AU2002356962B2 - Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation - Google Patents

Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation

Info

Publication number
AU2002356962B2
AU2002356962B2 AU2002356962A AU2002356962A AU2002356962B2 AU 2002356962 B2 AU2002356962 B2 AU 2002356962B2 AU 2002356962 A AU2002356962 A AU 2002356962A AU 2002356962 A AU2002356962 A AU 2002356962A AU 2002356962 B2 AU2002356962 B2 AU 2002356962B2
Authority
AU
Australia
Prior art keywords
adenosine
cell
subject
immune
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.)
Expired
Application number
AU2002356962A
Other versions
AU2002356962C1 (en
AU2002356962A1 (en
Inventor
Akio Ohta
Michail V. Sitkovsky
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.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority claimed from PCT/US2002/036829 external-priority patent/WO2003050241A2/en
Publication of AU2002356962A1 publication Critical patent/AU2002356962A1/en
Assigned to THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Amend patent request/document other than specification (104) Assignors: THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTHAND HUMAN SERVICES
Application granted granted Critical
Publication of AU2002356962B2 publication Critical patent/AU2002356962B2/en
Publication of AU2002356962C1 publication Critical patent/AU2002356962C1/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

METHODS FOR USING EXTRACELLULAR ADENOSINE INHIBITORS AND ADENOSINE RECEPTOR INHIBITORS TO ENHANCE IMMUNE RESPONSE AND
INFLAMMATION
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application Nos: 60/340,772 filed December 12, 2001 and 60/342,585 filed December 19, 2001 , both hereby incorporated by reference in their entirety.
FIELD
This application relates to the use of inhibitors of extracellular adenosine and/or inhibitors of adenosine receptors, such as adenosine receptor antagonists and agents that decrease formation or degrade extracellular adenosine, to enhance an immune response and inflammation, and in some examples modulate NF-kB activity.
BACKGROUND The inflammatory response helps eliminate harmful agents from the body, but inflammation is also a non-specific response that can harm healthy tissue. There is a wide range of pathogenic insults that can initiate an inflammatory response including infection, allergens, autoimmune stimuli, immune response to transplanted tissue, noxious chemicals, and toxins, ischemia/reperfusion, hypoxia, mechanical and thermal trauma, as well as growth of tumors. Inflammation is normally a localized action that results in expulsion or dilution of a pathogenic agent, resulting in isolation of the damaging agent and injured tissue. The cells involved in inflammation include leukocytes (i.e. the immune system cells— neutrophils, eosinophils, lymphocytes, monocytes, basophils, macrophages, B cells, dendritic cells, granulocytes and mast cells), the vascular endothelium, vascular smooth muscle cells, fibroblasts, and myocytes.
Adenosine modulates diverse physiological functions including induction of sedation, vasodilatation, suppression of cardiac rate and contractility, inhibition of platelet aggregability, stimulation of gluconeogenesis and inhibition oflipolysis (see, Stiles, Trends Pharmacol. Sci. 7:486, 1986; Williams, Ann. Rev. Pharmacol. Toxicol. 27:315, 1987; Ramkumar et al, Prog. Drug. Res'. 32:195, 1988). In addition, adenosine and some adenosine analogs that non-selectively activate adenosine receptor subtypes decrease neutrophil production of inflammatory oxidative products (Cronstein et al., Ann. N. Y. Acad. Sci. 451:291, 1985; Roberts et al, Biochem. J., 227:669, 1985; Schrier e. α/., -/. Immunol. 137:3284, 1986; Cronstein et al., Clinical Immunol. Immunopath. 42:76, 1987).
Based on biochemical and pharmacological criteria, four subtypes of adenosine receptors have been differentiated: A2a, A2b, A 1, and A3. Al and A3 inhibit, and A2a and A2b stimulate, adenylate cyclase, respectively (Stiles, ibid; Williams, ibid; see also U. S. Patent No. 5,441,883 for A3 receptors). Substantial progress has been made concerning the biochemical and pharmacological properties of these adenosine receptors such as ligand binding characteristics, glycosylation, and regulation. In addition to its effects on adenylate cyclase, adenosine opens potassium channels, reduces flux through calcium channels, and inhibits or stimulates phosphoinositide turnover through receptor-mediated mechanisms (Fredholm and Dunwiddie, Trends Pharmacol. Sci. 9: 130, 1988; Sebastiao et al, Br. J. Pharmacol. 100:55, 1990; Stiles, Clin. Res. 38:10, 1990; and Nakahata et al, J. Neurochem. 57:963, 1991). The cDNAs that encode the Al, A2, and A3 adenosine receptors have been cloned (Libert et al, Science 244:569, 1989; Maenhaet et al, Biochem. Biophys. Res. Commun. 173 : 1169, 1990; Libert et al. , EMBO J. 10: 1677, 1991 ; Mahan et al. , Molecular Pharmacol. 40: 1 , 1991; Reppert et al, Molec. Endo. 5:1037-1048, 1991; U.S. Patent No. 5,441,883). Molecular cloning of the adenosine receptors has revealed that they belong to the superfamily of G-protein coupled receptors.
SUMMARY It is disclosed herein that adenosine receptors play a non-redundant role in down-regulation of inflammation in vivo by acting as a physiological "STOP" (a termination mechanism) that can limit the immune response and thereby protect normal tissues form excessive immune damage during pathogenesis of different diseases. Adenosine receptors, such as A2a, A2b, and A3, are shown to down-regulate the immune response during inflammation and protect tissues from immune damage. Inhibition of signaling through the adenosine receptor can be used to intensify and prolong the immune response.
Methods are provided herein to increase an immune response. In one example, the method increases desirable and targeted tissue damage, such as damage of a tumor, for example cancer. Disclosed herein are methods of inhibiting one or more processes conducive to the production of extracellular adenosine and adenosine-triggered signaling through adenosine receptors. For example, enhancement of an immune response, local tissue inflammation, and targeted tissue destruction is accomplished by: inhibiting or reducing the adenosine-producing local tissue hypoxia; by degrading (or rendering inactive) accumulated extracellular adenosine; by preventing or decreasing expression of adenosine receptors on immune cells; and or by inhibiting/antagonizing signaling by adenosine ligands through adenosine receptors. The results disclosed herein demonstrate that by in vivo administration of agents that disrupt the "hypoxia -> adenosine accumulation -> immunosuppressive adenosine receptor signaling to immune cells" pathway in subjects suffering from various diseases (e.g. cancer and sepsis) can result in in vivo treatment of tumors or improved immunization.
In one example, the method includes administering one or more inhibitors of extracellular adenosine and or adenosine receptor inhibitors, such as an adenosine receptor antagonist. To increase the efficacy of a vaccine, one or more adenosine receptor inhibitors and/or inhibitors of extracellular adenosine can be administered in conjunction with the vaccine. In one example, one or more adenosine receptor inhibitors or inhibitors of extracellular adenosine are administered to increase an immune response/inflammation. In another example, a method is provided to achieve targeted tissue damage, such as for tumor destruction.
BRIEF DESCRIPTION OF THE FIGURES FIG. 1 A is a bar graph showing that pharmacologically activated cAMP-elevating receptors or increases in cAMP are capable of blocking inflammation in vivo. The differences between treated and untreated mice are statistically significant as indicated by the asterisk (*/><0.05).
FIG. IB is a bar graph showing that pharmacologically activated cAMP-elevating A2a receptors are capable of blocking inflammation in vivo. The differences between treated and untreated mice are statistically significant as indicated by the asterisk (*P<0.05).
FIGS. 2A and 2B are bar graphs showing cAMP levels in lymphoid cells from (A) wild- type (A2aR+/+) or (B) A2aR deficient (A2aR) mice treated with CGS21680 alone, or CGS21680 and ZM241385.
FIGS. 2C and 2D are bar graphs showing cAMP levels in lymphoid cells in (C) wild-type (A2aR+/+) or (D) A2aR deficient (A2aR" _) treated with FK, isoproterenol, or PGE2. The differences between treated and untreated mice are statistically significant as indicated by the asterisk (*P<0.05). FIGS. 3 A and 3B are dot plots showing serum levels of (A) ALT or (B) TNF-α in A2aR+/+ and A2aR~'~ mice at various time points.
FIG. 4A is a bar graph showing serum ALT levels in mice treated with different combinations of inflammatory stimuli,(Con-A) and A2 receptor antagonist ZM241385. * <0.05 versus A2aR+ + mice.
FIGS. 4B and 4C are scatter plots showing serum ALT levels in mice injected with (B) Pseudomonas Exotoxin A or (C) carbon tetrachloride.
FIG. 5 is a survival graph showing that A2a receptors protect against death from septic shock. *P<0.05.
FIG. 6A-D are scatter plots showing the serum levels of the indicated cytokines in A2aRT mice as compared with A2aR~7+ wild type mice subjected to endotoxic shock. *-°<0.05.
FIG. 7 is a digital image showing the results of a ribonuclease protection assay (RPA) demonstrating the increase of inflammation (by pro-inflammatory cytokines mRNA expression) in mice with inactivated A2a receptors, after treatment with inflammatory stimuli.
FIGS. 8A and 8B are digital images showing a ribonuclease protection assay (RPA) demonstrating the increase of inflammation (by pro-inflammatory cytokines mRNA expression) in mice with pharmacologically inactivated A2a receptors after treatment with inflammatory stimuli. FIG. 9 is a digital image showing the results of a nuclear extract electrophoretic mobility shift assay of macrophages that demonstrates that A2a receptors negatively regulate NF-kB translocation into the nucleus, and thereby its activity, in vivo.
FIGS. 10 A and 10B are digital images of Western blots showing that adenosine receptors negatively regulate NF-kB translocation by inhibiting phosphorylation of Ik-B by IKK kinase. FIG. 11 is a schematic showing the intracellular events following the activation of immune cells and the mechanism of A2a adenosine receptor and cAMP-mediated inhibition of NF-kB activities.
FIGS. 12A-12C are dot plots showing that adenosine receptor antagonists improve immunotherapy of cancer tumors by reducing the number of metastatic nodules.
FIGS. 13 and 14 are graphs showing that adenosine receptor antagonists improve the therapy of cancers by reducing the size/volume of tumors.
FIG.15 is a scatter plot showing IgGi concentration in mice injected subcutaneously with TNP-KLH alone (CFA) or with adenosine receptor antagonist theophylline (CFA + antagonist) and demonstrates the improvement of antibody production by theophylline in the immunization mixture.
FIG. 16 is a summary of the methods that can be used to enhance and/or prolong inflammatory responses by inhibiting or decreasing the endogenous anti-inflammatory processes.
SEQUENCE LISTING The nucleic acid sequence listed in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases. Only one strand of each nucleic acid sequence is shown, but the complementary strand is understood as included by any reference to the displayed strand.
SEQ ID NO: 1 shows the nucleotide sequence of a CpG oligonucleotide.
DETAILED DESCRIPTION OF SEVERAL SPECIFIC EMBODIMENTS
Abbreviations
Adoral Adenosine receptor A 1
Adora2a Adenosine receptor A2a Adora2b Adenosine receptor A2b
Adora3 Adenosine receptor A3
ADA Adenosine deaminase, adenosine degrading enzyme
ADA-PEG Polyethylene glycol-modified ADA to extend half life in vivo
ADA SCID Adenosine deaminase severe combined immunodeficiency ALT Alanine aminotransferase. Liver enzyme cAMP Cyclic adenosine monophosphate
CGS CGS21680
Con A: Concanavalin A
FK Forskolin H-E Haematoxylin and eosin
IL-12p40 Interleukin-12p40
IL-lβ Interleukin -lβ
IL-6 Interleukin-6 Iso Isoproterenol
LPS Lipopolysaccharide, bacterial endotoxin
PEA Pseudomonas exotoxin A
PGE2 Prostaglandin E2 TNF-α Tumor necrosis factor a
Terms
The following explanations of terms and methods are provided to better describe the present disclosure and to guide those of ordinary skill in the art in the practice of the present disclosure. As used herein and in the claims, the singular forms "a" or "an" or "the" include plural references unless the context clearly dictates otherwise. For example, reference to "an adenosine receptor antagonist" includes a plurality of such antagonists and reference to "the adenosine receptor" includes reference to one or more receptors and equivalents thereof known to those skilled in the art, and so forth. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. Unless explained otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Definitions of common terms in molecular biology can be found in Benjamin Lewin, Genes V, published by Oxford University Press, 1994 (ISBN 0-19-854287-9); Kendrew et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182- 9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk
Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8). Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below. The materials, methods, and examples are illustrative only and not intended to be limiting.
Adenosine: A ribonucleotide which includes the nitrogenous base, adenine, linked to the sugar, ribose.
Adenosine receptors: At least four subtypes of adenosine receptor (Adoral, Adora2a, Adora2b, and Adora3, also called Al, A2a, A2b, and A3, respectively) have been cloned. Adenosine receptors include both naturally occurring peptides, as well as adenosine receptor fragments and variants that retain full or partial adenosine receptor biological activity. Adenosine receptors are members of the G-protein coupled receptor (GPCR) superfamily, and are thought to mediate stimulation or inhibition of adenylyl cyclase activity, and hence cyclic AMP levels. The effects on cAMP levels of the methylxanthine antagonists of adenosine receptors (such as caffeine, and theophylline which are present in tea, coffee and cocoa) are known.
The Al receptor is linked to inhibition of adenylyl cyclase activity. However, there is also evidence for coupling (via G-proteins) to ion channels, and phospholipase C. In the nervous system, the Al adenosine receptor mediates inhibition of transmitter release and the reduction in neuronal activity. Blockade of this receptor in the heart leads to the accelerated, pronounced "pounding" observed after drinking large amounts of strong coffee (due to caffeine and theophylline). In one example, Al is shown as GenBank Accession No L22214.
A2a is almost exclusively coupled to stimulation of adenylyl cyclase activity. Its distribution m the CNS is very discrete, bemg heavily localized m the caudate and putamen bodies, and the nucleus accumbens and olfactory tubercle. In the periphery, the A2a receptor is present on platelets and is anti-aggregatory. In one embodiment, A2A is shown as GenBank Accession No. AH003248, and A2b is shown as GenBank Accession No. NM000676. A2a and A2b receptors cause similar cellular effects, but have different tissue distribution and requirements for the levels of extracellular adenosme needed for their activation. It appears that A2b receptor is activated by higher levels of adenosine then A2a receptor (Lmden, Ann Rev Pharmacol Toxicol 41 :775-87, 2001). A3 couples to inhibition of adenylyl cyclase activity. In one example, A3 is shown as GenBank Accession No. AH003597.
Adenosine receptor inhibitor: Any agent or composition that decreases the activity of an adenosine receptor. For example, such an inhibitor may decrease the activity of an adenosine receptor, as compared to the activity of the adenosine receptor m the absence of such an inhibitor. Examples include, but are not limited to, a pharmacological antagomst, a gene therapy agent, a ribozyme, an antisense ohgonucleotide, or another catalytic nucleic acid that selectively binds mRNA encoding an adenosine receptor. Adjuvant: Any agent that enhances or mcreases one or more immune-stimulating properties of another agent (such as a chemical compound or antigenic epitope) An adjuvant augments, stimulates, activates, potentiates, or modulates the immune response at the cellular or humoral level For example, addition of an adjuvant to a vaccine improves the immune response of a cell, such as a cell in a subject An adjuvant can be used so that less vaccine is needed to produce the immune response. One specific, non-limiting example of an adjuvant is Freund's adjuvant, which is a water-in-oil emulsion that contains an immunogen, an emulsifying agent and mycobacteπa. The classical agents (Freund's adjuvant, BCG, Corynebactertum parvum) contain bactenal antigens. Some adjuvants are endogenous (e g histamine, mterferon, transfer factor, tuftsin, ιnterleukm-1 and ιnterleukm-12). The mode of action of an adjuvant can be non-specific, resulting m increased immune responsiveness to a wide variety of antigens, or antigen-specific, i e affecting a restricted type of immune response to a narrow group of antigens. The therapeuhc efficacy of many biological response modifiers is related to their antigen-specific lmmunoadjuvanticity.
Agent: Any polypeptide, compound, small molecule, organic compound, salt, polynucleotide, peptidomimetic, or other molecule of interest Agonist An agent that has affinity for and stimulates physiologic activity of a receptor normally stimulated by one or more naturally occurring agents, thus triggering a biochemical response In one example, one molecule of agonist (A) bmds reversibly to a receptor molecule (R) to form an active agomst-receptor complex (AR), which generates a pharmacological response while the agomst remains bound
Antagonist An agent that tends to nullify the action of another, as a drug that bmds to a receptor without eliciting a biological response In one example, an antagomst is a chemical compound that is an antagomst for an adenosine receptor, such as the A2a, A2b, or A3 receptor Specific examples of adenosine receptor antagonists, mclude, but are not limited to ZM241385, MRS1220, 1,7, methylxantine (caffeine), theophylline, teobromin, SCH 58261 [7-(2-phenylethyl)-5- amιno-2-(2-furyl)-pyrazolo-[4,3-e]-l,2, 4-tπazolo[l,5-c]pyπmιdιne] (Schermg-Plough Research Institute, Milan, Italy), KW-6002 [(E)-l,3-dιethyl-8-(3,4-dιmethoxystyryl)-7-methyl-3,7-dιhydro-lH- puπne-2,6-dιone] (Kyowa Hakko Kogyo Co Ltd , Shizuoka, Japan), and ADA-PEG Particular non- limiting examples of antagomsts are descnbed m U S Patent Nos 5,565,566, 5, 545, 627, 5,981,524, 5,861,405, 6,066,642, 6,326,390, 5,670,501, 6,117,998, 6,232,297, 5,786,360, 5,424,297, 6,313,131, 5, 504,090, and 6,322,771 In another example, an adenosine receptor antagonist is an antisense ohgonucleotide, ribozyme, or other catalytic nucleic acid that selectively binds mRNA encoding the adenosme receptor
Animal: Living multi-cellular vertebrate orgamsms, a category that includes, for example, mammals and birds The term mammal includes both human and non-human mammals Similarly, the term "subject" mcludes both human and veterinary subjects
Antibody: Immunoglobulm molecules and lmmunologically active portions of immunoglobulm molecules, i.e. molecules that contam an antigen-binding site that specifically binds (lmmunoreacts with) an antigen A naturally occurring antibody (e g IgG) mcludes four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds However, the antigen-binding function of an antibody can be performed by fragments of a naturally- occurring antibody Thus, these antigen-bmding fragments are also designated by the term "antibody"
Examples of bmding fragments encompassed withm the term antibody include (I) an Fab fragment consisting of the VL, VH, CL and CHI domains, (n) an Fd fragment consisting of the VH and CHI domains, (in) an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (IV) a dAb fragment (Ward et al , Nature 341 544-6, 1989) which consists of a VH domain, (v) an isolated complimentanty determimng region (CDR), and (vi) an F(ab')2 fragment, a bivalent fragment compπsing two Fab fragments linked by a disulfide bridge at the hinge region Furthermore, although the two domains of the Fv fragment are coded for by separate genes, a synthetic linker can be made that enables them to be made as a smgle protem chain (known as single chain Fv (scFv), Bird et al Science 242 423-6, 1988, and Huston et al Proc Natl Acad Set 85 5879-83, 1988) by recombinant methods Such single chain antibodies are also included
In one example, antibody fragments are those which are capable of crosslinking their target antigen, e g , bivalent fragments such as F(ab')2 fragments Alternatively, an antibody fragment which does not itself crosslink its target antigen (e g , a Fab fragment) can be used m conjunction with a secondary antibody which serves to crosslink the antibody fragment, thereby crosslinking the target antigen. Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described for whole antibodies. An antibody is further intended to include bispecific and chimeric molecules that specifically bind the target antigen. "Specifically binds" refers to the ability of individual antibodies to specifically immunoreact with an antigen, such as a T cell surface molecule. The binding is a non-random binding reaction between an antibody molecule and an antigenic determinant of the T cell surface molecule. The desired binding specificity is typically determined from the reference point of the ability of the antibody to differentially bind the T cell surface molecule and an unrelated antigen, and therefore distinguish between two different antigens, particularly where the two antigens have unique epitopes. An antibody that specifically binds to a particular epitope is referred to as a "specific antibody." Antigen: A compound, composition, or agent capable of being the target of inducing a specific immune response, such as stimulate the production of antibodies or a T-cell response in a subject, including compositions that are injected or absorbed into a subject. An antigen reacts with the products of specific humoral or cellular immunity, including those induced by heterologous immunogens. The term "antigen" includes all related antigenic epitopes.
Antisense, Sense, and Antigene: Double-stranded DNA (dsDNA) has two strands, a 5' -> 3' strand, referred to as the plus strand, and a 3' -> 5' strand (the reverse compliment), referred to as the minus strand. Because RNA polymerase adds nucleic acids in a 5' -> 3' direction, the minus strand of the DNA serves as the template for the RNA during transcription. Thus, the RNA formed will have a sequence complementary to the minus strand and identical to the plus strand (except that U is substituted for T). Antisense molecules are molecules that are specifically hybridizable or specifically complementary to either RNA or the plus strand of DNA. Sense molecules are molecules that are specifically hybridizable or specifically complementary to the minus strand of DNA. Antigene molecules are either antisense or sense molecules directed to a dsDNA target.
Antisense oligonucleotide: A sequence of at least about 8 nucleotides, such as about at least 10, 12, 15, 20, 30 or 50 nucleotides, wherein the sequence is from a gene sequence (such as all or a portion of a cDNA or gene sequence, or the reverse complement thereof), arranged in reverse orientation relative to the promoter sequence in a transformation vector. In one example, the sequence is an adenosine receptor sequence (e.g. Genbank accession number L22214, AH003248, NM000676, and AH003597). Where the reverse complement of a adenosine receptor sequence is used to suppress expression of proteins from the adenosine receptor locus, the sense strand of adenosine or adenosine receptor locus or cDNA is inserted into the antisense construct. A reduction of adenosine receptor protein expression in a transgenic cell can be obtained by introducing into cells an antisense oligonucleotide based on an adenosine receptor locus, e.g. the adenosine receptor Al, A2a, A2b, or A3 locus, including the reverse complement of the adenosine receptor cDNA coding sequence, the adenosine receptor cDNA or gene sequence or flanking regions thereof. The introduced sequence need not be the full-length human adenosine receptor cDNA or gene or reverse complement thereof, and need not be exactly homologous to the equivalent sequence found m the cell type to be transformed Generally, however, where the introduced sequence is of shorter length, a higher degree of homology to the native adenosine or adenosme receptor locus sequence will be needed for effective antisense suppression The introduced antisense sequence in the vector can be at least 30 nucleotides in length, and improved antisense suppression will typically be observed as the length of the antisense sequence increases, such as when the sequence is greater than 100 nucleotides For suppression of the adenosme receptor gene itself, transcription of an antisense construct results m the production of RNA molecules that are the reverse complement of mRNA molecules transcribed from the endogenous adenosine receptor gene m the cell For suppression of protein expression from the opposite strand of the adenosme receptor locus, transcπption of an antisense construct results in the production of RNA molecules that are identical to the mRNA molecules transcribed from the endogenous adenosme or adenosine receptor gene, assuming the antisense construct was generated from sequence within the adenosine receptor gene rather than in a flanking region Antisense molecules made to target the sequence that is the reverse complement of the adenosine receptor locus will serve to suppress any abnormal expression of protems or peptides from the strand of the locus not encodmg the adenosme receptor cDNA
Autoimmune disorder: A disorder m which the immune system produces an immune response (e g a cell or a T cell response) against an endogenous antigen, with consequent mjury to tissues
Avidity: The overall strength of mteraction between two agents or molecules, such as an antigen and an antibody Avidity depends on both the affinity and the valency of interactions Therefore, the avidity of a pentamenc IgM antibody, with ten antigen binding sites, for a multivalent antigen can be much greater than the avidity of a dimeπc IgG molecule for the same antigen B cell or B lymphocyte: One of the two major types of lymphocyte The antigen receptor on B lymphocytes, sometimes called the B cell receptor, is a cell-surface lmmunoglobuhn On activation by an antigen, B cells differentiate into cells producmg antibody molecules of the same antigen-specificity as this receptor
Binding or stable binding: An ohgonucleotide binds or stably binds to a target nucleic acid if a sufficient amount of the ohgonucleotide forms base pairs or is hybπdized to its target nucleic acid to permit detection of that binding Binding can be detected by either physical or functional properties of the target oligonucleotide complex Binding between a target and an ohgonucleotide can be detected by any procedure known to one skilled in the art, including both functional and physical binding assays Bmding can be detected functionally by determining whether binding has an observable effect upon a biosynthetic process such as expression of a gene, DNA replication, transcription, translation and the like
Physical methods of detecting the binding of complementary strands of DNA or RNA include such methods as DNase I or chemical footpπnting, gel shift and affinity cleavage assays, Northern blotting, dot blotting and light absorption detection procedures. For example, one method that is widely used involves observing a change in the light absorption of a solution containing an oligonucleotide (or an analog) and a target nucleic acid at 220 to 300 nm as the temperature is slowly increased. If the oligonucleotide or analog has bound to its target, there is an increase in absorption at a characteristic temperature as the oligonucleotide (or analog) and target disassociate from each other, or melt.
The binding between an oligomer and its target nucleic acid is frequently characterized by the temperature (Tm) at which 50% of the oligomer is melted from its target. A higher (Tm) means a stronger or more stable complex relative to a complex with a lower (Tm). Biological samples: Suitable biological samples include samples containing genomic DNA,
RNA (including mRNA), and/or protein, obtained from cells of a subject. Examples include, but are not limited to, peripheral blood, urine, semen, saliva, tissue biopsy, surgical specimen, amniocentesis samples, derivatives and fractions of blood such as serum, and biopsy material.
Cancer: Malignant neoplasm that has undergone characteristic anaplasia with loss of differentiation, increase rate of growth, invasion of surrounding tissue, and is capable of metastasis. cDNA (complementary DNA): A piece of DNA lacking internal, non-coding segments (introns) and regulatory sequences that determine transcription. cDNA is synthesized in the laboratory by reverse transcription from messenger RNA extracted from cells.
Complementarity and percentage complementarity: Molecules with complementary nucleic acids form a stable duplex or triplex when the strands bind, (hybridize), to each other by forming Watson-Crick, Hoogsteen or reverse Hoogsteen base pairs. Stable binding occurs when an oligonucleotide remains detectably bound to a target nucleic acid sequence under the required conditions.
Complementarity is the degree to which bases in one nucleic acid strand base pair with the bases in a second nucleic acid strand. Complementarity is conveniently described by percentage, i.e. the proportion of nucleotides that form base pairs between two strands or within a specific region or domain of two strands. For example, if 10 nucleotides of a 15-nucleotide oligonucleotide form base pairs with a targeted region of a DNA molecule, that oligonucleotide is said to have 66.67% complementarity to the region of DNA targeted. In the present disclosure, "sufficient complementarity" means that a sufficient number of base pairs exist between the oligonucleotide and the target sequence to achieve detectable binding, and in the case of the binding of an antigen, disrupt expression of gene products (such as adenosine receptors). When expressed or measured by percentage of base pairs formed, the percentage complementarity that fulfills this goal can range from as little as about 50% complementarity to full, (100%) complementary. In general, sufficient complementarity is at least about 50%, for example at least 75%, 90%, 95%, 98% or even 100% complementarity.
A thorough treatment of the qualitative and quantitative considerations involved in establishing binding conditions that allow one skilled in the art to design appropriate oligonucleotides for use under the desired conditions is provided by Beltz et al Methods Enzymol 100:266-285, 1983, and by Sambrook et al (ed ), Molecular Cloning A Laboratory Manual, 2nd ed , vol 1-3, Cold Spπng Harbor Laboratory Press, Cold Sprmg Harbor, NY, 1989
Comprises A term that means "including " For example, "comprising A or B" means mcludmg A or B, or both A and B, unless clearly mdicated otherwise
Cytokine: Proteins made by cells that affect the behavior of other cells, such as lymphocytes In one example, a cytokine is a chemokme, a molecule that affects cellular trafficking DNA Deoxyπbonucleic acid DNA is a long chain polymer which compnses the genetic mateπal of most living orgamsms (some viruses have genes comprising πbonucleic acid (RNA)) The repeatmg umts in DNA polymers are four different nucleotides, each of which compnses one of the four bases, adenine, guamne, cytosine and thymine bound to a deoxyπbose sugar to which a phosphate group is attached Tnplets of nucleotides (refeπed to as codons) code for each ammo acid m a polypeptide The term codon is also used for the corresponding (and complementary) sequences of three nucleotides m the mRNA into which the DNA sequence is transcπbed Deletion The removal of a sequence of DNA, the regions on either side being j omed together
Differentiation: The process by which cells become more specialized to perform biological functions Differentiation is a property that is totally or partially lost by cells that have undergone malignant transformation Epitope: An antigenic determinant These are particular chemical groups or peptide sequences on a molecule that are antigemc, i e that elicit a specific immune response An antibody binds a particular antigemc epitope
Encode: A polynucleotide is said to "encode" a polypeptide if, in its native state or when manipulated by methods well known to those skilled m the art, it can be transcπbed and/or translated to produce the mRNA for and/or the polypeptide or a fragment thereof The anti-sense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom
Hybridization: Ohgonucleotides and their analogs hybπdize by hydrogen bonding, which mcludes Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bondmg, between complementary bases Generally, nucleic acid consists of nitrogenous bases that are either pyrimidines (cytosine (C), uracil (U), and thymine (T)) or purines (adenine (A) and guanine (G))
These nitrogenous bases form hydrogen bonds between a pyπmidine and a puπne, and the bondmg of the pyπmidine to the purme is referred to as "base pairing " More specifically, A will hydrogen bond to T or U, and G will bond to C "Complementary" refers to the base pairing that occurs between to distinct nucleic acid sequences or two distinct regions of the same nucleic acid sequence For example, a therapeutically effective ohgonucleotide can be complementary to an adenosine receptor- encoding mRNA, or an adenosine receptor-encoding dsDNA
"Specifically hybridizable" and "specifically complementary" are terms that indicate a sufficient degree of complementaπty such that stable and specific binding occurs between the ohgonucleotide (or its analog) and the DNA or RNA target The ohgonucleotide or ohgonucleotide analog need not be 100% complementary to its target sequence to be specifically hybridizable An ohgonucleotide or analog is specifically hybridizable when bmdmg of the ohgonucleotide or analog to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA, and there is a sufficient degree of complementanty to avoid non-specific bmding of the ohgonucleotide or analog to non-target sequences under conditions where specific bindmg is desired, for example under physiological conditions in the case of in vivo assays or systems Such binding is referred to as specific hybridization
Hybπdization conditions resultmg m particular degrees of strmgency will vary dependmg upon the nature of the hybndization method of choice and the composition and length of the hybridizing nucleic acid sequences Generally, the temperature of hybridization and the lomc strength (especially the Na+ concentration) of the hybndization buffer will determine the stnngency of hybndization, though waste times also influence stringency Calculations regarding hybndization conditions required for attaining particular degrees of stnngency are discussed by Sambrook et al (ed ), Molecular Cloning A Laboratory Manual, 2nd ed , vol 1-3, Cold Sprmg Harbor Laboratory Press, Cold Spπng Harbor, NY, 1989, chapters 9 and 11
For the purpose of the present disclosure, "stπngent conditions" encompass conditions under which hybridization will only occur if there is less than 25% mismatch between the hybridization molecule and the target sequence "Stπngent conditions" can be broken down into particular levels of stnngency for more precise definition Thus, as used herem, "moderate stringency" conditions are those under which molecules with more than 25% sequence mismatch will not hybπdize, conditions of "medium stringency" are those under which molecules with more than 15% mismatch will not hybridize, and conditions of "high stringency" are those under which sequences with more than 10% mismatch will not hybndize Conditions of "very high stnngency" are those under which sequences with more than 6% mismatch will not hybndize
Hypersensitivity: Immune responses to innocuous antigens that lead to symptomatic reactions upon re-exposure are called hypersensitivity reactions These can cause hypersensitivity diseases if they occur repetitively This state of heightened reactivity to an antigen is called hypersensitivity Hypersensitivity reactions are classified by mechanism type I hypersensitivity reactions involve IgE antibody triggering of mast cells, type II hypersensitivity reactions involve IgG antibodies against cell-surface or matrix antigens, type III hypersensitivity reactions involve antigen antibody complexes, and type IV hypersensitivity reactions are T cell-mediated
Immune cell: Any cell involved m a host defense mechamsm, such as cells that produces pro-inflammatory cytokines, and such as cells that participate in tissue damage and/or disease pathogenesis Examples include, but are not limited to T cells, B cells, natural killer cells, neutrophils, mast cells, macrophages, antigen-presenting cells, basophils, and eosinophils
Immune response A change m immunity, for example, a response of a cell of the immune system, such as a B cell or T cell, to a stimulus In one example, the response is specific for a particular antigen (an "antigen-specific response"). In one example, an immune response is a T cell response, such as a Thl, Th2, or Th3 response. In a particular example, an increased or enhanced immune response is an increase in the ability of a subject to fight off a disease, such as a viral infection or tumor. Immunoglobulins: A class of proteins found in plasma and other body fluids that exhibits antibody activity and binds with other molecules with a high degree of specificity; divided into five classes (IgM, IgG, IgA, IgD, and IgE) on the basis of structure and biological activity. Immunoglobulins and certain variants thereof are known and many have been prepared in recombinant cell culture (e.g. see U.S. Patent No. 4,745,055; U.S. Patent No. 4,444,487; WO 88/03565; EP 256,654; EP 120,694; EP 125, 023; Faoulkner et al, Nature 298:286, 1982; Morrison, . Immunol. 123:793, 1979; Morrison et al., Ann Rev. Immunol 2:239, 1984).
A native (naturally occurring) immunoglobulm is made up of four polypeptide chains. There are two long chains, called the "heavy" or "H" chains which weigh between 50 and 75 kilodaltons and two short chains called "light" or "L" chains weighing in at 25 kilodaltons. They are linked together by disulfide bonds to form a "Y" shaped molecule. Each heavy chain and light chain can be divided into a variable region and a constant region. An Fc region includes the constant regions of the heavy and the light chains, but not the variable regions.
Inhibitor of extracellular adenosine: Any agent or composition that decreases the activity or level of extracellular adenosine. Examples include, but are not limited to, agents that degrade extracellular adenosine, render extracellular adenosine inactive, and/or decrease or prevent the accumulation or formation of extracellular adenosine. Particular examples include, but are not limited to, enzymes such as adenosine deaminase, adenosine kinase, and adenosine kinase enhancers; oxygenation; redox-potential changing agents which diminish the degree of hypoxia-ischemia; and other catalytic agents that selectively bind and decrease or abolish the ability of endogenously formed adenosine to signal through adenosine receptors. Other examples include cell culture conditions that result in negative selection of cells with adenosine receptors and enrichment of cell populations without adenosine receptors.
Inflammation: When damage to tissue occurs, the body's response to the damage is usually inflammation. The damage can be due to trauma, lack of blood supply, hemonhage, autoimmune attack, transplanted exogenous tissue, or infection. This generalized response by the body includes the release of many components of the immune system (e.g. IL-1 and TNF), attraction of cells to the site of the damage, swelling of tissue due to the release of fluid and other processes.
Inflammation, the response of tissue to injury, is divided into two phases, termed acute and chronic. In the acute phase, inflammation is characterized by increased blood flow and vascular permeability, accumulation of fluid, and accumulation of leukocytes and inflammatory mediators
(e.g. cytokines). In the subacute/chronic phase, inflammation is characterized by the development of specific humoral and cellular immune responses to the pathogen(s) present at the site of tissue injury. During both the acute and chronic inflammatory processes, a variety of soluble factors are involved in leukocyte recruitment through increased expression of cellular adhesion molecules and chemoattrachon Many of these soluble mediators regulate the activation of both the resident cells (such as fibroblasts, endothehal cells, tissue macrophages, and mast cells) and newly recruited inflammatory cells (such as monocytes, lymphocytes, neutrophils, and eosinophils) Isolated: An "isolated" biological component (such as a nucleic acid or protein) has been substantially separated, produced apart from, or purified away from other biological components m the cell of the organism in which the component naturally occurs, i e other chromosomal and extrachromosomal DNA and RNA, and protems Nucleic acids and proteins that have been "isolated" thus mclude nucleic acids and protems punfied by standard purification methods The term also embraces nucleic acids and protems prepared by recombmant expression m a host cell as well as chemically synthesized nucleic acids
Leukocyte Cells in the blood, also termed "white cells," that are involved in defending the body against mfective orgamsms and foreign substances Leukocytes are produced in the bone marrow There are 5 main types of white blood cells, subdivided between 2 main groups polymorphonuclear leukocytes (neutrophils, eosinophils, basophils) and mononuclear leukocytes
(monocytes and lymphocytes) When an infection is present, the production of leukocytes increases
Lymphocytes: A type of white blood cell that is involved in the immune defenses of the body There are two mam types of lymphocytes B-cells and T-cells
Mammal: This term includes both human and non-human mammals Similarly, the term "subject" mcludes both human and vetennary subjects
Monoclonal antibody An antibody produced by a single clone of B-lymphocytes Monoclonal antibodies are produced by methods known to those of skill in the art, for instance by makmg hybrid antibody-forming cells from a fusion of myeloma cells with immune spleen cells
Natural killer (NK) cell: These are large, usually granular, non-T, non-B lymphocytes, which kill certain tumor cells NK cells are important in innate immunity to viruses and other lntracellular pathogens, as well as in antibody-dependent cell-mediated cytotoxicity (ADCC)
Neoplasm: An abnormal mass of tissue that results from excessive cell division hat is uncontrolled and progressive, also called a tumor Neoplasms can be begin (neither lnfiltrative nor cancerous) or malignant (invasive) Nucleic acid: A deoxynbonucleotide or πbonucleotide polymer m either single or double stranded form, and unless otherwise limited, encompasses known analogues of natural nucleotides that hybπdize to nucleic acids in a manner similar to naturally occurring nucleotides
Oligonucleotide: A linear polynucleotide sequence of up to about 200 nucleotide bases m length, for example a polynucleotide (such as DNA or RNA) which is at least 6 nucleotides, for example at least 15, 25, 50, 75, 100 or even 200 nucleotides long.
Operably linked: A first nucleic acid sequence is operably linked with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence For mstance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Generally, operably linked DNA sequences are contiguous and, where necessary to join two protein coding regions, in the same reading frame.
Pharmaceutical agent: A chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject or a cell.
"Incubating" includes a sufficient amount of time for an agent to interact with a cell. "Contacting" includes incubating an agent in solid or in liquid form with a cell.
Pharmaceutically acceptable carriers: The pharmaceutically acceptable carriers useful in this disclosure are conventional. Remington 's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 15th Edition (1975), describes compositions and formulations suitable for pharmaceutical delivery of adenosine receptor inhibitors and/or inhibitors of extracellular adenosine.
In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. For solid compositions (e.g., powder, pill, tablet, or capsule forms), conventional non-toxic solid earners can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
Polynucleotide: A linear nucleotide sequence, including sequences of greater than 100 nucleotide bases in length.
Polypeptide: Any chain of amino acids, regardless of length or post-translational modification (e.g. glycosylation or phosphorylation).
Preventing or treating a disease: "Preventing" a disease refers to inhibiting or decreasing the full development of a disease, for example in a person who is known to have a predisposition to a disease. An example of a person with a known predisposition is someone with a history of diabetes in the family, or who has been exposed to factors that predispose the subject to a condition, such as lupus or rheumatoid arthritis. "Treatment" refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition after it has begun to develop.
Probes and primers: Nucleic acid probes and primers can be readily prepared based on a nucleic acid sequence. A probe includes an isolated nucleic acid attached to a detectable label or reporter molecule. Typical labels include radioactive isotopes, enzyme substrates, co-factors, ligands, chemiluminescent or fluorescent agents, haptens, and enzymes. Methods for labeling and guidance in the choice of labels appropriate for various purposes are discussed, e.g. in Sambrook et al. (In Molecular Cloning: A Laboratory Manual, CSHL, New York, 1989) and Ausubel et al. (In Current Protocols in Molecular Biology, Greene Publ. Assoc. and Wiley-Intersciences, 1992). Primers are short nucleic acid molecules, such as DNA oligonucleotides at least 10 nucleotides in length, such as about at least 12, 15, 17, 20, or 25 nucleotides in length. Primers can be annealed to a complementary target DNA strand by nucleic acid hybridization to form a hybrid between the primer and the target DNA strand, and then the primer extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification of a nucleic acid sequence, e.g. by the polymerase chain reaction (PCR) or other nucleic-acid amplification methods known in the art.
Methods for preparing and using probes and primers are described, for example, in Sambrook et al. (In Molecular Cloning: A Laboratory Manual, CSHL, New York, 1989), Ausubel et al. (In Current Protocols in Molecular Biology, Greene Publ. Assoc. and Wiley-Intersciences, 1998), and Innis et al. (PCR Protocols, A Guide to Methods and Applications, Academic Press, Inc., San Diego, CA, 1990). PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5, ® 1991 , Whitehead Institute for Biomedical Research, Cambridge, MA). One of ordinary skill in the art will appreciate that the specificity of a particular probe or primer increases with its length. Thus, for example, a primer of 30 consecutive nucleotides of a adenosme receptor encoding nucleotide will anneal to a target sequence, such as another nucleic acid encoding an adenosine receptor, with a higher specificity than a corresponding primer of only 15 nucleotides. Thus, m order to obtain greater specificity, probes and primers can be selected that mclude at least 17, 20, 23, 25, 30, 35, 40, 45, 50 or more consecutive nucleotides of nucleotide sequence of interest. Purified: The term purified does not require absolute purity; rather, it is intended as a relative term. Thus, for example, a purified peptide or nucleic acid preparation is one m which the peptide or nucleic acid is more ennched than the peptide or nucleic acid is in its natural environment within a cell For example, a preparation is purified such that the protein or nucleic acid represents at least 50%, such as at least 70%, of the total peptide or nucleic acid content of the preparation.
Receptor: A molecular structure within a cell or on the surface of a cell, charactenzed by selective bmdmg of a specific substance and a specific physiological effect that accompanies the binding, for example, cell surface receptors for peptide hormones, neurotransmitters, immunoglobulins, small molecules, and cytoplasmic receptors for steroid hormones. An adenosine receptor is a cell surface receptor for adenosme, and includes, but is not limited to, A2 and A3 receptors
Recombinant: A recombmant nucleic acid is one that has a sequence that is not naturally occumng or has a sequence that is made by an artificial combination of two otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e g. by genetic engineering techniques. Similarly, a recombinant protem is one encoded for by a recombinant nucleic acid molecule. Ribozyme: Synthetic RNA molecules that possess highly specific endoribonuclease activity. The production and use of ribozymes are disclosed in U.S. Patent No. 4,987,071 to Cech and U.S. Patent No. 5,543,508 to Haselhoff. The inclusion of ribozyme sequences within antisense RNAs can be used to confer RNA cleaving activity on the antisense RNA, such that endogenous mRNA molecules that bind to the antisense RNA are cleaved, which in turn leads to an enhanced antisense inhibition of endogenous gene expression.
Specific binding agent: An agent that binds substantially only to a defined target. Thus an antibody or antibody fragment-specific binding agent binds substantially only the defined antibody or antibody fragment, or an antibody region within a protein, such as a fusion protein. As used herein, the term "adenosine receptor specific binding agent," includes anti-adenosine receptor antibodies (and functional antibody fragments thereof) and other agents (such as potential therapeutic agents) that bind substantially only to adenosine receptors.
Antibodies can be produced using standard molecular procedures described in a number of texts, including Harlow and Lane (Antibodies, A Laboratory Manual, CSHL, New York, 1988). The determination that a particular agent binds substantially only to the target protein or peptide can readily be made by using or adapting routine procedures. One suitable in vitro assay makes use of the Western blotting procedure (Harlow and Lane, Antibodies, A Laboratory Manual, CSHL, New York, 1988).
Shorter fragments of antibodies can also serve as specific binding agents. For instance, FAbs, Fvs, and single-chain Fvs (SCFvs) that bind to adenosine receptor are adenosine receptor- specific binding agents.
Subject: Living multi-cellular vertebrate organisms, a category that includes both human and non-human mammals.
T Cell: A white blood cell involved in the immune response. T cells include, but are not limited to, CD4+ T cells and CD8+ T cells. A CD4+ T lymphocyte is an immune cell that carries a marker on its surface known as "cluster of differentiation 4" (CD4). These cells, also known as helper T cells, help orchestrate the immune response, including antibody responses as well as killer T cell responses. CD8+ T cells carry the "cluster of differentiation 8" (CD8) marker. In one embodiment, a CD8 T cell is a cytotoxic T lymphocyte. In another example, a CD8 cell is a suppressor T cell.
Target sequence: A portion of ssDNA, dsDNA or RNA that, upon hybridization to a therapeutically effective oligonucleotide or oligonucleotide analog, results in the inhibition of gene expression, such as adenosine receptor gene expression. An antisense or a sense molecule can be used to target a portion of dsDNA, since both will interfere with the expression of that portion of the dsDNA. The antisense molecule can bind to the plus strand, and the sense molecule can bind to the minus strand. Thus, target sequences can be ssDNA, dsDNA, and RNA.
Therapeutically effective amount: A quantity of an agent or composition sufficient to achieve a desired effect in a subject being treated. For instance, this can be the amount necessary to increase activity of an immune cell and/or enhance an immune response in a subject. In one example, it is an amount that will inhibit viral, fungal, or bacterial replication or to measurably alter outward symptoms of the viral, fungal, or bacterial infection. In another example, it is an amount that will decrease or prevent further mmor growth. When administered to a subject, a dosage will generally be used that will achieve target tissue concentrations (for example, in lymphocytes) that has been shown to achieve in vitro inhibition of viral replication or reduction of mmor cells.
Therapeutically effective dose: A dose sufficient to prevent advancement, or to cause regression of the disease, for example a dose sufficient to reduce the volume or size of a tumor. In another example, it is an amount which is capable of relieving symptoms caused by a disease, such as pain or swelling.
Therapeutically effective adenosine receptor oligonucleotides and oligonucleotide analogs: Characterized by their ability to inhibit or decrease expression of one or more adenosine receptors. As described below, complete inhibition is not necessary for therapeutic effectiveness. Therapeutically effective oligonucleotides are characterized by their ability to inhibit or decrease the expression of one or more adenosine receptors. Inhibition is a reduction in adenosine receptor expression observed when compared to adenosine receptor production in the absence of the oligonucleotide or oligonucleotide analog. For example, an oligonucleotides may be capable of inhibiting the expression of adenosine receptors by at least 15%, 30%, 40%, 50%, 60%, or 70%, or more, and still be considered to be therapeutically effective. Therapeutically effective oligonucleotides and oligonucleotide analogs are additionally characterized by being sufficiently complementary to adenosine receptor-encoding nucleic acid sequences. As described herein, sufficient complementary means that the therapeutically effective oligonucleotide or oligonucleotide analog can specifically disrupt the expression of adenosine receptors, and not significantly alter the expression of genes other than adenosine receptors. Transduced and Transformed: A virus or vector "transduces" a cell when it transfers nucleic acid into the cell. A cell is "transformed" by a nucleic acid transduced into the cell when the DNA becomes stably replicated by the cell, either by incorporation of the nucleic acid into the cellular genome, or by episomal replication. As used herein, the term transformation encompasses all techniques by which a nucleic acid molecule might be introduced into such a cell, including fransfection with viral vectors, transformation with plasmid vectors, and introduction of naked DNA by electroporation, lipofection, and particle gun acceleration.
Treatment: Refers to both prophylactic inhibition of initial infection, and therapeutic interventions to alter the natural course of an untreated disease process, such as infection with a virus. Tumor: An abnormal mass of tissue that results from excessive cell division that is uncontrolled and progressive, also called a neoplasm. Tumors can be benign (neither infiltrative nor cancerous) or malignant (invasive). Vaccine: A dead or attenuated (non-pathogenic) form of a pathogen, or an antigen isolated from a pathogen, administered to a subject to induce adaptive immunity to the pathogen.
Vector: A nucleic acid molecule as introduced mto a host cell, thereby producing a transformed host cell. A vector can mclude nucleic acid sequences that permit it to replicate in the host cell, such as an ongin of replication. A vector can also include one or more selectable marker genes and other genetic elements known in the art. The term "vector" includes viral vectors, such as adenoviruses, adeno-associated viruses, vaccima, and retroviruses vectors.
The Stop Mechanism of Inflammation
It is disclosed herem that the adenosme receptor is a physiological "stop" mechamsm for inflammation in vivo, and as such, extracellular adenosine and adenosine receptors (such as A2a, A2b, and A3) are pharmacological and genetic targets for affecting inflammation and thereby altenng the immune response. Inhibition or reduction of extracellular adenosine or the adenosine receptor through the use an inhibitor of extracellular adenosme (such as an agent that prevents the formation of, degrades, renders mactive, and or decreases extracellular adenosme), and/or an adenosine receptor inhibitor (such as an adenosine receptor antagonist), as disclosed herem, is of use in generatmg an immune response, such as a macrophage, neutrophil, granulocyte, dendritic cell, T- and/or B cell- mediated response In addition, an inhibitor of extracellular adenosme and adenosme receptor inhibitors are of use in promoting acute or chronic inflammation. Inhibitors of the Gs protem mediated cAMP dependent lntracellular pathway and inhibitors of the adenosine receptor -triggered G, protem mediated intracellular pathways, can also be used to mcrease acute and chrome inflammation.
Inhibitors of: Adenosine Receptors, the Intracelluar cAMP Dependent Pathway and
Extracellular Adenosine
Disclosed herein are methods for increasing an immune response or inflammation, and methods for targeted tissue damage, by contacting an immune cell or administering to a subject, one or more agents that inhibit extracellular adenosine or adenosine receptor inhibitors, such as adenosine receptor antagonists. A summary is provided in FIG 16
An agent that inhibits extracellular adenosine mcludes agents that render extracellular adenosine non-functional (or decrease such function), such as a substance that modifies the structure of adenosine to nullify the ability of adenosine to signal through adenosine receptors. This can be, for example, an enzyme (e g adenosine deaminase) or another catalytic molecule that selectively binds and destroys the adenosine, thereby abolishing or significantly decreasing the ability of endogenously formed adenosine to signal through adenosine receptors and terminate inflammation. One agent that degrades extracellular adenosine is ADA-PEG, polyethylene glycol-modified ADA that has been used in treatment of patients with ADA SCID (Hershfield, Hum Mutat 5 107, 1995). In another example, an agent that inhibits extracellular adenosine includes agents that prevent or decrease formation of extracellular adenosine, and/or prevent or decrease the accumulation of extracellular adenosine.
Adenosine receptor inhibits include adenosine receptor antagonists. An antagomst is any substance that tends to nullify the action of another, as an agent that binds to a cell receptor without eliciting a biological response. In one example, the antagonist is a chemical compound that is an antagonist for an adenosine receptor, such as the A2a, A2b, or A3 receptor. In another example, the antagonist is a peptide, or a pepidomimetic, that binds the adenosine receptor but does not trigger a Gl protein dependent intracellular pathway. Suitable antagonists are described in U.S. Patent Nos. 5,565,566; 5, 545, 627, 5,981,524; 5,861,405; 6,066,642; 6,326,390; 5,670,501 ; 6,117,998; 6,232,297; 5,786,360; 5,424,297; 6,313,131, 5,504,090; and 6,322,771.
In another example, the antagonist is an antisense molecule or catalytic nucleic acid molecule (e.g. a ribozyme) that specifically binds mRNA encoding an adenosine receptor. In specific, non-limiting examples, the antisense molecule or catalytic nucleic acid molecule binds A2a, A2b, or A3. In a further example, an antisense molecule or catalytic nucleic acid targets biochemical pathways downstream of the adenosine receptor. For example, the antisense molecule or catalytic nucleic acid can inhibit an enzyme involved in the Gs protein- or Gi protein-dependent intracellular pathway.
Adenosine receptor protein expression in a host cell can be reduced by introducing into cells an antisense construct or another genetic sequence-targeting agent, based on an adenosine receptor locus, e.g. the adenosine receptor Al, A2a, A2b, or A3 locus (e.g. Genbank accession numbers L22214, AH003248, NM000676, and AH003597, respectively). An antisense construct includes the reverse complement of the adenosine receptor cDNA coding sequence, the adenosine receptor cDNA or gene sequence or flanking regions thereof. For antisense suppression, a nucleotide sequence from the adenosine receptor locus (e.g. all or a portion of the adenosine receptor cDNA or gene or the reverse complement thereof) is arranged in reverse orientation relative to the promoter sequence in a vector. The vector is then introduced into a cell of interest. Where the reverse complement of the reported sequences is used to suppress expression of proteins from the adenosine receptor locus, the sense strand of the disclosed adenosine or adenosine receptor locus or cDNA is inserted into the antisense construct. Without being bound by theory, it is believed that antisense RNA molecules bind to the endogenous mRNA molecules and thereby inhibit translation of the endogenous mRNA.
For suppression of an adenosine receptor gene, transcription of an antisense construct results in the production of RNA molecules that are the reverse complement of mRNA molecules transcribed from the endogenous adenosine receptor gene in the cell. The introduced sequence need not be the full-length human adenosine receptor cDNA or gene or reverse complement thereof, and need not be exactly homologous to the equivalent sequence found in the cell type to be transformed. Generally, however, where the introduced sequence is of shorter length, a higher degree of homology to the native adenosine or adenosine receptor locus sequence is needed for effective antisense suppression. In one example, the introduced antisense sequence in the vector is at least 10, such as at least 30 nucleotides m length Improved antisense suppression is typically observed as the length of the antisense sequence increases Shorter polynucleotide (oligonucleotides) can convemently be produced synthetically as well as in vivo. In specific aspects, the ohgonucleotide is at least 10 nucleotides, at least 15 nucleotides, at least 30, at least 100 nucleotides, or at least 200 nucleotides To inhibit the translation of the target RNA molecule, such as an adenosine receptor, the antisense molecule will ideally persist in the cell for a sufficient period to contact the target RNA. However, cells contains enzymes and other components that cause polynucleotides (such as an antisense molecule) to degrade. The antisense molecule can be engineered such that it is not degraded m the cell. This can be done, for example, by substituting the normally occurring phosphodiester linkage which connects the individual bases of the antisense molecule with modified linkages These modified linkages can, for example, be a phosphorothioate, methylphosphonate, phosphodithioate, or phosphoselenate. Furthermore, a single antisense molecule can contain multiple substitutions in various combinations The antisense molecule can also be designed to contam different sugar molecules For example the molecule can contam the sugars πbose, deoxyπbose or mixtures thereof, which are linked to a base The bases give nse to the molecules' ability to bmd complementaπly to the target RNA. Complementary bindmg occurs when the base of one molecule forms a hydrogen bond with another molecule Normally the base ademne (A) is complementary to thymidine (T) and uracil (U), while cytosine (C) is complementary to guamne (G). Therefore, the sequence ATCG of the antisense molecule will bond to TAGC of the target RNA. Additionally, the antisense molecule does not have to be 100% complementary to the target RNA to be effective.
The oligonucleotides can be DNA or RNA, or chimeπc mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. The ohgonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, and can include other appending groups such as peptides, or agents facilitating transport across the cell membrane (see, e g , Letsinger et al, PNAS USA 86 6553-6, 1989, Lemaitre et al , PNAS USA 84:648-52, 1987; PCT Publication No WO 88/09810) or blood-brain barrier (see, e g , PCT Publication No WO 89/10134), hybndization triggered cleavage agents (see, e g , Krol et al , BioTechmques 6:958-76, 1988) or intercalating agents (see, e g , Zon, Pharm Res. 5.539-49, 1988)
In a particular example, an adenosine receptor antisense polynucleotide is provided, for example as a single-stranded DNA Such a polynucleotide can include a sequence antisense to a sequence encoding an Al, A2a, A2b, or A3 receptor The ohgonucleotide can be modified at any position on its structure with substituents generally known in the art For example, a modified base moiety can be 5-fluorouracιl, 5-bromouracιl, 5-chlorouracιl, 5-ιodouracιl, hypoxanthine, xanthine, acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylamιnomethyl-2-thιourιdιne, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N~6- sopentenyladenine, 1-methylguanme, 1-methyhnosme, 2,2-dιmethylguanιne, 2-methyladenme, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, methoxyarninomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'- methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-S- oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4- thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-S-oxyacetic acid, 5-methyl-2- thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, and 2,6-diaminopurine.
In another example, the polynucleotide includes at least one modified sugar moiety such as arabinose, 2-fluoroarabinose, xylose, and hexose, or a modified component of the phosphate backbone, such as phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, or a formacetal or analog thereof.
The antisense polynucleotide can be conjugated to another molecule, for example a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent. A targeting moiety can also be included that enhances uptake of the molecule by tumor cells. The targeting moiety can be a specific binding molecule, such as an antibody or fragment thereof that recognizes a molecule present on the surface of the tumor cell.
Suppression of endogenous adenosine receptor locus expression can also be achieved using catalytic nucleic acids such as ribozymes. Ribozymes are synthetic RNA molecules that possess highly specific endoribonuclease activity. The production and use of ribozymes are disclosed in U.S. Patent No. 4,987,071 to Cech and U.S. Patent No. 5,543,508 to Haselhoff. Ribozymes can be synthesized and administered to a cell or a subject, or can be encoded on an expression vector, from which the ribozyme is synthesized in the targeted cell (as in PCT publication WO 9523225, and Beigelman et al. Nucl Acids Res. 23:4434-42, 1995). Examples of oligonucleotides with catalytic activity are described in WO 9506764, WO 9011364, and Sarver et al. (Science 247:1222-5, 1990). The inclusion of ribozyme sequences within antisense RNAs can be used to confer RNA cleaving activity on the antisense RNA, such that endogenous mRNA molecules that bind to the antisense RNA are cleaved, which in mm leads to an enhanced antisense inhibition of endogenous gene expression.
In addition, dominant negative mutant forms of an adenosine receptor can be used to block endogenous adenosine receptor activity. In this example, a nucleic acid encoding a dominant negative mutant form of an adenosine receptor is operably linked to a promoter. In one specific, non-limiting example, the promoter is an inducible promoter. A vector containing the promoter and the nucleic acid encoding the dominant negative adenosine receptor is than introduced into a cell.
In another example, local tissue accumulation of extracellular adenosine is inhibited using a preparation of adenosine deaminase (ADA). This can be, for example, an enzyme, adenosine deaminase or a ribozyme, or another catalytic molecule that selectively binds and destroys adenosine, thereby abolishing, or substantially decreasing, the ability of endogenously- formed adenosine to signal through adenosine receptors and terminate inflammation. The propagation of adenosine receptor-tnggered mtracellular signalmg cascade can also be affected by the use of specific inhibitors of enzymes and protems that are mvolved in regulation of synthesis and/or secretion of pro-inflammatory molecules, including modulators of nuclear transcription factors Suppression of adenosine receptor expression or expression of the Gs protem- or Gi protein- dependent mtracellular pathway, or the cAMP dependent mtracellular pathway, are also used to increase/enhance inflammation or the immune response m a vanety of situations (see below)
Increasing an Immune Response Methods are disclosed herem to enhance and prolong the pro-inflammatory response by blocking the natural, extracellular adenosine-dependent, endogenous anti-inflammatory processes in vivo using an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine In one example, the adenosine receptor is A2a, A2b, or the A3 receptor
Also disclosed herein is a method for mcreasing an activity of an immune cell Immune cells include, but are not limited to leukocytes (i e neutrophils, eosinophils, lymphocytes, monocytes, basophils, macrophages, B cells, T cells, dendntic cells, and mast cells), as well as other types of pro-inflammatory cytokine -producing cells In another example, the immune cell is a macrophage In yet another example, the immune cell is an antigen-presenting dendntic cell In a further embodiment, the immune cell is a natural killer cell In an additional example, the immune cell is a granulocyte The immune cell activity can be mcreased either in vivo or in vitro In one example, this method mcludes targeting adenosine receptors on any other cell that produces pro- inflammatory cytokines/molecules, including those that are not considered "classical immune cells "
Thus, in one specific, non-limiting example the cell is a B cell, and secretion of an immunoglobulm (e g IgG or IgM) is increased In another specific, non-hmitmg example the cell is a T cell and the activity is secretion of a cytokine (e g IL-2 or IL-4) is increased Similarly, in another embodiment the cell is either a helper T cell or a cytotoxic T cell, and either the helper T cell functions or the cytotoxic T cell functions are increased Without being bound by theory, cytotoxic T cell activities are increased due to longer expression of lethal hit dehvenng Fas Ligand molecules or due to better trafficking of immune cells toward the targeted tissue in vivo Without bemg bound by theory, T helper cell activities are enhanced because of prolonged secretion of cytokines
The method includes contacting the immune cell with an adenosine receptor inhibitor, such as an adenosine receptor antagonist, or an inhibitor of extracellular adenosine, thereby increasing the activity of the immune cell The immune cell can be mvolved in an acute immune response or in a chronic immune response One of skill in the art can readily identify methods of use in identifying an mcreased activity of an immune cell For example, secretion of cytokines can be measured by ELISA or PCR-based assays or in biological assays In one example, the increase in activity is measured as compared to a control Suitable controls include an immune cell not contacted with an adenosme antagonist, or a standard value
A method is disclosed herem for enhancing an immune response m a subject The method includes administering to the subject a therapeutically effective dose of an adenosine receptor inhibitor and or an inhibitor of extracellular adenosme, to enhance the immune response In one example, the immune response is a macrophage/monocyte or B cell response In another example the immune response is a T cell response
A method is provided herem for improving a T cell mediated immune response The method includes the administration of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine, to a subject In one embodiment, the subject is an immunosuppressed subject, such as a subject infected with an immunodeficiency virus (e g HIV-1 or HIV-2) The administration of the adenosme receptor inhibitor and or an inhibitor of extracellular adenosme results m an mcrease m a desned immune response and/or prolonged secretion of a cytokine of interest In another example, the subject is infected with a pathogen such as a bacterial, viral, or fungal pathogen Adenosme receptor inhibitors and/or inhibitors of extracellular adenosine are administered to facilitate pathogen destruction in the subject In one example, the subject is immunosupressed Immune deficiencies (e g deficiencies of one or more type of immune cells, or of one or more lmmunological factors) associated with immune deficiency diseases, immune suppressive medical treatment, acute and/or chronic infection, and aging can be treated using the methods described herein A general overview of immunosuppressive conditions and diseases can be found in Hamsons "Pnnciples of Internal Medicine," 14th Edition, McGraw-Hill, 1998, and particularly in chapter 86 (Pnnciples of Cancer Therapy), chapter 307 (Primary Immune Deficiency Diseases), and chapter 308 (Human Immunodeficiency Virus Diseases)
Many medical treatments can impair the immune system Corticosteroids, for example, can reduce cell-mediated immunity The predominant toxicity associated with cancer therapies (e g chemotherapy and radiotherapy) is destruction of proliferating cells, such as hematopoietic cells, responsible for maintenance of the immune and blood systems Likewise, immune suppression and depletion of the immune system is required for bone marrow transplantation, in which immune cells are eliminated and subsequently replaced with transplanted cells Certain known immunostimulants (e g erythropoietin and colony stimulating factors such as G-CSF, which is sometimes marketed under the name "Neupogen," U S Patent No 5,536,495) have been used previously to treat certain of these conditions by stimulating regeneration of the immune cells The immunostimulatory compounds and mixtures of the disclosure can be used to stimulate the immune systems of patients suffering from medical treatment or latrogemcally induced immune suppression, including those who have undergone bone manow transplants, chemotherapy, and/or radiotherapy
Other conditions are known in which the immune system is compromised or suppressed For example, activation of the immune system (via stimulation of T cell production) by adenosine receptor antagonist treatment can also be beneficial m aging subjects, in whom immune function is often compromised Similarly, other conditions are known in which the immune response is abnormal or undesirable Any of these conditions would also benefit from the methods disclosed herem, or application of the descnbed compositions In general, the need for treatment with one of the methods or compositions of this disclosure can be determined by examining the immune status of a test subject, and comparmg this immune status to a control or average immune state (a hypothetical "normal" subject) For example, bone marrow biopsies or peπpheral blood lymphocytes can be sampled to assess immune function Indications of reduced immune function mclude leukopema, for example neutrophema or lymphopema, or evidence of diminished white blood cell function Where the test subject has a reduced immunity condition, such as a reduction m a peripheral white blood cell count to below normal, for example 25% below normal, the immunostimulatory methods of the disclosure should be considered as treatments to improve the immune suppressed condition
Also disclosed are methods that can be used to enhance NF-kB activity in a subject, by administering to a subject an adenosme receptor inhibitor and/or an inhibitor of extracellular adenosme Enhancement of NF-kB activity, promotes transcπption of pro-inflammatory cytokines, such as IL-12p40 and TNF-α, thereby increasing an immune response
Vaccines
A method is provided for mcreasmg an immune response to an antigen by providing an adjuvant activity To increase an immune response to an antigen, the antigen is administered in conjunction with an inhibitor of extracellular adenosine, an adenosine receptor inhibitor, an inhibitor of the inttacelluar cAMP dependent pathway, and/or an inhibitor of intracellular Gi protein dependent pathway, which functions as an adjuvant
In one example, a method for prolongmg an immune response to a vaccme is provided The method includes administering an adenosine receptor inhibitor in conjunction with the vaccme, such as an adenosine receptor antagonist
Thus, disclosed herein is the use of an adenosine receptor inhibitor, such as an adenosine receptor antagonist, in adjuvant formulations Methods for the stimulation of an immune response to a particular antigen are thus also within the scope of the disclosure The host ammals to which the adjuvant and adjuvant-containing vaccine formulations of the present disclosure are usefully administered include human as well as non-human mammals
Typically, an antigen is employed in mixture with the adjuvant compounds of the disclosure Therapeutic adjuvant formulations are disclosed herein which, for example, include (I) at least one therapeutically effective antigen or vaccine, and (n) at least one adenosine receptor antagonist or adenosine degrading drug (e g ADA-PEG) Such therapeutic composition can for example include at least one antigenic agent such as
(A) live, heat killed, or chemically attenuated viruses, bacteria, mycoplasmas, fungi, and protozoa,
(B) fragments, extracts, subunits, metabolites and recombinant constructs of (A), (C) fragments, subumts, metabolites and recombmant constructs of mammalian protems and glycoproteins, (D) tumor-specific antigens, and (E) nucleic acid vaccines
The therapeutic composition can therefore utilize any suitable antigen or vaccine component m combination with an adenosine receptor antagonist e g an antigemc agent, such as antigens from pathogenic and non-pathogemc organisms, viruses, and fungi, m combination with an adjuvant compound of the disclosure
As a further example, such therapeutic compositions can suitably include proteins, peptides, antigens and vaccmes which are pharmacologically active for disease states and conditions such as smallpox, yellow fever, distemper, cholera, fowl pox, scarlet fever, diphtheria, tetanus, whooping cough, influenza, rabies, mumps, measles, foot and mouth disease, and poliomyelitis In the resulting vaccme formulation, compnsing (l) an antigen, and (u) at least one adenosine receptor inhibitor or and inhibitor of extracellular adenosme, the antigen and adjuvant compound are each present m an amount effective to elicit an immune response when the formulation is administered to a host animal, embryo, or ovum vaccinated therewith (see below)
Tumor Treatment The importance of lymphoid cells m tumor immunity has been repeatedly shown A cell- mediated host response to tumors includes the concept of lmmunologic surveillance, by which cellular mechamsms associated with cell-mediated immunity destroy newly transformed tumor cells after recognizing tumor-associated antigens (antigens associated with tumor cells that are not apparent on normal cells) This is analogous to the process of rejection of transplanted tissues from a non-identical donor In humans, the growth of mmor nodules has been inhibited in vivo by mixing suspensions of a patient's peripheral blood lymphocytes and of mmor cells, suggesting a cell- mediated reaction to the tumor In vitro studies have shown that lymphoid cells from patients with certain neoplasms show cyto toxicity against corresponding human mmor cells in culture These cytotoxic cells, which are generally T-cells, have been found with neuroblastoma, malignant melanomas, sarcomas, and carcinomas of the colon, breast, cervix, endometπum, ovary, testis, nasopharynx, and kidney Humoral antibodies that react with mmor cells in vitro have also been produced m response to a variety of animal tumors induced by chemical carcinogens or viruses Hybndoma technology in vitro permits the detection and production of monoclonal anti-mmor antibodies directed against a variety of animal and human neoplasms Antibody-mediated protection against mmor growth in vivo has been demonstrable m both leukemias and lymphomas
A method is provided herein to increase inflammatory actions of immune cells including tumor-infiltrating lymphocytes, and in some embodiments, to additionally promote the recruitment of other immune cells with anti-mmor activity to improve the destruction of the mmor (such as reducing the size or volume of the tumor) A method is provided to improve both natural anti-cancer immune response and adaptive lmmunotherapy of cancer by immune cells that recogmze tumor-associated antigens on the mmor cell surface In one example, a first agent is administered to a subject that has an affinity (tropism) for mmor cells. A second agent that is an adenosine receptor inhibitor (such as adenosine receptor antagonist) and/or an inhibitor of extracellular adenosine, is administered to the subject to promote the immune response against the mmor. Without being bound by theory, the first agent selectively accumulates in the tumor due to tropism for the to tumor cells or the local environment. The first agent initiates the death of some low proportion of mmor cells due to its own cytotoxicity against mmor cells.
In one example, the first agent induces cell death in the mmor cells. In an additional example, the first agent is a chemotherapeutic agent. In yet another embodiment, the first agent initiates an immune response directed against the tumor cells. In one example, the second agent is a genetic targeting agent used to mutate an adenosine receptor such that the receptor does not bind adenosine, or does not activate the biochemical pathway triggered by the adenosine receptor. It is shown herein that by triggering low levels of inflammation in targeted tissues (e.g. tumors) with a first agent, in addition to complementary inactivation of adenosine receptors or decreasing extracellular adenosine using genetic or pharmacological techniques, results in destruction of the tissue (e.g mmor).
In one example, the first agent is an immunotoxin that accumulates in the mmor due to their selective interactions with tumor-specific antigens. These reagents cause direct destmction of mmor cells, although destruction of the mmor is not complete. Without being bound by theory, the death of a portion of the mmor cells creates an inflammatory environment within the mmor and activates mmor infiltrating immune cells (macrophages and T cells). The natural inhibitory pathway which would prematurely terminate this anti-mmor activity will be then interrupted by the adenosine receptor inhibitor (such as adenosine receptor antagonist) or an inhibitor of extracellular adenosine (such as an extracellular adenosine degrading or disrupting agent). Thus, administration of a inhibitor of an adenosine receptor and/or an inhibitor of extracellular adenosine, exacerbates tumor cell death. In another example, the first compound initiates the anti-mmor process in vivo. A bi- functional immune cell activating reagent is coupled to an antibody that binds a mmor specific antigen and to a T cell or macrophage- activating ligand (e.g. anti-T cell receptor mAb or T cell-like receptor ligand, respectively). Without being bound by theory the first agent accumulates in the mmor due to its selective interactions with tumor-specific antigens. The first agent also directs activation of mmor infiltrating immune cells, which destroys mmor cells. This activation of immune cells and mmor cells death will creates an inflammatory environment within the mmor and also activates mmor infiltrating immune cells (e.g. macrophages and T cells). The second agent is an adenosine receptor inhibitor (such as an adenosine receptor antagonist) or an inhibitor of extracellular adenosine that exacerbates mmor cell death. In another embodiment, the first agent initiates an anti-mmor process in vivo is a population of T cells that are specific for mmor antigens, alone or in combination with other ligands that enhance antitumor activity of T cells (e.g. CTLA-4 ligand; Kuhns et al, Proc. Natl Acad. Sci. USA 97:12711, 2001) or in combination with the removal of CD25+ regulatory T cells. Depletion of either of these two lmmunoregulatory mechanisms improves anti-mmor CTL activity (Sutmuller et al, J Exp Med 94-823-32, 2001). Without bemg bound by theory, this activation of immune cells and mmor cells death creates an inflammatory environment within the tumor and activates mmor infiltrating immune cells (macrophages and T cells). In this example, the second agent is not an adenosme receptor antagomst or adenosme degradmg agent. Instead, it is the process of preparmg anti-tumor immune cells under conditions that lead to the loss of (or reduction of) adenosine receptors, and thereby renders these cells uninhabitable by tumor-associated adenosme. This process can mclude additional conditions, such as hypoxic incubators to mcrease endogeneous adenosine formation m cell cultures, or addition of adenosme analogs to provide selective negative pressure to prevent or decrease expansion of adenosine receptor-expressing immune cells
In an additional embodiment, the first agent is a cytotoxic compound that accumulates m mmor because of differences between mmor and normal tissue environment (e g differences m growth rate, redox potential or oxygen tension (hypoxia) or other chemical differences) Without bemg bound by theory, this compound causes mmor cell death and creates an inflammatory environment withm the mmor and further activates mmor infiltrating immune cells (macrophages and T cells). The second agent is an adenosine receptor inhibitor (such as an adenosine receptor antagomst) or an inhibitor of extracellular adenosine, that exacerbates mmor cell death by preventing or decreasing the inactivation of anti-mmor cells by adenosine
In yet another embodiment, the first agent is a compound that accumulates in mmor cells and is cytotoxic due to the increased proliferation of mmor cells Without being bound by theory, this compound causes mmor cell death and creates an inflammatory environment within the tumor. Tumor infiltrating immune cells (macrophages and T cells) are activated. The second agent is an adenosme receptor inhibitor (such as an adenosine receptor antagonist) or an inhibitor of extracellular adenosine (such as an extracellular adenosine degrading agent) that exacerbates mmor cell death.
Pharmaceutical Compositions and Administration Pharmaceutical compositions that include at least one adenosine receptor inhibitor, such as an adenosine receptor antagonist, and or at least one inhibitor of extracellular adenosme, can be formulated with an appropπate solid or liquid carrier, depending upon the particular mode of administration chosen The pharmaceutically acceptable earners and excipients useful in this disclosure are conventional For instance, parenteral formulations usually comprise injectable fluids that are pharmaceutically and physiologically acceptable fluid vehicles such as water, physiological saline, other balanced salt solutions, aqueous dextrose, glycerol or the like. Excipients that can be included are, for instance, other proteins, such as human serum albumin or plasma preparations. If desired, the pharmaceuttcal composition to be administered can also contain mmor amounts of non- toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffermg agents and the like, for example sodium acetate or sorbitan monolaurate Other medicinal and pharmaceutical agents, for instance another lmmunostimulant, also can be mcluded Immunoshmulants mclude, but are not limited to, IF A, COX-2 inhibitors, IL-12, saponins (e g QS-23), and N-acetyl-cysteine, for example
The dosage form of the pharmaceutical composition will be determined by the mode of administration chosen For mstance, in addition to mjectable fluids, topical and oral formulations can be employed Topical preparations can mclude eye drops, ointments, sprays and the like Oral formulations can be liquid (e g syrups, solutions or suspensions), or solid (e g powders, pills, tablets, or capsules) For solid compositions, conventional non-toxic solid earners can mclude pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate Actual methods of preparmg such dosage forms are known, or will be apparent, to those skilled in the art
The pharmaceutical compositions that compnse an adenosine receptor antagomst in some embodiments of the disclosure will be formulated m unit dosage form, suitable for individual administration of precise dosages For example, one possible unit dosage can contain from about 1 mg to about 1 g of adenosme receptor agomst or antagomst The amount of active compound(s) administered will be dependent on the subject bemg treated, the seventy of the affliction, and the manner of administration, and is best left to the judgment of the prescπbmg clinician Within these bounds, the formulation to be administered will contain a quantity of the active component(s) in amounts effective to achieve the desired effect m the subject bemg treated
The compounds of this disclosure can be administered to humans or other animals on whose cells they are effective in vaπous manners such as topically, orally, intravenously, intramuscularly, mtrapeπtoneally, lntranasally, transdermally, intradermally, mtrathecally, and subcutaneously The particular mode of administration and the dosage regimen will be selected by the attending clinician, taking into account the particulars of the case (e g the subject, the disease, the disease state involved, and whether the treatment is prophylactic) Treatment can involve daily or multi-daily doses of compound(s) over a period of a few days to months, or even years
A therapeutically effective amount of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine, can be the amount of adenosine receptor antagonist necessary to stimulate the immune system of a subject Specific immunostimulatory effects that can be caused by adenosine receptor antagonists as well as specific immunosuppressive effects that can be caused by adenosine receptor agonists are described herem In some embodiments, an immunostimulatory amount of an adenosine receptor antagomst is an amount sufficient to stimulate an immune response (for instance, any of the stimulatory responses discussed herein) without causing a substantial cytotoxic effect (e g without killing more than 10% of cells in a sample)
An effective amount of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine can be administered in a single dose, or m several doses, for example daily, during a course of treatment However, the effective amount of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine will be dependent on the specific agomst or antagonist applied, the subject being treated, the seventy and type of the affliction, and the manner of administration of the therapeutic(s). For example, a therapeutically effective amount of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine can vary from about 0.1 mg/Kg body weight to about 1 g/Kg body weight.
Site-specific administration of the disclosed compounds can be used, for instance by applying an adenosine receptor antagonist to a precancerous region, a region of tissue from which a neoplasm has been removed, or a region suspected of being prone to neoplastic development.
The present disclosure also includes combinations of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine, with one or more other agents useful in the treatment of an immune-related disorder, condition, or disease. For example, the compounds of this disclosure can be administered in combination with effective doses of other immunosuppressives, immunostimulants, anti-cancer agents, anti-inflammatories, anti-infectives, and/or vaccines. The term "administration in combination" or "co-administration" refers to both concurrent and sequential administration of the active agents. In one example, SEQ ID NO: 1 is co-administered with an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine. In another example SEQ ID NO: 1 is administered before or after administration of an adenosine receptor inhibitor and/or an inhibitor of extracellular adenosine.
Examples of agent that can be used in combination with the compounds of the disclosure are AS-101 (Wyeth-Ayerst Labs.), bropirimine (Upjohn), gamma interferon (Genentech), GM-CSF (Genetics Institute), IL-2 (Cetus or Hoffman-LaRoche), human immune globulin (Cutter Biological), IMREG (from Imreg of New Orleans, Louisiana), SK&F106528 (Genentech), TNF (Genentech), azathioprine, cyclophosphamide, chlorambucil, and methotrexate. Treatment of a subject using the immunostimulatory compositions of the disclosure can be indicated after (or while) the subject has received an anti-proliferative or other cytotoxic therapeutic treatment. Examples of anti- proliferatives compounds include the following: ifosamide, cisplatin, methotrexate, cytoxan, procarizine, etoposide, BCNU, vincristine, vinblastine, cyclophosphamide, gencitabine, 5-flurouracie, paclitaxel, and doxorubicin.
In some examples, a subject is adminsitered a cytotoxic treatment, then monitored for a period of time (usually in the range of days to weeks) to determine if the treatment leads to an immunosuppressive effect. Such monitoring can include monitoring peripheral blood for leukopenia or pancytopenia, and/or monitoring T cell function. A subject that displays an immune suppression will be a candidate for treatment using the therapeutic methods of the disclosed disclosure.
Methods for Screening for Adenosine Receptor Antagonists
Also disclose are methods for screening for adenosine receptor antagonist effects using a cell of the immune system. The method includes contacting an activated immune cell with the compound, and assaying the activity of the immune cell. An increase in activity or a prolonged period of activation of the immune cell indicates that the compound is an adenosine receptor antagonist, which is efficient in an in vivo setting. Such methods can also be used to screen for immunosuppressants and immunostimulants, wherein increased activity or a prolonged period of activation of the immune cell indicates that the compound is an immunostimulant, and wherein decreased activity or a reduction in the period of activation of the immune cell indicates that the compound is an immunosuppressant. Among other uses, functional assays of receptor antagonist function permit optimization of the dosage amounts of each receptor agonist or antagonist effective in therapeutic uses. These assays can also be used to test known adenosine receptor antagonists, as well as newly identified adenosine receptor antagonists or putative adenosine receptor antagonists for immunosuppressive or immunostimulatory bioactivity. Candidate agents can initially be screened for subsequent selection and testing in one or more of the assays described herein.
Adenosine receptor antagonist immunostimulatory activity is the ability of an adenosine receptor antagonist to enhance an immune response in an immune cell, an immune system, and/or more generally in a subject. More specifically, adenosine receptor antagonist-related immunostimulation includes those effects that can be seen when an adenosine receptor antagonist is applied to an in vitro system using a dosage of less than 2 μg/ml, and more particularly when less than 1 μg/ml (e.g., about 0.1 μg/ml to as little as 0.003 μg/ml or less). In an in vivo system, these effects are seen at an application level of about 1 μg to about 5 μg in a 20 g mouse, or about 50 to about 250 μg/Kg body weight. Specific immunostimulatory effects that can be involved include stimulation of T-cell production, stimulation of interleukin production (e.g. production of IL-1 and/or IL-12 by macrophages), and activiation of natural killer cells and/or macrophages.
Methods for examining adenosine receptor inhibitor-mediated immunostimulation include those disclosed herein, such as direct measurement of the activation or proliferation of one or more immune cell types, or increased (or decreased) interleukin production (e.g. IL-1 or IL-12). Secondary effects of immune stimulation can also be measured as described herein, for instance by examining the formation of tumors, the relative rate of growth of a mmor, or tumor metastasis, or by resistance of an organism treated with the test compound to viral or other infection.
EXAMPLE 1 Activation of A2a Receptors Reduces Concanavalin-A-Induced Liver Damage in vivo
This example describes methods that were used to demonstrate that pharmacological activation of A2a receptors by a selective A2a agonist prevents liver damage in a model of inflammatory liver injury.
Concanavalin A (Con-A)-induced liver injury in mice is mediated by T-cells, macrophages and pro-inflammatory cytokines TNF-α, IL-4 and IFN-γ, and represents a well-described in vivo inflammation model of viral and autoimmune hepatitis. Five B6 wild type mice were injected intravenously (i.v.) with 20 mg/kg Con A (type IV, Sigma, St. Louis, MO) in sterile PBS alone or co- injected intraperitoneally (i.p.) with CGS21680 (2 mg/kg), isoproterenol (100 mg kg, Sigma, St. Louis, MO), or prostaglandm E2 (PGE2, 5 mg/kg, Sigma, St. Louis, MO) just before Con A treatment. The extent of liver damage and inflammation were quantified by measurmg serum levels of liver enzyme alanine aminotransferase (ALT) and TNF-α at 1 5 hours, 4 hours, and 8 hours after Con-A injection TNF-α was measured using an ELISA kit (R&D systems, Minneapolis, MN) according to manufacturer suggestions. Serum ALT activity was determined using a coloπmetπc assay kit (Sigma, St Louis, MO). The data disclosed herein are expressed as mean +/- s e.m. Differences between groups were evaluated usmg Student's t-test.
As shown in FIG. 1A, pharmacological activation of several cAMP-elevating Gs-coupled receptors prevented or reduced liver damage. Similarly, activation of A2a, inhibited Con-A-mduced pro-inflammatory TNF-α accumulation in vivo The A2a agomst, CGS21680, also inhibited secretion of IL-12 and IFN-γ by activated macrophages and T cells in vitro as measured using an ELISA kit (R&D systems, Minneapolis, MN) (FIG. IB) Therefore, pharmacological activation of A2a receptors or other Gs-proteιn-coupled receptors in vivo prevents or reduces Con-A-induced liver damage and pro-inflammatory TNF-α accumulation
EXAMPLE 2 Effect of A2a Agonists and Antagonists on cAMP levels in Mouse Liver Mononuclear Cells in vitro This example describes methods that were used to demonstrate that A2a receptor signaling (cAMP accumulation) is decreased or even abolished m liver mononuclear cells and macrophages from A2aR '" mice but not in cells from A2aR+/+ httermates.
Littermates or age-matched wild type (A2aR+/+) and homozygous A2a receptor gene deficient mice (A2aR"A) were used m all experiments for better reproducibihty of results. C57BL/6- background A2a receptor-deficient mice have been described previously (Chen et al , J Neuroscience 19:9192-9200, 1999, Apasov et al, Br J Pharmacol 131 -43-50, 2000, and Armstrong et al , Biochem J 354 123-130, 2001). The A2a receptor genotypes of mice were determined by Southern blot analysis (Chen et al , J Neuroscience 19 9192-200, 1999)
Stimulation of cells and measurement of cAMP levels were earned out as described previously (Apasov et al , Br J Pharmacol 131.43-50, 2000, herein incoφorated by reference) Briefly, liver mononuclear cells were isolated from parenchymal hepatocytes and cell debris using Percoll (Amersham Pharmacia Biotech, Uppsala, Sweden) The resulting liver mononuclear cells (1 x 105 cells/200 μl) were incubated at 37°C for 30 minutes in the presence of 10 μM CGS21680, 1 μM ZM241385 (Tocπs, Ballwin, MO), 100 μM isoproterenol, 50 μM forskolm, or 1 μM PGE2 The cAMP levels were determined using a cAMP enzyme lmmunoassay kit according to the manufacturer's instructions (Amersham Pharmacia Biotech, Buckinghamshire, England, UK) As shown in FIG 2 A, the A2aR antagomst, ZM241385, inhibited AGORA2a agonist (CGS21680)-ιnduced cAMP increase in mononuclear cells from A2aR+/+ mouse livers By contrast, the A2aR agomst, CGS21680, did not increase cAMP in mononuclear cells from A2aR"A mouse livers (FIG. 2B). Cells from A2aR_ " mice retained the cAMP response to ligands of other Gs protein coupled receptors (FIG. 2D). Therefore, A2a receptors can downregulate inflammation when activated by agonists. Therefore, there is a deficiency in cAMP-elevating receptors in A2a deficient mice. As a result, these mice can be used to establish the role of A2a receptors in inflammation in vivo.
EXAMPLE 3 Accumulation of Inflammatory Cytokines and Liver Damage in A2a-Receptor-Deficient Mice
This example describes methods that were used to demonstrate that the absence of functional A2a receptors results in increased inflammation and exacerbated tissue damage, in response to administration of Con-A.
A2a+ + (n=5) and A2a receptor gene deficient (A2a"'") mice (n=5) were used. Mice were injected intravenously with a sub-optimal dose (12.5 mg/kg) of Con-A (see inset, FIG. 3A), and subsequently, serum levels of ALT and cytokines were measured at 1, 6, 8, 24, and 48 hours as described in EXAMPLE 1 (cytokines were measured using an ELISA kit (R&D systems) according to manufacturer suggestions).
As shown in FIG. 3A, a large increase in serum ALT levels in A2a"'" mice as compared to A2a+/+mice was observed following Con-A treatment. Even a sub-optimal dose of inflammatory stimuli, Con-A (see inset to FIG. 3 A) resulted in the death of two out of five A2aR''" mice within 48 hours, while all A2aR+ + controls survived. Low doses of Con-A, which caused only minimal or no liver damage in control A2aR+/+ mice, were sufficient to induce extensive inflammation and liver damage in A2aR mice, as evidenced accumulation of dead cells and leukocytes using TdT apoptosis test and haematoxylin and eosin (H-E) stain.
The effect of TNF-α on liver injury in A2aR+ + (n=5) and A2aR"'' (n=5) mice was compared by injecting mice with a combination of D-galactosamine (Sigma, St. Louis, MO) and TNF-α (PharMingen, San Diego, CA). D-galactosamine (700 mg/kg) was injected intraperitoneally 30 minutes before i.v. injection of recombinant mouse TNF-α (4-15 μg/kg). After six hours, mice were sacrificed and the liver damage was evaluated by measuring serum ALT levels as described in EXAMPLE 1. As shown in FIG. 3B, deficiency in A2a receptors did not affect the susceptibility of hepatocytes to in vivo damage by TNF-α. Therefore, differences between A2aR+ + and A2aR_/" mice were not explained by increased susceptibility of A2aR"A hepatocytes to TNF-α, since TNF-α was equally efficient in directly destroying hepatocytes in both A2aR" and A2aR+/+ mice in vivo (FIG. 3B). Furthermore, excessive and prolonged pro-inflammatory TNF-α accumulation was observed in the serum of A2aR"A mice compared to low or undetectable TNF-α levels in A2aR+ + mice (FIG. 3B). IFN-γ was also present at higher concentrations and for a greater duration in A2aR_ ' mice, although levels of IL-4 were not different between A2aR" " and A2aR+ + mice. Inactivation of A2a receptors in A2aR+ + wild type mice using the A2 receptor antagonist ZM241385, also increased inflammatory tissue damage in A2aR" mice (FIG. 4). In summary, other c AMP -triggering Gs-coupled receptors do not appreciably compensate for the lack of A2a receptors on immune cells of A2aR" mice, as demonstrated by the increased sensitivity of A2aR" _ mice to Con-A. Mice with genetically inactivated adenosine receptors indeed lack functional adenosine receptors. A2aR_ " mice have other fully functional receptors (e.g. prostaglandin or beta-adrenergic receptors), which may function as natural down-regulators of immune response. These data demonstrate that A2a receptor gene deficient mice can be used to implicate A2a receptors as non-redundant downregulators of immune response in vivo. These data also demonstrate that the signal transduction pathway leading to cAMP accumulation in these mice is functional, thereby completely excluding any possibility that an artifactual mutation exists in A2aR gene deficient mice.
EXAMPLE 4 Inactivation of A2a receptors in vivo Exacerbates Liver Damage
This example provides methods that were used to demonstrate the tissue-protecting properties of A2 adenosine receptors in other models of inflammatory liver injury and systemic inflammation. These results demonstrate that targeted tissue damage can be achieved in vivo, for example when the targeted tissue is a mmor, the first agent or drug is specific immune cell (e.g. T cells or NK-T cells), a toxin (e.g. PEA) or a cytotoxic agent (e.g. carbon tetrachloride).
B6 mice (n=5) were injected with 12.5 mg/kg of Con-A alone or in combination with A2aR antagonist ZM241385 (2 mg/kg). Pseudomonas Exotoxin A (PEA, 100 μg/kg i.v., Sigma, St. Louis, MO) was also used to induce liver injury as follows. Mice were injected with PEA alone (n=6) or in combination with an i.p. injection of ZM241385 (n=7) before and 12 hours after the PEA injection. Carbon tetrachloride (CCl4)-induced hepatotoxicity was determined by injecting (i.p.) A2aR+ + (n=7) and A2aR_ " (n=7) mice with CC14 (0.5 ml/kg, Sigma) dissolved in olive oil. Subsequent to the injections, serum ALT measurements were made as described in
EXAMPLE 1, which indicate the extent of liver damage. In addition, histological evaluations and an analysis of tissue damage and apoptotic cells (the detection of apoptotic cells by in situ staining of single strand breaks in nuclear DNA) were determined.
As shown in FIG. 4A, pharmacological inactivation of A2a receptors in vivo using an adenosine receptor antagonist, exacerbates Con-A-induced liver damage. Inactivation of A2 adenosine receptors in A2aR+/+ mice by antagonist ZM241385, exacerbated the T cell- and TNF-α- dependent acute hepatotoxicity of PEA (FIG. 4B). Increased liver injury was also observed in A2aR" '" mice during chemically (CCl4)-induced acute hepatotoxicity (FIG. 4C).
Therefore, enhanced and prolonged accumulation of pro-inflammatory cytokines and exaggerated liver damage occurs in A2a-receptor-deficient mice as compared to wild-type mice. These data demonstrate that adenosine receptors, such as A2a receptors, function in vivo as physiological downregulators of immune response/inflammation and function as protectors from excessive tissue damage. Genetic inactivation of A2a receptors results in much stronger, longer mflammatory response to very low, sub-optimal doses of pro-inflammatory stimuli This is evidenced by tissue damage and death of animals (virtually no tissue damage and only short duration of higher levels of pro-inflammatory cytokines were detected m normal wild type ttermates which express A2a receptors). Virtually no tissue damage was detected m normal wild type httermates given the same dose of pro-inflammatory stimuli, Con-A without the administration of the A2aR antagonist.
In summary, these results demonstrate that a targeted tissue (in this example it was liver, but other tumors can be targeted) can be reduced or destroyed by disengaging immunosuppressive "brakes" using two agents The first agent is target tissue-specific, such as cytotoxic cells with tropism to a mmor, which can mcrease T-cell dependent tissue damage; such as an immunotoxm with tropism to a tumor (FIG 4B, which can result in immunotoxm-dependent targeted tissue damage; and such as a toxic chemical agent with tropism to the tumor (FIG. 4C), which can result in chemotherapy-dependent targeted tissue damage. This agent initiates non-observable or low intensity inflammation. The second agent inactivates or decreases hypoxia, extracellular adenosine, and/or the presence of adenosine receptors. This second agent enhances the intensity and prolongs the duration of targeted tissue destruction.
EXAMPLE 5 Enhanced Accumulation of Pro-inflammatory Cytokines and Tissue Damage in Endotoxin-Treated A2a Receptor-Deficient Mice
This example descπbes methods further used to demonstrate the role of A2a adenosine receptors m down-regulating pro-inflammatory cytokine accumulation and tissue damage, using an in vivo septic shock model following subcutaneous and i.v. bacterial endotoxm (LPS) injection.
Lipopolysacchande (LPS, E coh 011 LB4; 3 mg/kg, Sigma) was mjected i.v. into A2aR"A (n=l 1) and A2aR+ + (n=10) mice. Survival was monitored for 96-120 hours after LPS injection.
Statistical analysis confirmed the higher and faster mortality of mice without adenosine receptors As shown in FIG 5, all A2aT mice were dead by 48-72 hours, whereas survival of A2a+/+ mice was 10- 20% at 120 hours or 96 hours
Endotoxic shock in male A2aR '" mice and age-matched A2aR+ J" mice was induced by i.v injection of 3 or 5 mg/kg LPS Subsequently, at 1 hour and 16 hours after injection, blood samples were obtained by retro-orbital bleeding In another group of mice, LPS was mjected (100 μg/kg) into a dorsal air pouch, which was prepared using sterile air essentially as described m Levy et al. (Nat Immunol 2 612, 2001). Serum cytokine levels were determined at different times after LPS injection using ELISA kits obtamed from R&D systems according to manufacturer's suggestions as follows TNF-α and IL-6 levels were measured at 1 hour, and IL-12p40 and IL-lβ levels at 3 hours. As shown in FIGS 6A-D, absence of A2a adenosine receptors dramatically increases the level of pro- inflammatory cytokines in A2aR7" mice as compared to wild-type mice after air pouch LPS injection (infected wound model) Therefore, A2a adenosine receptors protect agamst death from septic shock, as mice lacking adenosme receptors (due to genetic inactivation), die faster (FIG 5) and have higher levels of cytotoxic TNF-α (FIG 6A) in response to bactenal endotoxm These results demonstrate that A2a adenosme receptors are the natural and non-redundant "brakes" of inflammatory tissue damage.
EXAMPLE 6 Enhanced Accumulation of Pro-Inflammatory Cytokines mRNA in A2aR mice After Activation of Immune Cells Mutant (A2aR / ) or wild-type mice were injected (I p ) with 20 nmol CpG ohgonucleotide (5'-T*C*CATGACGTTCCTG*A*T*G*C*T-3', asterisk means phosphorothioate SEQ ID NO 1) This toll-receptor activating CpG DNA preparation stimulates the immune system After one hour, mRNA was extracted from spleenocytes and analyzed for cytokine gene expression using an RNase protection assay with commercial templates (mCK-2b and mCK-3b, Pharmingen, San Diego, CA)
As shown in FIG 7, NF-kB transcπbed cytokine mRNAs (such as TNF-α and IL- 12p40) are dramatically increased in the absence of adenosine receptors Therefore, A2a adenosme receptors are involved m down-regulating pro-inflammatory cytokine mRNA accumulation including IL-12 (which is important for the T cell iesponse), during in vivo activation of immune cells by CpG This provides further evidence that targeted inactivation of adenosine receptors can be used to enhance the immune response Since cytokine IL-12 is important in promoting T-cell dependent immune response, these data demonstrate that genetic targeting the "adenosine accumulatιon->adenosιne - >adenosme receptors-> signaling " pathway in immune cells can enhance an immune response, which can be used as an lmmunoenhancer to improve vaccines
EXAMPLE 7 Adenosine Receptor Antagonists Increase Expression of
Inflammatory Cytokines in CpG-Activated Immune Cells in vivo
This example descnbes methods used to determine the role of adenosine receptor antagonists on NF-KB activity and expression of inflammatory cytokines such as TNF and IL-12p40
C57BL/6 mice were pretreated with ZM241385 (10 mg/kg I p ) 15 minutes before administration of SEQ ID NO 1, and the expression of cytokine mRNA in the spleen was compared with the mice treated with CpG alone, using the methods described m EXAMPLE 6
As shown in FIG 7, in the adenosine receptor mutant mice (adenosine receptors genetically inactivated), there was higher mRNA expression of NF-kB-regulated pro-inflammatory cytokines (TNFα, IL-12p40) Similarly, as shown in FIGS 8A and 8B, higher expression of NF-kB-regulated pro-inflammatory cytokines is observed in ZM241385-treated mice (adenosine receptors pharmacologically inactivated) Therefore, admimsteπng an adenosine receptor antagomst can be used to mcrease or enhance the transcription of pro-inflammatory cytokines following CpG activation, due to enhanced or increased NF-kB activity EXAMPLE 8
Adenosine Receptors Decrease NF-kB nuclear translocation and Cytokine mRNA
Transcription by Inhibiting IKK-Mediated Phosphorylation of IkB To further demonstrate the role of adenosine receptors on IKK- mediated phosphorylation, which is necessary for NF-kB nuclear translocation, which is needed for expression of cytokines, the following methods were used.
Mutant (A2aR ) and wild-type mice were injected (i.v.) with CpG to activate immune cells as described in EXAMPLE 6. One hour after the injection, nuclear extracts from peritoneal macrophages were isolated using standard methods. Nuclear extracts were compared in electrophoretic mobility shift assay (EMSA) for binding to specific DNA sequences according to routine EMSA methods, to determine the extent of NF-kB translocation into the nucleus of macrophages.
As shown in FIG. 9, CpG-induced NF-kB translocation into the nucleus is increased in the absence of adenosine A2a receptors. This demonstrates that A2a receptors negatively regulate NF- kB translocation into the nucleus, and thereby its activity, in vivo.
To determine the role of IkB phoshorylation, C57BL/6 mice were administered (i.v.) 5 nMol of CpG (SEQ ID NO: 1) in the presence or absence of the adenosine receptor agonist CGS21680 (2 mg/kg). After 20 minutes, macrophages were isolated as described above, and subjected to Western blot analysis using Ab that recognize IkB and Ab can distinguish the phosphorylated form of IkB
(IkB-P). As shown in FIG. 11A, phosphorylation of IkB was decreased or inhibited in the presence of the adenosine receptor agonist CGS21680, following immune stimulation with CpG. As shown in FIG. 1 IB, in control panels the parallel samples had similar levels of IkB as shown in Western blots. Therefore, NF-kB activity is regulated by adenosine receptors due to the inhibition of (or decrease of) phosphorylation of IkB by IKK. In the absence of phosphorylation of IkB, NF-kB cannot translocate into the nucleus, and therefore NF-kB cannot induce mRNA expression of inflammatory cytokines such as IL-12p40 and TNF-α. Enhancement of NF-kB transcription factor nuclear translocation by inhibition of A2 adenosine receptor-mediated signaling is explained by prevention of cAMP-induced inhibition of phosphorylation of IkB by IKK. As summarized in FIG. 11, the presence of active adenosine receptors, such as A2a, decreases or inhibits inflammation by blocking IKK-mediated IkB phosphorylation, and NF-kB nuclear translocation, thereby inhibiting or decreasing mRNA expression of pro-inflammatory cytokines.
EXAMPLE 9
Adenosine Receptor Antagonists Decrease Tumor Growth
To determine if a mmor self-protection mechanism could be defeated by reducing the presence of adenosine, the following methods were used. Because tumors are hypoxic, and hypoxia is conducive to adenosine accumulation in the brain, heart, and in solid mmors, it is possible that adenosine inhibits or prevents anti-mmor immune cells (such as T-killer cells) from contacting the mmor, thus preventing the mmor from being acted upon by the anti-mmor cells. For example, the presence of adenosine may inhibit or decrease the signaling of CTL chemokines receptors (which may result in a decreased attraction to a tumor and/or a decrease or inhibition of chemotaxis); inhibit or decrease motility of CTLs; inhibit or decrease production of inflammatory cytokines by CTL; inhibit or decrease the formation of CTL/tumor conjugates; inhibit or decrease FasL/granule exocytosis; and/or inhibit or decrease a lethal hit by CTL. As a result, if adenosine is blocked or reduced, then anti-mmor cells may be more effective in reducing a mmor. BALB/c mice were inoculated i.v. with CMS4 mmor cells (Methylcholanthrene-induced sarcoma, 2.5 x 105 cells) on day zero to induce the formation of lung tumors. Ten-days later, antigen- specific T-killer cells (CTL) cells (5 x 105 or 1 x 106 cells) were injected into the mice (i.v.) in the presence or absence of an i.p. injection of ZM241385 (10 mg/kg/day) or administered a relatively nonselective antagonist of A2a and A2b receptors, 1,3,7-thrimethylxantine (caffeine, 0.1% w/v) via drinking water to inactivate A2 Receptors on the CTL-cell surface, since it is through these receptors the mmor signals T-killer cells and thereby stop them from delivering the lethal "hit" to mmor cells. The lung mmors were subsequently examined on days 17, day 18 and day 24, by sacrificing the mice and evaluating their lungs for the number and size of metastasis by visual inspection.
As shown in FIGS. 12A-C, administration of an adenosine receptor antagonist, such as ZM241385 or caffeine, greatly improves immunotherapy of cancer tumors, as evidenced by a decrease in the number of metastatic nodules in the lung. In contrast, CTL cells alone were not as capable of efficiently reducing tumor metastasis.
To demonstrate that similar results are obtained with non-immunogeneic mmors, such as breast mmors, the following methods were used. BALB/c mice were inoculated subcutaneously with 1 x 105 non-immunogeneic 4T1 breast mmor cells (American Type Culture Collection, Manassas, VA, Catalog No. CRL-2539) on day zero to induce the formation of breast mmors. When injected into BALB/c mice, 4T1 cells spontaneously produce highly metastatic mmors that can metastasize to the lung, liver, lymph nodes and brain while the primary mmor is growing in situ. Seven days after tamor inoculation, mice were injected daily (i.p.) with ZM241385 (10 mg/kg) or caffeine (20 mg/kg). Time-dependent changes of mmor diameter and mmor volume were subsequently calculated.
As shown in FIG. 13, adenosine receptor antagonists slow the growth of non-immunogeneic 4T1 breast mmor cells (s.c), indicating that administration of an adenosine receptor antagonists to a subject having a mmor can reduce the number and or size of one or more tumors in the subject. These results also indicate that adenosine receptor antagonists reduce mmor growth by impairing angiogenesis. EXAMPLE 10 Adenosine Receptor Antagonists Decrease Tumor Size
This example describes methods that were used to improve anti-mmor vaccination by co- administering an adenosine receptor antagonist. BALB/c mice (immunocompromised nude mice; or immunocompetent C57BL/6 mice) were inoculated subcutaneously with B 16 melanoma cells (American Type Culture Collection, Manassas, VA), or B16-H2-Kd transfected mmor cells (increases the immunogenicity of the cells) on day zero to induce the formation of melanomas. The treatment with adenosine receptor antagonists started 28 days after injection of mmor cells, when the mmor reached 7-9 mm in diameter. As shown in FIG. 14, daily i.p. treatments with ZM241385 (0.2 mg/mouse) and caffeine (0.4 mg/mouse) resulted in mmor retardation that became significant after 3-7 days of treatment. The delay in mmor growth was greater in immunocompetent C57BL/6 mice, and less so in immunocompromised nude mice. However, as the mmor increases in size, both antagonists slow down mmor growth even in immunocompromised animals. This indicates that administration of adenosine receptor antagonists to a subject having a mmor can reduce the number and/or size of one or more mmors in the subject. In addition, adenosine receptor antagonists appear to improve both anti-mmor immunity and preventing or decreasing angiogenesis.
EXAMPLE 11 Adenosine Receptor Antagonists Improve Immune Response to
Subcutaneous and Intra-Peritoneal Vaccination
This example describes methods used to determine if an immune response to vaccines can be improved by co-administering an adenosine receptor antagonist, such as A2a and A3 antagonists, and whether any effects is altered if the vaccine is delivered via different delivery routes. TNP-KLH (100 μg, Sigma), a model antigen, was injected i.p. or subcutaneously into the footpad of A2aR+ + mice along with an adenosine receptor antagonist (about 1 mg/kg of theophylline (an A2a antagonist), ZM241385 (an A2a antagonist), or MRS 1220 (an A3 antagonist, Sigma)). The antigen was prepared for injection as follows. DNP-KLH (1.0 mg/ml, Biosearch Technologies, Inc. catalog no. T-5060-5) was prepared in PBS (e.g. dissolve 4.0 mg of purified DNP-KLH in PBS and raise volume to 4.0 ml). Complete Freund's Adjuvant (2.0 ml, CFA; Sigma F-5881) was vortexed and mixed with 2.0 ml of the TNP-KLH solution at 4°C. The CFA/KLH mixture was drawn into a 3- ml glass syringe with a 19-gauge needle. The syringe was attached to a double-ended locking hub connector or a plastic 3-way stopcock. An empty 2-ml glass syringe was attached to the other end and the mixture forced back and forth from one end to the other repeatedly. When the mixture was white and homogeneous, the connector or stopcock was disconnected, a 25 gauge needle attached, and tested for emulsion by placing a drop on the surface of 50 ml of cold water in a 100-ml beaker. The drop should hold together; if not, repeat mixing. 200μl (100 μg) was injected intraperitoneally (i.p.) into each mouse. Samples of blood were obtained by retro-orbital bleeding at 7, 14, and 21 days after TNP- KLH and theophylline injection. Serum levels of anti-TNP-specific IgGl, IgG2a, IgG2b, IgG3, and IgM were estimated using an ELISA kit (Sigma) according to the manufacturer's instructions.
Serum levels of anti-TNP-specific IgGl were markedly increased at seven days in mice co- injected with TNP-KLH and ZM241385 or MRS1220, as compared to control. IgG2 was also improved on day 7. Therefore, blocking the endogenous anti-inflammatory pathway using adenosine receptor antagonists improves immune response to vaccination (as evidenced by higher titers of antigen-specific immunoglobulin IgGi), when adenosine receptor antagonists are administered with a vaccine. Similarly, serum levels of anti-TNP-specific IgGl were markedly increased at seven days in mice co-injected i.p. (FIG. 15) or subcutaneously with TNP-KLH and theophyllin, as compared to control. Therefore, co-administration of an adenosine receptor antagonist with a vaccine potentiates the immune response to the vaccine, when vaccination is intra-peritoneal or subcutaneous. Pharmacological inactivation of adenosine receptors, such as A2a and A3, using antagonists results in higher titers of antigen-specific immunoglobulins IgGi, which such antagonists are administered with a vaccine.
EXAMPLE 12 Administration of Adenosine Receptor Antagonists with Anti-Cancer Agents This example describes methods that can be used to facilitate the treatment of cancer in a subject using one or more adenosine receptor antagonists, alone or in combination with anti-cancer agents. This protocol serves as an example of such a treatment method, and is not limiting. Those of skill in the art can modify the protocol to suit the needs of the subject, and to optimize for the particular agents used. Subjects can, but need not, have received previous chemo- radio- or gene therapeutic treatments. Optimally, the patient will exhibit adequate bone manow function (defined as peripheral absolute granulocyte count of >2,000/mm3 and platelet count of 100,000/mm3.
Adenosine receptor antagonists are administered orally or parenterally in dosage unit formulations containing standard, well known non-toxic physiologically acceptable carriers, adjuvants, and vehicles as desired. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intra-arterial injection, or infusion techniques. Adenosine receptor antagonists can be administered in dosages of about 0.1 mg/kg to about 1 g/kg, depending on the antagonist used. For example, adenosine receptor antagonists can be administered to a subject at a dose of least 0.5 mg/kg of body weight, such as 3 - 10 mg/kg. The adenosine receptor antagonists can be delivered to the patient before, after or concunently with the other anti-cancer agents. A typical treatment course can include about six doses delivered over a 7 to 21 day period.
Alternatively, a treatment course can include daily doses delivered over a 7 to 21 day period. Upon election by the clinician, the regimen can be continued six doses every three weeks or on a more frequent (daily, twice daily, four times a day, etc.) or less frequent (monthly, bimonthly, quarterly, etc.) basis. Of course, these are only exemplary times for treatment, and the skilled practitioner will readily recognize that many other time-courses are possible. The adenosine receptor antagonists can be combined with any of a number of conventional chemotherapeutic regimens.
Regional delivery of adenosine receptor antagonists is an efficient method for delivering a therapeutically effective dose to counteract the clinical disease. Likewise, the chemotherapeutic agents can be directed to a particular affected region. Alternatively, systemic delivery of either or both agents can be appropriate.
Clinical responses can be defined by an acceptable measure. For example, a complete response can be defined by the disappearance of all measurable disease for at least a month. A partial response can be defined by a 20% or greater, such as 50% or greater, such as 75% or greater, reduction of the sum of the products of perpendicular diameters of all evaluable mmor nodules or at least 1 month with no tumor sites showing enlargement. Similarly, a mixed response can be defined by a reduction of the product of perpendicular diameters of all measurable lesions by 20% or greater, such as 50% or greater, with progression in one or more sites. Of course, the above-described treatment regimes can be altered by those of skill in the art, who will be able to take the information disclosed in this specification and optimize treatment regimes.
EXAMPLE 13 Using Adenosine Receptor Antagonists as an Adjuvant
This example describes a protocol for using one or more adenosine receptor antagonists as an adjuvant by administering the adenosine receptor antagonist to a subject in combination with a vaccine. This protocol is intended to serve as an example of such a method, and is not limiting. Those of skill in the art will be able to modify the protocol to suit the needs of the subject, and to optimize for the particular antagonists and vaccines used.
Adenosine receptor antagonists are administered orally, topically, or parenterally in dosage unit formulations containing standard, well known non-toxic physiologically acceptable earners, adjuvants, and vehicles as desired. Adenosine receptor antagonists can be administered in dosages of about 0.1 mg/kg to about 1 g/kg, depending on the antagonist used. The adenosine receptor antagonists can be delivered to the patient before, after or concunently with the vaccine.
A typical vaccination course can comprise a single dose. Optionally, the course can be repeated every twelve weeks or on a more frequent (monthly, weekly, etc.) or less frequent (biannually, annually, every three years, every ten years, etc.) basis. Of course, these are only exemplary times for vaccination, and the skilled practitioner will readily recognize that many other time-courses are possible. The adenosine receptor antagonists can be combined with any of a number of conventional vaccines.
Clinical responses can be defined by an acceptable measure. For example, TNF-α, IFN-γ, IL-4, IL-6, IL-lβ and IL-12p40 levels in blood or serum samples can be determined using commercially available ELISA kits according to manufacmrer suggestions. Alternately, antibodies to the vaccine can be measured in blood or serum samples using ELISA kits.
Of course, the above-described treatment regimes can be altered by those of skill in the art. In view of the many possible embodiments to which the principles of our disclosure may be applied, it should be recognized that the illustrated embodiments are only examples of the disclosure and should not be taken as a limitation on the scope of the disclosure. Rather, the scope of the disclosme is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.

Claims (54)

We claim.
1. A method for enhancing an immune response in a subject, comprising administering a therapeutically effective dose of an adenosine receptor inhibitor, and/or an inhibitor of extracellular adenosme, to the subject, thereby enhancing the immune response
2. The method of claim 1, wherein enhancing the immune response compnses increasing an activity of an immune cell m the subject.
3 The method of claim 1, wherem the subject has a neoplasm, and enhancing the immune response results in a reduction m a neoplasm cell volume and/or a reduction a number of neoplasm cells in the subject.
4. The method of claim 1, wherem the adenosine receptor inhibitor is an adenosine receptor antagomst.
5. The method of claim 4, wherem the adenosine receptor antagonist is an A2a, A2b, or A3 adenosine receptor antagonist
6 The method of claim 4, wherem the adenosine receptor antagomst is ZM241385, MRS1220, 1,7, methylxantine (caffeine), theophilline, teobromm, SCH5826, KW-6002, or ADA- PEG.
7. The method of claim 4, wherem the adenosme receptor antagonist is a ribozyme, an antisense oligonucleotide, or a catalytic nucleic acid that selectively binds mRNA encodmg the adenosine receptor.
8 The method of claim 1, wherem the inhibitor of extracellular adenosine is an agent that mcreases endogeneous adenosine kinase activity or an agent that increases endogeneous adenosine deaminase activity, thereby decreasing extracelluar adenosine, wherein decreasing the extracelluar adenosine enhances the immune response in the subject.
9. The method of claim 1, wherein the inhibitor of extracellular adenosine is an oxygenahon agent, a redox-potential changing agent, an adenosine-accumulation-reducmg agent, adenosine deaminase, adenosine kinase or an adenosine kinase enhancer
10 The method of claim 2, wherem the immune cell is a cell that produces one or more pro- inflammatory cytokines
11. The method of claim 2, wherein the immune cell is a macrophage, granulocyte, monocyte, neutrophil, dendritic cell, T cell, B cell, or natural killer cell.
12. The method of claim 11, wherein the immune cell is a B cell and wherein the activity is antibody production.
13. The method of claim 11, wherein the immune cell is a T cell and wherein the activity is an increase in efficiency of tumor-infiltrating lymphocytes (TIL).
14. The method of claim 11, wherein the immune cell is a macrophage, granulocyte, monocyte, or dendritic cell, and wherein the activity is pro-inflammatory cytokine production.
15. The method of claim 1, further comprising administering an agent that decreases inflammation-associated local tissue hypoxia and/or the redox stams of molecules in an inflamed local tissue environment, to the subject.
16. The method of claim 2, wherein the activity of the immune cell results in an increase in inflammation.
17. The method of claim 1, wherein the immune response is a macrophage, monocyte, neutrophil, granulocyte, dendritic cell, T cell, B cell, or a natural killer cell response.
18. The method of claim 1, wherein the immune response is a pro-inflammatory cytokine response.
19. The method of claim 18, wherein the pro-inflammatory cytokine response is an increase in IL-12p40 and or TNF-α mRNA expression.
20. The method of claim 1, further comprising administering a vaccine to the subject, wherein inhibiting an adenosine receptor and/or inhibiting extracellular adenosine, enhances an immune response stimulated by the vaccine.
21. The method of claim 20, wherein the vaccine comprises an antigenic polypeptide or an antigenic epitope thereof.
22. The method of claim 20, wherein the vaccine is a viral vaccine.
23. The method of claim 22, wherein the viral vaccine is a live, attenuated, or heat killed vaccine.
24. The method of claim 20, wherein the vaccine comprises T-killer cells.
25. The method of claim 22, wherein the T-killer cells can decrease an amount of virally infected cells in a subject.
26. The method of claim 1, wherein the subject is infected with a pathogenic agent comprising a virus, bacterium, or fungus.
27. The method of claim 1, wherein the subject is immunocompromised.
28. The method of claim 27, wherein the subject is infected with an immunodeficiency virus.
29. The method of claim 28, wherein the immunodeficiency virus is HIV-1 or HIV-2.
30. The method of claim 27, wherein the subject is receiving immunosuppressive therapy.
31. The method of claim 2, wherein the neoplasm is a mmor greater than 2 mm in diameter, and wherein the mmor has local hypoxia areas.
32. The method of claim 31, wherein the neoplasm is a mmor of the lung, breast, skin, or liver.
33. The method of claim 3, further comprising administering to the subject a therapeutically effective dose of an anti-neoplastic agent.
34. The method of claim 33, wherein the anti-neoplastic agent selectively targets cells of the neoplasm.
35. The method of claim 34, wherein the anti-neoplastic agent is a nucleic acid that encodes a protein that promotes cell death.
36. The method of claim 36, wherein the anti-neoplastic agent is an alkylating drug, a folate antagonist, a purine antagonist, a pyrimidine antagonist, a spindle poison, a podophyllotoxin, an antibiotic, a nitrosurea, an inorganic ion, a biologic response modifier, an enzyme, or a hormone.
37. The method of claim 3, wherein the subject is first administered radiation or a radioactive isotope to increase damage of the neoplasm.
38. The method of claim 1, further comprising administering a cytotoxic T-lymphocyte
(CTL) or lymphokine-activated killer (LAK) cell to the subject.
39. A method for screening for an adenosine receptor antagonist, comprising: contacting an immune cell with an agent; and assaying for increased activity of the immune cell as compared to a control in the absence of the agent, wherein increase activity of the immune cell indicates that the agent is an adenosine receptor antagonist.
40. The method of claim 39, wherein the immune cell is a cell that produces one or more pro-inflammatory cytokines.
41. The method of claim 39, wherein the immune cell is a macrophage, granulocyte, monocyte, neutrophil, dendritic cell, T cell, B cell, or natural killer cell.
42. The method of claim 39, wherein increased activity comprises increased cyclic AMP, increased cytokines, increased apoptotic cell death, and/or morphological changes.
43. A method of preparing a tumor defense-resistant immune cell or an anti- viral immune cell comprising: incubating an immune cell under culture conditions that decrease a number of A2a adenosine receptors, wherein the culture conditions comprise incubation with adenosine receptor antagonists or adenosine deaminase activity inhibitors, thereby increasing an amount of adenosine in the culture conditions.
44. The method of claim 43, wherein the immune cell is a CTL or LAK cell.
45. A method for reducing a volume of a mmor and/or reducing a number of tumor cells in a subject, comprising administering mmor defense-resistant immune cells generated using the method of claim 43, to the subject, thereby reducing a volume of a mmor and or reducing a number of mmor cells in the subject.
46. A method for enhancing an immune response to a virus in a subject, comprising administering anti- viral immune cells generated using the method of claim 43, to the subject, thereby enhancing an immune response to a virus in the subject.
47. A method for disrupting a blood supply of a mmor in a subject, comprising: administering to the subject a therapeutically effective dose of an adenosine receptor inhibitor, and or an inhibitor of extracellular adenosine, thereby disrupting the blood supply of the tumor.
48. The method of claim 47, wherein disruption of the blood supply of the mmor results in a reduction of a volume of the mmor and or a reduction in a number of mmor cells in the subject.
49. A method of enhancing NF-kB activity in a subject, comprising: administering to the subject a therapeutically effective dose of a adenosine receptor inhibitor, and or an inhibitor of extracellular adenosine. thereby enhancing NF-kB activity in the subject.
50. A method for enhancing an immune response in a subject, comprising administering a therapeutically effective dose of an agent that decreases hypoxia to the subject, thereby enhancing the immune response.
51. The method of claim 50, wherein the agent that decreases hypoxia is oxygenation, ad the immune response decreases the volume of a mmor in the subject, or enhances an immune response to a virus in the subject.
52. A method for increasing expression of IL-12p40 in an immune cell, comprising contacting the immune cell with an adenosine receptor antagonist.
53. The method of claim 52, further comprising contacting the immune cell with SEQ ID NO: 1.
54. A method for increasing one or more immunoenhancing properties of SEQ ID NO: 1, comprising co-administering an adenosine receptor antagonist with SEQ ID NO: 1.
AU2002356962A 2001-12-12 2002-11-14 Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation Expired AU2002356962C1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US34077201P 2001-12-12 2001-12-12
US60/340,772 2001-12-12
US34258501P 2001-12-19 2001-12-19
US60/342,585 2001-12-19
PCT/US2002/036829 WO2003050241A2 (en) 2001-12-12 2002-11-14 Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation

Publications (3)

Publication Number Publication Date
AU2002356962A1 AU2002356962A1 (en) 2003-06-23
AU2002356962B2 true AU2002356962B2 (en) 2007-04-19
AU2002356962C1 AU2002356962C1 (en) 2008-05-01

Family

ID=26992233

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002356962A Expired AU2002356962C1 (en) 2001-12-12 2002-11-14 Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation

Country Status (8)

Country Link
US (5) US8080554B2 (en)
EP (1) EP1465634B1 (en)
JP (1) JP2005516917A (en)
AU (1) AU2002356962C1 (en)
CA (1) CA2470104C (en)
DK (1) DK1465634T3 (en)
ES (1) ES2528384T3 (en)
WO (1) WO2003050241A2 (en)

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003050241A2 (en) 2001-12-12 2003-06-19 The Government Of The United States Of America As Represented By The Secretary, Department Of Healthand Human Services Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
CA2533926C (en) * 2003-07-31 2013-10-15 Endacea, Inc. Methods and compositions for producing antigenic responses
CN1865275B (en) * 2005-05-17 2011-06-15 长春华普生物技术有限公司 Artificial single-chain deoxynucleotide having therapeutic effect to human B cell tumour
CA2644996A1 (en) * 2006-03-02 2007-09-13 Cv Therapeutics, Inc. A2a adenosine receptor antagonists
US20070208042A1 (en) 2006-03-03 2007-09-06 Sherwood Services Ag Method of using vasoconstrictive agents during energy-based tissue therapy
US7767686B2 (en) * 2006-03-03 2010-08-03 Covidien Ag Method of using adenosine receptor blockers during tissue ablation
EP2111231A4 (en) * 2007-02-13 2010-12-15 Univ Northeastern Methods and compositions for improving immune responses
MX2009011320A (en) 2007-04-20 2009-12-15 Enzon Pharmaceuticals Inc Enzymatic anticancer therapy.
PL2147116T3 (en) * 2007-04-20 2016-08-31 Leadiant Biosciences Ltd Stable recombinant adenosine deaminase
US20080262001A1 (en) * 2007-04-23 2008-10-23 Adenosine Therapeutics, Llc Agonists of a2a adenosine receptors for treating recurrent tumor growth in the liver following resection
WO2009033161A1 (en) * 2007-09-07 2009-03-12 The John Hopkins University Adenosine receptor agonists and antagonists to modulate t cell responses
WO2010040003A2 (en) * 2008-10-01 2010-04-08 The Scripps Research Institute Human a2a adenosine receptor crystals and uses thereof
US8883500B2 (en) * 2008-12-05 2014-11-11 Northeastern University Method of preparing adenosine-resistant anti-tumor T lymphocytes for adoptive immunotherapy
US9308211B2 (en) 2009-06-16 2016-04-12 Infirst Healthcare Limited Drug combinations and uses in treating a coughing condition
GB0919893D0 (en) * 2009-11-13 2009-12-30 Biocopea Ltd Drug composition and its use in therapy
US10016437B2 (en) 2009-06-16 2018-07-10 Infirst Healthcare Limited Drug combinations and uses in treating a coughing condition
US9314465B2 (en) 2009-06-16 2016-04-19 Infirst Healthcare Limited Drug combinations and uses in treating a coughing condition
US9782428B2 (en) 2013-03-18 2017-10-10 Northeastern University Method for generation of broadly neutralizing anti-pathogen antibodies
WO2014153424A1 (en) * 2013-03-19 2014-09-25 La Jolla Institute For Allergy And Immunology Reducing diabetes in patients receiving hmg-coa reductase inhibitors (statins)
US20180153984A1 (en) * 2015-04-30 2018-06-07 The Regents Of The University Of California Adjuvant particles comprising adenosine receptor antagonists
KR102114562B1 (en) 2015-08-11 2020-05-26 노파르티스 아게 5-bromo-2,6-di- (1H-pyrazol-1-yl) pyrimidin-4-amine for use in the treatment of cancer
WO2017123675A1 (en) 2016-01-11 2017-07-20 Synlogic, Inc. Microorganisms programmed to produce immune modulators and anti-cancer therapeutics in tumor cells
US20200078400A1 (en) 2016-12-03 2020-03-12 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
CN110248678A (en) 2016-12-03 2019-09-17 朱诺治疗学股份有限公司 The method for adjusting CAR-T cell
CN110913875A (en) 2017-01-06 2020-03-24 同生运营公司 Microorganisms programmed to produce immune modulators and anti-cancer therapeutics in tumor cells
JP7284707B2 (en) 2017-04-07 2023-05-31 ジュノー セラピューティクス インコーポレイテッド Engineered Cells Expressing Prostate-Specific Membrane Antigen (PSMA) or Modified Forms Thereof and Related Methods
JP2020522489A (en) 2017-06-02 2020-07-30 ジュノー セラピューティクス インコーポレイテッド Articles of manufacture and methods for treatment with adoptive cell therapy
AU2018291032A1 (en) 2017-06-29 2020-01-16 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
CA3066109A1 (en) * 2017-07-12 2019-01-17 Synlogic Operating Company, Inc. Microorganisms programmed to produce immune modulators and anti-cancer therapeutics in tumor cells
CN110997681B (en) 2017-08-01 2022-11-15 默克专利股份公司 Thiazolopyridine derivatives as adenosine receptor antagonists
ES2940408T3 (en) 2017-08-21 2023-05-08 Merck Patent Gmbh Benzoimidazole derivatives as adenosine receptor antagonists
AU2018320672B2 (en) 2017-08-21 2023-09-07 Merck Patent Gmbh Quinoxaline derivatives as adenosine receptor antagonists
US20210132042A1 (en) 2017-11-01 2021-05-06 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
KR20200116077A (en) 2017-11-01 2020-10-08 주노 쎄러퓨티크스 인코퍼레이티드 Chimeric antigen receptor and coding polynucleotide specific for B cell maturation antigen
EP3703688A2 (en) 2017-11-01 2020-09-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
MA51210A (en) 2017-12-01 2020-10-07 Juno Therapeutics Inc METHODS FOR DOSING AND MODULATING GENETICALLY MODIFIED CELLS
MA51184A (en) 2017-12-15 2020-10-21 Juno Therapeutics Inc ANTI-CCT5 BINDING MOLECULES AND RELATED METHODS OF USE
WO2019160829A1 (en) * 2018-02-13 2019-08-22 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes (tils) with adenosine a2a receptor antagonists and therapeutic combinations of tils and adenosine a2a receptor antagonists
WO2020033624A1 (en) * 2018-08-09 2020-02-13 The Research Foundation For The State University Of New York Targeting pathogenic b cells in autoimmunity
SG11202104081XA (en) 2018-10-25 2021-05-28 Merck Patent Gmbh 5-azaindazole derivatives as adenosine receptor antagonists
CN112867715B (en) 2018-10-25 2024-03-05 默克专利股份公司 5-azaindazole derivatives as adenosine receptor antagonists
EP3873937A2 (en) 2018-11-01 2021-09-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
JP2022512917A (en) 2018-11-01 2022-02-07 ジュノー セラピューティクス インコーポレイテッド Treatment method using a chimeric antigen receptor specific for B cell maturation antigen
MX2021005734A (en) 2018-11-16 2021-09-10 Juno Therapeutics Inc Methods of dosing engineered t cells for the treatment of b cell malignancies.
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
AR117844A1 (en) 2019-01-22 2021-09-01 Merck Patent Gmbh THIAZOLOPYRIDINE DERIVATIVES AS ANTAGONISTS OF THE ADENOSINE RECEPTOR
EP3917570A1 (en) 2019-01-29 2021-12-08 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
CN113795267A (en) * 2019-03-12 2021-12-14 艾库斯生物科学有限公司 Oncogene driven treatment of cancer
CA3135487A1 (en) * 2019-03-29 2020-10-08 Arcus Biosciences, Inc. Treatment of cancer utilizing an identified adenosine fingerprint
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH423800A (en) 1959-04-03 1966-11-15 Ciba Geigy Process for the preparation of new pyrazolo (3,4-d) pyrimidines
US4444487A (en) 1979-07-02 1984-04-24 Xerox Corporation Multiple-flash fuser
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4745055A (en) 1985-05-07 1988-05-17 California Biotechnology Inc. Fused protein for enzyme immunoassay system
ATE143052T1 (en) 1986-07-07 1996-10-15 Centocor Inc CHIMERIC MUINE-HUMAN IMMUNOGLOBULIN SPECIFIC FOR TUMOR-ASSOCIATED 17-1A ANTIGEN
US4902505A (en) 1986-07-30 1990-02-20 Alkermes Chimeric peptides for neuropeptide delivery through the blood-brain barrier
GB8626412D0 (en) 1986-11-05 1986-12-03 Clark M R Antibodies
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5116742A (en) 1986-12-03 1992-05-26 University Patents, Inc. RNA ribozyme restriction endoribonucleases and methods
US5565566A (en) 1987-04-24 1996-10-15 Discovery Therapeutics, Inc. N6 -substituted 9-methyladenines: a new class of adenosine receptor antagonists
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US5543508A (en) * 1987-12-15 1996-08-06 Gene Shears Pty. Limited Ribozymes
US5298508A (en) * 1988-07-19 1994-03-29 The United States Of America As Represented By The Department Of Health And Human Services Irreversible inhibitors of adenosine receptors
US5441883A (en) 1992-03-03 1995-08-15 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education On Behalf Of The Oregon Health Sciences University A3 adenosine receptor, DNA, and uses
US5424297A (en) 1992-04-27 1995-06-13 University Of Virginia Alumni Patents Foundation Adenosine dextran conjugates
US5391568A (en) * 1992-07-10 1995-02-21 American Health Foundation Inhibition of lung tumorigenesis by administration of a polyphenol
WO1994025462A1 (en) 1993-05-03 1994-11-10 The United States Of America, Represented By The 8-substituted 1,3,7-trialkyl-xanthine derivatives as a2-selective adenosine receptor antagonists
AU7623194A (en) 1993-09-03 1995-03-22 Vpi Holdings Ltd. Oligonucleotides with rna cleavage activity
WO1995010267A1 (en) * 1993-10-08 1995-04-20 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Use of nitric oxide-releasing compounds as hypoxic cell radiation sensitizers
CA2183992A1 (en) 1994-02-23 1995-08-31 Dan T. Stinchcomb Method and reagent for inhibiting the expression of disease related genes
US5504090A (en) 1994-03-30 1996-04-02 Trustees Of The University Of Pennsylvania Compositions and methods for the prevention and treatment of ischemia-reperfusion organ injury
US5536495A (en) 1994-04-15 1996-07-16 Foster; Preston F. Use of G-CSF to reduce acute rejection
EP0772619B2 (en) 1994-07-15 2010-12-08 The University of Iowa Research Foundation Immunomodulatory oligonucleotides
US5670501A (en) 1994-09-01 1997-09-23 Discovery Therapeutics, Inc. N-substituted 9-alkyladenines
JP4431638B2 (en) 1996-01-29 2010-03-17 アメリカ合衆国 Dihydropyridine-, pyridine-, benzopyran-one- and triazoloquinazoline derivatives, their preparation and their use as adenosine receptor antagonists
US5786360A (en) 1996-11-19 1998-07-28 Link Technology Incorporated A1 adenosine receptor antagonists
US5859019A (en) * 1997-03-07 1999-01-12 Trustees Of The University Of Pennsylvania Methods for protecting against cardiac ischemia by administering adenosine A2a receptor antagonists
US6180133B1 (en) * 1997-11-25 2001-01-30 Watson Pharmaceuticals, Inc. Antioxidant composition for topical/transdermal prevention and treatment of wrinkles
AU4573799A (en) * 1998-06-19 2000-01-05 Beth Israel Deaconess Medical Center Dietary supplement for individuals under stress
ATE290910T1 (en) 1998-07-16 2005-04-15 Univ Pennsylvania USE OF A3-ADENOSINE ANTAGONISTS FOR THE PRODUCTION OF A MEDICINAL PRODUCT FOR LOWERING THE INTERNAL EYE PRESSURE
US6326390B1 (en) 1998-08-25 2001-12-04 King Pharmaceuticals Reseach And Development, Inc. Use of adenosine A3 receptor antagonists to inhibit tumor growth
US6277878B1 (en) * 1998-09-07 2001-08-21 Pfizer Inc Substituted indole compounds as anti-inflammatory and analgesic agents
US6232297B1 (en) 1999-02-01 2001-05-15 University Of Virginia Patent Foundation Methods and compositions for treating inflammatory response
US6313131B1 (en) 1999-02-16 2001-11-06 Upsher-Smith Laboratories, Inc. Method of kidney treatment
US6316435B2 (en) * 1999-02-24 2001-11-13 Supergen, Inc. Combination therapy for lymphoproliferative diseases
US6147878A (en) * 1999-02-25 2000-11-14 Lucent Technologies Inc. Secure latch locking clip
US6322771B1 (en) 1999-06-18 2001-11-27 University Of Virginia Patent Foundation Induction of pharmacological stress with adenosine receptor agonists
US7309688B2 (en) * 2000-10-27 2007-12-18 Johnson & Johnson Consumer Companies Topical anti-cancer compositions and methods of use thereof
AU784878B2 (en) 1999-12-02 2006-07-13 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A1 A2A and A3 receptors and uses thereof
US20020115635A1 (en) 2001-02-21 2002-08-22 Pnina Fishman Modulation of GSK-3beta activity and its different uses
WO2003050241A2 (en) 2001-12-12 2003-06-19 The Government Of The United States Of America As Represented By The Secretary, Department Of Healthand Human Services Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
EP2111231A4 (en) 2007-02-13 2010-12-15 Univ Northeastern Methods and compositions for improving immune responses
WO2009033161A1 (en) 2007-09-07 2009-03-12 The John Hopkins University Adenosine receptor agonists and antagonists to modulate t cell responses

Similar Documents

Publication Publication Date Title
US11471528B2 (en) Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
AU2002356962B2 (en) Methods for using extracellular adenosine inhibitors and adenosine receptor inhibitors to enhance immune response and inflammation
JP7236483B2 (en) Combination of PD-1 antagonists and CpG-C type oligonucleotides for the treatment of cancer
US20230330182A1 (en) Combined preparations for the treatment of cancer or infection
JP4980530B2 (en) Pharmaceutical composition comprising an adenosine receptor agonist or antagonist
WO2017040620A1 (en) Antagonistic pdl1 aptamers and their applications in cancer therapy
JP2023542093A (en) Use of thiauranib in combination with immune checkpoint inhibitors in antitumor therapy
Oyajobi et al. Receptor activator of NF‐κB ligand, macrophage inflammatory protein‐1α, and the proteasome: Novel therapeutic targets in myeloma
Parrondo et al. IRAK-4 inhibition: emavusertib for the treatment of lymphoid and myeloid malignancies
CN115243719A (en) CTB006 and Ponatinib combined application
CN114929241A (en) Methods of treating cancer with STING agonists
WO2019149219A1 (en) Use of anti-cd3 immunotoxin combined with anti-pd-l1 single domain antibody in treatment of cancer
US20240091351A1 (en) FOCAL IONIZING RADIATION AND CD47/SIRPa DISRUPTION ANTICANCER COMBINATION THERAPY
Thomas Emerging drugs for adult acute lymphoblastic leukaemia
TW202327584A (en) New oral pharmaceutical composition for cancer therapy
JP2021050164A (en) Cancer immunopotentiator, and potentiated anticancer agent containing the same
Anaissie Infectious complications in patients with chronic lymphocytic Leukemia
CN116194105A (en) Methods, therapies and uses for treating cancer
CN114225023A (en) Application of anti-CSF-1R antibody and anti-PD-L1 antibody
WO2018223136A1 (en) Methods and formulation for enhancing response to radiotherapy
Mohamad et al. PL11. 05 THE ROLE OF DENDRITIC CELL GENOTYPE IN THE INDUCTION OF T-CELL
Barnes Potential Applications of New Drugs in the Management of Childhood Asthma