured before and after antigen challenge or detection of lower levels of antigen-specific IgE in the ISS-ODN treated host as compared to an antigen-challenged control.

35. The method according to claims 1 or 10 wherein reduction or suppression of inflammation is measured by assaying inflammatory infiltrate from the host for a reduction in granulocyte counts in inflammatory infiltrate of an affected host tissue as measured in an antigen challenged host before and after ISS-ODN administration or detection of lower levels of granulocyte counts in an ISS-ODN treated host as compared to an antigen-challenged control.

36. A kit for use in-reducing or preventing inflammation in an antigen-sensitized host tissue, as well as in boosting the immune responsiveness of a host to a sensitizing antigen, comprising an immunostimulatory oligonucleotide (ISS-ODN) in a sterile vial, a device for delivering the ISS-ODN directly into a host tissue and at least one assay reagent for use in measuring any of the following values as indicators that the desired reduction or prevention of inflammation or boost in immune responsiveness has been achieved in an ISS-ODN treated host: (1) a reduction in levels of IL-4, IL-5 and/or IL-10 measured before and after antigen challenge; or detection of lower (or absent) levels of IL-4, IL-5 and/or IL-10 in an ISS-ODN treated host as compared to an antigen-primed, or primed and challenged, control; (2) an increase in levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) before and after antigen challenge; or detection of higher levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) in an ISS ODN treated host as compared to an antigen-primed, or primed and challenged, control; (3) IgG2a antibody production in an ISS-ODN treated host; or (4) a reduction in levels of antigen-specific IgE as measured before and after antigen challenge; or detection of lower (or absent) levels of antigen-specific IgE in an ISS-ODN treated host as compared to an antigen-primed, or primed and challenged, control.

37. A kit for use in reducing or preventing inflammation in an antigen-sensitized host tissue comprising an immunostimulatory oligonucleotide (IS S-ODN) in a sterile vial, a device for delivering the ISS-ODN directly into a host tissue and at least one assay reagent for use in measuring lymphocyte proliferation, IgG2a antibody levels, serum cytokine levels and/or granuloctye counts in inflammatory infiltrate of an affected host tissue.
Description



FIELD OF THE INVENTION

[0002] The invention relates to methods and oligonucleotide compositions for use in reducing or suppressing granulocyte-mediated inflammation in a host tissue and in modulating the host's immune responsiveness to an antigen.

HISTORY OF THE RELATED ART

[0003] In vertebrates, endothelial cell adhesion by granulocytes (eosinophils, basophils, neutrophils and mast cells) is followed by the release of inflammatory mediators, such as leukotrienes, major basic protein and histamine. In susceptible individuals, the resulting inflammation can damage affected host tissues.

[0004] The most common pathologic inflammatory condition is asthma, which is characterized by marked eosinophil infiltration into respiratory airways, followed by inflammation-induced tissue damage. Other pathologic inflammatory conditions associated with granulocyte infiltration into affected tissues include nasal polyposis, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, eosinophilic fasciitis, idiopathic hypereosinophilic syndrome and cutaneous basophil hypersensitivity, as well as inflammation and fibrosis resulting from increased production of granulocyte-stimulatory cytokines, such as interleukin (IL)-5 and certain interferons (INF).

[0005] Routine treatment of such conditions is typically directed toward inhibiting the activity of inflammatory mediators released after granulocyte adhesion to endothelia (e.g., by delivering a corticoid composition to the affected tissues). Where the identity of an inflammation inducing antigen is known, some immune protection against further antigen challenge can be provided through immunization. However, although effective in stimulating production of neutralizing antibodies, canonical immunization does not effectively stimulate longer term cellular immunity. Moreover, antigen immunization stimulates host production of IL-4 and IL-5. IL-5 encourages granulocyte adhesion to endothelia while IL-4 induces immunoglobulin switching to the IgE isotype at the risk of anaphylaxis.

SUMMARY OF THE INVENTION

[0006] The invention provides means to rapidly suppress antigen-stimulated inflammation in a mammalian host by suppressing granulocyte infiltration into a host tissue. The invention also provides immunization-free means to provide protection to an antigen-sensitized mammalian host against subsequent antigen challenge without risk of anaphylaxis. These aims are achieved by the invention through delivery of an immunostimulatory oligonucleotide (ISS-ODN) to the host without codelivery of an immunizing antigen.

[0007] Surprisingly, ISS-ODN have anti-inflammatory properties in addition to their immunostimulatory properties. ISS-ODN are therefore useful in the treatment and prevention of inflammation associated with antigen-stimulated granulocyte infiltration of tissue, such as occurs in the respiratory passages of asthmatics during an asthma attack. Advantageously, delivery of ISS-ODN according to the invention suppresses antigen-stimulated granulocyte infiltration into host tissue even before the ISS-ODN affect the host's immune response to the antigen. Thus, the invention provides an antigen-independent method to reduce antigen-stimulated inflammation by suppressing cellular adhesion, thereby avoiding the release of inflammatory mediators which would be stimulated through granulocyte-binding of endothelial cells.

[0008] An example of a therapeutic application for the invention is in the control of asthma, whereby the ISS-ODN are delivered into pulmonary tissue intranasally or by systemic routes. In asthmatics, eosinophil infiltration of lung tissue occurs mainly during the late phase of an allergic response to a respiratory allergen. Canonical immunotherapy can modulate the host immune response to the allergen and eventually stem the tide of eosinophils into the host airways. However, practice of the invention suppresses eosinophil infiltration of host airways well before the host immune system responds to the respiratory allergen, thereby providing a form of protection against the airway narrowing and respiratory tissue damage which characterize an acute asthma attack.

[0009] In another aspect, the invention provides means to shift a present host cellular immune response to an antigen away from a Th2 phenotype and into a Th1 phenotype. To this end, ISS-ODN are delivered by any route through which antigen-sensitized host tissues will be contacted with the ISS-ODN. ISS-ODN administered in this fashion boost both humoral (antibody) and cellular (Th1 type) immune responses of the host. Unlike canonical immunotherapy immunity is stimulated by this method of the invention even when no additional antigen is introduced into the host. Thus, use of the method to boost the immune responsiveness of a host to subsequent challenge by a sensitizing antigen without immunization avoids the risk of immunization-induced anaphylaxis, suppresses IgE production in response to the antigen challenge and eliminates the need to identify the sensitizing antigen for use in immunization. An especially advantageous use for this aspect of the invention is treatment of localized allergic responses in target tissues where the allergens enter the body, such as the skin and mucosa.

[0010] Suppression of the Th2 phenotype according to the invention is also a useful adjunct to canonical immunotherapy to reduce antigen-stimulated IL-4 and IL-5 production. Thus, the invention encompasses delivery of ISS-ODN to a host to suppress the Th2 phenotype associated with conventional antigen immunization (e.g., for vaccination or allergy immunotherapy).

[0011] The shift to a Th1 phenotype achieved according to the invention is accompanied by increased secretion of IFN .alpha., .beta., and .gamma., as well as IL-12 and IL-18. Each of these cytokines enhance the host's immune defenses against intracellular pathogens, such as viruses. Thus, the invention encompasses delivery of ISS-ODN to a host to combat pathogenic infection.

[0012] Angiogenesis is also enhanced in the Th1 phenotype (ostensibly through stimulation by IL-12). Thus, the invention encompasses delivery of ISS-ODN to a host to stimulate therapeutic angiogenesis to treat conditions in which localized blood flow plays a significant etiological role, such as in diabetic retinopathy.

[0013] Pharmaceutically acceptable compositions of ISS-ODN are provided for use in practicing the methods of the invention. The ISS-ODN of the invention include DNA or RNA oligonucleotides which are enriched with CpG dinucleotides, including those which are comprised of the primary structure 5'-Purine-Purine-[C]-[G]-Pyrimidine-Pyrimidine-3'.

[0014] Where appropriate to the contemplated course of therapy, the ISS-ODN may be administered with other anti-inflammatory or immunotherapeutic agents. Thus, a particularly useful composition for use in practicing the method of the invention is one in which an anti-inflammatory agent (e.g., a glucocorticoid) or immunotherapeutic agent (e.g., an antigen, cytokine or adjuvant) is mixed with, or conjugated to, an ISS-ODN.

[0015] The ISS-ODN can also be provided in the form of a kit comprising ISS-ODN and any additional medicaments, as well as a device for delivery of the ISS-ODN to a host tissue and reagents for determining the biological effect of the ISS-ODN on the treated host.

BRIEF DESCRIPTION OF DRAWINGS

[0016] FIG. 1 is a chart which summarizes aspects of the mammalian immune system.

[0017] FIG. 2 is a graph of data which confirm a shift from a Th2 to a Th1 phenotype (as measured by IgG2A production) in mice treated with an ISS-ODN 3 days before antigen challenge.

[0018] FIGS. 3a and 3b are graphs of data which confirm the induction of a Th2 phenotype (as measured by IgG1 production) in mice treated with a mutant, inactive ISS-ODN 3 days before antigen challenge.

[0019] FIG. 4 is a graph of data which confirm Th1-associated suppression of antigen-specific IgE in antigen-sensitized, ISS-ODN (pCMV-LacZ, a plasmid containing two copies of the DY1018 ISS-ODN) treated mice as compared to antigen-sensitized (control) mice.

[0020] FIG. 5 is a graph of data which confirm suppression of IL-4 secretion by ISS-ODN as compared to a control.

[0021] FIG. 6 is a graph of data which confirm suppression of IL-5 secretion by ISS-ODN as compared to a control.

[0022] FIG. 7 is a graph of data which confirm suppression of IL-10 secretion by ISS-ODN as compared to a control.

[0023] FIG. 8 is a graph of data which confirm stimulation of INF-.gamma. secretion by ISS-ODN as compared to a control.

[0024] FIG. 9 is a graph of data demonstrating an ISS-ODN mediated shift to a Th1 phenotype (as indicated by IFN.gamma. levels) in animals treated with ISS-ODN before antigen challenge (asterisked bars) or after antigen challenge.

[0025] FIG. 10 is a graph of data demonstrating an ISS-ODN mediated boost in immune responsiveness (as indicated by increases in CD4+ lymphocyte proliferation) in animals treated with ISS-ODN before antigen challenge (asterisked bars) or after antigen challenge.

DETAILED DESCRIPTION OF THE INVENTION

[0026] A. Anti-Inflammatory and Immunotherapeutic Methods of the Invention

[0027] 1. Therapeutic Effects of the Methods of the Invention

[0028] The main therapeutic goals which may be achieved through practice of the methods of the invention are treatment of inflammation and boosting of host immune responsiveness with a Th1 phenotype against a sensitizing antigen. Both goals are achieved by delivering ISS-ODN to an antigen-sensitized host; i.e., a mammal whose immune system has been primed to respond to challenge by a sensitizing antigen. For purposes of this disclosure, "sensitizing antigen" refers to an exogenous, immunogenic protein, peptide, glycoprotein, lipid or polysaccharide. For reference, a chart summarizing aspects of mammal antigen immunity is appended as FIG. 1.

[0029] The anti-inflammatory method of the invention is useful in suppressing the onset of, and in reducing, acute granulocyte-mediated inflammation in an antigen-sensitized host. Specifically, treatment of an antigen-sensitized (primed) host before subsequent antigen challenge suppresses antigen-stimulated infiltration of host tissue by granulocytes (especially, eosinophils and basophils). Similarly, treatment of an antigen-sensitized host on or after antigen challenge reduces antigen-stimulated infiltration of host tissue by granulocytes. Advantageously, the anti-inflammatory impact of ISS-ODN delivered according to the invention is rapid, taking effect even before the ISS-ODN would be expected to impact the host's immune responsiveness to the sensitizing antigen. The invention therefore provides the host with fairly immediate protection against tissue damage from granulocyte-mediated inflammation.

[0030] For example, as shown by the data in Example II, antigen-sensitized animal models of allergic asthma treated with ISS-ODN without concurrent antigen challenge experienced as much as a 90% reduction of eosinophil infiltration into respiratory tissue as compared to control animals and animals treated only with an inactive ISS-ODN mutant. Significantly, reduction of eosinophil infiltration in previously challenged mice, or suppression of eosinophil infiltration in primed, unchallenged mice, was obtained within as little as 24 hours of delivery of the ISS-ODN. The effect of the ISS-ODN on eosinophil infiltration is therefore independent of the later-developing host immune response to the sensitizing antigen. Being antigen independent, the ISS-ODN can be utilized as inflammation suppressors before antigen challenge or during a period when the risk of antigen challenge is present (e.g., during an allergy season). Importantly, as shown in Examples IV and VI, ISS-ODN can be used according to the invention to prevent inflammation or an immune response on subsequent antigen challenge in an antigen-primed host as well as to reduce inflammation or other antigen-stimulated immune responses after antigen challenge.

[0031] Although the invention is not limited to any mechanism of action, it is probable that the anti-inflammatory activity of ISS-ODN is at least in part a consequence of IL-5 suppression. However, suppression of granulocyte accumulation in host tissue is achieved more rapidly (within 24 hours) than immune activation of cytokine-secreting lymphocytes would be expected to occur. It is therefore also possible that ISS-ODN administered according to the invention physically interfere with granulocyte adhesion to endothelial, perhaps by blocking VCAM-1 endothelial receptors, their eosinophilic ligand (VLA-4) or by lysing granulocytes. Whatever the mechanism, ISS-ODN suppression of granulocyte accumulation according to the invention appears to be independent of ISS-ODN stimulation of the host immune system.

[0032] The immunotherapeutic method of the invention produces a vaccination-like immune response to challenge by a sensitizing antigen without concurrent exposure of the host to the antigen. Immune stimulation achieved through practice of the invention is comparable to the immune stimulation which occurs on vaccination of a host with a sensitizing antigen. Thus, the methods of the invention provides means to immunize a host against a sensitizing antigen without deliberate antigen challenge.

[0033] Advantageously, the immune response stimulated according to the invention differs from an immunization response in that the latter develops in a Th2 phenotype while the former develops in a Th1 phenotype. In this regard, it is helpful to recall that CD4+ lymphocytes generally fall into one of two distinct subsets; i.e., the Th1 and Th2 cells. Th1 cells principally secrete IL-2, IFN.gamma. and TNF.beta. (the latter two of which mediate macrophage activation and delayed type hypersensitivity) while Th2 cells principally secrete IL-4 (which stimulates production of IgE antibodies), IL-5 (which stimulates granulocyte infiltration of tissue), IL-6 and IL-10. These CD4+ subsets exert a negative influence on one another; i.e., secretion of Th1 lymphokines inhibits secretion of Th2 lymphokines and vice versa.

[0034] Factors believed to favor Th1 activation resemble those induced by viral infection and include intracellular pathogens, exposure to IFN-.beta., IFN-.alpha., IFN.gamma., IL-12 and IL-18 and exposure to low doses of antigen. Th1 type immune responses also predominate in autoimmune disease. Factors believed to favor Th2 activation include exposure to IL-4 and IL-10, APC activity on the part of .beta.-lymphocytes and high doses of antigen. Active Th1 (IFN.gamma.) cells enhance cellular immunity and are therefore of particular value in responding to intracellular infections, while active Th2 cells enhance antibody production and are therefore of value in responding to extracellular infections (at the risk of anaphylactic events associated with IL-4 stimulated induction of IgE antibody production). Thus, the ability to shift host immune responses from the Th1 to the Th2 repertoire and vice versa has substantial clinical significance for controlling host immunity against antigen challenge (e.g., in infectious and allergic conditions).

[0035] To that end, the methods of the invention shift the host immune response to a sensitizing antigen toward a Th1 phenotype (Example IV). Consequently, antigen-stimulated/Th2 associated IL-4, IL-5 and IL-10 secretion (Example VI), IL-5 stimulated granulocyte infiltration of antigen-sensitized tissue (Examples II and III) and IL-4 stimulated production of IgE (Example V) are suppressed, thereby reducing the host's risk of prolonged allergic inflammation and minimizing the risk of antigen-induced anaphylaxis. Although the invention is not limited to any particular mechanism of action, it is conceivable that ISS-ODN facilitate uptake of exogenous antigen by antigen presenting cells for presentation through host MHC Class I processing pathways. Whatever the mechanism of action, use of ISS-ODN to boost the host's immune responsiveness to a sensitizing antigen and shift the immune response toward a Th1 phenotype avoids the risk of immunization-induced anaphylaxis, suppresses IgE production in response to a sensitizing antigen and eliminates the need to identify the sensitizing antigen for use in immunization.

[0036] With reference to the invention, ISS-ODN mediated "reduction of inflammation" (in a primed, antigen-challenged host), "prevention of inflammation" (in a primed host before antigen challenge) and "boosting of immune responsiveness in a Th1 phenotype" in an ISS-ODN treated host are evidenced by any of the following events:

[0037] (1) a reduction in levels of IL-4 measured before and after antigen-challenge; or detection of lower (or even absent) levels of IL-4 in a treated host as compared to an antigen-primed, or primed and challenged, control;

[0038] (2) an increase in levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) before and after antigen challenge; or detection of higher levels of IL-12, IL-18 and/or IFN (.alpha., .beta. or .gamma.) in an ISS-ODN treated host as compared to an antigen-primed or, primed and challenged, control;

[0039] (3) IgG2a antibody production in a treated host; or

[0040] (4) a reduction in levels of antigen-specific IgE as measured before and after antigen challenge; or detection of lower (or even absent) levels of antigen-specific IgE in an ISS-ODN treated host as compared to an antigen-primed, or primed and challenged, control.

[0041] Also, with respect to reduction and prevention of inflammation in particular, an especially meaningful indicia of the efficacy of the inventive method in a treated host is:

[0042] (5) a reduction in granulocyte counts (e.g., of eosinophils or basophils, depending on which cell type is most involved in the condition affecting the host) in inflammatory infiltrate of an affected host tissue as measured in an antigen-challenged host before and after ISSODN administration, or detection of lower (or even absent) levels of eosinophil or basophil counts in a treated host as compared to an antigen-primed, or primed and challenged, control.

[0043] Exemplary methods for determining such values are described further in the Examples.

[0044] 2. Methods and Routes for Administration of ISS-ODN to a Host

[0045] The ISS-ODN of the invention are administered to a host using any available method and route suitable for drug delivery, including ex vivo methods (e.g., delivery of cells incubated or transfected with an ISS-ODN) as well as systemic or localized routes. However, those of ordinary skill in the art will appreciate that methods and localized routes which direct the ISS-ODN into antigen-sensitized tissue will be preferred in most circumstances to systemic routes of administration, both for immediacy of therapeutic effect and avoidance of oligonucleotide degradation in vivo.

[0046] The entrance point for many exogenous antigens into a host is through the skin or mucosa. Thus, delivery methods and routes which target the skin (e.g., for cutaneous and subcutaneous conditions) or mucosa (e.g., for respiratory, ocular, lingual or genital conditions) will be especially useful. Those of ordinary skill in the clinical arts will be familiar with, or can readily ascertain, means for drug delivery into skin and mucosa. For review, however, exemplary methods and routes of drug delivery useful in the invention are briefly discussed below.

[0047] Intranasal administration means are particularly useful in addressing respiratory inflammation, particularly inflammation mediated by antigens transmitted from the nasal passages into the trachea or bronchioli. Such means include inhalation of aerosol suspensions or insufflation of the polynucleotide compositions of the invention. Nebulizer devices suitable for delivery of polynucleotide compositions to the nasal mucosa, trachea and bronchioli are well-known in the art and will therefore not be described in detail here. For general review in regard to intranasal drug delivery, those of ordinary skill in the art may wish to consult Chien, Novel Drug Delivery Systems, Ch. 5 (Marcel Dekker, 1992).

[0048] Dermal routes of administration, as well as subcutaneous injections, are useful in addressing allergic reactions and inflammation in the skin. Examples of means for delivering drugs to the skin are topical application of a suitable pharmaceutical preparation, transdermal transmission, injection and epidermal administration.

[0049] For transdermal transmission, absorption promoters or iontophoresis are suitable methods. For review regarding such methods, those of ordinary skill in the art may wish to consult Chien, supra at Ch. 7. Iontophoretic transmission may be accomplished using commercially available "patches" which deliver their product continuously via electric pulses through unbroken skin for periods of several days or more. Use of this method allows for controlled transmission of pharmaceutical compositions in relatively great concentrations, permits infusion of combination drugs and allows for contemporaneous use of an absorption promoter.

[0050] An exemplary patch product for use in this method is the LECTRO PATCH trademarked product of General Medical Company of Los Angeles, Calif. This product electronically maintains reservoir electrodes at neutral pH and can be adapted to provide dosages of differing concentrations, to dose continuously and/or to dose periodically. Preparation and use of the patch should be performed according to the manufacturer's printed instructions which accompany the LECTRO PATCH product; those instructions are incorporated herein by this reference.

[0051] Epidermal administration essentially involves mechanically or chemically irritating the outermost layer of the epidermis sufficiently to provoke an immune response to the irritant. An exemplary device for use in epidermal administration employs a multiplicity of very narrow diameter, short tynes which can be used to scratch ISS-ODN coated onto the tynes into the skin. The device included in the MONO-VACC old tuberculin test manufactured by Pasteur Merieux of Lyon, France is suitable for use in epidermal administration of ISS-ODN. Use of the device is according to the manufacturer's written instructions included with the device product; these instructions regarding use and administration are incorporated herein by this reference to illustrate conventional use of the device. Similar devices which may also be used in this embodiment are those which are currently used to perform allergy tests.

[0052] Ophthalmic administration (e.g., for treatment of allergic conjunctivitis) involves invasive or topical application of a pharmaceutical preparation to the eye. Eye drops, topical cremes and injectable liquids are all examples of suitable milieus for delivering drugs to the eye.

[0053] Systemic administration involves invasive or systemically absorbed topical administration of pharmaceutical preparations. Topical applications as well as intravenous and intramuscular injections are examples of common means for systemic administration of drugs.

[0054] 3. Dosing Parameters for ISS-ODN

[0055] A particular advantage of the ISS-ODN of the invention is their capacity to exert anti-inflammatory and immunotherapeutic activity even at relatively minute dosages. Although the dosage used will vary depending on the clinical goals to be achieved, a suitable dosage range is one which provides up to about 1-1000 .mu.g of ISS-ODN/ml of carrier in a single dosage. In view of the teaching provided by this disclosure, those of ordinary skill in the clinical arts will be familiar with, or can readily ascertain, suitable parameters for administration of ISS-ODN according to the invention.

[0056] In this respect, it should be noted that the anti-inflammatory and immunotherapeutic activity of ISS-ODN in the invention is essentially dose-dependent. Therefore, to increase ISS-ODN potency by a magnitude of two, each single dose is doubled in concentration. Clinically, it may be advisable to administer the ISS-ODN in a low dosage (e.g., about 1 .mu.g/ml to about 50 .mu.g/ml), then increase the dosage as needed to achieve the desired therapeutic goal. Alternatively, a target dosage of ISS-ODN can be considered to be about 1-10 .mu.M in a sample of host blood drawn within the first 24-48 hours after administration of ISS-ODN. Based on current studies, ISS-ODN are believed to have little or no toxicity at these dosage levels.

[0057] B. ISS-ODN Anti-Inflammatory Compositions

[0058] 1. ISS-ODN Structure

[0059] Functionally, ISS-ODN enhance the cellular and humoral immune responses in a host, particularly lymphocyte proliferation and the release of cytokines (including IFN) by host monocytes and natural killer (NK) cells. Immunostimulation by synthetic ISS-ODN in vivo occurs by contacting host lymphocytes with, for example, ISS-ODN oligonucleotides, ISS-ODN oligonucleotide-conjugates and ISS-containing recombinant expression vectors (data regarding the activity of ISS-ODN conjugates and ISS-ODN vectors are set forth in co-pending, commonly assigned U.S. patent applications Serial No., 60/028,118 and Ser. No. 08/593,554; data from which is incorporated herein by reference to demonstrate ISS-ODN immunostimulatory activity in vivo). Thus, while native microbial ISS-ODN stimulate the host immune system to respond to infection, synthetic analogs of these ISS-ODN are useful therapeutically to modulate the host immune response not only to microbial antigens, but also to tumor antigens, allergens and other substances. Structurally, ISS-ODN are non-coding oligonucleotides 6 mer or greater in length which may include at least one unmethylated CpG motif. The relative position of each CpG sequence in ISS-ODN with immunostimulatory activity in certain mammalian species (e.g., rodents) is 5'-CG-3' (i.e., the C is in the 5' position with respect to the G in the 3' position). Many known ISS-ODN flank the CpG motif with at least two purine nucleotides (e.g., GA or AA) and at least two pyrimidine nucleotides (5'-Purine-Purine[C]-[G]-Pyrimidine-Pyri- midine-3'). CpG motif-containing ISS-ODN are believed to stimulate B lymphocyte proliferation (see, e.g., Krieg, et al., Nature, 374:546-549, 1995).

[0060] The core hexamer structure of the foregoing ISS-ODN may be flanked upstream and/or downstream by any number or composition of nucleotides or nucleosides. However, ISS-ODN are at least 6 mer in length, and preferably are between 6 and 200 mer in length, to enhance uptake of the ISS-ODN into target tissues. Those of ordinary skill in the art will be familiar with, or can readily identify, reported nucleotide sequences of known ISS-ODN. For ease of reference in this regard, the following sources are especially helpful:

[0061] Yamamoto, et al., Microbiol. Immunol., 36:983 (1992)

[0062] Ballas, et al., J. Immunol., 157:1840 (1996)

[0063] Klinman, et al., J. Immunol., 158:3635 (1997)

[0064] Sato, et al., Science, 273:352 (1996)

[0065] Each of these articles are incorporated herein by reference for the purpose of illustrating the level of knowledge in the art concerning the nucleotide composition of ISS-ODN.

[0066] In particular, ISS-ODN useful in the invention include those which have the following hexameric nucleotide sequences:

[0067] 1. ISS-ODN having "CpG" dinucleotides; and,

[0068] 2. Inosine and/or uracil substitutions for nucleotides in the foregoing hexamer sequences for use as RNA ISS-ODN.

[0069] For example, DNA based ISS-ODN useful in the invention include those which have the following hexameric nucleotide sequences:

[0070] AACGTT, AGCGTC, GACGTT, GGCGTT, AACGTC, AGCGTC, GACGTC, GGCGTC, AACGCC, AGCGCC, GACGCC, GGCGCC, AGCGCT, GACGCT, GGCGCT, TTCGAA, GGCGTT and AACGCC (respectively, SEQ.ID.Nos. 1-18).

[0071] ISS-ODN may be single-stranded or double-stranded DNA, single or double-stranded RNA and/or oligonucleosides. The ISS-ODN may or may not include palindromic regions. If present, a palindrome may extend only to a CpG motif, if present, in the core hexamer sequence, or may encompass more of the hexamer sequence as well as flanking nucleotide sequences.

[0072] The nucleotide bases of the ISS-ODN which flank the CpG motif of the core hexamer and/or make up the flanking nucleotide sequences may be any known naturally occurring bases or synthetic nonnatural bases (e.g., TCAG or, in RNA, UACGI). Oligonucleosides may be incorporated into the internal region and/or termini of the ISS-ODN using conventional techniques for use as attachment points for other compounds (e.g., peptides). The base(s), sugar moiety, phosphate groups and termini of the ISS-ODN may also be modified in any manner known to those of ordinary skill in the art to construct an ISS-ODN having properties desired in addition to the described activity of the ISS-ODN. For example, sugar moieties may be attached to nucleotide bases of ISS-ODN in any steric configuration.

[0073] In addition, backbone phosphate group modifications (e.g., methylphosphonate, phosphorothioate, phosphoroamidate and phosphorodithioate internucleotide linkages) can confer anti-microbial activity on the ISS-ODN and enhance their stability in vivo, making them particularly useful in therapeutic applications. A particularly useful phosphate group modification is the conversion to the phosphorothioate or phosphorodithioate forms of the ISS-ODN oligonucleotides. In addition to their potentially anti-microbial properties, phosphorothioates and phosphorodithioates are more resistant to degradation in vivo than their unmodified oligonucleotide counterparts, making the ISS-ODN of the invention more available to the host.

[0074] 2. Synthesis of, and Screening for, ISS-ODN

[0075] ISS-ODN can be synthesized using techniques and nucleic acid synthesis equipment which are well-known in the art. For reference in this regard, see, e.g., Ausubel, et al., Current Protocols in Molecular Biology, Chs. 2 and 4 (Wiley Interscience, 1989); Maniatis, et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Lab., New York, 1982); U.S. Pat. No. 4,458,066 and U.S. Pat. No. 4,650,675. These references are incorporated herein by reference for the sole purpose of demonstrating knowledge in the art concerning production of synthetic oligonucleotides. Because the ISS-ODN is non-coding, there is no concern about maintaining an open reading frame during synthesis.

[0076] ISS-ODN may be incorporated into a delivery vector, such as a plasmid, cosmid, virus or retrovirus, which may in turn code for therapeutically beneficial polypeptides, such as cytokines, hormones and antigens. Incorporation of ISS-ODN into such a vector does not adversely affect their activity.

[0077] Alternatively, ISS-ODN may be isolated from microbial species (especially mycobacteria) using techniques well-known in the art, such as nucleic acid hybridization. Preferably, such isolated ISS-ODN will be purified to a substantially pure state; i.e., to be free of endogenous contaminants, such as lipopolysaccharides. ISS-ODN isolated as part of a larger polynucleotide can be reduced to the desired length by techniques well known in the art, such as by endonuclease digestion. Those of ordinary skill in the art will be familiar with, or can readily ascertain, techniques suitable for isolation, purification and digestion of polynucleotides to obtain ISS-ODN of potential use in the invention.

[0078] Confirmation that a particular oligonucleotide has the properties of an ISS-ODN useful in the invention can be obtained by evaluating whether the ISS-ODN affects cytokine secretion and IgG antibody isotype production as described in Section A.I, above. Details of in vitro techniques useful in making such an evaluation are given in the Examples; those of ordinary skill in the art will also know of, or can readily ascertain, other methods for measuring cytokine secretion and antibody production along the parameters taught herein.

[0079] The techniques for making phosphate group modifications to oligonucleotides are known in the art and do not require detailed explanation. For review of one such useful technique, the an intermediate phosphate triester for the target oligonucleotide product is prepared and oxidized to the naturally occurring phosphate triester with aqueous iodine or with other agents, such as anhydrous amines. The resulting oligonucleotide phosphoramidates can be treated with sulfur to yield phophorothioates. The same general technique (excepting the sulfur treatment step) can be applied to yield methylphosphoamidites from methylphosphonates. For more details concerning phosphate group modification techniques, those of ordinary skill in the art may wish to consult U.S. Pat. Nos. 4,425,732; 4,458,066; 5,218,103 and 5,453,496, as well as Tetrahedron Lett. at 21:4149 (1995), 7:5575 (1986), 25:1437 (1984) and Journal Am. Chem Soc., 93:6657 (1987), the disclosures of which are incorporated herein for the sole purpose of illustrating the standard level of knowledge in the art concerning preparation of these compounds.

[0080] A colloidal dispersion system may be used for targeted delivery of the ISS-ODN to an inflamed tissue. Colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. The preferred colloidal system of this invention is a liposome.

[0081] Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles (LUV), which range in size from 0.2-4.0 .mu.m can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules. RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al., Trends Biochem. Sci., 6:77, 1981). In addition to mammalian cells, liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells. In order for a liposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1) encapsulation of the genes encoding the antisense polynucleotides at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino, et al., Biotechniques, 6:682, 1988).

[0082] The composition of the liposome is usually a combination of phospholipids, particularly high-phasetransition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations.

[0083] Examples of lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides. Particularly useful are iacylphosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated. Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine.

[0084] The targeting of liposomes can be classified based on anatomical and mechanistic factors. Anatomical classification is based on the level of selectivity, for example, organ-specific, cell-specific, and organelle-specific. Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries. Active targeting, on the other hand, involves alteration of the liposome by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the liposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.

[0085] The surface of the targeted delivery system may be modified in a variety of ways. In the case of a liposomal targeted delivery system, lipid groups can be incorporated into the lipid bilayer of the liposome in order to maintain the targeting ligand in stable association with the liposomal bilayer. Various well known linking groups can be used for joining the lipid chains to the targeting ligand (see, e.g., Yanagawa, et al., Nuc. Acids Symp. Ser., 19:189 (1988), Grabarek, et al., Anal. Biochem., 185:131 (1990); Staros, et al., Anal. Biochem., 156:220 (1986) and Boujrad, et al., Proc. Natl. Acad. Sci USA, 90:5728 (1993), the disclosures of which are incorporated herein by reference solely to illustrate the standard level of knowledge in the art concerning conjugation of oligonucleotides to lipids). Targeted delivery of ISS-ODN can also be achieved by conjugation of the ISS-ODN to a the surface of viral and non-viral recombinant expression vectors, to an antigen or other ligand, to a monoclonal antibody or to any molecule which has the desired binding specificity.

[0086] Examples of other useful conjugate partners include any immunogenic antigen (including allergens, live and attenuated viral particles and tumor antigens), targeting peptides (such as receptor ligands, antibodies and antibody fragments, hormones and enzymes), non-peptidic antigens (coupled via a peptide linkage, such as lipids, polysaccharides, glycoproteins, gangliosides and the like) and cytokines (including interleukins, interferons, erythropoietin, tumor necrosis factor and colony stimulating factors). Such conjugate partners can be prepared according to conventional techniques (e.g., peptide synthesis) and many are commercially available.

[0087] Those of ordinary skill in the art will also be familiar with, or can readily determine, methods useful in preparing oligonucleotide-peptide conjugates. Conjugation can be accomplished at either termini of the ISS-ODN or at a suitably modified base in an internal position (e.g., a cytosine or uracil). For reference, methods for conjugating oligonucleotides to proteins and to oligosaccharide moieties of Ig are known (see, e.g., O'Shannessy, et al., J. Applied Biochem., 7:347 (1985), the disclosure of which is incorporated herein by reference solely to illustrate the standard level of knowledge in the art concerning oligonucleotide conjugation). Another useful reference is Kessler: "Nonradioactive Labeling Methods for Nucleic Acids", in Kricka (ed.), Nonisotopic DNA Probe Techniques (Acad.Press, 1992)).

[0088] Briefly, examples of known, suitable conjugation methods include: conjugation through 3' attachment via solid support chemistry see, e.g., Haralambidis, et al., Nuc. Acids Res., 18:493 (1990) and Haralambidis, et al., Nuc. Acids Res., 18:501 (1990) [solid support synthesis of peptide partner]; Zuckermann, et al., Nuc. Acids Res., 15:5305 (1987), Corey, et al., Science, 238:1401 (1987) and Nelson, et al., Nuc. Acids Res., 17:1781 (1989) [solid support synthesis of oligonucleotide partner]).

[0089] Amino-amino group linkages may be performed as described in Benoit. et al., Neuromethods, 6:43 (1987), while thiol-carboxyl group linkages may be performed as described in Sinah, et al., Oligonucleotide Analogues: A Practical Approach (IRL Press, 1991). In these latter methods, the oligonucleotide partner is synthesized on a solid support and a linking group comprising a Protected amine, thiol or carboxyl group opposite a phosphoramidite is covalently attached to the 5'-hydroxyl (see, e.g., U.S. Pat. Nos. 4,849,513; 5,015,733; 5,118,800 and 5,118,802).

[0090] Linkage of the oligonucleotide partner to a peptide may also be made via incorporation of a linker arm (e.g., amine or carboxyl group) to a modified cytosine or uracil base (see, e.g., Ruth, 4th Annual Congress for Recombinant DNA Research at 123). Affinity linkages (e.g., biotin-streptavidin) may also be used (see, e.g., Roget, et al, Nuc. Acids Res., 17:7643 (1989)).

[0091] Methods for linking oligonucleotides to lipids are also known and include synthesis of oligophospholipid conjugates see, e.g., Yanagawa, et al., Nuc. Acids Symp. Ser., 19:189 (1988)), synthesis of oligo-fatty acids conjugates (see, e.g., Grabarek, et al, Anal. Biochem., 185:131 (1990)) and oligo-sterol conjugates (see, e.g., Boujrad, et al., Proc. Natl. Acad. Sci USA, 90:5728 (1993)).

[0092] Each of the foregoing references is incorporated herein by reference for the sole purpose of illustrating the level of knowledge and skill in the art with respect to oligonucleotide conjugation methods.

[0093] Co-administration of a peptide drug with an ISS-ODN according to the invention may also be achieved by incorporating the ISS-ODN in cis or in trans into a recombinant expression vector (plasmid, cosmid, virus or retrovirus) which codes for any therapeutically beneficial protein deliverable by a recombinant expression vector. If incorporation of an ISS-ODN into an expression vector for use in practicing the invention is desired, such incorporation may be accomplished using conventional techniques which do not require detailed explanation to one of ordinary skill in the art. For review, however, those of ordinary skill may wish to consult Ausubel, Current Protocols in Molecular Biology, supra.

[0094] Briefly, construction of recombinant expression vectors (including those which do not code for any protein and are used as carriers for ISS-ODN) employs standard ligation techniques. For analysis to confirm correct sequences in vectors constructed, the ligation mixtures may be used to transform a host cell and successful transformants selected by antibiotic resistance where appropriate. Vectors from the transformants are prepared, analyzed by restriction and/or sequenced by, for example, the method of Messing, et al., (Nucleic Acids Res., 9-309, 1981), the method of Maxam, et al., (Methods in Enzymology, 65:499,1980), or other suitable methods which will be known to those skilled in the art. Size separation of cleaved fragments is performed using conventional gel electrophoresis as described, for example, by Maniatis, et al., (Molecular Cloning, pp. 133-134, 1982).

[0095] Host cells may be transformed with expression vectors and cultured in conventional nutrient media modified as is appropriate for inducing promoters, selecting transformants or amplifying genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

[0096] If a recombinant expression vector is utilized as a carrier for the ISS-ODN of the invention, plasmids and cosmids are particularly preferred for their lack of pathogenicity. However, plasmids and cosmids are subject to degradation in vivo more quickly than viruses and therefore may not deliver an adequate dosage of ISS-ODN to substantially inhibit ISS-ODN immunostimulatory activity exerted by a systemically administered gene therapy vector. Of the viral vector alternatives, adenoassociated viruses would possess the advantage of low pathogenicity. The relatively low capacity of adeno-associated viruses for insertion of foreign genes would pose no problem in this context due to the relatively small size in which ISS-ODN of the invention can be synthesized.

[0097] Other viral vectors that can be utilized in the invention include adenovirus, adeno-associated virus, herpes virus, vaccinia or an RNA virus such as a retrovirus. Retroviral vectors are preferably derivatives of a murine, avian or human HIV retrovirus. Examples of retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), and Rous Sarcoma Virus (RSV). A number of additional retroviral vectors can incorporate multiple genes. All of these vectors can transfer or incorporate a gene for a selectable marker so that transduced cells can be identified and generated.

[0098] Since recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance can be provided, for example, by using helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within the LTR. These plasmids are missing a nucleotide sequence that enables the packaging mechanism to recognize an RNA transcript for encapsidation. Helper cell lines that have deletions of the packaging signal include, but are not limited to, T2, PA317 and PA12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such helper cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion can be produced.

[0099] By inserting one or more sequences of interest into the viral vector, along with another gene which encodes the ligand for a receptor on a specific target cell, for example, the vector can be rendered target specific. Retroviral vectors can be made target specific by inserting, for example, a polynucleotide encoding a sugar, a glycolipid, or a protein. Preferred targeting is accomplished by using an antibody to target the retroviral vector. Those of skill in the art will know of, or can readily ascertain without undue experimentation, specific polynucleotide sequences which can be inserted into the retroviral genome to allow target specific delivery of the retroviral vector containing ISS-ODN.

[0100] C. Pharmaceutical Compositions of ISS-ODN

[0101] If to be delivered without use of a vector or other delivery system, ISS-ODN will be prepared in a pharmaceutically acceptable composition. Pharmaceutically acceptable carriers preferred for use with the ISS-ODN of the invention may include sterile aqueous of non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like. A composition of ISS-ODN may also be lyophilized using means well known in the art, for subsequent reconstitution and use according to the invention.

[0102] Absorption promoters, detergents and chemical irritants (e.g., keratinolytic agents) can enhance transmission of an ISS-ODN composition into a target tissue. For reference concerning general principles regarding absorption promoters and detergents which have been used with success in mucosal delivery of organic and peptide-based drugs, see Chien, Novel Drug Delivery Systems, Ch. 4 (Marcel Dekker, 1992).

[0103] Examples of suitable nasal absorption promoters in particular are set forth at Chien, supra at Ch. 5, Tables 2 and 3; milder agents are preferred. Suitable agents for use in the method of this invention for mucosal/nasal delivery are also described in Chang, et al., Nasal Drug Delivery, "Treatise on Controlled Drug Delivery", Ch. 9 and Table 3-4B thereof, (Marcel Dekker, 1992). Suitable agents which are known to enhance absorption of drugs through skin are described in Sloan, Use of Solubility Parameters from Regular Solution Theory to Describe Partitioning-Driven Processes, Ch. 5, "Prodrugs: Topical and Ocular Drug Delivery" (Marcel Dekker, 1992), and at places elsewhere in the text. All of these references are incorporated herein for the sole purpose of illustrating the level of knowledge and skill in the art concerning drug delivery techniques.

[0104] D. Kits for Use in Practicing the Methods of the Invention

[0105] For use in the methods described above, kits are also provided by the invention. Such kits may include any or all of the following: ISS-ODN (conjugated or unconjugated); a pharmaceutically acceptable carrier (may be pre-mixed with the ISS-ODN) or suspension base for reconstituting lyophilized ISS-ODN; additional medicaments; a sterile vial for each ISS-ODN and additional medicament, or a single vial for mixtures thereof; device(s) for use in delivering ISS-ODN to a host; assay reagents for detecting indicia that the anti-inflammatory and/or immunostimulatory effects sought have been achieved in treated animals and a suitable assay device.

[0106] Examples illustrating the practice of the invention are set forth below. The examples are for purposes of reference only and should not be construed to limit the invention, which is defined by the appended claims. All abbreviations and terms used in the examples have their expected and ordinary meaning unless otherwise specified.

EXAMPLE I

Murine Model for the Airway Hyperreactivity of Allergic Asthma

[0107] Sensitizing-antigen challenged mice of different strains model the airway hyperreactivity seen in allergic asthma. Suitable murine strains for use in modeling the disease include Balb/c mice (which are biased genetically toward the Th2 phenotype and produce enhanced concentrations of IL-4 and IL-5 in response to antigen challenge to CD4+ lymphocytes), C57BL/6 mice (which are IL-5 deficient, for detailed study of IL-5 induced tissue damage in asthma) and W/W.sup.v mice (which are mast cell deficient, for detailed study of mast cell activation in asthma).

[0108] Disease modeling mice are conveniently prepared by intraperitoneal or subcutaneous injection of a model sensitizing antigen, ovalbumin ("OVA") in carrier (e.g., sterile saline or a carrier with adjuvant, such as alum), followed by antigen challenge with aerosolized antigen. For example, mice may be immunized with 25 .mu.g OVA by subcutaneous injection (with or without adjuvant) weekly for 4-6 weeks, then challenged with 2 or 3 weekly aerosolizations of OVA at a concentration of 50 mg/ml in phosphate buffered saline (PBS) delivered in 20 minute intervals or at a concentration of 10 mg/ml 0.9% saline daily for about a week (in three 30 minute intervals daily). Nebulizer devices for use in the aerosolization are available from Aerotech II, CIS-US, Bedford, Mass., with a nasal chamber adapted for murine nasal passages (e.g., a nose-only chamber from Intox Products, Albuquerque, N. Mex.). When driven by compressed air at a rate of 10 liters/min., the devices described produce aerosol particles having a median aerodynamic diameter of 1.4 .mu.m.

[0109] Control mice are preferably littermates which are protein-antigen challenged without prior immunization. For farther details concerning this animal model, those of skill in the art may wish to refer to Foster, et al. J. Exp. Med., 195-201, 1995; and, Corry, et al., J. Exp. Med., 109-117, 1996.

EXAMPLE II

Reduction of Eosinophil Accumulation in Lung Tissue in a Murine Asthma Model by Administration of ISS-ODN

[0110] BALB/c mice, 6-10 weeks of age, were prepared as models of allergic asthma as described in Example I (subcutaneous injection of OVA followed by antigen challenge at a concentration of 50 mg OVA/ml PBS). Prior to each inhalation with OVA according to this scheme, sets of 8 mice each were treated as described in the Table below. Control mice were antigen challenged but untreated and naive mice were not challenged with antigen. All ISS doses were 100 .mu.g per administration. Dexamethasone (a conventional steroidal anti-inflammatory used in the treatment of asthma) doses were 5 mg/kg/mouse. Priming doses of antigen were 25 .mu.g OVA adsorbed to alum in 0.2 ml phosphate buffered saline (PBS). Challenge doses of antigen were 10 ml of 50 mg OVA/ml PBS. IN=intranasal; IP=intraperitoneal; SC=subcutaneous and N/A=not applicable.

1 Set # Material Received Route and Timing 1 Naive mice (no antigen) N/A 2 DY1018 (ISS-ODN) IN, 1 day before the first inhalation 3 DY1018 IN, 1 day before the second inhalation 4 DY1018 IN, with the second inhalation 5 DY1018 IN, 2 days after the second inhalation 6 DY1018 IP, 1 day before the first inhalation 7 DY1018 IP, 1 day before the second inhalation 8 DY1018 IP, with the second inhalation 9 DY1018 IP, 2 days after the second inhalation 10 DY1018 IT, 2 days after the second inhalation 11 DY1019 (M-ISS-ODN) IN, 2 days after inhalation 12 DY1019 IP, 2 days after the second inhalation 13 DY1019 IT, 2 days after the second inhalation 14 Dexamethansone SC, 2 days after the second inhalation 15 Dexamethansone SC, 7 days after the second inhalation 16 control mice (antigen only) N/A

[0111] DY1018 has the nucleotide sequence:

[0112] 5'-TGACTGTGAACGTTCGAGATGA-3' (SEQ.ID.No. 19) with a phosphothioate backbone as the nucleotide sequence:

[0113] 5'-TGACTGTGAAGGTTGGAGATGA-3' (SEQ.ID.No. 20) with a phosphothioate backbone.

[0114] On day 32, each mouse was bled by tail snip (approximately 50 .mu.l volume) into a 0.1 mM solution of PBS and EDTA. Red blood cells in solution were lysed with 150 mM NH.sub.4Cl and 10 mM KHCO.sub.2O in dH.sub.2O then stained (Wright-Giesma stain). Lung lavage from each mouse was obtained after sacrifice by canalization of the trachea and lavage with 800 microliters PBS, then the lavage product was stained. Bone marrow samples from each mouse were obtained by flushing of extracted femur marrow with PBS. Histological specimens of lung and trachea tissue were obtained from the right lower lobe of the lung and trachea. Specimens were frozen, sectioned to a 5 micron width and stained with DAB peroxidase.

[0115] Results are expressed in the Table below as percent eosinophils compared to total leukocytes (inflammatory infiltrate) in each sample, except for the "lung" results, which represent the number of eosinophils per microscopic field (5 randomly selected fields were evaluated for each sample). In summary, the control mice had an average of 67% eosinophils in the lung/trachea tissue samples, while mice who received the mutant ISS-ODN (M-ISS-ODN; DY1019) had 52% and 88% (.+-.12%) average accumulation of eosinophils in lung tissue after IP and IN administration, respectively. The higher values for the mice treated with M-ISS-ODN after antigen challenge is most likely owing to the immunoinhibitory properties of DY1019 (see, the co-pending, commonly owned U.S. patent application entitled "Inhibitors of DNA Immunostimulatory Sequence Activity"; Eyal Raz, inventor; filed Jun. 6, 1997 (Serial No. 60/048,793)). Mouse sets 7 and 8 therefore model an partially immune incompetent host with allergic asthma.

[0116] In startling contrast, the mice pre-treated with the DY1018 ISS-ODN delivered intranasally had less than about 10% eosinophil accumulation in the lung and trachea when treated after antigen challenge and only about 19% eosinophil accumulation when treated before antigen challenge. These values represent up to an 80% reduction in eosinophil accumulation compared to the control mice and more than a 90% reduction in comparison to M-ISS-ODN (IN) treated mice.

[0117] The IP ISS-ODN treated mice fared even better, with a 6% eosinophil accumulation in the lung and trachea on treatment before and after antigen challenge. This value represents an 86% reduction in eosinophil accumulation as compared to the control mice and an 91% reduction as compared to M-ISS-ODN (IP) treated mice.

[0118] These data indicate that the IL-5 stimulated eosinophil accumulation in lung tissue which characterizes the late phase of allergic asthma is inhibited by the ISS-ODN therapeutic methods of the invention.

2 Lung and Broncheoalveolar Tracheal Set # Bone Marrow Lavage Blood Tissue 1 (naive) 3 .+-. 2 0 2 .+-. 1 2 .+-. 1 2 (ISS) 5 .+-. 1 10 .+-. 2 3 .+-. 1 8 .+-. 1 3 (ISS) 12 .+-. 1 17 .+-. 4 6 .+-. 2 19 .+-. 5 4 (ISS) 5 .+-. 1 3 .+-. 1 2 .+-. 1 6 .+-. 1 5 (ISS) 8 .+-. 2 4 .+-. 3 3 .+-. 1 6 .+-. 4 6 (ISS) 10 .+-. 1 10 .+-. 1 4 .+-. 1 16 .+-. 4 7 (M-ISS) 13 .+-. 1 51 .+-. 3 10 .+-. 1 88 .+-. 12 8 (M-ISS) 13 .+-. 1 43 .+-. 3 10 .+-. 1 52 .+-. 14 9 (control) 3 .+-. 2 42 .+-. 4 14 .+-. 3 67 .+-. 5

EXAMPLE III

Antigen Independent Reduction of Eosinophil Accumulation in Lung Tissue

[0119] To evaluate whether the eosinophil suppression demonstrated by the data in Example II is dependent upon immune stimulation by the ISS-ODN, mice were sensitized to OVA using a conventional, Th2 stimulatory adjuvant (alum), treated with ISS-ODN or a control, and measured for eosinophil suppression before ISS-ODN stimulation of the mouse immune system would be expected to occur.

[0120] More specifically, groups of four mice were immunized with 25 .mu.g OVA in 1 mg alum by subcutaneous injection on days 1, 7, 14 and 21. This immunization protocol is known to stimulate a Th2 type response to the antigen in mice. On day 27, one group of animals received 100 .mu.g of the DY1018 ISS-ODN described in Example I by intraperitoneal administration. A control group received the mutant DY1019 M-ISS-ODN described in Example I by the same route.

[0121] On day 28, the animals in each group received 10 mg OVA/ml phosphate buffered saline by inhalation for 30 minutes. On day 30, some of the animals in each group received a second injection of ISS-ODN or M-ISS-ODN and the animals who had not been treated on day 27 were treated with ISS-ODN or M-ISS-ODN. The inhalation challenge with OVA was repeated on day 31 and the animals were sacrificed for eosinophil counting within 24 hours.

[0122] The results of this experiment are set forth in the Table below. Animals that received two treatments with ISS-ODN on days 27 and 30 had only 5.8%, eosinophils in the broncheo-alveolar fluid (BALF) lavage on day 32, even though immune stimulation by the ISS-ODN would be minimal so shortly after treatment. Even after only one treatment with ISS-ODN (on day 30), eosinophil accumulation in the BALF the treated animals was limited to 10.3%. In contrast, the control animals twice treated with M-ISS-ODN had 42.3% eosinophils in extracted BALF.

3 Treated on Animals Day 28 Blood Bone Marrow BALF ISS-ODN Yes 1.9% .+-. 0.8 5.8% .+-. 2.5 5.8% .+-. 2.8 M-ISS-ODN Yes 9.8% .+-. 2.1 13.0% .+-. 0.9 42.3% .+-. 3.5 ISS-ODN No 3.5% .+-. 0.6 10.5% .+-. 1.4 10.3% .+-. 1.3

[0123] These data establish that practice of the invention can inhibit allergic inflammation in animals and that the inhibition can occur as quickly as one day after treatment.

EXAMPLE IV

Selective Induction of a Th1 Response in a Host After Administration of an ISS-ODN Containing Plasmid

[0124] In mice, IgG 2A antibodies are serological markers for a Th1 type immune response, whereas IgG 1 antibodies are indicative of a Th2 type immune response. Th2 responses include the allergy-associated IgE antibody class; soluble protein antigens tend to stimulate relatively strong Th2 responses. In contrast, Th1 responses are induced by antigen binding to macrophages and dendritic cells.

[0125] To determine which response, if any, would be produced by mice who received ISS-ODN according to the invention, nine groups of Balb/c mice were immunized with 10 .mu.g .beta.-galactosidase protein (conjugated to avidin; Sigma, St. Louis, Mo.) to produce a model allergic phenotype and treated as follows:

4 Mouse Group ISS-ODN Treatment 1 None (.beta.-gal) 2 DY1018 (ISS-ODN) injected with the antigen 3 DY1018 injected 72 hrs. after the antigen (same site) 4 DY1019 (M-ISS-ODN) injected with the antigen 5 DY1019 injected 72 hrs. after the antigen (same site)

[0126] At 2 week intervals, any IgG 2a and IgG 1 to (.beta.-galactosidase present in the serum of each mouse were measured by enzyme-linked immunoabsorbent assay (using antibodies specific for the IgG 1 and IgG 2A subclasses) on microtiter plates coated with the enzyme.

[0127] As shown in FIG. 2, only the mice who received the ISS-ODN produced high titers of IgG 2A antibodies, which increased in number over a period of 12 weeks. As shown in FIG. 3, immunization of the mice with the antigen itself or with the mutant ISS-ODN induced production of relatively high titers of IgG 1 antibodies. The data shown in the FIGURES comprise averages of the values obtained from each group of mice.

[0128] These data indicate that a selective Th1 response is induced by administration of an ISS-ODN according to the invention to an antigen-challenged host. Further, the data indicate that ISS-ODN administration according to the invention biases the immune system toward the Th1 phenotype on antigen challenge, even when the ISS-ODN are administered before antigen challenge (in this instance, 72 hours before challenge).

EXAMPLE V

Suppression of IgE Antibody Response to Antigen by Immunization with Antigen-Encoding Polynucleotides

[0129] To demonstrate the IgE suppression achieved through stimulation of a Th1 type cellular immune response in preference to a Th2 type cellular immune response, five to eight week old Balb/c mice were immunized with one of two recombinant expression vectors: ISS-ODN containing pCMV-Lac-Z (which contains two copies of nucleotide sequences similar to the DY1018 ISS-ODN) or a control plasmid, pCMV-BL. A third group of the mice received injections of antigen (.beta.-galactosidase). Plasmid DNA was purified and its endotoxin content reduced to 0.5-5 ng/1 mg DNA by extraction with TRITON X-114 (Sigma, St. Louis, Mich.). Before inoculation, pDNA was precipitated in ethanol, washed with 70% ethanol and dissolved in pyrogen free normal saline.

[0130] Immunization was by intradermal injection of plasmid DNA loaded onto separate tynes of a MONOVACC.RTM. multiple tyne device (Connaught Lab, Inc., Swiftwater, Pa.). Briefly, the tyne devices were prepared after extensive washing in DDW and overnight soaking in 0.5% SDS (sulfated dodecyl saline), washed again in DDW, soaked overnight in 0.1N NaOH, washed again in DDW and dried at 37.degree. C. for 8 hours. Six .mu.l of plasmid DNA dissolved in normal saline were pipetted onto the tynes of the tyne device just prior to each inoculation described below. The total amount of pDNA loaded on the device per inoculation was 25 .mu.g each of pCMV-Lac-Z and pCMV-BL. For purposes of estimating actual doses, it was assumed that less than 10% of the pDNA solution loaded onto the tyne device was actually introduced on injection of the tynes into intradermal tissue.

[0131] Each mouse was treated 3 times with 2 inoculations of each plasmid in a one week interval injected intradermally at the base of the tail. Another group of mice received a single intradermal injection in the base of the tail of 10 .mu.g of .beta. galactosidase protein (dissolved in 50 .mu.l of normal saline) in lieu of pDNA.

[0132] Toward inducing an IgE antibody response to subsequent sensitizing-antigen challenge, each group of mice was injected once intraperitoneally with 0.1 ml of phosphate buffered saline (PBS) solution containing 1 .mu.g of antigen (.beta. galactosidase; Calbiochem, San Diego, Calif.) and 3 mg of ALUM aluminum hydroxide as adjuvant (Pierce Chemical, Rockford, Ill.) 14 weeks after the initial immunization. Total IgE was assayed in sera from the mice 4 times over the subsequent 4 consecutive weeks.

[0133] IgE was detected using a solid phase radioimmunoassay (RAST) in a 96 well polyvinyl plate (a radioisotopic modification of the ELISA procedure described in Coligan, "Current Protocols In Immunology", Unit 7.12.4, Vol. 1, Wiley & Sons, 1994), except that purified polyclonal goat antibodies specific for mouse C chains were used in lieu of antibodies specific for human Fab. To detect anti-Lac-Z IgE, the plates were coated with .beta.-galactosidase (10 .mu.g/ml). The lowest IgE concentration measurable by the assay employed was 0.4 ng of IgE/ml.

[0134] Measuring specifically the anti-antigen response by each group of mice, as shown in FIG. 4, anti-Lac-Z IgE levels in the ISS-ODN containing plasmid injected mice were consistently low both before and after boosting (averaging about 250 CPM in RAST), while the protein injected mice developed high levels of anti-Lac-Z, particularly after the first antigen booster injection, when anti-Lac-Z levels in the mice rose to an average of about 3000 CPM. Consistent with acquisition of tolerance, anti-Lac-Z IgE levels in the protein injected mice declined over time, but continued to rise in the control mice who had not received any immunization to .beta.-galactosidase.

[0135] These data show that the ISS-ODN containing plasmid injected mice developed an antigen specific Th1 response to the plasmid expression product with concomitant suppression of IgE production, while tolerance was acquired in the protein injected mice only after development of substantially higher levels of antigen specific IgE antibodies.

EXAMPLE VI

IL-4, IL-5, IL-10 and INF.gamma. Levels, and CD4+ Lymphocyte Proliferation, in Mice After Delivery of ISS-ODN

[0136] BALB/c mice were injected intravenously with 100 .mu.g of DY1018, DY1019 or a random sequence control (DY1043) then sacrificed 24 hrs later. Splenocytes were harvested from each mouse.

[0137] 96 well microtiter plates were coated with anti-CD3 antibody (Pharmingen, La Jolla, Calif.) at a concentration of 1 .mu.g/ml of saline. The anti-CD3 antibody stimulates T cells by delivering a chemical signal which mimics the effects of binding to the T cell receptor (TCR) complex. The plates were washed and splenocytes added to each well (4.times.10.sup.5/well) in a medium of RPMI 1640 with 10% fetal calf serum. Supernatants were obtained at days 1, 2 and 3.

[0138] Th2 cytokine (IL-4, IL-5 and IL-10) levels were assayed in the supernatants using a commercial kit; Th1 cytokine (INF.gamma.) levels were assayed with an anti-INF.gamma. murine antibody assay (see, e.g., Coligan, "Current Protocols in Immunology", Unit 6.9.5., Vol. 1, Wiley & Sons, 1994). Relatively high levels of IL-4 and IL-10 with low levels of INF-.gamma. would be expected in mice with a Th2 phenotype, while relatively low levels of IL-4 and IL-10 with high levels of INF-.gamma. would be expected in mice with a Th1 phenotype. Relatively high levels of IL-5 characterize a proinflammatory milieu, while the converse is true of relatively low levels of IL-5.

[0139] As shown in FIGS. 5 and 6, levels of anti-CD3 stimulated IL-4 and IL-10 secretion in DY1018 treated mice were substantially lower than in the control mice. Levels in the DY1019 mice were intermediate. Levels of pro-inflammatory IL-5 were reduced in DY1018 treated mice to a comparable extent (FIG. 7).

[0140] Levels of T cell proliferation in response to antigen challenge were greatly reduced in DY1018 (ISS-ODN) treated mice as compared to DY1019 (mutant ISS-ODN) treated and control mice. This suppression of T cell proliferation was reversible on administration of IL-2, demonstrating that the suppression was due to Th2 anergy in the ISS-ODN treated mice (see, Table below).

5 Treatment Control (CPM) ISS-ODN (CPM) M-ODN (CPM) OVA (50 .mu.g/ml) 40680 .+-. 5495 15901 .+-. 4324 42187 .+-. 13012 OVA + IL-2 (1.5 65654 .+-. 17681 42687 .+-. 6329 79546 .+-. 10016 ng/ml) OVA-IL-2 (15 60805 .+-. 19181 57002 .+-. 10658 60293 .+-. 5442 ng/ml)

[0141] Levels of Th1 stimulated IFN-.gamma. secretion were greatly increased in the DY1018 treated mice, but substantially reduced in the DY1019 treated mice (as compared to the control), indicating stimulation of a Th2-type milieu in the latter mice (FIG. 8). Additional data demonstrating these results are shown in the Table below. "b/f" in the Table refers to before; "1st", "2nd" and "each" refer to administration of the compound before the 1st or 2nd antigen challenge.

[0142] Importantly, treatment of mice before antigen challenge is even more effective in shifting the immune response on antigen challenge to a Th1 phenotype than is post-challenge treatment. As shown in FIGS. 9 and 10, antigen primed (but unchallenged) animals injected with ISS-ODN DY1019 72 hours before antigen challenge (with .beta. galactosidase) mounted a more robust Th1-type immune response to the antigen than did their post-challenge treated littermates or littermates treated pre-challenge with a mutant, inactive oligonucleotide (DY1019), as measured by increased IFN.gamma. secretion (FIG. 9) and CD4+ lymphocyte proliferation (FIG. 10).

6 Set # IL-5(pg/ml) IFN-.gamma.(pg/ml) 1 (naive) <20 <20 2 (ISS) in b/f 1st 466 .+-. 40 246 .+-. 86 3 (ISS) in b/f 2nd 531 .+-. 109 168 .+-. 22 4 (ISS) in with 2nd 575 .+-. 90 98 .+-. 44 5 (ISS) in b/f each 200 .+-. 66 443 .+-. 128 6 (ISS) ip; b/f 1st 190 .+-. 52 664 .+-. 61 7 (ISS) ip; b/f 2nd 421 .+-. 102 252 .+-. 24 8 (ISS) ip; with 2nd 629 .+-. 110 104 .+-. 15 9 (ISS) ip; b/f each 121 .+-. 18 730 .+-. 99 10 (ISS) it; b/f each 191 .+-. 49 610 .+-. 108 11 (M-ISS) in; b/f each 795 .+-. 138 31 .+-. 22 12 (M-ISS) ip; b/f each 820 .+-. 122 33 .+-. 33 13 (M-ISS) it; b/f each 657 .+-. 52 102 .+-. 57 14 (steroid) sc; b/f each 424 .+-. 90 <20 15 (steroid) sc; daily 252 .+-. 96 <20 16 (control) not treated 750 .+-. 124 24 .+-. 21

[0143] Further, ISS-ODN administered according to the invention suppress Th2 cytokine release from Th2 sensitized mouse cells (splenocytes harvested from OVA-primed mice, then incubated for 72 hours with 100 .mu.g/ml OVA in vitro). ISS-ODN treatment took place either 1 (-1) or 3 (-3) days before sacrifice. These data are shown below:

7 Group IL-3 (pg/ml) IL-5 (pg/ml) IFN-.gamma. (pg/ml) Control 1299 .+-. 89 657 .+-. 52 <20 ISS-ODN (-1) 309 .+-. 26 112 .+-. 18 <20 ISS-ODN (-3) 463 .+-. 48 144 .+-. 27 <20 ISS-ODN (-1) 964 .+-. 81 508 .+-. 77 <20

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed