AU2013203959A1 - Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs - Google Patents

Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs Download PDF

Info

Publication number
AU2013203959A1
AU2013203959A1 AU2013203959A AU2013203959A AU2013203959A1 AU 2013203959 A1 AU2013203959 A1 AU 2013203959A1 AU 2013203959 A AU2013203959 A AU 2013203959A AU 2013203959 A AU2013203959 A AU 2013203959A AU 2013203959 A1 AU2013203959 A1 AU 2013203959A1
Authority
AU
Australia
Prior art keywords
antigen
brm
immune response
effective dose
cells
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
AU2013203959A
Inventor
Adiran Ion Bot
Thomas Kundig
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.)
Mannkind Corp
Original Assignee
Mannkind Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2012201956A external-priority patent/AU2012201956B2/en
Application filed by Mannkind Corp filed Critical Mannkind Corp
Priority to AU2013203959A priority Critical patent/AU2013203959A1/en
Publication of AU2013203959A1 publication Critical patent/AU2013203959A1/en
Priority to AU2016259332A priority patent/AU2016259332A1/en
Abandoned legal-status Critical Current

Links

Abstract

CkNRFPobI\DCC\SCG4249I-..I.D OC./ A412 The present invention includes use of a biological response modifier (BRM) to modulate an immune response in a subject while avoiding or limiting common side-effects associate with BRM use. The BRMs of the present invention can be injected into a secondary lymphatic organ of the subject or into an area of relatively high drainage into a secondary lymphatic organ of the subject. The modulated immune response can be general or antigen-specific.

Description

H-\eg\Im o~veIAnfRPofbhDCC\SCG\424335_I .doc-9/0J2013 METHODS TO TRIGGER, MAINTAIN AND MANIPULATE IMMUNE RESPONSES BY TARGETED ADMINISTRATION OF BIOLOGICAL RESPONSE MODIFIERS INTO LYMPHOID ORGANS This is a divisional of Australian Patent Application No. 2012201956, which is a divisional of Australian Patent Application No. 2005321940, the entire contents of which are incorporated herein by reference. CROSS REFERENCE TO RELATED APPLICATIONS This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 60/640,727, filed on December 29, 2004, entitled METHODS TO TRIGGER, MAINTAIN AND MANIPULATE IMMUNE RESPONSES BY TARGETED ADMINISTRATION OF BIOLOGICAL RESPONSE MODIFIERS INTO LYMPHOID ORGANS; the disclosure of which is incorporated herein by reference in its entirety. BACKGROUND OF THE INVENTION Field of the Invention [00011 Embodiments described herein relate to methods to improve the efficacy and therapeutic index of a broad range of biological response modifiers that function as potentiators or modulators of immune responses mediated by B cells, CD4' T cells, CD8* T cells (including helper, regulatory, and/or cytotoxic T lymphocytes), as well as NK and NKT cells, by administering such biological response modifiers into secondary lymphoid organs such as lymph nodes. Description of the Related Art [00021 Immune responses can include the generation of antibodies, T helper (Th) cells and cytotoxic T lymphocytes (CTLs). Administration of biological response modifiers (BRMs) offers the possibility of manipulating immune responses since the co-stimulatory environment can decisively influence the quality and magnitude of immune responses. There are three classes of such molecules: [00031 1. Cytokines, chemokines and co-stimulating molecules. [0004] 2. Molecules that trigger cytokine, chemokine formation and modulate the expression of co-stimulating molecules. 100051 3. Antagonists or agonists for cytokines, chemokines and co-stimulating molecules or their receptors, for example, including antibodies, siRNA and antisense oligos. - 1 - WO 2006/071934 PCT/US2005/047250 [OO06] Current immunization protocols typically deliver antigens and BRMs to non-lymphoid areas. Examples of such delivery include intravenous (i.v.), intramuscular (i.m.), subcutaneous (sc.), intradermual (i.d.) injection, and the like. Also, such non-lymphatic delivery generally requires that the BRMs are delivered at high concentrations or in large quantities. One problem with the non-lymphatic delivery of BRMs is that there are often severe adverse effects (including adverse clinical events). The undesirable side effects associated with non-lymphatic administration of many potent BRMs include toxicity and inflammation, and are due to excessive stimulation of signal transduction pathways that, although resulting in immune modulation, have collateral effects. In the short term such activation of the immune system may lead to flu-like symptoms including fever, muscle pain, joint pain, fatigue, malaise, and/or nausea and the like. Over the- long term more serious symptoms may develop. For example, both, interferon-alpha and interferon-beta have been associated with autoimmune phenomena (Weber RW, Curr Opin Allergy Clin Immunol. 4:277-83, 2004 (incorporated herein by reference in its entirety)). Also, repeated administration of vaccine adjuvants such as CpG has been found to cause lymphoid follicle destruction and immunosuppression (Heikenwalder M, et al., Nat Med. 10(2):187-92, 2004 (incorporated herein by reference in its entirety)). [0007] The toxicity of treatment with co-stimulatory cytokines, exemplified by the major T cell growth hormone IL-2 is associated with severe toxicity resembling the clinical manifestations of septic shock. A study by Atkins et al., analyzed the side-effects of such treatment in 270 patients. (Atkins, MB, et al., Journal of Clinical Oncology, 17(7): 2105, 1999 (incorporated herein by reference in its entirety)). In the study, hypotension occurred in the majority (64%) of patients. Other observed symptoms included supraventricular tachycardia (17% of patients), adult respiratory distress syndrome (4% of patients), respiratory failure, coma (1% of patients), elevations of creatinine levels, and elevated bilirubin levels. Moreover, nausea, vomiting, and mental status changes were common among the observed patients. Infections occurred frequently (15% of patients), and life threatening sepsis occured in 3% of the patients. In addition, 2.2% of the 270 patients observed in the study died from treatment-related toxicity. Severe toxicity was also observed for another co-stimulatory cytoldne, IL-12, in a phase I study where fever/chills, fatigue, nausea, vomiting, headache, anemia, neutropenia, lymphopenia, hyperglycemia, thrombocytopenia, and hypoalbuminemia were observed. In a phase I study, of the -2- WO 2006/071934 PCT/US2005/04725G 17 patients receiving IL- 12 in the phase II study, 12 patients were hospitalized and 2 patients died, (Reviewed in M. Colombo and G. Trinchieri, Cytokine & Growth Factor Reviews, 13(2):155-168, 2002 (incorporated herein by reference in its entirety)). Thus, many potentially useful BRMs cannot be effectively used because of the severe side effects, particularly when delivered non-lymphatically (e.g., intravenously, intramuscularly, subcutaneously or intradermally). [00081 For example, signal transduction initiated by toll-like receptors (TLRs); such as TLR9 - a natural receptor for umethylated CpG motifs - results in activation of transcription factors including NF-kB,. with substantial impact on the innate and adaptive immunity. However, practical application of new adjuvants such as CpG oligodeoxynuclotides (CpG ODNs) remains a challenge since prominent systemic activation of NF-kB may result in severe side effects reminiscent of septic shock, thus limiting their therapeutic index (TI) and practical effectiveness. Similarly, non-lymphatic use of other ligands for TLRs such as double stranded RNAs (dsRNAs) can result in activation of pathways resulting in generation of TNF-alpha, thus mimicking a toxic/septic syndrome and precluding safe and effective clinical use of such reagents. Indeed, although the immunopotentiating effects of dsRNA have long been known from in vitro studies, attempts to us it as an adjuvant in vivo have repeatedly failed due to its toxic effects, Comparably based concern over toxicity has limited the clinical use of imiquimod and resiquimod to topical applications. [00091 As another example, 1I-2, IL-12, and interferons, despite the potential benefit of their potent effects on T cell proliferation and innate immunity, are extremely toxic when delivered non-lymphatically, Further, antibodies that are strong modulators of T cell response and blockers of T cell tolerance, such as anti CTLA4, are quite toxic and result in autoinmune syndromes when delivered non lymphatically. Finally, antibodies that block the generation of T1 cells (such as anti IFN-gamma, anti-IL-12) and cytokines that promote the generation of T regulatory responses (such as TGF-beta, IL-10, IL-4), have potential toxic effects (e.g., impairing immune responses), and/or limited efficacy due to pharmacokinetics when delivered non-lymphatically. In fact, due to considerations primarily related to the TI, for 70 years only one adjuvant, alum, was approved for large scale vaccinations despite substantial progress in the area of immune manipulation. [0010] Methods to improve on the safety and efficacy of BRM use are the desired and described below. -3- WO 2006/071934 PCT/US2005/047250 Summary of the Invention [0011] Some embodiments relate to methods of improving the therapeutic index of a BRM. The methods can include, for example, the steps of administering a BRM to a secondary lymphoid organ of a subject in a lymphatically effective dose wherein the lymphatically effective dose avoids or reduces a BRM-related adverse clinical event as compared to use of a non-lymphatically effective dose of the BRM. The modulated immune response can be antigen-specific. The antigen can be endogenously present in the secondary lymphoid organ of the subject The method further can include detecting little or no systemic toxicity or inflammation, for example, by detecting the presence or absence of toxicity or inflammation. Preferably, a lymphatically effective dose is at least 10-fold less than a non lymphatically effective dose. The administering step can include co-administration of the antigen with the BRM. The administering step can include administering the antigen proximal in time to the BRM. The administering step can include administering a lymphatically effective dose of a BRM directly to a secondary lymphoid organ of the subject, The administering step can include administering the antigen to an identified area of relatively high lymphatic drainage that drains to a secondary lymphoid organ of the subject. The administering step can include administration directly to a lymph node or lymph vessel. The modulation of the immune response can include an increased or a decreased response to the antigen. The modulation of the immune response can include a shift toward a humoral response to the antigen, a shift in relative proportions of individual antibody isotypes of antigen-specific antibodies, a shift toward a cellular response to the antigen, a shift toward a Ti response to the antigen, a shift toward a T2 to the antigen, a shift toward a CTL response to the antigen, a shift toward a T helper cell response to the antigen, a shift toward a T regulatory cell response to the antigen, or any other modulation. The BRM can include a cytokine, for example, IL-12, IL-18, GM-CSF, flt3-ligand, interferons, TNF-alpha, and the like. The BRM can include a chemokine, for example, IL-8, MlP-3alpha, MIP-lalpha, MCP-1, MCP-3, RANTES, and the like. The BRM can include a toll-like receptor ligand, for example, dsRNA, or a DNA comprising a CpG sequence. The DNA can be, for example, an oligonucleotide, a plasmid, or the like. The toll-like receptor ligand can be, for example, peptidoglycan, LPS, LPS analogues, flagellin, lipoteichoic acid, imiquiniode, resiquimod, microbial nucleic acids, CpG-containing oligonucleotides, ds RNA, polyl:C, and the like. The -4- WO 2006/071934 PCT/S2005/04725 BRM can also comprise a small molecule, antibody, or engineered soluble ligand that acts as an agonist or an antagonist specific for a target selected from the group consisting of cellular receptors, co-stimulatory receptors, cytokine receptors, chenoline receptors, signal transduction elements, and transcriptional regulators. The BRM can include an antibody specific for a co-stimulatory receptor, for example, anti-CD40, anti-CTLA4, anti-CD3, anti-CD28, and anti-OX40; or other agonists of these receptor molecules. Similarly the BRM can be an antagonist of an inhibitory receptor, for example an anti-CD25 antibody. Further, the BRM can be a molecule that increases the activity of T regulatory cells, such as TOF-, IL-10, 11-4 antibodies or antagonists for pro-inflammatory cytokines. Preferably, the antigen is not the BRM. The antigen can include a tumor-associated antigen, for example. Also, the antigen can include a microbial antigen, for example, one associated with a protist, fungi, bacterium, virus, prion, or the like. The antigen can also include an allergen, a toxin or toxoid, a disease-matched antigen, and the like. [0012] Other embodiments relate to methods of modulating an antigen specific immune response, the method comprising the step of administering a BRM to a secondary lymphoid organ of a subject in a dose sufficient to obtain a modulated immune response. [00131 Other embodiments relate to methods of modulating an immune response to an antigen that include, for example, co-administering a BRM and said antigen to a secondary lymphatic organ whereby the antigen-specific immune response is modulated, wherein the modulation does not consist essentially of an augmented CTL response. [00141 Still further embodiments relate to methods of generating an immune response while minimizing adverse side effects of a biological response modifier, which methods can include, for example, administering a BRM to a secondary lymphoid organ, wherein the BRM can include a toll-like receptor ligand. [0015] Some embodiments can alternatively include observing less severe side effects than would result from a non-lymphatically effective dose. [0016] In some aspects, a dose of BRM is delivered that is effective at triggering, maintaining or manipulating an immune response, while having little or no systemic or local toxicity. [0017] Some embodiments relate to use of a BRM in the manufacture of a medicament suitable for administration to a secondary lymphoid organ of a subject. -5- WO 2006/071934 PCT/US2005/04725@ {0018] Some embodiments relate to use of a BRM in the manufacture of a medicament suitable for modulating an immune response in a subject without causing a BRM-related adverse clinical event. [0019] Some embodiments relate to use of a BRM in the manufacture of a medicament that increases the therapeutic index of the BRM and that is suitable for modulating an immune response in a subject. [0020] Some embodiments relate to use of a BRM and an antigen in the manufacture of a medicament.for modulating an immune response in a-subject that is specific to said antigen and that is suitable for injection into a secondary lymphoid organ of a subject. [0021] Other embodiments relate to a composition comprising a lymphatically effective dose of a BRM, wherein said lymphatically effective dose comprises an amount of a BRM that is relatively nontoxic and wherein said lymphatically effective dose is less than a non-lyphatically effective dose. [0022] Other embodiments relate to a composition comprising a lymphatically effective dose of a BRM, wherein said lymphatically effective dose comprises an amount of a BRM that is relatively non-toxic and wherein the amount of the BRM is insufficient to be a non-lyphatically effective dose. [0023] Other embodiments relate to a composition comprising a lymphatically effective dose of a BRM and an antigen, wherein said lymphatically effective dose comprises an amount of a BRM that is relatively non-toxic and wherein the amount of the BRM is insufficient to be a non-lyphatically effective dose. In preferred embodiments the lymphatically effective dose of a BRM used in such a composition is at least ten-fold less than the corresponding non-lyphatically effective dose. Brief Descrition of the Drawings [0024] Figure 1 shows that the induction of acute phase reaction (A) and increased spleen cellularity as an indication of splenomegaly (B) occurred at a dose of lOnM of CpG ODN irrespective of the tested route, but did not occur at lower doses. [0025] Figure 2 presents the effect of titrated CpG ODN treatment on splenocyte subset composition. [0026] Figure 3 shows the effect of CpG ODN treatment on in vivo maturation of dendritic cells measured as CD80 and CD86 expression. -6- WO 2006/0}71934 PCT/U1S2005/047259 [0027] Figure 4 shows the induction of cytotoxic responses against a model antigen (OVA) by co-administration of antigen and CpG ODN at doses devoid of systemic acute phase reaction. 10028] Figure 5 shows the induction of immunity to a cancer antigen by intralymph node co-administration of a selected peptide epitope with two different BRMs. [0029] Figure 6 shows the induction of functional immunity resulting in the clearance of human tumor cells within lung parenchyma, by intra lymph node co administration of a selected peptide epitope with two different BRUs. [0030] Figure 7 shows decreased immune responsiveness to the self PSMA 288-297 epitope as compared to the non-self PSMA 730-739 epitope by subcutaneous immunization in combination with IFA. [0031] Figures SA-8E show induction of cytotoxic response by co administration of various peptides with adjuvant (polyI:C) into lymph nodes using self and non-self epitopes. 10032] Figure 8A shows lysis induced by immunization with PSMA 288 297 on T2 cells pulsed with the peptide. {0033] Figure 8B shows lysis induced by immunization with PSMA 730 739 on T2 cells pulsed with the peptide. [0034] Figure 8C shows lysis induced by immunization with PRA E 425-433 and PRAME 300-309 on T2 cells pulsed with the peptide. [0035] Figure 8D shows lysis induced by immunization with NY-ESO-I 157-165 on T2 cells pulsed with the peptide on the left and on 624 cells transformed to express a GFP/NY-ESO-1 fusion protein on the right. [0036] Figure SE shows lysis induced by immunization with PSMA 288 297 on a PSMA-expressing human tumor cell line (LNCap). [0037] Figure 9 shows the induction of antibody response against PLA2 by co-adminisfration of antigen and CpG ODN at doses devoid of systemic acute phase reaction. [0038] Figure 10 shows an isotype profile of anti-PLA2 serum antibodies in CBA/J mice vaccinated on day 1, 15 and 29 with 0.1 ptg PLA2 and the indicated Toll-like receptor ligands or the reference adjuvant AI(OH)3. [0039] Figure 11 shows intracellular IFN-y in the CD4+ and CD8+ T cells of mice injected with phospholipase A2 and various TLR ligands. -7- WO 2006/071934 PCT/US2005/047250 [0040] Figure 12 shows the levels of secretion of cytokines in vitro (in mice injected with phospholipase A2 and various TLR ligands). [0041] Figure 13 shows the percent of CD8+ and CD44+ cells positive for intracellular IFN-y following in vitro restimulation with antigen (following intranodal or subcutaneous administration ofimniquimod during immunization). [00421 Figure 14 depicts a model explaining the efficacy and improved TI by co-administration of antigen and BRMs into lymph nodes. Detailed description of the preferred embodiments Definitions [0043] Non-lymphatic administration - In preferred embodiments, non lymphatic administration means delivery in a manner such that the effects of the delivered substance are seen throughout the body. For example, this can include intravenous administration, as well as administration subcutaneously, intradermally, intramuscularly, orally, or in any other manner so that the agent can be absorbed into the tissues or fluids of the body .generally. In other embodiments, non-lymphatic administration can mean delivery in a manner such that the effects of the delivered substance are seen in substantial portions, regions, tissues, systems, or the like, of the body, even if not seen in all parts thereof [0044] Intralymphatic administration - In preferred embodiments, intralymphatic administration means delivery in a manner such that effects are focused generally in the lymphatic system; that is, the agent is preferentially absorbed into one or more organs of the lymphatic system, In various embodiments, intralymphatic administration can include administration(s) directly into a primary or secondary lymphoid organ, or into a lymphatic vessel or to a site of relatively high lymphatic drainage, In preferred embodiments delivery is into a secondary lymphoid organ or into a vessel or to a site of relatively high lymphatic drainage. In some embodiments, administering a BRM to a secondary lymphoid organ of a subject can be done directly by administering a BRM directly to the secondary lymphoid organ or indirectly by administering the BRM to an identified area of relatively high lymphatic drainage. [0045] Therapeutic Index (TI) - In some embodiments this can be defined quantitatively as the ratio of LD 5 o to ED 5 0 . EDSo is the dose of a drug that is pharmacologically effective for 50% of the population exposed to the drug or a 50% response in a biological system that is exposed to the drug, LD 5 0 is the chemical dose -8- WO 2006/071934 PCT/US2005/047250 lethal to 50 percent of a test population In other preferred embodiments the term also can be defined qualitatively to indicate the difference between the dose at which an agent becomes medically useful and the dose at which undesirable effects (adverse clinical events) become apparent, or impair or eliminate usefulness, without reference to any particular quantitative measurement. [0046] Secondary lymphoid organs - organs where lymphocytes reside and encounter antigen. Examples include lymph nodes, adenoids and tonsils, the appendix, the spleen, and peyer's patches of the gut. Generally, primary lymphoid organs are those in which lymphocytes develop (thymus, bursa, bone marrow), [0047] Non-lymphatically effective dose - The amount of a BRM or antigen effective to modulate an immune response to an antigen in order to obtain a desired outcome when using non-lymphatic administration. In alternative embodiments the immune response is characterized without reference to antigen specificity. [0048] Lynphatically effective dose - The amount of a BRM or antigen effective to modulate an immune response to obtain a desired outcome when using intralymphatic administration, In some embodiments, the immune response is characterized without reference to antigen specificity. In other embodiments, the immune response is antigen-specific. Generally a lymphatically effective dose can be small enough to reduce or avoid adverse clinical events that can occur from use of a non-lymphatically effective dose. [00491 Modulate an immune response - Any change in an immune response. The change can involve an increase, decrease, shortening, prolongation, shift in time of appearance, peak, or disappearance, of any parameter of the response, or a change in the balance of components of the response that can result in a desired outcome. The response can comprise B cells, CD4* T cells, CD8* T cells (including helper, regulatory, and / or cytotoxic Tjlymphocytes), NK cells, and NKT cells, and any of the soluble molecules they produce in response to antigenic stimulation. In preferred embodiments the change in an immune response is specific to one or more specific antigens. 100501 BRM-related adverse clinical event - Any systemic or localized side-effect arising from administration of a BRM that is undesired including without limitation acute phase reaction, flu-like symptoms, fever, chills, muscle pain, joint pain, fatigue, malaise, nausea, vomiting, headache, anemia, neutropenia, -9- WO 2006/071934 PCT/US2005/047250D lymphopenia, hyperglycemia, thrombocytopenia, hypoalbuminenia, systemic immune activation, splenomegaly, lymphoadenopathy, toxicity, septic shock, hypotension, supraventricular tachycardia, adult respiratory distress syndrome, mental status changes, elevated bilirubin levels, elevated creatinine levels, and the like. [0051] Desired outcome - Any positive effect on the disease or condition being treated. In some embodiments, the positive effect comprises a decrease in the severity, frequency, or duration of one or more symptoms caused by the disease or condition being treated. In other embodiments, the positive effect comprises a decrease in the mortality of the disease or condition being treated. In some embodiments, the positive effect comprises remission of the disease or condition being treated. In yet other embodiments, the positive effect comprises elimination of the disease or condition being treated. In still other embodiments the desired outcome comprises a detectable increase, decrease, shift, or other modulation of an immune response as compared to what is, could be, or would be expected to be, obtained without use of a BRM. [0052] Embodiments described herein relate to methods to trigger, maintain, and manipulate immune responses by targeted administration of biological response modifiers (BRMs) to lymphoid organs. . Some embodiments relate to methods to improve the efficacy and therapeutic index (TI) of a broad range of BRMs by administering the BRMs into secondary lymphoid organs, such as lymph nodes. The BRMs can act as potentiators or modulators of specific immune responses mediated, for example, by B cells, T helper (Th) cells and / or cytotoxic T lymphocyte (CTL) cells. Many BRMs are underutilized or not utilized at all in many contexts because of their adverse side effects. Further embodiments relate to methods of utilizing such BRMs while avoiding some or all of their adverse side effects. [0053] Some embodiments relate to methods that are based upon the unexpected observation that intralymphatic administration of a BRM results in an improved therapeutic index for the BRM, such that immunological effectiveness can be obtained while using doses small enough to avoid clinically relevant toxicity. It was not clear a priori whether administration to secondary lymphoid organs (where the interaction between various cell populations important for the generation of immune responses is optimal) of BRMs, as described herein, with various types of antigens would result in improvement of TI or efficacy of vaccination or active immunotherapy, since the normal sequence of antigen exposure and subsequent -10- WO 2006/071934 PCT/US2005/047250 interaction between antigen presenting cells (APCs), T and B cells is different. Thus, it was previously unknown whether inadvertent stimulation or over-stimulation of immune cells by BRMs might have resulted in decreased responses or toxicity. As described herein it has been discovered that this is not the case. Some embodiments relate to a methodology to address these issues and take full advantage of classes of existing or potential BRMs that although potent, pose safety considerations when delivered non-lymphatically. [00541 In some preferred embodiments, the methods can include the administration of BRMs to secondary lymphoid organs, or administration into the lymphatic vessels draining into such secondary organs. In the lympboid organs, antigen presenting cells (APCs), innate immune cells, B cells, Th cells and CTLs are in geographic proximity, for example, within the same microenvironment. Thus, the predominant locus for generation of immune responses is within secondary lymphoid organs where the proximity between these cells is optimal. Some embodiments relate to methods of delivering BRM at dose that triggers, maintains, manipulates or otherwise modulates an immune response, and which also avoids localized toxicity. [0055] In some embodiments the methods can include co-administration of antigen with the BRM. However, in other embodiments the methods relate to modulating the immune response to antigen that is already present, but unrecognized or only recognized in a suboptimal fashion by the immune response within the lymphoid environment. One example is metastatic disease in draining lymph nodes of a tumoral process. In such embodiments the antigen preferably is not co administered, while in some embodiments, it can be co-administered. [00561 In some embodiments the immune response is characterized without reference to a specific antigen. Instead the modulation is of the general character of an immune response such as a shift toward or away from a TI or T2 response, or changing the absolute or relative amount of an antibody isotype or subset of T cells, or stimulating or inhibiting the proliferation or activity of any of the cell types of the immune system (including, for example, B cells, T cells, NK cell, NKT cells, and antigen presenting cells and the like). [0057] Administration into the secondary lymphoid organs can result in increased TI by increasing the concentration of BRM in the microenvironment where the antigen is processed and presented to immune cells, while minimizing non lymphatic exposure to BRMs. Both antibody and T cell responses (such as Th cells -11- WO 2006/071934 PCT/US2005/047250 and CTLs) can be increased or optimized by this methodology, without undesired non-lymphatic exposure to problematic amounts of BRMs. In fact, the overall efficacy of combinations of antigen and BRM can be substantially increased by this methodology. In general, such methods can be used to trigger, amplify, suppress, or restore immune responses against microbial and tumor-associated antigens, in prophylactic or therapeutic fashion. [0058] Potential target microbes include, without limitation, hepatitis viruses (e.g., C, B and delta), herpes viruses, HIV, HTLV, HPV, EBV, and the like. Potential target tumor antigens include, without limitation, cancer-testes antigens (e.g., NYESO-1, PRAME, SSX2, MAGE, LAGE, GAGE, etc.), tissue specific antigens (e.g., Melan-A, tyrosinase, gp100, PSMA, etc.) and oncofetal antigens (e.g., CEA, etc.), associated with carcinomas, sarcomas and other types of tumors, and the like. Numerous exemplary epitopes, epitope analogs, and combinations or antigens from tumor antigens and methods of using the same are found in U.S.'Application Nos. 10/117,937, filed on April 4, 2002, 10/657,022, filed on Septebmer 5, 2003, 11/067,159 (filed on February 25, 2005, Pub. No. 20050221440), 11/067,064 (filed on February 25; 2005, Pub. No. 20050142144) all entitled EPITOPE SEQUENCES; Application No. 10/8714708 and Provisional Application No. 60/580,969, both filed on June 17, 2004 and entitled COMBINATIONS OF TUMOR ASSOCIATED ANTIGENS IN COMPOSITIONS FOR VARIOUS TYPES OF CANCERS; and U.S. Provisional Application Nos, 60/581,001 and 60/580,962, both filed on June 17, 2004, and respectively entitled SSX-2 ANALOGS and NY-ESO PEPTIDE ANALOGS, each of which is incorporated herein by reference in its entirety. Vectors or formulations to deliver antigens can include peptides, recombinant proteins, viruses or bacteria, recombinant nucleic acids or cells encompassing the above listed agents, and the like. [0059] Exemplary BRMs can include, without limitation, cytoidnes such as IL-12, IL-18, GM-CSF, flt3 ligand (flt3L), interferons, TNF-alpha, and the like; and chenokines such as IL-8, MIP-3alpha, MIP-lalpha, MCP-1, MCP-3, RANTES, and the like. In addition, BRMs can be, without limitation, molecules that trigger cytokine or chemokine production, such as ligands for TLRs (peptidoglycans, LPS or analogues, CpG ODNs, dsRNAs, small molecules that bind to TLRs such as imiquimode, and the like). Antibodies, small organic molecules, and engineered soluble ligands that bind to co-stimulatory molecules (anti-CD40, CTLA-4, anti -12- WO 2006/071934 PCT/US2005/04725@ OX40, and the like), cellular receptors, cytokine receptors, chemokine receptors, signal transduction elements, or transcriptional regulators can be used as BRMs in the context described herein. Finally, BRMs can include small interfering RNA (siRNA) that alter expression of proteins which in turn are regulators of the immune system. [0060] Further, the disclosed methods can, for example, be used to modulate or suppress undesired immune responses associated with inflammatory, allergic or autoirmmune disorders, by enabling immune deviation, generation of T regulatory cells or immune tolerance. For example, one or more BRMs which suppress certain immune responses or which modulate responses in a manner that is favourable for the above-mentioned disorders can be administered alone or in combination with an antigen to a secondary lymphoid organ. The one or more BRMs can be delivered in a low amount, which can minimize or avoid the adverse effects normally associated with the one or more BRMs. For Ti-mediated autoimmune diseases, antigens can include epitopes derived from insulin, GAD65, myelin basic protein, proteolipid protein, MOG, collagen II, heat shock proteins, etc. BRMs may be Kr,4, TGF-beta, 1L-10, and the like; or molecules that trigger their production. In addition, BRMs may be antibodies that enable the mechanisms listed above. Examples include anti-IFN-gamma, anti-IL-12, and including their use in combination with the BRMs listed above to amplify the overall effect. For T2 mediated diseases, the BRMs that can be used include Ti-activating antigens, including those listed in the previous section. Together, such methods can be applied prophylactically or therapeutically in diseases such as autoimmune diabetes, multiple sclerosis, rheumatoid arthritis and various allergies including asthmatic disorder with allergic aetiology [0061] As used herein BRM can refer to any molecule that modulates the activity of the immune system, or the cells thereof through an interaction other than with an antigen receptor. BRM is also commonly applied to complex biological preparations comprising such molecules in which the active entity, or entities, without regard for whether the active component(s) of the mixture had been defined, Examples of complex biological preparations used as BRMs include OK-432, PSK, AIL and lentinan. In preferred embodiments of the invention the active component(s) of such a mixture are defined. In other preferred embodiments of the invention BRMs sourced from complex biological preparations are at least partially purified, or substantially purified, for example OK-PSA (Okamoto et al., Journal of the National -13- WO 2006/071934 PCT/US2005/047250) Cancer Institute, 95:316-326, 2003 which is incorporated herein by reference in its entirety) or AILb-A (Okamoto et al., Clinical and Diagnostic Laboratory Immunology, 11:483-495, 2004 which is incorporated herein by reference in its entirety). In particularly preferred embodiments the BRM is of defined molecular composition. BRMs include immunopotentiating adjuvants that activate pAPC or T cells including, for example: TLR ligands, endocytic-Pattem Recognition Receptor (PRR) igands, quillaja saponins, tucaresol, cytoidnes, and the like. Some preferred adjuvants are disclosed in Marciani, D.L Drug Discovery Today 8:934-943, 2003, which is incorporated herein by reference in its entirety. In one review, it was reported that a BRM was injected into a lymph node (Sadao et al,, "Locoregional immunotheraphy-Topics at the 13t and 140" Meeting of the Japanese Research Society for Surgical Cancer Immunology," Biotherapy 9(7):845-851 (1995) (incorporated herein by reference in its entirety)). However, the report provided minimal detail and no guidance regarding the exact response caused by the injection. The report did not disclose any antigen specific immune response, for example, a B cell or T cell response due to the injection. [0062] One class of BRM includes mostly small organic natural or synthetic molecules, which exert immune modulating effects by stimulating pathways of innate immunity. It has been shown that macrophages, dendritic and other cells carry so-called Toll-like receptor (TLRs), which recognize pathogen-associated molecular patterns (PAMPs) on micro-organisms (Thoma-Uszynsld, S. et al., "Induction of direct antimicrobial activity through mammalian toll-like receptors," Science 2001. 291:1544-1547; Aldra, S., "Mammalian Toll-like receptors," Curr Opin Immunol 2003. 15: 5-11; each of which is incorporated herein by reference in its entirety). Ligation of the TLRs activates NF-xB and leads to the production of several important mediators of innate immunity, e.g., IFN-o, IFN-y, TNF-oU, I-I, IL 6, IL-12 and IL-18, and induces the expression of co-stimulatory molecules, ie., B7.1 (CD80), B7.2 (CD86), and CD40 on antigen-presenting cells. It is the presence of these molecules along with presentation of microbial antigens that activates the CD4 T cells required to initiate most adaptive immune responses. [00631 So far, eleven mammalian TLRs have been described together with a multitude of natural and synthetic ligands, e.g,, lipopolysaccharide, various glycans and mannans, the mycobacterial extract muramyl dipeptide, bacterial flagellin, -14- WO 2006/071934 PCTI/US2005/947250 bacterial dsDNA (which contains CpG motifs), viral and synthetic dsRNA (e.g., polyl:C) and viral ssRNA (Ahmad-Nejad, P. et al., Eur J Immunol 2002. 32: 1958 1968 ; Chamaillard, M., et al., Nat Immunol 2003. 4: 702-707; McSorley, S. 1, et al., Jmunol 2002. 169: 3914-3919; Krieg, A. M.,. Annu Rev Immunol 2002. 20: 709 760; Heil, F., et al., Science 2004. 303: 1526-1529; Diebold, S. S.et al., Science 2004. 303: 1529-1531; each of which is incorporated herein by reference in its entirety). CpG especially (TLR-9 ligand) has shown widespread experimental application and clinical potential as adjuvant by allowing efficient maturation of antigen-presenting cells and subsequent activation of antigen-specific lymphocytes (Krieg, A. M., Annu Rev Immunol 2002. 20: 709-760; Weigel, B. J.et al., Clin Cancer Res 2003. 9: 3105 3114; Verthelyi, D.et al., Aids 2004. 18: 1003-1008; Storni, T, et al., Jlmnunol 2004. 172: 1777-1785; each of which is incorporated herein by reference in its entirety). Furthermore, a new generation of purely synthetic anti-viral imidazoquinolines, e.g., imiquimod and resiquimod, has been found to stimulate the cellular path of immunity by binding the TLRs 7 and 8 (Hemmi, H. et al, Nat Immunol 3:196-200, 2002; Dummer, R. et al., Dermatology 207:116-118, 2003; Kadowai, N. et al. JExp Med. 194:863-9, 2001; each of which is incorporated herein by reference in its entirety). Others suggest that these compounds are better viewed as shifting, as opposed to stimulating immune responses (Brugnolo, F. et al. JAllergy Clin Immunol 111:380-8, 2003, which is incorporated herein by reference in its entirety). 10064] The administration of a BRM with an antigen was studied. T cell responses against protein antigens are of interest for treatment and immunization against microbes and tumors. The methods can include the co-administration of a model antigen such as OVA with CpG ODN, which was studied to determine whether their administration results in favorable immune response (CTL) at doses devoid of detectable systemic acute phase reaction. Furthermore, as described more fully herein, melan-A 26-35, is a major, well-described epitope derived from a tumor-associated antigen expressed on melanoma cells, and is a model antigen for co-administered studies with the BRMs listed above. An example of particular BRMs that are potent immune modulators, and also associated with safety concerns when delivered non lymphatically, are unmethylated CpG oligodeoxynuclotides (CpG ODN) and synthetic dsRNA (polyl:C) that bind to TLR9 and TLR3, respectively, on APC and innate immune cells. -15- WO 2006/071934 PCT/US2005/04725D [0065] Since a major potential application of CpG ODNs consists in cancer immunotherapy, it was administered into lymph nodes at low doses (around 0.1 mole) to determine whether it augments immunization with the Melan-A peptide epitope. Mice carrying a transgene that expresses a human-mouse chimeric MHC class I molecule (displaying the human A2 allotype) were immunized with peptide alone or peptide admixed with CpG ODN. The peptide used was the A27L analogue of the A2-restricted Melan-A 26-35 epitope. The analogue is cross-reactive with the natural peptide and displays decreased Kf along with increased immunogenicity. t0066] In addition, the methodologies described herein can be utilized in the area of allergic desensitisation and in generation of antibody responses against a broad range of microbial antigens. Such methods can include the co-administration of an antigen, such as PLA2, along with CpG ODN, which was studied to determine whether such administration results in favorable immune response (antibodies) at doses low in or lacking in detectable systemic acute phase reaction. [00671 Additional variations will be apparent to one of sdil in the art. Various embodiments can specifically include or exclude the use of particular BRMs, antigens, forms of antigen, mode of administration, etc. For example, some embodiments exclude the use of BRMs OIC-432, lentinan, PSKT, IL-2, TNF, and IFN gamma, individually. Other embodiments exclude the use of all of the same or more than one of the same, for example, 2, 3, 4, or 5 of the BRMs. [0068] Additional methodology, compositions, peptides, and peptide analogues are disclosed in U.S. Provisional Application No. 60/581,001, filed June 17, 2004, , and 11/156,253, filed on June 17, 2005, both entitled "SSX-2 PEPTIDE ANALOGS;" U.S. Provisional Application No. 60/5 80,962, filed June 17, 2004,, and 11/155,929, filed on June 17, 2005, both entitled NY-ESO PEPTIDE ANALOGS; U.S. Patent Application No. 09/999,186, filed November 7, 2001, entitled METHODS OF COMMERCIALIZING AN ANTIGEN; U.S. Provisional Application No. 60/640,402, filed on December 29, 2004, and U.S. Patent Application No / __, (Attorney Docket No. MANNK.047A) filed on even date as the instant application, both entitled, METHODS TO ELICIT, ENHANCE AND SUSTAIN IMMUNE RESPONSES AGAINST MHC CLASS I- RESTRICTED EPITOPES, FOR PROPHYLACTIC OR THERAPEUTIC PURPOSES; and U.S. Provisional Application No. 60/640,821, filed on December 29, 2004, and U.S. Patent Application No / _ , (Attorney Docket No. MANNK.048A filed on even date as the instant -16- WO 2006/071934 PCT/US2005/047250 application), both entitled METHODS TO BYPASS CD4+ CELLS IN THE INDUCTION OF AN IMMUNE RESPONSE, each of which is hereby incorporated by reference in its entirety. Beneficial epitope selection principles for immunotherapeutics are disclosed in U.S. patent application Nos. 09/560,465 (filed on April 28, 2000), 10/026,066 (filed on December 7, 2001; Pub. No. 20030215425 Al), 10/005,905 (filed on November 7, 2001), 10/895,523 (filed on Iuly 20, 2004, Pub. No. 20050130920), and 10/896,325 (filed on July 20, 2004) all entitled EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS; U.S. Pat. No. 6,861,234(filed on April 28, 2000) and U.S. patent application No. 10/956,401 (filed on October 1, 2004, Pub. No. 20050069982 Al) both entitled METHOD OF EPITOPE DISCOVERY; U.S. patent application Nos. 09/561,571 (filed on April 28, 2000) entitled EPITOPE CLUSTERS; 10/094,699 (filed on March 7, 2002; Pub. No. 20030046714 Al) and 11/073,347 (filed on June 30, 2005) both entitled ANTI NEOVASCULATURE PREPARATIONS FOR CANCER; and 10/117,937 (filed on April 4, 2002; Pub. No. 20030220239 Al) and 10/657,022 (filed on September 5, 2003; Publication No. 20040180354 Al), and PCT Application No. PCT/US2003/027706 (Pub. No. W004022709A2) all entitled EPITOPE SEQUENCES, and each of which is hereby incorporated by reference in its entirety. Aspects of the overall design of vaccine plasmids are disclosed in U.S. Patent applications 09/561,572 (filed on April 28, 2000) and 10/225,568 (filed on August 20, 2002, Pub, No. 20030138808) both entitled EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS and U.S. Pat. App. Nos. 10/292,413 (filed on November 7, 2002; Pub. No.20030228634 Al), 10/777,053 (filed on February 10, 2004, Pub. No. 20040132088), 10/837,217 (filed on April 30, 2004) all entitled EXPRESSION VECTORS ENCODING EPITOPES OF TARGET ASSOCIATED ANTIGENS AND METHODS FOR THEIR DESIGN; 10/225,568 (filed on August 20, 2002; Pub No. 2003-0138808), PCT Application No. PCT/US2003/026231 (Pub. No. WO 2004/018666) and U.S. Patent No. 6,709,844 and U.S. patent application No. 10/437,830 (filed on May 13, 2003, Pub. No. 20030180949) both entitled AVOIDANCE OF UNDESIRABLE REPLICATION iNTERMEDIATES IN PLASMIND PROPAGATION, each of which is hereby incorporated by reference in its entirety. [0069] Specific antigenic combinations of particular benefit in directing an immune response against particular cancers are disclosed in U.S. Provisional -17- WO 20061071934 PCT/US2005/047250 Application No. 60/479,5 54, filed on June 17, 2003, and U.S. -Patent Application No. 10/871,708, filed on June 17, 2004, PCT Patent Application No. PCT/US2004/019571, U.S. Provisional Application No. 60/640,598, filed on December 29, 2004, and U.S. Patent Application No / _, (Attorney Docket No. MANNK.049A), filed on even date herewith, all entitled COMBINATIONS OF TUMOR-ASSOCIATED ANTIGENS IN VACCINES FOR VARIOUS TYPES OF, each of which is also hereby incorporated by reference in its entirety. t00701 Intranodal administration for the generation of CTL is taught in U.S. Patent Application Nos. 09/380,534; 09/776,232 (Pub. No. 20020007173 Al) now U.S. Patent No. 6,977,074; / (Attorney Dockey No. MANNK.OOICP2CI, filed on December 19, 2005; in PCT Application No. PCTUS98/14289 (Pub. No. W09902183A2) each entitled A METHOD OF INDUCING A CTL RESPONSE and in U.S. Application No. 10/871,707, filed on June 17, 2004, entitled, METHODS TO ELICIT, ENHANCE AND SUSTAIN IMMUNE RESPONSES AGAINST MHC CLASS I-RESTRICTED EPITOPES, FOR PROPHYLACTIC OR THERAPEUTIC PURPOSES, each of which is hereby incorporated by reference in its entirety. [0071] Intranodal administration of allergen for allergy desensitization is taught in U.S. Patent No. 6,773,695 entitled MODULATION OF ALLERGIC RESPONSE, which is hereby incorporated by reference in its entirety. [0072] The integration of diagnostic techniques to assess and monitor immune responsiveness with methods of immunization is discussed more fully in Provisional U.S. Patent Application No. 60/580,964, filed on June 17, 2004, and U.S. Patent Application No. 11/155,928, filed on June 17, 2005, both entitled IMPROVED EFFICACY OF ACTIVE IMMUNOTHERAPY BY INTEGRATING DIAGNOSTIC WITH THERAPEUTIC METHODS, each of which is hereby incorporated by reference in its entirety. [0073] Additional relevant disclosure is present in U.S. Patent App. Nos. 11/155,288 (filed on June 17, 2005) entitled COMBINATIONS OF TUMOR ASSOCIATED ANTIGENS IN COMPOSITIONS FOR VARIOUS TYPES OF CANCERS, 11/156,369 and 60/691,889 (both filed on June 17, 2005) both entitled "EPITOPE ANALOGS," 60/691,579 (filed on June 17, 2005) entitled METHODS AND COMPOSITIONS TO ELICIT MULTIVALENT IMUNE RESPONSES AGAINST DOMINANT AND SUBDOMINANT EPITOPES, EXPRESSED ON -18- WO 2006/071934 PCT[US2005/047250 CANCER CELLS AND TUMOR STROMA, and 60/691,581 (filed on June 17, 2005) entitled MULTIVALENT ENTRAIN-AND-AMPLIFY IMMUNOTHERAPEUTICS FOR CARCINOMA, [0074] The compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990. 100751 The compositions of the present invention can be prepared in combination with an acceptable pharmaceutical carrier. Suitable carriers can contain inert ingredients that do not interact with the compound. Techniques for the preparation of pharmaceutical compositions are well known in the art and for example described in Remington's Pharmaceutical Sciences (Mack Publishing Company, Easton, Pa.,) (which is incorporated herein in its entirety). Suitable pharmaceutical carriers for intravenous and other parenteral administration include, for example, sterile water, physiological saline, bacteriostatic saline (i.e., saline containing about 0.9% mg/mil benzyl alcohol), phosphate-buffered saline, Hank's solution, or Ringer's lactate. [0076] The term "carrier" defines a chemical compound that facilitates the delivery or incorporation of a compound into cells or tissues. 10077] The term "diluent" defines a liquid, for pharmaceuticals generally an aqueous solution, that will dissolve and can be used to dilute a compound of interest (active agent) as well as stabilize the biologically active form of the compound. Salts dissolved in buffered solutions are utilized as diluents in the art. One commonly used buffered solution is phosphate buffered saline because it mimics the salt conditions of human blood. Since buffer salts can control the pH of a solution at low concentrations, a buffered diluent rarely modifies the biological activity of a compound. {0078] The term "physiologically acceptable" defines a carrier or diluent that does not abrogate the biological activity' and properties of a compound (active agent) and is compatible with the functioning of a living organism to which it will be delivered. -19- WO 2006/071934 PCT/US2005/047250 [0079] For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. The compositions may take such forus as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0080) The exact formulation, route of administration and dosage for the pharmaceutical compositions of the present invention dan be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl et aL 1975, in "The Phannacological Basis of Therapeutics", Ch. 1 p. 1, which is incoporated herein in its entirety). Typically, the dose range of the composition administered to the patient can be from about 0.5 to 1000 mg/kg of the patient's body weight. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the patient. Note that for particular compounds disclosed herein, suitable human dosage can be inferred from ED 50 or ID 50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals. Example 1 High doses of CpGs (10 nmol) administered either subcutaneously or intralymphatically induce significant acute phase response and splenomegaly. [00811 It is well known that microbial components such as endotoxin and bacterial DNA can induce severe adverse effects, e.g., systemic immune activation, splenomegaly, lymphoadenopathy and septic shock (reviewed in Schmidt, U., H. Wagner, and T. Miethke, "CpG-DNA upregulates the major acute-phase proteins SAA and SAP," Cell Microbiol 1:61, 1999 (which is incorporated herein in its entirety)). These adverse effects are usually preceded by a so-called acute-phase response that, similar to measuring CRP in humans, can be quantified by measuring the acute phase protein serum amyloid A (SAA). SAA is produced by the liver upon stimulation by proinflammatory cytoldnes such as IL-1, IL-6, and TNF-ct. 100821 Mice were subcutaneously (s.c.) or intralymphatically (i.i) injected with titrated amounts of CpG ODN 1668pt (SEQ. MD. No. 1) (10, 1, 0.1 and 0.01 nmol) and 24 hours later analyzed for the presence of enhanced concentrations of the acute phase protein serum amyloid A (mSAA) in the blood, mSAA levels were -20- WO 2006/071934 PCT/US2005/047250 assessed by ELISA. Elevated serum levels of SAA were observed in mice receiving CpG doses of 10 nmol, regardless of the route of administration. CpG doses lower than 1 nmol did not induce a measurable acute phase response (Figure 1 A). The means ± SEM (n = 3) are shown. These results show that irrespective of the administration route, an acute phase reaction was observed only at the highest dose of CpG (10 nmol). [0083] Since the administration of CpGs at doses of 10 - 20 nmol has been reported to induce splenomegaly with changes in cell composition (Sparwasser, T,, L. et al., "Immunostimulatory CpG-oligodeoxynucleotides cause extramedullary marine hemopoiesis," J Immunol 162:2368, 1999 (which is incorporated herein in its entirety)) these changes were assessed as an additional parameter of adverse effects. C57BL/6 mice were treated with different doses of CpG and euthanized 8 days later for analysis of the spleen. Similar to mSAA levels, splenomegaly and changes in cell composition were observed only with the high doses of CpG (Figure 1 B). The means ± SEM (n= 3) are shown. At 10 nmole, splenocyte numbers increased about 2-3-fold for both immunization routes. Only moderate changes were observed for B lymphocytes (Figure 2 A). In contrast, the relative amounts of CD4 and CD8 T cells as well as NK cells were substantially reduced in a dose-dependent manner for both the subcutaneous and intralymphatic route of CpG administration (Figure 2 B-D). The means + SEM (n - 3) are shown. Hence, these results confirm that administration of CpG shows considerable adverse effects at the "recommended" doses around 10 nmol for ODN 1668pt, whereas 0.1 and 0.01 nmol did not induce any detectable side effects, Example2 Intralymphatic administration of CpG enhances activation of dendritic cells. [00841 To assess CpG-induced activation of professional APCs, C57BL/6 mice received titrated amounts of CpG QDN 1668pt either subcutaneously by injection into the inguinal region or by direct injection into the inguinal lymph node. After one day the inguinal lymph nodes were collected and DCs were isolated by Collagenase D digestion and positive isolation using anti-CD11c magnetic beads. Activation of DCs was assessed by measuring up-regulation of CD80 and CD86 (Figures. 3A and B). Values represent the mean fluorescence intensity (MI) measured by flow cytometry. As control, mice were immunized i.ln. with saline -21- WO 20061071934 PCT/US2005/0472S solution. Lymph node cells pooled from three mice per group were used for the analysis. At least 1.5 x 10 4 CD11c cells were acquired per condition. High CpG doses of 10 and I nmol significantly enhanced expression of CD80 and CD86, regardless of the route of administration. At low CpG doses of 0.1 and 0.01 nmol, however, only direct injection into the inguinal lymph node induced significant DC maturation. Note that an intralymphatic control injection with saline induced slight upregulation of CD80 and CD86. This may be explained by the physical lymph node damage produced by the needle. Thus, although both sites of injection are very close to each other, direct intralymphatic administration reduced the dose of CpG that is necessary for DC maturation by 10-50 fold. Example 3 Cytotoxicity induced by ovalbumin together with low amounts of CpG ODN (0.01 nmol)given intralymphatically. [0085] The optimal intralymphatic dose of CpG for CTL induction was evaluated and compared to the optimal subcutaneous dose, C57BL/6 mice were injected with OVA alone or in combination with CpG ODN 1668pt. CD8 T cell responses were tested for direct ex vivo CTL activity seven days later using 51 Cr release assays on EL-4 target cells pulsed with the ovalbunin peptide epitope (SEQ. I0 No. 2) at various effector to target cell ratios. Two dilution series of effector cells per mouse were perfonned, The means + SEM (n - 3) are shown (see Figure 4). While mice vaccinated intralymphatically showed significant induction of cytotoxic CD8 T cells even at 0.01 nmol CpG, subcutaneous vaccination did not elicit CTL activity in this assay system. The CTL activity was correlated with the frequency of CD8 lymphocytes staining positive for intracellular IFNy as measured by flow cytometry (data not shown). These data demonstrate a potent generation of cytotoxic immunity against a dominant epitope bome by a model protein antigen, by intralymph node co-administration of CpG ODNs at doses that were not associated with systemic acute phase reaction. -22- WO 2006/071934 PCTWUS2005/047250 Example 4 Intranodal delivery of antigen plus low dose CpG ODN or polyI:C increases a CD8+ T cell response against a tumor-associated antigen. [OO86] H-2 class I-negative, HLA-A2.1-transgenic (HED-1) mice (Pascolo, S., et al., . Exp Med. 185:2043-2051, 1997 (which is incorporated herein in its entirety)) were housed under pathogen-free conditions and used for evaluation of the immunogenicity of HLA-A2.1-restricted human tumor-associated cytotoxic T lymphocyte (CTL) epitopes. Female mice 8-12. weeks of age were used for intralymphatic immunization and for isolation of splenocytes for in vivo cytotoxicity studies. HHD-1 mice were immunized with 25 pL of a I mg/nL solution in PBS of Melan-A peptide (SEQ. 11D. No. 3) alone, plus 0.1 nmole of a CpG ODN (Sigma) (SEQ. lD. No. 4) , or plus 25pg polyL:C, a synthetic double stranded RNA, by injection, bilaterally, into the inguinal lymph nodes. A total of four rounds of injections were given, on days 0, 3, 14 and 17. [0087] The immune response was measured by tetramer and CD8 double staining of splenocytes from the immunized mice. Mononuclear cells isolated from peripheral blood after density centrifugation (Lympholyte Marmmal, Cedarlane Labs) with HLA-A*0201 MART1 (SEQ. 11D. No. 3)-PE MHC tetramer (Beckman Coulter) and FITC conjugated rat anti-mouse CD8a (Ly-2) monoclonal antibody (BD Biosciences). Data was collected using a BD FACS Calibur flow cytometer and analysed using Celiquest software by gating on the lymphocyte population and calculating the percent of tetramer+ cells within the CD8* CTL population. The results (see Figure 5) were expressed as means ± SEM of % tetramer stained cells within the CD8+ T cell population (A), and with specific dot plot plots representative for each group (B). Splenocytes from naive transgenic mice were included as controls, The data show that co-administration of a peptide with a dose of CpG ODN that is not associated with systemic acute phase reaction, nor with observed mortality or morbidity, results in potent immune response against a tumor-associated antigen. Similarly, use of polyl:C, at doses not associated with observed mortality or morbidity, was also capable of supporting induction of a robust antigen specific response. -23- WO 2006/071934 PCT[US2005/04725o Example 5 Intranodal delivery of low dose BRM plus antigen induces effective CTL immunity against a tumor-associated antigen. [0088] The effectiveness of CTL generated as described in Example 4 was assessed as in vivo clearance of tumor cells from pulmonary tissue in mice challenged with Melan-A+ tumor cells, Human melanoma tumor target cells, 624.38 (Melan-A*, HLA-A2t) were cultured in RPMI medium supplemented with 10% fetal bovine serum (Hyclone), 0.1mM non-essential amino acids, and 0.3 mg/mL L-glutamine in a 5% C02 incubator at 37CC. The HLA-A2- subclone, 624.28, was grown under the same conditions and served as a negative control. Specific targets were prepared by incubating 108 624.38 cells with 2.5 pM CFSE in PBS for 20 minutes (CFSeI ) and 10 8 HLA-A2~, 624.28 target controls were labelled with 0.25 pM CFSE (CFSE"). Each immunized ID-1 mouse was intravenously injected in the tail vein with a mixture of 10 7 CFSEk 624.38 and i 7 CFSB' 624.28 cells followed by an identical injection 3 hours later. Un-immunized HHD-1 mice were also injected and served as naive controls. After 14 hours, peripheral blood was harvested via cardiac puncture and the mononuclear cells were isolated by density centrifugation and stained with tetramer as described above. The lungs were surgically removed, minced, and treated with 0.1% collagenase buffer for 2 hours. The mononuclear cells were then isolated by density centrifugation and CFSE positive cells were analysed by FACS. The in vivo specific lysis of human melanoma target cells in the lung was calculated by the following formula: [(1-%CFSEbi / %CFSE'*) - (1- %CFSEhi Control / %CFSE" Control)] [0089] The histograms shown in Figure 6 were representative for the three experimental groups. Immunization with antigen alone, though generating a readily detectable response by tetramer staining, resulted in only minimal tumor cell lysis. In contrast inclusion of a BRM in the protocol led to substantial tumor cell lysis. Thus co-administration of a peptide with a dose of CpG ODN that is not associated with systemic acute phase reaction, nor with observed mortality or morbidity, results in potent cytolytic response against human tumor cells. Similarly, use of polyI:C, at -24- WO 2006/071934 PCTIUS2005/047250 doses not associated with observed mortality or morbidity, was also capable of supporting induction of substantial lytic activity. Example 6 Induction of cytotoxic immunity by co-administration of defined self or non-self MHC class I restricted antigen plus adjuvant into the lymph nodes. [0090] HHD-A2 transgenic mice (n=4/group) were immunized with various peptides (see Table 1) admixed with polyL:C, by direct inoculation into the inguinal lymph nodes (12.
5 g peptide + 12.5ptg of adjuvant, in 25pl of PBS / each inguinal lymph node at day 0, 3, 14 and 17). One week after the final administration, splenocytes were stimulated ex vivo with 10tg/ml of peptide in presence of 5U/mi of rIL-2 and tested in a standard cytotoxic assay, against 5'Cr-labeled target cells (T2 cells) uncoated or coated with cognate peptide, at various Effector:Target ratios. Alternatively, in the case of PSMA 288-297 epitope, cytotoxicity was measured against PSMA expressing human tumor cells (LNCap) cells coated or not with 10ig/ml of MHC class I blocking antibody (W6/32). Cytotoxicity resulting from immiAzation with the NY-ESO-1 157-165 epitope was also assessed against 624.28 and 624.38 cells that had been transformed to express a GFP/NY-ESO-1 fusion protein. Table I Antigen (peptide) Identity in m00se9 Hu-PSMA 288-297 Self: conserved between mouse and h unan SEQ. ID. No. 5 Hu-PSMA 730-739 Non-self: not conserved between mouse and human SEQ. IfD. No. 6 Hu-PRAME 425-433 Non-self:, not conserved between mouse and human SEQ. ID. No. 7 Hu-PRAME 300-309 Non-self not conserved between mouse and human SEQ.IED.No. 8 Hu-NY-ESO-1 157-165 Non-self: not conserved between mouse and human SEQ. ID. No. 9 [0091T Alternatively, mice were immunized with the PSMA 288-297 or PSMA 730-739 epitope peptide in IFA and peptide by subcutaneous injection (5gg peptide in 100 l, twice, at day 0 and 7). One week after the last administration, splenocytes were stimulated ex vivo with 5gg (Figure 7, top panel), 10lg (Figure 7, -25- WO 2006/071934 PCT/US2005/047250 middle panel) and 20pg/ml (Figure 7, bottom panel) of peptide for 3 days and the IFN-gamna concentration in cell supernatants measured by ELISA. [0092] The results show that irrespective of the self or non-self nature of the epitope (reflected in the overall diminished responsiveness to self epitope by a conventional immunization procedure - Figure 7), co-administration of peptide, and polyl:C (synthetic dsRNA) into the lymph nodes triggered a specific cytotoxic response recognizing target cells pulsed with peptide.(Figure 8A-C and left half of D) or tumor cells expressing the target antigen (Figure 8E and right half of D). Example 7 Enhanced IgG antibody responses against PLA2 by intra lymph node co administration of CpG ODN and antigen. [0093] The doses of CpG required to enhance Th.-dependent IgG immune responses against an allergen, and whether intralymphatic administration could reduce the required dose of CpG were evaluated. PLA2, the major allergen from bee venom, was used as a model allergen. CBA/J mice were immunized biweekly with 10, 1, 0.1 and 0.01 nmol ODN-1668pt together with 0.1 gg of PLA2 either intralymphatically (A) or subcutaneously (B). Three weeks after the third injection, sera of mice were collected and PLA2-specific IgG was determined by ELISA (Figure 9). Values represent means and standard deviations obtained from three mice per group. One representative of two similar experiments is shown. In subcutaneously immunized mice, the highest IgG titers were induced by using CpG doses of 10 and 1 nmol, which is consistent with the doses described in the literature (Krieg, A. M., "CpG motifs in bacterial DNA and their immune effects," Annu Rev Immunol 20:709, 2002 (which is incorporated herein in its entirety)), If CpG doses were lowered to 0.1 and 0.01 rnol an adjuvant effect was no longer detectable. The picture was reversed for the intralymphatic immunization route, where highest -antibody titers were observed for low CpG doses of 0.1 and 0.01 nmol and no antibody responses were detectable with the high CpG dose of 10 nmol. Note that PLA2 given alone did not generate significant IgG titers (not shown). These data demonstrate that subcutaneous administration of CpG required significantly higher doses than intralymphatic administration. Importantly, the optimal adjuvant effect of CpG for intralymphatic -26- WO 2006/071934 PCT/US2005/047250 immunization was achieved at doses that did not cause detectable systemic side effects. [0094] This example shows a potent generation of specific IgG antibodies against PLA2 by intra lymph node co-administration of CpG ODNs, at lower doses that were not associated with systemic acute phase reaction. Examle 8 Enhanced IgG antibody responses against PLA2 by intra lmph node co administration of various TLR-ligands and antigen. [0095] To evaluate the effect of various TLR-ligands on the antibody response, CBA/J mice were immunized with three injections at two week intervals of 0.1 gg phospholipase A2 (PLA2) and a TLR ligand; specifically S. aureus peptidoglycan (PGN), Ecoli lipopolysaccharide (LPS), polyriboinosinic polyribocytidylic acid (PolyI:C), lipoteichoic acid (LTA), flagellin, phosphorothioate modified CpG ODN 1826 (SEQ. ]ID. No. 10), and the experimental TLR-7/8-binding compound 3M003 (a gift from 3M Corporation, St. Paul, MN) (see table 2). The compounds were mixed within an hour before injection into the inguinial lymph node. Table 2. Description of adjuvants used for immunizations TLR-L Adjuvant Dose number Description & Source LTA 1.25 pg 2 Lipoteichoic acid from St aureus PGN 2 pg 2 & 6 Peptidoglycan from St aureus LPS 2 pg 2 & 4 Lipopolysaccharide from E. coli Poly(I:C) 2 pg 3 Synthetic analogue of dsRNA Flagellin 3 pg 5 S. thyphimurium 3M003 2 pg 7 & 8 Small synthetic antiviral imidazoqionoline CpG I nmole 9 Synthetic dsDNA with repeating C and G motives Ai(OH) 3 60 pg * - Aluminium hydroxide * injected amount with respect to Al" [00961 Figure 10 illustrates PLA2-specific IgG2a and IgGI antibodies in sera as well as the ratio of IgG2a to IgGI titers (determined by ELISA) at different time points (Fig. 10); the ratio of IgG2a to IgGI antibodies in murine serum is -27- WO 2006/071934 PCT/US2005/0472503 typically used as a qualitative measure for the relative strength of Th1 (IgG2a) and Th2 (IgG1) type immune responses. The sera were obtained over 13 weeks and analyzed by ELISA for anti-PLA2 IgG2a (Fig. lOA) and IgG1 (Fig. 10B). The ratio of IgG2a to IgGi titers was calculated as a measure for Thl/Th2 immune response balance (Fig. 10C). The serological titers were defined as the inverse of the highest dilution yielding an absorbance higher than that of a negative serum plus three standard deviations and expressed as geometric means t standard error (n =3-4). Pre-immunization levels of anti-PLA2 antibodies were measured on sera from five mice taken the day before first injection. The results from one representative out of two immunization experiments are shown. t0097] Seven weeks after the first and three weeks after last injection notable sero-conversion was observed in mice receiving LPS, PolyI:C or CpG. CpG induced IgG2a (Fig. I OA), LPS predominantly IgGI (Fig. 10B), and PolyI:C induced high antibodies titers of both IgGl and IgG2a isotypes. This is also highlighted in the resulting isotype ratios (Fig. 10C). Other TLR ligands tested also showed IgGI and IgG2a antibodies in early sera compared to background levels. When PLA2 was administered together with the conventional adjuvant aluminium hydroxide, a balanced profile of IgG2a and IgG1 antibodies was produced, but the titers were lower than those produced by PolyI:C- or LPS-containing vaccines. Also, the ratio of IgG2a to IgGl was shifted towards Th2 when compared to PolyI:C, CpG and peptidoglycan (Fig. 10C). None of the vaccines induced detectable IgE antibodies against PLA2 upon intralymphatic injection (data not shown). [0098] The kinetics of the antibody production was also affected by the adjuvant. PolyI:C produced its maximum levels of antibodies within three weeks of the final boost injection (seven weeks after initiating immunization). LPS caused a delay of three weeks for both the IgG1 and the IgG2a isotypes, and the IgG2a titer increased relatively more than the IgG1. In seven-week sera from CpG-injected mice, IgG1 was hardly detectable but increased at 10 and 13 weeks indicating a delay in the IgG1 antibody production. However, the serum concentration of IgGI was geometrically only 30-50% of the titers induced by LPS or PolyI:C. As with LPS, peak titers were delayed when peptidoglycan and flagellin were used as the BRM. Peptidoglycan induced predominantly IgG2a, whereas flagellin induced both IgG2a and IgG1. -28- WO 2006/071934 PCT/US2005/047250 Example 9. Modulated Cellular Responses against PLA2 by intra lymph node co-administration of various TLR-ligands and antigen. [0099] After the last bleeding (ten weeks after the third injection), mice from selected groups described in Example 8, above, (aluminium hydroxide, PolyI:C, LPS, CpG and 3M003) received another boost injection of PLA2 and adjuvant. Two weeks later spleen cells were isolated, and analysed directly for intracellular IFN-y by flow cytometry (Fig. 11) or analyzed for IFN-y, II-4 and IL-10 secretion by ELISA after four days cultivation with 10 pig/ml PLA2 (Fig. 12). For Figure 11, intracellular IFN y was measured by flow cytometry after four hours stimulation of cells with PdBu and ionomycin (both at 500 ng/nl) in the presence of brefeldin A (10pg/"l). The cells were then incubated with anti-CD 16/CD32 for Fc-recptor blocking and stained on ice and in PBS/FCS 2% with fluorescent-labelled Abs against CD4, CD8 and CD44 (all Abs from BD Biosciences (Franklin Lakes, NJ). Cells were subsequently fixed with 4% parafornaldehyde and permeabilised with 0.1% Nonidet P-40 before staining with Ab against IFN-y. Four-color cytometry were performed on a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA). Cells were gated on CD4 or CD8 positive lymphocytes and the results expressed as means (± S.D.) for three mice, as percent of T-cell subset (A) or as absolute numbers in the spleen (B). One representative out of two immunization experiments is shown. For Figure 12, after homogenisation of spleens and lysis of red blood cells, 5x10 5 splenocytes were cultured with 20 pig/ml PLA2 protein in 200 pl IMDM (Gibco) supplemented with L-glutamine (4 mM), FCS (10%), mercaptoethanol (75 pM), as well as streptomycin and ampicillin, Supernatants were collected after 96 hours incubation and IL-4, IL-10 and IFN-y concentrations determined by BLISA from R&D Systems (Abingdon, United Kingdom). Cytokine concentrations in supernatants as determined by ELISA after subtraction of spontaneous cytoidne secretion was determined upon in vitro stimulation with an irrelevant antigen (ovalbumin). Statistical differences between the different groups were indicated when significant (P < 0.01). One representative out of two immunization experiments is shown, [01001 A higher percentage of IFN-y producing cells were elicited from CD8 positive T cells (Fig. 11B) than from CD4-positive T cells (Fig. 11A), with CpG -29- WO 2006/071934 PCT/US2005/047250 showing the strongest effect The same was evident with regard to the absolute number of cytokine-producing cells (Fig. 11 C-D). [0101] The capacity to produce IFN-y correlated with the amount of cytokine secreted as measured by ELISA. The highest amounts of ]FN-y were produced by cells from mice immunized using PolyI:C and CpG (Fig. 12A), the concentrations measured being significantly higher than those obtained from mice immunized with LPS or 3M003 (P <0.01). Interestingly, whereas only marginal anti-PLA2 antibodies could be detected after immunizing mice with PLA2 and 3M003, this vaccine regime apparently generated a substantial number of IFN-y producing cells as observed both in FACS (Fig, 11) and in ELISA (Fig. 12). CpG caused significantly reduced IL-4 secretion (Fig. 11 B) when compared with 3M003 and PolyLC (P<0.01) or LPS (P<0.05), despite triggering the strongest IL-10 production (Fig. 12C). Example 10 Imiquimod exhibits adjuvant activity when administered intranodally in amounts ineffective when administered subcutaneously. [0102] Iniquimod, 1-(2-methylpropyl)-IH-imidazol {4,5-c] quinolin-4-amine, is a topical immune response modifier that is approved for the use in the treatment of human genital warts. Imiquimod binds to TLR-7 in mice and TLR-7 and TLR-8 in humans. Such Imidazoquinoline binding leads to activation of plasmacytoid dendritic cells (DCs). Subsequent rapid maturation of the DCs is associated with upregulation of expression of several DC cell surface components, including the co-stimulatory molecules CD40, CD80, and CD86, as well as MHC class I and 11 molecules, and chemokine receptors. TLR binding also stimulates the production of a characteristic profile of proinflammatory cytoldnes including ILl, IL-6, IL-12, interferon-y, and TNF-a. These characteristics make Itiquimod a very potent adjuvant candidate. Not surprisingly, however, Imiquimod shows significant toxicity when systemically administered, including fatigue, malaise, fever, headache, and lymphocytopenia (Witt, P.L. et al., Cancer Res. 53(21):5176-80, 1993 (incorporated herein by reference in its entirety)).. Even if only topically used on the skin of children in a manner that allows for enhanced penetration, fever may be observed (Campanelli, A. et al. JAm Acad Dermatol. 52(1):E1, 2005 (incorporated herein by reference in its entirety)). [0103] C57BL/6 mice were immunized with the immunodominant MHC class I binding epitope of the lymphocytic choriomeninigitis virus glycoprotein, p33 (SEQ. -30- WO 2006/071934 PCT/US2005/047250 ID. No. 11). On day 8 after immunization mice were bled to analyze p33-specifio CDS+ T cell function by FACS staining of CD8, CD44 and intracellular interferon-y production after a 4-hour in vitro stimulation with p33. All groups of mice received the same dose of 10 [g p33. The adjuvant hiiquimod, however, was administered by different routes and in different doses. Mice were either injected intralymphatically with 250pg or 25pg of Iniquimod, or they were injected subcutaneously with 250pg or 25pg of hniquimod. Control -mice were immunized with p33, but received no Imiquimod. As seen in Figure 13, 250ptg of imiquimod enhanced induction of p33 specific CD8+ T cells only, when administered intralymphatically, but not after subcutaneous administration, Thus, intralymphatic administration of liniquimod enhanced its adjuvant activity. 101041 Without being bound by any particular model, the foregoing examples can be understood as resulting from the co-localization of antigen and immune modulator within the same microenvironment. In this view, very low amounts of immune modulator are associated with extremely high ratios between local and systemic (non-lymphatic) bioavailability of such compounds when delivered into lymphoid organs, resulting in substantial local effect with minimal systemic impact. See Figure 14. More specifically, intralymph node delivery of minute amounts of BRM results in appropriate local bioavailability and immune activity since the lymphoid organs are the primary location where the immune response is initiated or amplified (Figure 14, upper row diagrams), In contrast, delivery of such minute amounts of BRM via intramuscular, subcutaneous or intravenous route, will result in reduced localization of BRM into lymphoid organs, simply because systemic clearance mechanisms or PK profile will play a more prominent role (Figure 14, lower row diagrams), Thus, to achieve a similar potency, more BRM needs to be administered via a conventional route (subcutaneous, intramuscular). Since the toxicity profile is dependent on the dosage, targeted intralymphatic delivery result in an overall improved therapeutic index. tO01] Thus, sub-nanomole doses of CpG ODN were extremely effective in promoting immunity against a tumor associated antigen, Melan-A, when the delivery route was intralymphatic. On average, in A2-transgenic mice immunized with the CpG peptide cocktail, approximately one out of five CD8+ T cells were tetramner positive - which represents a considerable expansion of the specific CD8+ T -31- WO 2006/071934 PCT/US2005/047250 cell population. Peptide immunization alone was not very effective in inducing a robust T cell population expansion, even when its inherently poor pharmacokinetics profile was mitigated by intralymphatic delivery; this may be due to limited co expression of immune activating receptors on APC in the absence of adjuvant usage. Notably, the co-administration of CpG ODN obviated the need for Th response in the process of induction of anti-tumoral immunity. In addition, it resulted in effective surveillance of non-lymphatic organs, as exemplified by the significant clearance of A2+ melanoma target cells in transgenic mice previously immunized via the intranodal route. [0106] In various embodiments, numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used to describe and claim certain embodiments of the invention can be understood as being modified by the term "about." Accordingly, in some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters can be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. The numerical values presented in some embodiments of the invention may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. (0107] In various embodiments, the terms "a" and "an" and "the" and similar referents used in the context of describing a particular embodiment of the invention (especially in the context of certain of the following claims) can be construed to cover both the singular and the plural, The recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein, Methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. "such as") provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed, No language in the specification should be construed as indicating that any non-claimed element is essential to the practice of the invention. -32- WO 2006/071934 PCT/US2005/047250 [0108 Groupings of alternative elements or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other members of the group or other elements found herein. It is contemplated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is herein deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims. [0109] . Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations on those preferred embodiments will become apparent to those of ordinary sIdil in the art upon reading the foregoing description. It is contemplated that skilled artisans can employ such variations as appropriate, and the invention can be practiced otherwise than specifically described herein. Accordingly, many embodiments of this invention include all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context. [0110] Furthermore, numerous references have been made to patents and printed publications throughout this specification. Each of the above cited references and printed publications are herein individually incorporated by reference in their entirety. [01111 Accordingly, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the present invention. Other modifications that can be employed are within the scope of various embodiments of the invention. Thus, by way of example, but not of limitation, alternative configurations of the present invention may be utilized in accordance with the teachings herein. Accordingly, the present invention is not limited to that precisely as shown and described. -33- C:\NRPonbI\DCCSCG424E15- .DOC-uzf4I/2 12 {0112] The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates. [0113] Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises' and 'comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. - 33a -

Claims (23)

1. Use of a BRM in the manufacture of a medicament for treatment or immunization against microbes and tumors for administration to a secondary lymphoid organ of a subject, wherein the BRM is administered in a lymphatically effective dose to obtain a modulated antigen-specific immune response and the lymphatically effective dose avoids or reduces a BRM-related adverse clinical event as compared to use of a non lymphatically effective dose of the BRM and wherein the therapeutic index of the BRM is improved, and wherein the BRM is quillaja saponin, and wherein the lymphatically effective dose is at least 10-fold less than the non-lymphatically effective dose.
2. The use of claim 1 wherein little or no BRM-related adverse clinical event results,
3. The use of claim 1 wherein the antigen is co-administered with the BRM.
4. The use of claim 1 wherein the antigen is administered proximal in time to the BRM.
5. The use of claim I wherein the BRM is administered to an identified area of relatively high lymphatic drainage.
6. The use of claim 1 wherein the antigen is administered to an identified area of high lymphatic drainage that drains to the secondary lymphoid organ of the subject.
7. The use of claim 1 wherein the antigen is endogenously present in the secondary lymphoid organ of the subject.
8. The use of claim 1 wherein the medicament is administered directly to a lymph node or lymph vessel.
9. The use of claim 1 wherein the modulated immune response comprises an increased response to the antigen. -34- H:\scgkniinvoven\N4R~onb\DCC\SCG\c617_1.decOL-14/211i3
10. The use of claim 1 wherein the modulated immune response comprises a decreased response to the antigen.
11. The use of claim I wherein the modulated immune response comprises a shift toward a humoral response to the antigen.
12. The use of claim 1 wherein the modulated immune response comprises a shift in relative proportions of individual antibody isotypes.
13. The use of claim 1 wherein the modulated immune response comprises a shift toward a cellular response to the antigen.
14. The use of claim 1 wherein the modulated immune response comprises a shift toward a TI response.
15. The use of claim 1 wherein the modulated immune response comprises a shift toward a T2.
16. The use of claim 13 wherein the modulated immune response comprises a shift toward a CTL response to the antigen.
17. The use of claim 13 wherein the modulated immune response comprises a shift toward a T helper cell response to the antigen.
18. The use of claim 13 wherein the modulated immune response comprises a shift toward a T regulatory cell response to the antigen.
19. The use of claim 1 wherein the antigen comprises a disease-matched antigen.
20. The use of claim 1, wherein the modulated immune response does not consist essentially of an augmented CTL response. - 35 - H:\scg\1menvoven\ P rlDC CI G\5 17_ ILdc-10iD4)20 13
21. A composition comprising a lymphatically effective dose of a BRM, wherein said lymphatically effective dose comprises an amount of a BRM that is relatively nontoxic and wherein said lymphatically effective dose is less than a non-lymphatically effective dose, and wherein said BRM is quillaja saponin.
22. The composition of claim 21, wherein the amount of the BRM is insufficient to be a non-lymphatically effective dose.
23. The composition of any one of claims 21 to 22, further comprising an antigen. -36-
AU2013203959A 2004-12-29 2013-04-11 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs Abandoned AU2013203959A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2013203959A AU2013203959A1 (en) 2004-12-29 2013-04-11 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
AU2016259332A AU2016259332A1 (en) 2004-12-29 2016-11-16 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/640,727 2004-12-29
AU2012201956A AU2012201956B2 (en) 2004-12-29 2012-04-04 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
AU2013203959A AU2013203959A1 (en) 2004-12-29 2013-04-11 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2012201956A Division AU2012201956B2 (en) 2004-12-29 2012-04-04 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2016259332A Division AU2016259332A1 (en) 2004-12-29 2016-11-16 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs

Publications (1)

Publication Number Publication Date
AU2013203959A1 true AU2013203959A1 (en) 2013-05-09

Family

ID=48229932

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013203959A Abandoned AU2013203959A1 (en) 2004-12-29 2013-04-11 Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs

Country Status (1)

Country Link
AU (1) AU2013203959A1 (en)

Similar Documents

Publication Publication Date Title
AU2005321940B2 (en) Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
AU2013316679B2 (en) Cell penetrating peptides
US9017654B2 (en) Preparation for vaccination, vaccination method and use of a vaccination preparation
ES2350043T3 (en) COMPOSITIONS TO PRODUCE, INCREASE AND MAINTAIN IMMUNE RESPONSES AGAINST RESTRICTED MHC CLASS I EPITOPES, FOR PROPHYLACTIC OR THERAPEUTIC PURPOSES.
JP5097400B2 (en) Combined vaccine
EP1872793B1 (en) Polyamino acid for use as adjuvant
Kanagavelu et al. Soluble multi-trimeric TNF superfamily ligand adjuvants enhance immune responses to a HIV-1 Gag DNA vaccine
AU2008265911B2 (en) Use of TLR agonists and/or type 1 interferons to alleviate toxicity of TNF-R agonist therapeutic regimens
Von Beust et al. Improving the therapeutic index of CpG oligodeoxynucleotides by intralymphatic administration
US10238747B2 (en) Method for inducing an immune response against avian, swine, spanish, H1N1, H5N9 influenza viruses and formulations thereof
CA2453880A1 (en) Use of glycosylceramides as adjuvants for vaccines against infections and cancer
WO2006083289A2 (en) Methods and compositions for enhancement of immunity by in vivo depletion of immunosuppressive cell activity
CZ304884B6 (en) Composition for inducing enhanced immune response against peptide or protein antigen in a mammal and use thereof in the preparation of a vaccine
ES2902835T3 (en) Improved cell compositions and methods for cancer therapy
US20040022760A1 (en) Methods of using Flt3-ligand in immunization protocols
JP2017510631A (en) Vaccine compositions and methods for restoring NKG2D pathway function against cancer
JP2005528899A (en) Method for generating antigen-presenting cells
AU2012201956B2 (en) Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
AU2016259332A1 (en) Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
AU2013203959A1 (en) Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
US20020022017A1 (en) Regulation of systemic immune responses utilizing soluble CD40
TWI398262B (en) Immunogenic peptides of tumor associated antigen l6 and uses thereof in cancer therapy
US20100310603A1 (en) TEM8 as an Adjuvant and Uses Thereof

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted