WO2014165916A1 - Methods and compositions for inducing an immune response - Google Patents

Methods and compositions for inducing an immune response Download PDF

Info

Publication number
WO2014165916A1
WO2014165916A1 PCT/AU2014/000390 AU2014000390W WO2014165916A1 WO 2014165916 A1 WO2014165916 A1 WO 2014165916A1 AU 2014000390 W AU2014000390 W AU 2014000390W WO 2014165916 A1 WO2014165916 A1 WO 2014165916A1
Authority
WO
WIPO (PCT)
Prior art keywords
ckr
antigen
subject
chemokine receptor
inhibitor
Prior art date
Application number
PCT/AU2014/000390
Other languages
French (fr)
Inventor
Shaun Mccoll
Ervin KARA
Mohammed Alsharifi
Iain COMERFORD
Original Assignee
Adelaide Research & Innovation Pty Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2013901227A external-priority patent/AU2013901227A0/en
Application filed by Adelaide Research & Innovation Pty Ltd filed Critical Adelaide Research & Innovation Pty Ltd
Publication of WO2014165916A1 publication Critical patent/WO2014165916A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6081Albumin; Keyhole limpet haemocyanin [KLH]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present disclosure relates to methods and compositions for inducing an immune response.
  • Vaccination is the administration of antigenic material to stimulate the immune system to develop adaptive immunity, typically to a pathogen. Vaccination represents one of the most effective means of treating infectious diseases. However, in spite of the success of some vaccines, such as vaccines against smallpox and polio, there remains a need to improve vaccination.
  • vaccines may also contain one or more adjuvants.
  • adjuvants have a number of disadvantages, including for example one or more of variable efficacy, restricted adjuvanticity and adverse side-effects.
  • problems including for example one or more of variable efficacy, restricted adjuvanticity and adverse side-effects.
  • the present disclosure relates to methods and compositions for inducing an immune response.
  • Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising:
  • Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising administering to the subject the selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide an immunogenic composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of an immunogenic composition.
  • Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to the selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a vaccine comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of a vaccine.
  • Certain embodiments of the present disclosure provide a nucleic acid vaccine comprising (i) a nucleic acid encoding an antigen and a nucleic acid encoding a chemokine receptor CCX-CKR inhibitor; and/or a nucleic acid encoding an antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a method of improving an immune response to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a method of reducing the amount of a selected antigen exposed to a subject to obtain a desired immunological response, the method comprising exposing the subject to a chemokine receptor CCX- CKR inhibitor.
  • Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor as an adjuvant.
  • composition comprising:
  • Certain embodiments of the present disclosure provide a combination product comprising the following components:
  • chemokine receptor CCX-CKR inhibitor a chemokine receptor CCX-CKR inhibitor
  • chemokine receptor CCX-CKR inhibitor a chemokine receptor CCX-CKR inhibitor
  • Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in an animal subject, the method comprising exposing the animal subject to the selected antigen, wherein the animal subject comprises a reduced chemokine receptor CCX-CKR activity.
  • Certain embodiments of the present disclosure provide a method of producing a hybridoma cell, the method comprising fusing a B cell with reduced chemokine receptor CCX-CKR activity with an immortalised cell.
  • Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody, the method comprising producing an antibody from a hybridoma comprising a B-cell and an immortalised cell, wherein the B cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity.
  • Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising reduced chemokine receptor CCX-CKR activity.
  • Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising a knock out and/or a knock down of a chemokine receptor CCX-CKR gene.
  • Certain embodiments of the present disclosure provide a method of improving protective immunity against an infectious agent in a subject, the method comprising exposing the subject to a selected antigen associated with the infectious agent and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a method of improving efficacy of a vaccine comprising a selected antigen, the method comprising exposing a subject to be vaccinated against the selected antigen to a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a method of improving high affinity antibody production to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • Figure 3 shows wild-type or CCX-CKR " " mice were immunised with Semliki Forrest Virus ( ⁇ -SFV via the intravenous route) and peripheral blood was collected on the indicated days post-immunisation.
  • Figure 4 shows wild-type or CCX-CKR " mice were immunised with Semliki Forrest and peripheral blood was collected on the indicated days post-immunisation.
  • the present disclosure relates to methods and compositions for inducing an immune response.
  • Certain embodiments of the present disclosure have one or more combinations of advantages.
  • some of the advantages of certain embodiments disclosed herein include one or more of the following: enhancing the efficacy of existing vaccines; allowing some vaccines to be delivered at lower doses; extending the availability of some vaccines; improving the induction of a humoral immune response; improving the induction of an adaptive immune response; improving the induction of a cytotoxic T lymphocyte (CTL) mediated immune response; improving the generation of high affinity antigen-specific antibodies; improving the generation of antibodies that are protective; improving the generation of neutralising antibodies; providing animal systems that are useful in producing high quality, high affinity monoclonal antibodies; and reducing the cost of vaccines.
  • CTL cytotoxic T lymphocyte
  • the present disclosure is based, at least in part, on the recognition that inhibition of the expression of the atypical chemokine receptor CCX-CKR enhances high affinity antibody production upon immunisation which are protective in a model virus infection.
  • Certain embodiments of the present disclosure provide a method of inducing an immune response in a subject.
  • Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising: inhibiting activity of a chemokine receptor CCX-CKR in the subject; and exposing the subject to the selected antigen.
  • the method is used for one or more of the following: to improve an immune response to the selected antigen in the subject; to vaccinate the subject against the selected antigen; to improve protective immunity in the subject against an infectious agent comprising the selected antigen or a related antigen; to improve efficacy of a vaccine comprising the selected antigen; to improve high affinity antibody to the selected antigen production in the subject; and to reduce the amount of the selected antigen exposed to the subject to obtain a desired immunological response.
  • the immune response comprises a humoral immune response.
  • the humoral immune response comprises a primary immune response.
  • the humoral immune response comprises a primary immune response and a secondary immune response.
  • the immune response comprises an adaptive immune response. In certain embodiments, the immune response comprises a CTL mediated immune response.
  • the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
  • a bacterial antigen e.g., a viral antigen
  • a fungal antigen e.g., a fungal antigen
  • a protozoan antigen e.g., a metazoan antigen
  • cancer antigen e.g., a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
  • an antigen derived from an allergen e.
  • an autoantigen e.
  • Other types of antigens are contemplated.
  • the selected antigen is derived from an infectious agent.
  • infectious agents include bacteria, viruses, fungi, pathogens, microorganisms, prions, protozoa, and metazoa.
  • the selected antigen is associated with an infectious agent.
  • the selected antigen is antigenically related to an antigen from an infectious agent.
  • the selected antigen comprises an exogenous antigen.
  • the selected antigen comprises all or part of a coat, a capsule, a cell wall, a flagellum, a fimbrae, and a toxin of a bacterium, a virus, and/or other pathogen or microorganism.
  • the selected antigen comprises an endogenous antigen, an autoantigen, and/or a tumor antigen.
  • the selected antigen is an antigen that is antigenically related to another antigen.
  • the selected antigen comprises a single antigen. In certain embodiments, the selected antigen comprises two or more antigens.
  • the immune response comprises induction of one or more of IgM, IgG, IgA and IgE antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of IgM antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of one or more of IgG, IgA and IgE antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of IgM and/or IgG antibodies to the selected antigen.
  • the antibodies comprise an affinity of at least 10 6 M _1 , at least 10 7 M _1 , at least 10 8 M _1 , at least 10 9 M _1 , at least 10 10 ⁇ _1 , at least 10 ⁇ ⁇ _1 , or at least 10 12 M _1 to the selected antigen.
  • antibody is to be understood to mean an immunoglobulin molecule with the ability to bind an antigenic region of another molecule, and includes monoclonal antibodies, polyclonal antibodies, multivalent antibodies, chimeric antibodies, multispecific antibodies, diabodies and fragments of an immunoglobulin molecule or combinations thereof that have the ability to bind to the antigenic region of another molecule with the desired affinity including a Fab, Fab', F(ab')2, Fv, a single- chain antibody (scFv) or a polypeptide that contains at least a portion of an immunoglobulin (or a variant of an immunoglobulin) that is sufficient to confer specific antigen binding, such as a molecule including one or more CDRs.
  • an immunoglobulin is a tetrameric molecule, each tetramer being composed of two identical pairs of polypeptide chains, each pair having one light chain and one heavy chain.
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids that is primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Human light chains are classified as K and ⁇ light chains.
  • Heavy chains are classified as ⁇ , ⁇ , ⁇ , , or ⁇ and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D” region of about 10 more amino acids.
  • the variable regions of each light/heavy chain pair form the antibody binding site, with the result that an intact immunoglobulin has two binding sites.
  • the variable regions further include hypervariable regions that are directly involved in formation of the antigen binding site. These hypervariable regions are usually referred to as Complementarity Determining Regions (CDR).
  • CDR Complementarity Determining Regions
  • FR Framework Regions In both light and heavy chains there are three CDRs (CDR-I to CDR-3) and four FRs (FR-I to FR-4).
  • the subject is human subject.
  • the subject is a mammalian subject, a primate, a livestock animal (such as a horse, a cow, a sheep, a goat, a pig), a domestic animal (such as a dog or a cat) and other types of animals such as monkeys, rabbits, mice, rats and laboratory animals.
  • the subject is an animal used for raising antibodies.
  • the subject is an animal used for producing a hybridoma cell. Use of any of the aforementioned animals as animal models is also contemplated. Veterinary applications are contemplated.
  • the subject is a subject to be vaccinated. In certain embodiments, the subject is immunologically naive to a selected antigen.
  • the immune response comprises the induction of high affinity antibodies to the selected antigen.
  • the high affinity antibodies comprise an affinity of at least 10 9 ⁇ _1 , ⁇ "1 , lO' ⁇ 'or 10 12 ⁇ _1 to the selected antigen.
  • the CCX-CKR gene (also referred to as CCRL1 or CCR11) encodes a receptor of the G protein-coupled receptor family, and is a receptor for C-C type chemokines. In humans, the receptor is encoded by the gene with the Genbank accession number AF 110640. The equivalent receptor in other species may be determined by a known method. The amino acid sequence of the human receptor has the Genbank accession number AF110640 1. Methods for identifying a CCX-CKR gene and/or receptor include for example nucleic acid and protein alignment programs, such as BLAST. The UniProt accession number for the corresponding mouse gene is Q924I3.
  • a chemokine receptor CCX-CKR is a human CCX- CKR receptor. In certain embodiments, the chemokine receptor CCX-CKR is a mammalian receptor. In certain embodiments, the CCX-CKR is an animal receptor. Examples of mammalian and animal receptors are as described herein.
  • a chemokine receptor CCX-CKR is a mouse CCX- CKR receptor.
  • Chemokine receptors CCX-CKR in other species are contemplated, including for example a horse, a cow, a sheep, a goat, a pig, a dog, a rat, a cat, a primate, a monkey, and a rabbit.
  • the CCX-CKR receptor is a homolog, paralog or ortholog of a receptor. In certain embodiments, the CCX-CKR receptor is a variant and/or a fragment of the receptor, such as a variant of the receptor arising from an alternatively spliced transcript.
  • inhibitor refers to a treatment, exposure or intervention that results directly and/or indirectly in a reduction of activity or function, including for example a decrease in activity, an inhibitory alteration in the timing and/or location of activity, or otherwise provide inhibitory control over activity.
  • inhibitor refers to an agent, treatment, or intervention that results directly and/or indirectly in a reduction of activity and/or function.
  • activity refers to the function of a species (eg a molecule) and includes, for example, the level, the specificity, the ability to interact (directly and/or indirectly) with and/or modify other species, the ability to signal, and the ability to cause changes (directly and/or indirectly) in other cellular and/or non- cellular events.
  • Examples of modulating the activity of a species include, for example, changes in the level of the species, changes in the localisation of the species, changes in the synthesis and/or degradation rates of the species, changes in the timing of activity, changes in the ability to interact with other species (such as a change in the ability of a ligand and a receptor to interact), changes in the chemical composition of the species, changes in signalling, and changes in cellular and/or non-cellular events affected by the species.
  • examples of inhibition include (i) directly and/or indirectly altering the activity of a receptor, such as altering the level of expression of the receptor, altering localisation of the receptor, partially or completely removing the gene for the receptor, altering internationalisation of the receptor, altering timing of receptor function, altering synthesis and/or degradation of the receptor, altering binding of a receptor to a ligand; (ii) a gene knockout and/or a gene knock down; (iii) altering the activity of a signalling pathway associated with an receptor; (iv) altering the level and/or altering the activity of a ligand that binds to the receptor, such as altering the synthesis of the ligand.
  • Other forms of inhibition are contemplated.
  • the inhibiting of activity of a chemokine CCX-CKR receptor comprises a gene knock out and/or a gene knock down.
  • Methods for producing a gene knockout or a gene knock down are known.
  • gene knock outs are as described in "Gene Knockout Protocols” (2001), edited by Martin J. Tymms, Ismail Kola, Human Press Inc.
  • Gene knock downs are as described in "Regulation of Gene Expression by Small RNAs” (2009) edited by Rajesh K. Gaur, John J. Rossi, CRC Press.
  • the gene knock out is a homozygous gene knock out. In certain embodiments, the gene knock out is a heterozygous gene knock out. [0068] In certain embodiments, a gene knock out and/or the gene knock down comprises a knock out and/or a knock down of a chemokine receptor CCX-CKR gene. In certain embodiments, a gene knock out and/or the gene knock down comprises a knock out and/or a knock down of a gene that regulates the expression of a chemokine receptor CCX-CKR gene. The knock out and/or a knock down of other genes is contemplated.
  • the subject comprises a gene knock out and/or a gene knock down.
  • the subject comprises a homozygous gene knock down.
  • the subject comprises a heterozygous gene knock down. Examples include a mouse comprising a homozygous knock out in a chemokine receptor CCX-CKR gene.
  • the inhibiting of activity of a chemokine receptor CCX-CKR comprises exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • the inhibitor comprises a selective inhibitor. In certain embodiments, the inhibitor comprises a non-selective inhibitor.
  • inhibitors include a drug, a small molecule, a protein, a polypeptide, a lipid, a carbohydrate, a nucleic acid, an oligonucleotide, a ribozyme, a biologic, an aptamer, a peptide, a non-peptide, a cofactor, a ligand, a ligand mimetic, a receptor, an enzyme, a kinase, a phosphatase, a cytokine, a growth factor, a metal ion, a chelate, an antisense nucleic acid, an inhibitor RNA, a microRNA, a siRNA, an antibody or antigen binding part thereof, an antibody mimetic, an amino acid, an antagonist, an inhibitor, or a suppressor.
  • a drug a small molecule, a protein, a polypeptide, a lipid, a carbohydrate, a nucleic acid, an oligonucleotide,
  • An inhibitor of a chemokine receptor CCX-CKR may be synthesized or produced by a method known in the art, or obtained commercially.
  • the chemokine receptor inhibitor comprises one or more of an antagonist, a small interfering RNA, a microRNA, an antisense RNA, a ligand, a ligand mimetic, or an antibody and/or an antigen binding part thereof.
  • the chemokine receptor inhibitor comprises one or more of an antagonist to a chemokine receptor CCX-CKR, a small interfering RNA for a chemokine receptor CCX-CKR, a microRNA for a chemokine receptor CCX-CKR, an antisense RNA for a chemokine receptor CCX-CKR, a ligand, or a ligand mimetic for a chemokine receptor CCX-CKR, or an antibody and/or an antigen binding part thereof for a chemokine receptor CCX-CKR.
  • the inhibitor is an inhibitor of ligand binding to a chemokine receptor CCX-CKR.
  • examples of such inhibitors include a ligand antagonist or an antibody antagonist.
  • the inhibitor reduces binding of a ligand to a chemokine receptor CCX-CKR.
  • the inhibitor inhibits the binding of a ligand to a chemokine receptor CCX-CKR.
  • the inhibitor is an inhibitor of one or more of CCL25 binding, CCL19 binding and CCL21 binding to a chemokine receptor CCX-CKR.
  • the inhibitor inhibits the activity of a ligand for a chemokine receptor CCX-CKR.
  • the inhibitor is an inhibitor of one or more of CCL25 activity, CCL19 activity and CCL21 activity.
  • the inhibitor inhibits the synthesis of a ligand for a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor inhibits the synthesis of one or more of CCL25, CCL19 and CCL21.
  • the inhibitor inhibits the processing of a species that forms a ligand for a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor inhibits the processing of precursor of one or more of CCL25, CCL19 and CCL21.
  • the inhibitor is an inhibitor of expression of a chemokine receptor CCX-CKR expression.
  • examples of such inhibitors include an antisense RNA to a chemokine receptor CCX-CKR mRNA or a small interfering RNA to a chemokine receptor.
  • the inhibitor modulates internalisation of a chemokine receptor CCX-CKR.
  • the inhibitor promotes internalisation of a chemokine receptor CCX-CKR.
  • the inhibitor inhibits internalisation of a chemokine receptor CCX-CKR
  • the inhibitor blocks internalisation of a chemokine receptor CCX-CKR
  • the inhibitor is an antisense nucleic acid, such as an antisense RNA.
  • the inhibitor is a small interfering RNA.
  • the inhibitor is a microRNA.
  • nucleic acid refers to an oligonucleotide or a polynucleotide and includes for example DNA, RNA and be may be single stranded, double stranded or triple stranded.
  • the nucleic acid may be any type of nucleic acid, including for example a non-naturally occurring nucleic acid or a nucleic acid of genomic origin, cDNA origin (derived from a mRNA), a microRNA, derived from a virus, or of synthetic origin.
  • the nucleic acid may also be an isolated nucleic acid or a purified nucleic acid.
  • isolated refers to a nucleic acid that has been separated from its natural environment. For example, an isolated nucleic acid or may be in a substantially purified state, being substantially free of other substances with which it is associated in nature or in vivo.
  • the chemokine receptor CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof. In certain embodiments, the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
  • the inhibitor comprises an antibody and/or an antigen binding fragment thereof. In certain embodiments, the inhibitor comprises a neutralising antibody. In certain embodiments, the inhibitor comprises an antagonist antibody. In certain embodiments, the inhibitor comprises an antibody that promotes or inhibits internalisation of a chemokine receptor CCX-CKR.
  • the antigen-binding fragment comprises a Fab, Fab', F(ab')2, Fd, Fv, a single-chain antibody (scFv), a chimeric antibody, a diabody or a polypeptide that contains at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding.
  • a Fab fragment is a monovalent fragment consisting of the VL, VH, CL and CH I domains.
  • a F(ab') 2 fragment is a bivalent fragment including two Fab fragments linked by a disulphide bridge at the hinge region.
  • a Fd fragment consists of the VH and CH I domains.
  • a Fv fragment consists of the VL and VH domains of a single arm of an antibody.
  • a dAb consists of a VH domain.
  • a single chain antibody (scFv) is an antibody in which VL and VH regions are paired to form a monovalent molecule via a synthetic linker that enable them to be made as a single protein chain.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites.
  • Antibody fragments that contain specific binding sites may be generated by a known method.
  • Methods for producing antigen-binding fragments or portions of antibodies are known in the art, for example as described in “Antibody Engineering: Methods and Protocols” (2004) ed. by B.K.C. Lo Humana Press, herein incorporated by reference; and “Antibody Engineering: A Practical Approach” (1996) ed. by J. McCafferty, H.R. Hoogenboom and DJ. Chriswell Oxford University Press, herein incorporated by reference.
  • F(ab') 2 fragments can be produced by pepsin digestion of the antibody molecule, and Fab fragments can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity, as described for example in Huse, W. D. et al. (1989) Science 254: 1275-1281, herein incorporated by reference.
  • Antibodies may be generated using known methods. For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others, may be immunized by injection with an appropriate antigen. Depending on the host species, various adjuvants may be used to increase an immunological response. Such standard adjuvants include Freund's adjuvant, mineral gels such as aluminium hydroxide, and surface-active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • standard adjuvants include Freund's adjuvant, mineral gels such as aluminium hydroxide, and surface-active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • the antibody is a polyclonal antibody.
  • a polyclonal antibody is a mixture of antibodies having different antigen specificities. Methods for producing and isolating polyclonal antibodies are known. In general, polyclonal antibodies are produced from B-lymphocytes. Typically polyclonal antibodies are obtained directly from an immunized subject, such as an immunized animal.
  • the antibody is a monoclonal antibody.
  • Monoclonal antibodies may be prepared using a technique that provides for the production of antibody molecules by continuous isolated cells in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. Methods for the preparation of monoclonal antibodies include for example Kohler et al. (1975) Nature 256:495-497, herein incorporated by reference; Kozbor et al. (1985) J. Immunol. Methods 81 :31-42, herein incorporated by reference; Cote et al. (1983) Proc. Natl. Acad. ScL 80:2026-2030, herein incorporated by reference; and Cole et al. (1984) MoT Cell Biol. 62: 109-120, herein incorporated by reference.
  • the antibody and/or an antigen binding fragment thereof comprises an isolated antibody. In certain embodiments, the antibody and/or an antigen binding fragment thereof comprises a purified antibody. Methods for producing and isolating polyclonal and monoclonal antibodies are known. The term "isolated" refers to an antibody that has been separated from its natural environment.
  • the antibody has an isotype selected from the group consisting of IgGl, IgG2a, IgG2b, IgG3, IgM and IgA.
  • the antibody and/or an antigen binding fragment thereof is a mouse antibody and/or an antigen binding fragment thereof, a human antibody and/or an antigen binding fragment thereof, or a humanized antibody and/or an antigen binding fragment thereof.
  • Humanized antibodies, or antibodies adapted for non-rejection by other mammals may be produced by a suitable method known in the art, including for example resurfacing or CDR grafting. In resurfacing technology, molecular modeling, statistical analysis and mutagenesis are combined to adjust the non-CDR surfaces of variable regions to resemble the surfaces of known antibodies of the target host.
  • the antibody may be generated as described in U.S. Pat. No. 6, 180,370, herein incorporated by reference; WO 92/22653, herein incorporated by reference; Wright et al. (1992) Critical Rev. in Immunol. 12(3,4): 125-168, herein incorporated by reference; and Gu et al. (1997) Thrombosis and Hematocyst 77(4):755-759), herein incorporated by reference.
  • Humanized antibodies typically have constant regions and variable regions other than the complementarity determining regions (CDRs) derived substantially or exclusively from a human antibody and CDRs derived substantially or exclusively from the non- human antibody of interest.
  • CDRs complementarity determining regions
  • chimeric antibodies for example the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, may be performed by a suitable method.
  • chimeric antibodies may be produced as described in Morrison, S. L. et al. (1984) Proc. Natl. Acad. Sci 81 :6851-6855; Neuberger, M. S. et al. (1984) Nature 312:604-608,; and Takeda, S. et al. (1985) Nature 314:452-454,.
  • Immunoassays may be used for screening to identify antibodies and/or antigen binding fragments thereof having the desired specificity.
  • Antibody molecules and antigen binding fragments thereof may also be produced recombinantly by methods known in the art, for example by expression in E.coli expression systems. For example, a method for the production of recombinant antibodies is as described in US patent 4,816,567, herein incorporated by reference. Antigen binding fragments may also be produced by phage display technologies or using peptide libraries, which are known.
  • the antibody and/or an antigen binding fragment thereof binds to a human a chemokine receptor CCX-CKR. In certain embodiments, the antibody and/or an antigen binding fragment thereof binds to a mammalian chemokine receptor CCX-CKR. In certain embodiments, the antibody and/or an antigen binding fragment thereof is a neutralising antibody to a chemokine receptor CCX-CKR.
  • the inhibiting of activity of a chemokine receptor CCX-CKR comprises exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • exposing refers to directly and/or indirectly contacting and/or treating a subject with an agent.
  • an inhibitor of a chemokine receptor may be administered to a subject to expose the subject to the inhibitor, or an agent may be administered to a subject that results in the production of an inhibitor in the subject, thereby exposing the subject to the inhibitor.
  • one or more cells may be removed from a subject and contacted directly or indirectly with an inhibitor, and the cells then introduced back into the same or another subject to effect exposure to an inhibitor.
  • a subject may be exposed to a nucleic acid expressing an inhibitor, for example by way of a nucleic acid vector encoding an inhibitor.
  • Methods for the construction of vectors expressing an inhibitor are known and include for example in Sambrook, J, Fritsch, E.F. and Maniatis, T. Molecular Cloning: A Laboratory Manual 2nd. ed. Cold Spring Harbor Laboratory Press, New York. (1989).
  • Methods for the delivery of nucleic acids to a subject are known and include for example [00108]
  • the subject will be exposed to an effective amount of an agent.
  • the term "effective amount" as used herein refers to that amount of an agent that is sufficient to illicit the desired response or outcome. The effective amount will vary depending upon a number of factors, including for example the specific activity of the agent being used and the characteristics of the subject.
  • a subject is exposed to an inhibitor of a chemokine receptor CCX-CKR in an amount ranging from one of the following selected ranges: 1 ⁇ g/kg to 100 mg/kg; 1 ⁇ g/kg to 10 mg/kg; 1 ⁇ g/kg to 1 mg/kg; 1 ⁇ g/kg to 100 ⁇ g/kg; 1 ⁇ g/kg to 10 ⁇ g/kg; 10 ⁇ g/kg to 100 mg/kg; 10 ⁇ g/kg to 10 mg/kg; 10 ⁇ g/kg to 1 mg/kg; 10 ⁇ g/kg to 100 ⁇ g/kg; 100 ⁇ g/kg to 100 mg/kg; 100 ⁇ g/kg to 10 mg/kg; 100 ⁇ g/kg to 1 mg/kg; 1 mg/kg to 10 mg/kg; and 10 mg/kg to 100 mg/kg body weight.
  • Other amounts are contemplated.
  • the exposing the subject to a chemokine receptor comprises administering a chemokine receptor CCX-CKR inhibitor to the subject.
  • the inhibitor is administered to the subject to produce a concentration of an inhibitor of a chemokine receptor CCX-CKR of 0.1 nM or greater, 0.5 nM or greater, 1 nM or greater, 5 nM or greater, 10 nM or greater, 50 nM or greater, 100 nM or greater, 500 nM or greater, 1 uM or greater, 5 uM or greater, 10 uM or greater , 100 uM or greater, 500 uM or greater, 1 mM or greater, or 10 mM or greater.
  • an antibody and/or an antigen binding fragment thereof is administered to the subject to produce a concentration in one of the aforementioned ranges.
  • Methods for administering antibodies are known and include for example Therapeutic Antibodies (2008) Edited By Yuti Chernajovsky, Ahuva Nissim. Springer Publishing.
  • the inhibitor of a chemokine receptor CCX-CKR is administered to the subject in an amount ranging from one of the following selected ranges: 1 ⁇ g/kg to 100 mg/kg; 1 ⁇ g/kg to 10 mg/kg; 1 ⁇ g/kg to 1 mg/kg; 1 ⁇ g/kg to 100 ⁇ /13 ⁇ 4; 1 ⁇ /13 ⁇ 4 to 10 ⁇ /13 ⁇ 4; 10 ⁇ /13 ⁇ 4 to 100 mg/kg; 10 ⁇ g/kg to 10 mg/kg; 10 ⁇ g/kg to 1 mg/kg; 10 ⁇ g/kg to 100 ⁇ g/kg; 100 ⁇ g/kg to 100 mg/kg; 100 ⁇ g/kg to 10 mg/kg; 100 ⁇ g/kg to 1 mg/kg; 1 mg/kg to 10 mg/kg; and 10 mg/kg to 100 mg/kg body weight.
  • an antibody and/or an antigen binding fragment thereof may be administered to the subject to produce in one of the aforementioned ranges.
  • a, antibody may be administered at a concentration of 0.5, 1, 2, 3, 4, 5, 10, 15 or 20 mg/kg body weight. Other amounts are contemplated.
  • the dose and frequency of administration may be determined by one of skill in the art.
  • the inhibitor of a chemokine receptor CCX-CKR is administered to a subject to inhibit the activity of the receptor in the subject.
  • Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising administering to the subject the selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • the inhibitor may be administered to the subject in a suitable form.
  • administering includes for example administering the inhibitor, or administering a prodrug of the inhibitor, or a derivative of the inhibitor that will form a therapeutically effective amount of the inhibitor within the body of the subject.
  • routes of administration that are systemic (e.g., via injection such as intravenous injection, orally in a tablet, pill, capsule, or other dosage form useful for systemic administration of pharmaceuticals), topical (e.g., creams, solutions, and the like, including solutions such as mouthwashes, for topical oral administration), and administration utilising nucleic acids expressing the inhibitor, such as viral vectors.
  • Other routes of administration are contemplated.
  • the inhibitors may be administered alone or may be delivered in a mixture with other therapeutic agents and/or agents that enhance, stabilise or maintain the activity of the inhibitor.
  • an inhibitor is administered in conjunction with an administration vehicle.
  • an administration vehicle e.g., pill, tablet, implant, injectable solution, etc.
  • the inhibitor may contain the inhibitor and additional agent(s).
  • the inhibitor is administered orally.
  • the inhibitor is administered via injection, such as intravenous injection.
  • the inhibitor is administered parenterally.
  • the inhibitor is administered by direct introduction to the lungs, such as by aerosol administration, by nebulized administration, and by being instilled into the lung.
  • the inhibitor is administered by implant. In certain embodiments, the certain embodiments, the inhibitor is administered by subcutaneous injection, intraarticular ly, mucosally, rectally, intranasally, intraocularly, vaginally, or transdermally.
  • the subject is exposed to the selected antigen.
  • Methods for exposing a subject to an antigen are known.
  • the amount of antigen exposed to the subject is in the range of 5 micrograms to 250 micrograms of antigen per dose.
  • the amount of antigen comprises 5 ⁇ g or more, 10 ⁇ g or more, 20 ⁇ g or more, 50 ⁇ g or more, 100 ⁇ g or more, 200 ⁇ g or more, or 2 0 ⁇ g or more.
  • the dose depends on the subject to be treated, capacity of the subject's immune system to generate a protective immune response, and the degree of protection desired.
  • exposing of the selected antigen to the subject may be by conventional routes such intravenous, subcutaneous, intradermally, intraperitoneal, or mucosal routes.
  • the exposure may be by parenteral injection, for example, a subcutaneous or intramuscular injection.
  • the exposure may also be by expressing a selected antigen from for example a virus or a nucleic acid vector, such as a nucleic acid vaccine.
  • the exposing of the selected antigen to the subject comprises administration of the selected antigen.
  • the inhibiting of activity of the chemokine receptor occurs at any one or more of before, concurrent with, or after exposure of the subject to the selected antigen.
  • administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject prior to exposure of the subject to the selected antigen.
  • administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject concurrent with exposure of the subject to the selected antigen.
  • a composition comprising an inhibitor of chemokine receptor CCX-CKR activity and a selected antigen may be administered to a subject.
  • administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject after exposure of the subject to the selected antigen.
  • the methods as described herein may be used to improve an immune response to a selected antigen. Methods for determining the improvement in an immune response are known.
  • Certain embodiments of the present disclosure provide a method of improving an immune response to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • the methods as described herein may be used to reduce the amount of a selected antigen exposed to a subject to obtain a desired immunological response.
  • the amount of antigen needed to obtain a desired immunological response may be reduced by 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, or 90% or more.
  • Certain embodiments of the present disclosure provide a method of reducing the amount of a selected antigen exposed to a subject to obtain a desired immunological response, the method comprising exposing the subject to a chemokine receptor CCX- CKR inhibitor.
  • the methods as described herein may be used to improve protective immunity against an infectious agent in a subject.
  • Certain embodiments of the present disclosure provide a method of improving protective immunity against an infectious agent in a subject, the method comprising exposing the subject to a selected antigen associated with the infectious agent and a chemokine receptor CCX-CKR inhibitor.
  • infectious agents include bacteria, viruses, fungi, protozoa, metazoan, pathogens and/or microorganisms.
  • the methods as described herein may be used to improve efficacy of a vaccine.
  • Methods for determining the efficacy of a vaccine include for example determination of the incidence of a disease among subjects who have received a vaccine as compared to the incidence in unvaccinated subjects.
  • Certain embodiments of the present disclosure provide a method of improving efficacy of a vaccine comprising a selected antigen, the method comprising exposing a subject to be vaccinated against the selected antigen to a chemokine receptor CCX-CKR inhibitor.
  • the methods as described herein may be used to improve high affinity antibody production.
  • High affinity antibodies are described herein.
  • Certain embodiments of the present disclosure provide a method of improving high affinity antibody production to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
  • the methods as described herein may be used to increase CTL activity.
  • compositions comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • the composition is an immunogenic composition.
  • the composition is a vaccine composition.
  • the composition is a pharmaceutical composition.
  • Certain embodiments of the present disclosure provide an immunogenic composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Immunogenic compositions may be produced by a known method.
  • compositions may be produced as described in "Development of Vaccines: From Discovery to Clinical Testing” (2011) edited by Manmohan Singh, Indresh K. Srivastava Wiley Publishing.
  • the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
  • chemokine receptor CCX-CKR inhibitors are as described herein.
  • the inhibitor comprises one or more of an antagonist (eg a peptide antagonist or a non-peptide antagonist), a small interfering RNA, a microRNA, an antisense RNA, a ligand for a chemokine receptor CCX-CKR, an antibody and/or an antigen binding part thereof.
  • an antagonist eg a peptide antagonist or a non-peptide antagonist
  • small interfering RNA eg. a small interfering RNA, a microRNA, an antisense RNA, a ligand for a chemokine receptor CCX-CKR, an antibody and/or an antigen binding part thereof.
  • the chemokine receptor CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof, as described herein. In certain embodiments, the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
  • the antibody comprises a polyclonal antibody, as described herein. In certain embodiments, the antibody comprises a monoclonal antibody, as described herein.
  • An example of an immunogenic composition is as follows:
  • the selected antigen may be adsorbed on 0.5 mg aluminium as aluminium hydroxide.
  • the composition may be administered by intramuscular administration.
  • the composition further comprises one or more excipients.
  • Excipients are known in the art and are as described, for example, in Grabenstein JD. ImmunoFacts: Vaccines and Immunologic Drugs - 2012 (37th revision). St Louis, MO: Wolters Kluwer Health, 2011.
  • excipients include antibiotics (eg neomycin and/or polymyxin B; gentamicin); formaldehyde; stabilizers (eg monosodium glutamate, 2-phenoxyethanol, lactose, sucrose (both sugars), glycine, gelatin, preservatives (eg. thiomersal, phenoxyethanol, and formaldehyde); and diluents (eg sterile saline or sterile water).
  • antibiotics eg neomycin and/or polymyxin B; gentamicin
  • formaldehyde eg monosodium glutamate, 2-phenoxyethanol, lactose, suc
  • the composition further comprises an adjuvant.
  • adjuvants are as described herein.
  • Common adjuvants include aluminum hydroxide, aluminum potassium sulfate, other mineral salts, oil emulsions, particulate adjuvants, and microbial derivatives.
  • additional adjuvants include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS-21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E.
  • coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl-muramyl-L-threonyl-D- isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-( - 2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80
  • composition comprising:
  • Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising exposing the subject to an immunogenic composition as described herein.
  • Certain embodiments of the present disclosure provide a combination product comprising the following components:
  • chemokine receptor CCX-CKR inhibitor a chemokine receptor CCX-CKR inhibitor
  • components are provided in a form for separate or coadministration to induce an immune response to the selected antigen in a subject.
  • Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to the selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • a selected antigen as described herein and a chemokine receptor CCX-CKR inhibitor as described herein may used in a vaccine.
  • the selected antigen is an antigen that is antigenically related to another antigen.
  • Certain embodiments of the present disclosure provide a vaccine comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of a vaccine.
  • the vaccine comprises a nucleic acid vaccine, such as a DNA vaccine.
  • Nucleic acid vaccines and methods for constructing and administering nucleic acid vaccines are known. Nucleic acid vaccines are as described, for example, in “DNA vaccines: methods and protocols" 2003 by W. Mark Saltzman, Hong Shen, Janet L. Brandsma, Humana Press inc. Methods for cloning are described in, for example, Sambrook J. et al. Molecular Cloning: A Laboratory Manual (3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (200), herein incorporated by reference.
  • nucleic acid vaccine comprising a nucleic acid encoding an antigen and a nucleic acid encoding a chemokine receptor CCX-CKR inhibitor.
  • nucleic acid vaccine comprising a nucleic acid encoding an antigen and a chemokine receptor CCX-CKR inhibitor.
  • Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to a nucleic acid vaccine as described herein.
  • the method comprises exposing the subject to one or more adjuvants.
  • Adjuvants are as described herein and include aluminum hydroxide, aluminum potassium sulfate, other mineral salts, oil emulsions, particulate adjuvants, and microbial derivatives.
  • additional adjuvants include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS- 21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E.
  • coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl- muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D- isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D- isoglutaminyl-L-alanine-2-( -2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)- ethylamine (CGP 19835 A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80
  • Certain embodiments of the present disclosure provide a combination product comprising the following components:
  • chemokine receptor CCX-CKR inhibitor a chemokine receptor CCX-CKR inhibitor
  • the selected antigen is present in a vaccine composition, an immunogenic composition, a therapeutic formulation, or a medicament.
  • the selected antigen is prepared into a composition.
  • the selected antigen is mixed with one or more excipients which are pharmaceutically acceptable and compatible with the selected antigen to form a composition.
  • excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • a composition may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, and/or pH buffering agents.
  • the selected antigen is mixed with one or more adjuvants.
  • additional adjuvants which may be effective include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS-21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E.
  • coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N- acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L- alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L- alanyl-D-isoglutaminyl-L-alanine-2-(l'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835 A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80
  • a chemokine receptor CCX-CKR inhibitor may itself be used as an adjuvant, based on the fact that inhibition of the expression of the atypical chemokine receptor CCX-CKR enhances the immune response to an antigen.
  • Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor as an adjuvant.
  • the methods described herein are used to produce an antibody to the selected antigen in an animal or a human subject.
  • Antibodies are as described herein.
  • the methods described herein are used to produce a CTL response.
  • Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in a subject, the method comprising exposing the 1 subject to the selected antigen, wherein the subject comprises a reduced chemokine receptor CCX-CKR activity.
  • the subject is an animal subject.
  • Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in an animal subject, the method comprising exposing the animal subject to the selected antigen, wherein the animal subject comprises a reduced chemokine receptor CCX-CKR activity.
  • Animal subjects are as described herein.
  • the animal subject comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene.
  • Certain embodiments of the present disclosure provide the use of an animal subject with reduced chemokine receptor CCX-CKR activity to produce an antibody.
  • CCX-CKR " " mice may be used to produce monoclonal antibodies to a selected antigen.
  • Certain embodiments of the present disclosure provide a method of producing a hybridoma cell.
  • Certain embodiments of the present disclosure provide a method of producing a hybridoma cell, the method comprising fusing a B cell with reduced chemokine receptor CCX-CKR activity with an immortalised cell. Methods for producing hybridoma cells are known.
  • the B cell comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene. In certain embodiments, the B cell comprises a homozygous knock out in a chemokine receptor gene. Methods for creating gene knock out or gene knock down in cells are known.
  • Certain embodiments of the present disclosure provide a hybridoma produced by a method as described herein.
  • a typical protocol is as follows: Animals (e.g. mice) are first exposed to the selected antigen. Usually this is done by a series of injections of the antigen, over the course of several weeks. Once splenocytes are isolated from the mammal's spleen, the B cells may be fused with immortalised myeloma cells. The myeloma cells are generally selected to ensure they are not secreting antibody themselves and that they lack the hypoxanthine-guanine phosphoribosyltransferase (HGPRT) gene, making them sensitive to HAT medium. The fusion may be accomplished, for example, using polyethylene glycol or Sendai virus.
  • HGPRT hypoxanthine-guanine phosphoribosyltransferase
  • Fused cells are incubated in HAT medium for roughly 10 to 14 days. Aminopterin blocks the pathway that allows for nucleotide synthesis and unfused myeloma cells die, as they cannot produce nucleotides by the de novo or salvage pathways because they lack HGPRT. Removal of the unfused myeloma cells is necessary because they have the potential to outgrow other cells, especially weakly established hybridomas. Unfused B cells die as they have a short life span. In this way, only the B cell-myeloma hybrids survive, since the HGPRT gene coming from the B cells is functional. These cells produce antibodies and are immortal. The incubated medium is then diluted into multi-well plates to such an extent that each well contains only one cell. Since the antibodies in a well are produced by the same B cell, they will be directed towards the same epitope, and are thus monoclonal antibodies.
  • the next stage is a rapid primary screening process, which identifies and selects only those hybridomas that produce antibodies of appropriate specificity.
  • the hybridoma culture supernatant, secondary enzyme labeled conjugate, and chromogenic or fluorescent substrate, are then incubated, and the formation of a colored product indicates a positive hybridoma.
  • immunocytochemical screening or flow cytometry can also be used.
  • the B cell that produces the desired antibodies can be cloned to produce many identical daughter clones. Supplemental media containing interleukin-6 are essential for this step. Once a hybridoma colony is established, it will continually grow in culture medium like RPMI-1640 (with antibiotics and fetal bovine serum) and produce antibodies.
  • Multiwell plates are used initially to grow the hybridomas, and after selection, are changed to larger tissue culture flasks. This maintains the well-being of the hybridomas and provides enough cells for cryopreservation and supernatant for subsequent investigations.
  • the culture supernatant can yield 1 to 60 ⁇ g/ml of monoclonal antibody, which is maintained at -20 °C or lower until required.
  • Certain embodiments of the present disclosure provide a modified hybridoma.
  • Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising reduced chemokine receptor CCX-CKR activity. Methods for reducing chemokine receptor activity are as described herein.
  • Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising a knock out or a knock down of a chemokine receptor CCX-CKR gene.
  • Certain embodiments of the present disclosure provide a hybridoma comprising reduced chemokine receptor CCX-CKR activity. Methods for reducing chemokine receptor CCX-CKR activity are described herein.
  • Certain embodiments of the present disclosure provide an antibody produced from a hybridoma cell as described herein. Methods for producing antibodies from hybridoma cells are known.
  • Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody from a hybridoma cell as described herein.
  • Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody, the method comprising producing an antibody from a hybridoma comprising a B-cell and an immortalised cell, wherein the B cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity.
  • Standard techniques may be used for recombinant DNA technology, oligonucleotide synthesis, antibody production, peptide synthesis, tissue culture and transfection. Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein.
  • mice were immunised with ovalbumin in Alum-OH gel and peripheral blood was taken at the indicate time post-immunisation. Immunoglobulin-specific ELISAs were conducted to assess the effect on anti-OVA antibody production. Knock-out mice were produced as described in Comerford I.et al (2010) Blood 116(20): 4130-40.
  • OVA immunisation and ELISA 8-10 week old female CCX-CKR + + and CCX- CKR " " mice were immunised with 100 ⁇ g ovalbumin (OVA) in aluminum hydroxide gel (100 ⁇ / ⁇ ⁇ ) intraperitoneally. Serum was harvested on days 0 (pre-bleed prior to immunisation), 14 and 21 post-immunisation. Direct ELISAs were used to quantify serum titers of anti-OVA antibodies. Briefly, 96-well plates were coated with 10 ⁇ g/mL OVA protein in 0.05M carbonate buffer (Na 2 C0 3 + NaHC0 3 ; pH9.6) overnight and subsequently blocked with 2% skim milk/PBS.
  • mice were immunised with P-KLH in Alum and peripheral blood was taken at the indicate time post-immunisation. Immunoglobulin- specific ELISAs were conducted to assess the effect on high ( P (5) ) and low (NP (16) ) affinity and anti- P antibody production.
  • NP-KLH immunisation and ELISA 8-10 week old female CCX-CKR + + and CCX-CKR " mice were immunised with 100 ⁇ g NP24-KLH precipitated in alum (100 uL/mouse) intraperitoneally. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 14 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of high-affinity and global-affinity anti-NP antibodies.
  • mice were immunised with Semliki Forrest Virus ( ⁇ - SFV via the intravenous route) and peripheral blood was collected on the indicated days post-immunisation.
  • Anti-SFV-specific ELISAs were conducted to quantify anti-SFV antibody production (top and middle panels) and the ability of the antibodies to neutralise SFV by preventing its ability to infect Vero cells was determined (bottom panel).
  • mice 8-10 week old female CCX-CKR + + and CCX-CKR " " mice were immunised with two doses known to not induce protective antibody responses in wild-type B6 mice: 2.5 x 10 8 and 2.5 x 10 7 PFU-equivalent of gamma (y)-irradiated (50KGy) semliki forest virus (SFV) (y-SFV) intravenously. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 15 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of anti-SFV antibodies.
  • Wild-type or CCX-CKR " " mice were immunised with Semliki Forrest and peripheral blood was collected on the indicated days post-immunisation.
  • Anti-SFV- specific ELISAs were conducted to quantify anti-SFV antibody production (top panel) and the ability of the antibodies to neutralise SFV by preventing its ability to infect cells was determined (bottom panel).
  • mice 8-10 week old female CCX-CKR + + and CCX-CKR " ' " mice were immunised with 2.5 x 10 9 PFU-equivalent of gamma-irradiated ( ⁇ ) (50KGy) semliki forest virus (SFV) ( ⁇ -SFV) intravenously. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 15 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of anti-SFV antibodies. Briefly, 96-well plates were coated with boric acid- inactivated SFV in 0.05M carbonate buffer (Na 2 C0 3 + NaHC0 3 ; pH9.6) overnight and subsequently blocked with 3% BSA/PBS.
  • 0.05M carbonate buffer Na 2 C0 3 + NaHC0 3 ; pH9.6
  • Hybridoma cells from mice deficient in CCX-CKR activity may be produced using the protocol set out below.
  • Gerbu Adjuvant MM #3001A, 6x1 ml, Accurate Chemical & Scientific Corp.
  • mice Female mice, 8-10 weeks of age
  • Dispense PEG along sides of tube Does not need to be lml/min slow, just slow enough to keep from blasting cells. Do not touch cell/PEG slurry with pipette. After dispensing PEG, mix by swirling tube.
  • An immunogenic composition may be produced as follows:
  • the selected antigen may be adsorbed on 0.5 mg aluminium as aluminium hydroxide.
  • the composition may be administered by intramuscular administration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present disclosure relates to methods and compositions for inducing an immune response to a selected antigen in a subject. Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising inhibiting activity of a chemokine receptor CCX-CKR in the subject and exposing the subject to the selected antigen.

Description

METHODS AND COMPOSITIONS FOR INDUCING AN IMMUNE RESPONSE PRIORITY CLAIM
[001] This application claims priority to Australian provisional patent application number 2013901227 filed on 10 April 2013, the contents of which are hereby incorporated by reference.
FIELD
[002] The present disclosure relates to methods and compositions for inducing an immune response.
BACKGROUND
[003] Vaccination is the administration of antigenic material to stimulate the immune system to develop adaptive immunity, typically to a pathogen. Vaccination represents one of the most effective means of treating infectious diseases. However, in spite of the success of some vaccines, such as vaccines against smallpox and polio, there remains a need to improve vaccination.
[004] For example, for many infectious diseases it has not been possible to develop an effective vaccine, as an effective immune response to the pathogen cannot be induced.
[005] It would also be advantageous to improve the efficacy of many vaccines. Having the ability to enhance the efficacy of existing vaccines would be highly desirable as it would make a vaccine more effective in terms of stimulating protective immunity, allow a vaccine to be delivered at lower doses and extend the availability of the vaccine.
[006] To boost the immune response, vaccines may also contain one or more adjuvants. However, adjuvants have a number of disadvantages, including for example one or more of variable efficacy, restricted adjuvanticity and adverse side-effects. [007] Accordingly, there is a need to provide improved methods and compositions for inducing an immune response.
SUMMARY
[008] The present disclosure relates to methods and compositions for inducing an immune response.
[009] Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising:
inhibiting activity of a chemokine receptor CCX-CKR in the subject; and exposing the subject to the selected antigen.
[0010] Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising administering to the subject the selected antigen and a chemokine receptor CCX-CKR inhibitor.
[0011] Certain embodiments of the present disclosure provide an immunogenic composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
[0012] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of an immunogenic composition.
[0013] Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to the selected antigen and a chemokine receptor CCX-CKR inhibitor.
[0014] Certain embodiments of the present disclosure provide a vaccine comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
[0015] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of a vaccine. [0016] Certain embodiments of the present disclosure provide a nucleic acid vaccine comprising (i) a nucleic acid encoding an antigen and a nucleic acid encoding a chemokine receptor CCX-CKR inhibitor; and/or a nucleic acid encoding an antigen and a chemokine receptor CCX-CKR inhibitor.
[0017] Certain embodiments of the present disclosure provide a method of improving an immune response to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[0018] Certain embodiments of the present disclosure provide a method of reducing the amount of a selected antigen exposed to a subject to obtain a desired immunological response, the method comprising exposing the subject to a chemokine receptor CCX- CKR inhibitor.
[0019] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor as an adjuvant.
[0020] Certain embodiments of the present disclosure provide a composition comprising:
(i) a chemokine receptor CCX-CKR inhibitor;
(ii) one or more selected antigens;
(iii) an excipient; and
optionally one or more adjuvants.
[0021] Certain embodiments of the present disclosure provide a combination product comprising the following components:
a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to induce an immune response to the selected antigen in a subject. [0022] Certain embodiments of the present disclosure provide a combination product comprising the following components:
a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to vaccinate a subject.
[0023] Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in an animal subject, the method comprising exposing the animal subject to the selected antigen, wherein the animal subject comprises a reduced chemokine receptor CCX-CKR activity.
[0024] Certain embodiments of the present disclosure provide a method of producing a hybridoma cell, the method comprising fusing a B cell with reduced chemokine receptor CCX-CKR activity with an immortalised cell.
[0025] Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody, the method comprising producing an antibody from a hybridoma comprising a B-cell and an immortalised cell, wherein the B cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity.
[0026] Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising reduced chemokine receptor CCX-CKR activity.
[0027] Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising a knock out and/or a knock down of a chemokine receptor CCX-CKR gene.
[0028] Certain embodiments of the present disclosure provide a method of improving protective immunity against an infectious agent in a subject, the method comprising exposing the subject to a selected antigen associated with the infectious agent and a chemokine receptor CCX-CKR inhibitor. [0029] Certain embodiments of the present disclosure provide a method of improving efficacy of a vaccine comprising a selected antigen, the method comprising exposing a subject to be vaccinated against the selected antigen to a chemokine receptor CCX-CKR inhibitor.
[0030] Certain embodiments of the present disclosure provide a method of improving high affinity antibody production to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[0031] Other embodiments are disclosed herein.
BRIEF DESCRIPTION OF THE FIGURES
[0032] Certain embodiments are illustrated by the following figures. It is to be understood that the following description is for the purpose of describing particular embodiments only and is not intended to be limiting with respect to the description.
[0033] Figure 1 shows wild-type or CCX-CKR" " mice were immunised with ovalbumin in Alum-OH gel and peripheral blood was taken at the indicate time post- immunisation. Immunoglobulin- specific ELISAs were conducted to assess the effect on anti-OCA antibody production. Data represent mean +/- sem, n=4 from one experiment that was repeated with similar results.
[0034] Figure 2 shows wild-type or CCX-CKR" " mice were immunised with NP-KLH in Alum and peripheral blood was taken at the indicate time post-immunisation. Immunoglobulin-specific ELISAs were conducted to assess the effect onhigh (NP(5)) and low (NP(16)) affinity and anti-NP antibody production. Data represent mean +/- sem, n=4.
[0035] Figure 3 shows wild-type or CCX-CKR" " mice were immunised with Semliki Forrest Virus (γ-SFV via the intravenous route) and peripheral blood was collected on the indicated days post-immunisation. Anti-SFV-specific ELISAs were conducted to quantify anti-SFV antibody production (top and middle panels) and the ability of the antibodies to neutralise SFV by preventing its ability to infect Vero cells was determined (bottom panel). Data represent mean +/- sem, n=4 per dose. [0036] Figure 4 shows wild-type or CCX-CKR " mice were immunised with Semliki Forrest and peripheral blood was collected on the indicated days post-immunisation. Anti-SFV-specific ELISAs were conducted to quantify anti-SFV antibody production (top panel) and the ability of the antibodies to neutralise SFV by preventing its ability to infect cells was determined (bottom panel). Data represent mean +/- sem, n=X.
DETAILED DESCRIPTION
[0037] The present disclosure relates to methods and compositions for inducing an immune response.
[0038] Certain embodiments of the present disclosure have one or more combinations of advantages. For example, some of the advantages of certain embodiments disclosed herein include one or more of the following: enhancing the efficacy of existing vaccines; allowing some vaccines to be delivered at lower doses; extending the availability of some vaccines; improving the induction of a humoral immune response; improving the induction of an adaptive immune response; improving the induction of a cytotoxic T lymphocyte (CTL) mediated immune response; improving the generation of high affinity antigen-specific antibodies; improving the generation of antibodies that are protective; improving the generation of neutralising antibodies; providing animal systems that are useful in producing high quality, high affinity monoclonal antibodies; and reducing the cost of vaccines. Other advantages of certain embodiments of the present disclosure are also disclosed herein.
[0039] The present disclosure is based, at least in part, on the recognition that inhibition of the expression of the atypical chemokine receptor CCX-CKR enhances high affinity antibody production upon immunisation which are protective in a model virus infection.
[0040] Certain embodiments of the present disclosure provide a method of inducing an immune response in a subject.
[0041] Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising: inhibiting activity of a chemokine receptor CCX-CKR in the subject; and exposing the subject to the selected antigen.
[0042] In certain embodiments, the method is used for one or more of the following: to improve an immune response to the selected antigen in the subject; to vaccinate the subject against the selected antigen; to improve protective immunity in the subject against an infectious agent comprising the selected antigen or a related antigen; to improve efficacy of a vaccine comprising the selected antigen; to improve high affinity antibody to the selected antigen production in the subject; and to reduce the amount of the selected antigen exposed to the subject to obtain a desired immunological response.
[0043] In certain embodiments, the immune response comprises a humoral immune response. In certain embodiments, the humoral immune response comprises a primary immune response. In certain embodiments, the humoral immune response comprises a primary immune response and a secondary immune response.
[0044] In certain embodiments, the immune response comprises an adaptive immune response. In certain embodiments, the immune response comprises a CTL mediated immune response.
[0045] In certain embodiments, the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule. Other types of antigens are contemplated.
[0046] In certain embodiments, the selected antigen is derived from an infectious agent. Examples of infectious agents include bacteria, viruses, fungi, pathogens, microorganisms, prions, protozoa, and metazoa. In certain embodiments, the selected antigen is associated with an infectious agent. In certain embodiments, the selected antigen is antigenically related to an antigen from an infectious agent.
[0047] In certain embodiments, the selected antigen comprises an exogenous antigen. In certain embodiments, the selected antigen comprises all or part of a coat, a capsule, a cell wall, a flagellum, a fimbrae, and a toxin of a bacterium, a virus, and/or other pathogen or microorganism.
[0048] In certain embodiments, the selected antigen comprises an endogenous antigen, an autoantigen, and/or a tumor antigen.
[0049] In certain embodiments, the selected antigen is an antigen that is antigenically related to another antigen.
[0050] In certain embodiments, the selected antigen comprises a single antigen. In certain embodiments, the selected antigen comprises two or more antigens.
[0051] In certain embodiments, the immune response comprises induction of one or more of IgM, IgG, IgA and IgE antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of IgM antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of one or more of IgG, IgA and IgE antibodies to the selected antigen. In certain embodiments, the immune response comprises induction of IgM and/or IgG antibodies to the selected antigen.
[0052] In certain embodiments, the antibodies comprise an affinity of at least 106M_1, at least 107M_1, at least 108M_1, at least 109M_1, at least 1010Μ_1, at least 10ηΜ_1, or at least 1012M_1 to the selected antigen.
[0053] The term "antibody" is to be understood to mean an immunoglobulin molecule with the ability to bind an antigenic region of another molecule, and includes monoclonal antibodies, polyclonal antibodies, multivalent antibodies, chimeric antibodies, multispecific antibodies, diabodies and fragments of an immunoglobulin molecule or combinations thereof that have the ability to bind to the antigenic region of another molecule with the desired affinity including a Fab, Fab', F(ab')2, Fv, a single- chain antibody (scFv) or a polypeptide that contains at least a portion of an immunoglobulin (or a variant of an immunoglobulin) that is sufficient to confer specific antigen binding, such as a molecule including one or more CDRs. [0054] In this regard, an immunoglobulin is a tetrameric molecule, each tetramer being composed of two identical pairs of polypeptide chains, each pair having one light chain and one heavy chain. The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids that is primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as K and λ light chains. Heavy chains are classified as μ, Δ, γ, , or ε and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 10 more amino acids. The variable regions of each light/heavy chain pair form the antibody binding site, with the result that an intact immunoglobulin has two binding sites. The variable regions further include hypervariable regions that are directly involved in formation of the antigen binding site. These hypervariable regions are usually referred to as Complementarity Determining Regions (CDR). The intervening segments are referred to as Framework Regions (FR). In both light and heavy chains there are three CDRs (CDR-I to CDR-3) and four FRs (FR-I to FR-4).
[0055] In certain embodiments, the subject is human subject. In certain embodiments, the subject is a mammalian subject, a primate, a livestock animal (such as a horse, a cow, a sheep, a goat, a pig), a domestic animal (such as a dog or a cat) and other types of animals such as monkeys, rabbits, mice, rats and laboratory animals. In certain embodiments, the subject is an animal used for raising antibodies. In certain embodiments, the subject is an animal used for producing a hybridoma cell. Use of any of the aforementioned animals as animal models is also contemplated. Veterinary applications are contemplated.
[0056] In certain embodiments, the subject is a subject to be vaccinated. In certain embodiments, the subject is immunologically naive to a selected antigen.
[0057] In certain embodiments, the immune response comprises the induction of high affinity antibodies to the selected antigen. In certain embodiments, the high affinity antibodies comprise an affinity of at least 109Μ_1, ΙΟ^Μ"1, lO'^'or 1012Μ_1 to the selected antigen. [0058] The CCX-CKR gene (also referred to as CCRL1 or CCR11) encodes a receptor of the G protein-coupled receptor family, and is a receptor for C-C type chemokines. In humans, the receptor is encoded by the gene with the Genbank accession number AF 110640. The equivalent receptor in other species may be determined by a known method. The amino acid sequence of the human receptor has the Genbank accession number AF110640 1. Methods for identifying a CCX-CKR gene and/or receptor include for example nucleic acid and protein alignment programs, such as BLAST. The UniProt accession number for the corresponding mouse gene is Q924I3.
[0059] In certain embodiments, a chemokine receptor CCX-CKR is a human CCX- CKR receptor. In certain embodiments, the chemokine receptor CCX-CKR is a mammalian receptor. In certain embodiments, the CCX-CKR is an animal receptor. Examples of mammalian and animal receptors are as described herein.
[0060] In certain embodiments, a chemokine receptor CCX-CKR is a mouse CCX- CKR receptor. Chemokine receptors CCX-CKR in other species are contemplated, including for example a horse, a cow, a sheep, a goat, a pig, a dog, a rat, a cat, a primate, a monkey, and a rabbit.
[0061] In certain embodiments, the CCX-CKR receptor is a homolog, paralog or ortholog of a receptor. In certain embodiments, the CCX-CKR receptor is a variant and/or a fragment of the receptor, such as a variant of the receptor arising from an alternatively spliced transcript.
[0062] The term "inhibiting" as used herein refers to a treatment, exposure or intervention that results directly and/or indirectly in a reduction of activity or function, including for example a decrease in activity, an inhibitory alteration in the timing and/or location of activity, or otherwise provide inhibitory control over activity.
[0063] The term "inhibitor" as used herein refers to an agent, treatment, or intervention that results directly and/or indirectly in a reduction of activity and/or function. [0064] The term "activity" as used herein refers to the function of a species (eg a molecule) and includes, for example, the level, the specificity, the ability to interact (directly and/or indirectly) with and/or modify other species, the ability to signal, and the ability to cause changes (directly and/or indirectly) in other cellular and/or non- cellular events. Examples of modulating the activity of a species include, for example, changes in the level of the species, changes in the localisation of the species, changes in the synthesis and/or degradation rates of the species, changes in the timing of activity, changes in the ability to interact with other species (such as a change in the ability of a ligand and a receptor to interact), changes in the chemical composition of the species, changes in signalling, and changes in cellular and/or non-cellular events affected by the species.
[0065] In relation to inhibiting activity of a CCX-CKR receptor, examples of inhibition include (i) directly and/or indirectly altering the activity of a receptor, such as altering the level of expression of the receptor, altering localisation of the receptor, partially or completely removing the gene for the receptor, altering internationalisation of the receptor, altering timing of receptor function, altering synthesis and/or degradation of the receptor, altering binding of a receptor to a ligand; (ii) a gene knockout and/or a gene knock down; (iii) altering the activity of a signalling pathway associated with an receptor; (iv) altering the level and/or altering the activity of a ligand that binds to the receptor, such as altering the synthesis of the ligand. Other forms of inhibition are contemplated.
[0066] In certain embodiments, the inhibiting of activity of a chemokine CCX-CKR receptor comprises a gene knock out and/or a gene knock down. Methods for producing a gene knockout or a gene knock down are known. For example, gene knock outs are as described in "Gene Knockout Protocols" (2001), edited by Martin J. Tymms, Ismail Kola, Human Press Inc. Gene knock downs are as described in "Regulation of Gene Expression by Small RNAs" (2009) edited by Rajesh K. Gaur, John J. Rossi, CRC Press.
[0067] In certain embodiment, the gene knock out is a homozygous gene knock out. In certain embodiments, the gene knock out is a heterozygous gene knock out. [0068] In certain embodiments, a gene knock out and/or the gene knock down comprises a knock out and/or a knock down of a chemokine receptor CCX-CKR gene. In certain embodiments, a gene knock out and/or the gene knock down comprises a knock out and/or a knock down of a gene that regulates the expression of a chemokine receptor CCX-CKR gene. The knock out and/or a knock down of other genes is contemplated.
[0069] In certain embodiments, the subject comprises a gene knock out and/or a gene knock down. In certain embodiments, the subject comprises a homozygous gene knock down. In certain embodiments, the subject comprises a heterozygous gene knock down. Examples include a mouse comprising a homozygous knock out in a chemokine receptor CCX-CKR gene.
[0070] In certain embodiments, the inhibiting of activity of a chemokine receptor CCX-CKR comprises exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[0071] In certain embodiments, the inhibitor comprises a selective inhibitor. In certain embodiments, the inhibitor comprises a non-selective inhibitor.
[0072] Examples of inhibitors include a drug, a small molecule, a protein, a polypeptide, a lipid, a carbohydrate, a nucleic acid, an oligonucleotide, a ribozyme, a biologic, an aptamer, a peptide, a non-peptide, a cofactor, a ligand, a ligand mimetic, a receptor, an enzyme, a kinase, a phosphatase, a cytokine, a growth factor, a metal ion, a chelate, an antisense nucleic acid, an inhibitor RNA, a microRNA, a siRNA, an antibody or antigen binding part thereof, an antibody mimetic, an amino acid, an antagonist, an inhibitor, or a suppressor.
[0073] An inhibitor of a chemokine receptor CCX-CKR may be synthesized or produced by a method known in the art, or obtained commercially.
[0074] In certain embodiments, the chemokine receptor inhibitor comprises one or more of an antagonist, a small interfering RNA, a microRNA, an antisense RNA, a ligand, a ligand mimetic, or an antibody and/or an antigen binding part thereof. [0075] In certain embodiments, the chemokine receptor inhibitor comprises one or more of an antagonist to a chemokine receptor CCX-CKR, a small interfering RNA for a chemokine receptor CCX-CKR, a microRNA for a chemokine receptor CCX-CKR, an antisense RNA for a chemokine receptor CCX-CKR, a ligand, or a ligand mimetic for a chemokine receptor CCX-CKR, or an antibody and/or an antigen binding part thereof for a chemokine receptor CCX-CKR.
[0076] In certain embodiments, the inhibitor is an inhibitor of ligand binding to a chemokine receptor CCX-CKR. Examples of such inhibitors include a ligand antagonist or an antibody antagonist. In certain embodiments, the inhibitor reduces binding of a ligand to a chemokine receptor CCX-CKR.
[0077] In certain embodiments, the inhibitor inhibits the binding of a ligand to a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor is an inhibitor of one or more of CCL25 binding, CCL19 binding and CCL21 binding to a chemokine receptor CCX-CKR.
[0078] In certain embodiments, the inhibitor inhibits the activity of a ligand for a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor is an inhibitor of one or more of CCL25 activity, CCL19 activity and CCL21 activity.
[0079] In certain embodiments, the inhibitor inhibits the synthesis of a ligand for a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor inhibits the synthesis of one or more of CCL25, CCL19 and CCL21.
[0080] In certain embodiments, the inhibitor inhibits the processing of a species that forms a ligand for a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor inhibits the processing of precursor of one or more of CCL25, CCL19 and CCL21.
[0081] In certain embodiments, the inhibitor is an inhibitor of expression of a chemokine receptor CCX-CKR expression. Examples of such inhibitors include an antisense RNA to a chemokine receptor CCX-CKR mRNA or a small interfering RNA to a chemokine receptor. [0082] In certain embodiments, the inhibitor modulates internalisation of a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor promotes internalisation of a chemokine receptor CCX-CKR. In certain embodiments, the inhibitor inhibits internalisation of a chemokine receptor CCX-CKR In certain embodiments, the inhibitor blocks internalisation of a chemokine receptor CCX-CKR
[0083] In certain embodiments, the inhibitor is an antisense nucleic acid, such as an antisense RNA. In certain embodiments, the inhibitor is a small interfering RNA. In certain embodiments, the inhibitor is a microRNA. Methods for producing and delivering therapeutic nucleic acids, such as antisense nucleic acids, microRNAs and siRNAs are known, for example "Therapeutic Oligonucleotides" (2008) edited by Jens Kurreck, RSC Publishing.
[0084] The term "nucleic acid" refers to an oligonucleotide or a polynucleotide and includes for example DNA, RNA and be may be single stranded, double stranded or triple stranded. The nucleic acid may be any type of nucleic acid, including for example a non-naturally occurring nucleic acid or a nucleic acid of genomic origin, cDNA origin (derived from a mRNA), a microRNA, derived from a virus, or of synthetic origin. The nucleic acid may also be an isolated nucleic acid or a purified nucleic acid. The term "isolated" refers to a nucleic acid that has been separated from its natural environment. For example, an isolated nucleic acid or may be in a substantially purified state, being substantially free of other substances with which it is associated in nature or in vivo.
[0085] In certain embodiments, the chemokine receptor CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof. In certain embodiments, the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
[0086] In certain embodiments, the inhibitor comprises an antibody and/or an antigen binding fragment thereof. In certain embodiments, the inhibitor comprises a neutralising antibody. In certain embodiments, the inhibitor comprises an antagonist antibody. In certain embodiments, the inhibitor comprises an antibody that promotes or inhibits internalisation of a chemokine receptor CCX-CKR. [0087] In certain embodiments, the antigen-binding fragment comprises a Fab, Fab', F(ab')2, Fd, Fv, a single-chain antibody (scFv), a chimeric antibody, a diabody or a polypeptide that contains at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding.
[0088] A Fab fragment is a monovalent fragment consisting of the VL, VH, CL and CH I domains. A F(ab')2 fragment is a bivalent fragment including two Fab fragments linked by a disulphide bridge at the hinge region. A Fd fragment consists of the VH and CH I domains. A Fv fragment consists of the VL and VH domains of a single arm of an antibody. A dAb consists of a VH domain. A single chain antibody (scFv) is an antibody in which VL and VH regions are paired to form a monovalent molecule via a synthetic linker that enable them to be made as a single protein chain. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites.
[0089] Antibody fragments that contain specific binding sites may be generated by a known method. Methods for producing antigen-binding fragments or portions of antibodies are known in the art, for example as described in "Antibody Engineering: Methods and Protocols" (2004) ed. by B.K.C. Lo Humana Press, herein incorporated by reference; and "Antibody Engineering: A Practical Approach" (1996) ed. by J. McCafferty, H.R. Hoogenboom and DJ. Chriswell Oxford University Press, herein incorporated by reference. For example, F(ab')2 fragments can be produced by pepsin digestion of the antibody molecule, and Fab fragments can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity, as described for example in Huse, W. D. et al. (1989) Science 254: 1275-1281, herein incorporated by reference.
[0090] Antibodies may be generated using known methods. For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others, may be immunized by injection with an appropriate antigen. Depending on the host species, various adjuvants may be used to increase an immunological response. Such standard adjuvants include Freund's adjuvant, mineral gels such as aluminium hydroxide, and surface-active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
[0091] In certain embodiments, the antibody is a polyclonal antibody. A polyclonal antibody is a mixture of antibodies having different antigen specificities. Methods for producing and isolating polyclonal antibodies are known. In general, polyclonal antibodies are produced from B-lymphocytes. Typically polyclonal antibodies are obtained directly from an immunized subject, such as an immunized animal.
[0092] In certain embodiments, the antibody is a monoclonal antibody. Monoclonal antibodies may be prepared using a technique that provides for the production of antibody molecules by continuous isolated cells in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. Methods for the preparation of monoclonal antibodies include for example Kohler et al. (1975) Nature 256:495-497, herein incorporated by reference; Kozbor et al. (1985) J. Immunol. Methods 81 :31-42, herein incorporated by reference; Cote et al. (1983) Proc. Natl. Acad. ScL 80:2026-2030, herein incorporated by reference; and Cole et al. (1984) MoT Cell Biol. 62: 109-120, herein incorporated by reference.
[0093] In certain embodiments, the antibody and/or an antigen binding fragment thereof comprises an isolated antibody. In certain embodiments, the antibody and/or an antigen binding fragment thereof comprises a purified antibody. Methods for producing and isolating polyclonal and monoclonal antibodies are known. The term "isolated" refers to an antibody that has been separated from its natural environment.
[0094] In certain embodiments, the antibody has an isotype selected from the group consisting of IgGl, IgG2a, IgG2b, IgG3, IgM and IgA.
[0095] In certain embodiments, the antibody and/or an antigen binding fragment thereof is a mouse antibody and/or an antigen binding fragment thereof, a human antibody and/or an antigen binding fragment thereof, or a humanized antibody and/or an antigen binding fragment thereof. [0096] Humanized antibodies, or antibodies adapted for non-rejection by other mammals, may be produced by a suitable method known in the art, including for example resurfacing or CDR grafting. In resurfacing technology, molecular modeling, statistical analysis and mutagenesis are combined to adjust the non-CDR surfaces of variable regions to resemble the surfaces of known antibodies of the target host. Strategies and methods for the resurfacing of antibodies, and other methods for reducing immunogenicity of antibodies within a different host are known, for example as described in US patent 5,639,641. Humanized forms of the antibodies may also be made by CDR grafting, by substituting the complementarity determining regions of, for example, a mouse antibody, into a human framework domain.
[0097] Methods for humanizing antibodies are known. For example, the antibody may be generated as described in U.S. Pat. No. 6, 180,370, herein incorporated by reference; WO 92/22653, herein incorporated by reference; Wright et al. (1992) Critical Rev. in Immunol. 12(3,4): 125-168, herein incorporated by reference; and Gu et al. (1997) Thrombosis and Hematocyst 77(4):755-759), herein incorporated by reference.
[0098] Humanized antibodies typically have constant regions and variable regions other than the complementarity determining regions (CDRs) derived substantially or exclusively from a human antibody and CDRs derived substantially or exclusively from the non- human antibody of interest.
[0099] Techniques developed for the production of "chimeric antibodies", for example the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, may be performed by a suitable method. For example, chimeric antibodies may be produced as described in Morrison, S. L. et al. (1984) Proc. Natl. Acad. Sci 81 :6851-6855; Neuberger, M. S. et al. (1984) Nature 312:604-608,; and Takeda, S. et al. (1985) Nature 314:452-454,.
[00100] Immunoassays may be used for screening to identify antibodies and/or antigen binding fragments thereof having the desired specificity.
[00101] Antibody molecules and antigen binding fragments thereof may also be produced recombinantly by methods known in the art, for example by expression in E.coli expression systems. For example, a method for the production of recombinant antibodies is as described in US patent 4,816,567, herein incorporated by reference. Antigen binding fragments may also be produced by phage display technologies or using peptide libraries, which are known.
[00102] In certain embodiments, the antibody and/or an antigen binding fragment thereof binds to a human a chemokine receptor CCX-CKR. In certain embodiments, the antibody and/or an antigen binding fragment thereof binds to a mammalian chemokine receptor CCX-CKR. In certain embodiments, the antibody and/or an antigen binding fragment thereof is a neutralising antibody to a chemokine receptor CCX-CKR.
[00103] In certain embodiments, the inhibiting of activity of a chemokine receptor CCX-CKR comprises exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[00104] The term "exposing", and related terms such as "expose" and "exposure", refers to directly and/or indirectly contacting and/or treating a subject with an agent.
[00105] For example, an inhibitor of a chemokine receptor may be administered to a subject to expose the subject to the inhibitor, or an agent may be administered to a subject that results in the production of an inhibitor in the subject, thereby exposing the subject to the inhibitor.
[00106] In another example, one or more cells may be removed from a subject and contacted directly or indirectly with an inhibitor, and the cells then introduced back into the same or another subject to effect exposure to an inhibitor.
[00107] In another example, a subject may be exposed to a nucleic acid expressing an inhibitor, for example by way of a nucleic acid vector encoding an inhibitor. Methods for the construction of vectors expressing an inhibitor are known and include for example in Sambrook, J, Fritsch, E.F. and Maniatis, T. Molecular Cloning: A Laboratory Manual 2nd. ed. Cold Spring Harbor Laboratory Press, New York. (1989). Methods for the delivery of nucleic acids to a subject are known and include for example [00108] Typically, the subject will be exposed to an effective amount of an agent. The term "effective amount" as used herein refers to that amount of an agent that is sufficient to illicit the desired response or outcome. The effective amount will vary depending upon a number of factors, including for example the specific activity of the agent being used and the characteristics of the subject.
[00109] In certain embodiments, a subject is exposed to an inhibitor of a chemokine receptor CCX-CKR in an amount ranging from one of the following selected ranges: 1 μg/kg to 100 mg/kg; 1 μg/kg to 10 mg/kg; 1 μg/kg to 1 mg/kg; 1 μg/kg to 100 μg/kg; 1 μg/kg to 10μg/kg; 10 μg/kg to 100 mg/kg; 10 μg/kg to 10 mg/kg; 10 μg/kg to 1 mg/kg; 10 μg/kg to 100 μg/kg; 100 μg/kg to 100 mg/kg; 100 μg/kg to 10 mg/kg; 100 μg/kg to 1 mg/kg; 1 mg/kg to 10 mg/kg; and 10 mg/kg to 100 mg/kg body weight. Other amounts are contemplated.
[00110] In certain embodiments, the exposing the subject to a chemokine receptor comprises administering a chemokine receptor CCX-CKR inhibitor to the subject.
[00111] Examples of administration routes are as described herein.
[00112] In certain embodiments, the inhibitor is administered to the subject to produce a concentration of an inhibitor of a chemokine receptor CCX-CKR of 0.1 nM or greater, 0.5 nM or greater, 1 nM or greater, 5 nM or greater, 10 nM or greater, 50 nM or greater, 100 nM or greater, 500 nM or greater, 1 uM or greater, 5 uM or greater, 10 uM or greater , 100 uM or greater, 500 uM or greater, 1 mM or greater, or 10 mM or greater.
[00113] In certain embodiments, an antibody and/or an antigen binding fragment thereof is administered to the subject to produce a concentration in one of the aforementioned ranges. Methods for administering antibodies are known and include for example Therapeutic Antibodies (2008) Edited By Yuti Chernajovsky, Ahuva Nissim. Springer Publishing.
[00114] In certain embodiments, the inhibitor of a chemokine receptor CCX-CKR is administered to the subject in an amount ranging from one of the following selected ranges: 1 μg/kg to 100 mg/kg; 1 μg/kg to 10 mg/kg; 1 μg/kg to 1 mg/kg; 1 μg/kg to 100 μ§/1¾; 1 μ§/1¾ to 10μ§/1¾; 10 μ§/1¾ to 100 mg/kg; 10 μg/kg to 10 mg/kg; 10 μg/kg to 1 mg/kg; 10 μg/kg to 100 μg/kg; 100 μg/kg to 100 mg/kg; 100 μg/kg to 10 mg/kg; 100 μg/kg to 1 mg/kg; 1 mg/kg to 10 mg/kg; and 10 mg/kg to 100 mg/kg body weight. In certain embodiments, an antibody and/or an antigen binding fragment thereof may be administered to the subject to produce in one of the aforementioned ranges. For example, a, antibody may be administered at a concentration of 0.5, 1, 2, 3, 4, 5, 10, 15 or 20 mg/kg body weight. Other amounts are contemplated. The dose and frequency of administration may be determined by one of skill in the art.
[00115] In certain embodiments, the inhibitor of a chemokine receptor CCX-CKR is administered to a subject to inhibit the activity of the receptor in the subject.
[00116] Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising administering to the subject the selected antigen and a chemokine receptor CCX-CKR inhibitor.
[00117] The inhibitor may be administered to the subject in a suitable form. In this regard, the term "administering" includes for example administering the inhibitor, or administering a prodrug of the inhibitor, or a derivative of the inhibitor that will form a therapeutically effective amount of the inhibitor within the body of the subject. The term include routes of administration that are systemic (e.g., via injection such as intravenous injection, orally in a tablet, pill, capsule, or other dosage form useful for systemic administration of pharmaceuticals), topical (e.g., creams, solutions, and the like, including solutions such as mouthwashes, for topical oral administration), and administration utilising nucleic acids expressing the inhibitor, such as viral vectors. Other routes of administration are contemplated.
[00118] The inhibitors may be administered alone or may be delivered in a mixture with other therapeutic agents and/or agents that enhance, stabilise or maintain the activity of the inhibitor. In certain embodiments, an inhibitor is administered in conjunction with an administration vehicle. In certain embodiments, an administration vehicle (e.g., pill, tablet, implant, injectable solution, etc.) may contain the inhibitor and additional agent(s). [00119] In certain embodiments, the inhibitor is administered orally. In certain embodiments, the inhibitor is administered via injection, such as intravenous injection. In certain embodiments, the inhibitor is administered parenterally. In certain embodiments, the inhibitor is administered by direct introduction to the lungs, such as by aerosol administration, by nebulized administration, and by being instilled into the lung. In certain embodiments, the inhibitor is administered by implant. In certain embodiments, the certain embodiments, the inhibitor is administered by subcutaneous injection, intraarticular ly, mucosally, rectally, intranasally, intraocularly, vaginally, or transdermally.
[00120] In certain embodiments, the subject is exposed to the selected antigen. Methods for exposing a subject to an antigen are known.
[00121] Typically, the amount of antigen exposed to the subject is in the range of 5 micrograms to 250 micrograms of antigen per dose. In certain embodiments, the amount of antigen comprises 5μg or more, 10μg or more, 20μg or more, 50μg or more, 100μg or more, 200μg or more, or 2 0μg or more. The dose depends on the subject to be treated, capacity of the subject's immune system to generate a protective immune response, and the degree of protection desired.
[00122] For example, exposing of the selected antigen to the subject may be by conventional routes such intravenous, subcutaneous, intradermally, intraperitoneal, or mucosal routes. The exposure may be by parenteral injection, for example, a subcutaneous or intramuscular injection. The exposure may also be by expressing a selected antigen from for example a virus or a nucleic acid vector, such as a nucleic acid vaccine.
[00123] In certain embodiments, the exposing of the selected antigen to the subject comprises administration of the selected antigen.
[00124] In certain embodiments, the inhibiting of activity of the chemokine receptor occurs at any one or more of before, concurrent with, or after exposure of the subject to the selected antigen. [00125] In certain embodiments, administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject prior to exposure of the subject to the selected antigen. In certain embodiments, administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject concurrent with exposure of the subject to the selected antigen. For example, a composition comprising an inhibitor of chemokine receptor CCX-CKR activity and a selected antigen may be administered to a subject. In certain embodiments, administration of an inhibitor of chemokine receptor CCX-CKR activity may be administered to a subject after exposure of the subject to the selected antigen.
[00126] In certain embodiments, the methods as described herein may be used to improve an immune response to a selected antigen. Methods for determining the improvement in an immune response are known.
[00127] Certain embodiments of the present disclosure provide a method of improving an immune response to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[00128] In certain embodiments, the methods as described herein may be used to reduce the amount of a selected antigen exposed to a subject to obtain a desired immunological response. For example, the amount of antigen needed to obtain a desired immunological response may be reduced by 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, or 90% or more.
[00129] Certain embodiments of the present disclosure provide a method of reducing the amount of a selected antigen exposed to a subject to obtain a desired immunological response, the method comprising exposing the subject to a chemokine receptor CCX- CKR inhibitor.
[00130] In certain embodiments, the methods as described herein may be used to improve protective immunity against an infectious agent in a subject.
[00131] Certain embodiments of the present disclosure provide a method of improving protective immunity against an infectious agent in a subject, the method comprising exposing the subject to a selected antigen associated with the infectious agent and a chemokine receptor CCX-CKR inhibitor.
[00132] Methods for determining the extent of protective immunity are known.
[00133] Examples of infectious agents are described herein and include bacteria, viruses, fungi, protozoa, metazoan, pathogens and/or microorganisms.
[00134] In certain embodiments, the methods as described herein may be used to improve efficacy of a vaccine. Methods for determining the efficacy of a vaccine are known and include for example determination of the incidence of a disease among subjects who have received a vaccine as compared to the incidence in unvaccinated subjects.
[00135] Certain embodiments of the present disclosure provide a method of improving efficacy of a vaccine comprising a selected antigen, the method comprising exposing a subject to be vaccinated against the selected antigen to a chemokine receptor CCX-CKR inhibitor.
[00136] In certain embodiments, the methods as described herein may be used to improve high affinity antibody production. High affinity antibodies are described herein.
[00137] Certain embodiments of the present disclosure provide a method of improving high affinity antibody production to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
[00138] In certain embodiments, the methods as described herein may be used to increase CTL activity.
[00139] Certain embodiments of the present disclosure provide a composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor. [00140] In certain embodiments, the composition is an immunogenic composition. In certain embodiments, the composition is a vaccine composition. In certain embodiments, the composition is a pharmaceutical composition.
[00141] Certain embodiments of the present disclosure provide an immunogenic composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
[00142] Immunogenic compositions may be produced by a known method. For example, compositions may be produced as described in "Development of Vaccines: From Discovery to Clinical Testing" (2011) edited by Manmohan Singh, Indresh K. Srivastava Wiley Publishing.
[00143] Examples of selected antigens are as described herein. In certain embodiments, the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
[00144] Examples of chemokine receptor CCX-CKR inhibitors are as described herein. In certain embodiments, the inhibitor comprises one or more of an antagonist (eg a peptide antagonist or a non-peptide antagonist), a small interfering RNA, a microRNA, an antisense RNA, a ligand for a chemokine receptor CCX-CKR, an antibody and/or an antigen binding part thereof.
[00145] In certain embodiments, the chemokine receptor CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof, as described herein. In certain embodiments, the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
[00146] In certain embodiments, the antibody comprises a polyclonal antibody, as described herein. In certain embodiments, the antibody comprises a monoclonal antibody, as described herein.
[00147] An example of an immunogenic composition is as follows:
One ml adult dose, as a sterile suspension in isotonic saline: Selected Antigen: 5 -250 μg
Neutralising CCX-CKR Antibody: 10-500 μg
The selected antigen may be adsorbed on 0.5 mg aluminium as aluminium hydroxide. The composition may be administered by intramuscular administration.
[00148] In certain embodiments, the composition further comprises one or more excipients. Excipients are known in the art and are as described, for example, in Grabenstein JD. ImmunoFacts: Vaccines and Immunologic Drugs - 2012 (37th revision). St Louis, MO: Wolters Kluwer Health, 2011. Examples of excipients include antibiotics (eg neomycin and/or polymyxin B; gentamicin); formaldehyde; stabilizers (eg monosodium glutamate, 2-phenoxyethanol, lactose, sucrose (both sugars), glycine, gelatin, preservatives (eg. thiomersal, phenoxyethanol, and formaldehyde); and diluents (eg sterile saline or sterile water).
[00149] In certain embodiments, the composition further comprises an adjuvant. Examples of adjuvants are as described herein.
[00150] Common adjuvants include aluminum hydroxide, aluminum potassium sulfate, other mineral salts, oil emulsions, particulate adjuvants, and microbial derivatives. Examples of additional adjuvants include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS-21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E. coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl-muramyl-L-threonyl-D- isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-( - 2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80 emulsion. [00151] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of an immunogenic composition. Immunogenic compositions are as described herein.
[00152] Certain embodiments of the present disclosure provide a composition comprising:
(i) a chemokine receptor CCX-CKR inhibitor;
(ii) one or more selected antigens; and
(iii) optionally, one or more excipients and one or more adjuvants.
[00153] Certain embodiments of the present disclosure provide a method of inducing an immune response to a selected antigen in a subject, the method comprising exposing the subject to an immunogenic composition as described herein.
[00154] Certain embodiments of the present disclosure provide a combination product comprising the following components:
a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to induce an immune response to the selected antigen in a subject.
[00155] Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to the selected antigen and a chemokine receptor CCX-CKR inhibitor.
[00156] Methods of vaccinating a subject against a selected antigen are known, and are as described herein. For example, vaccines and methods of vaccination are as described in "Vaccine Protocols" (2003) 2nd Edition, edited by Andrew P. Robinson, Michael J. Hudson, Martin P. Cranage, Humana Press.
[00157] In certain embodiments, a selected antigen as described herein and a chemokine receptor CCX-CKR inhibitor as described herein may used in a vaccine. [00158] In certain embodiments, the selected antigen is an antigen that is antigenically related to another antigen.
[00159] Certain embodiments of the present disclosure provide a vaccine comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
[00160] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor in the preparation of a vaccine.
[00161] In certain embodiments, the vaccine comprises a nucleic acid vaccine, such as a DNA vaccine. Nucleic acid vaccines, and methods for constructing and administering nucleic acid vaccines are known. Nucleic acid vaccines are as described, for example, in "DNA vaccines: methods and protocols" 2003 by W. Mark Saltzman, Hong Shen, Janet L. Brandsma, Humana Press inc. Methods for cloning are described in, for example, Sambrook J. et al. Molecular Cloning: A Laboratory Manual (3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (200), herein incorporated by reference.
[00162] Certain embodiments of the present disclosure provide a nucleic acid vaccine comprising a nucleic acid encoding an antigen and a nucleic acid encoding a chemokine receptor CCX-CKR inhibitor.
[00163] Certain embodiments of the present disclosure provide a nucleic acid vaccine comprising a nucleic acid encoding an antigen and a chemokine receptor CCX-CKR inhibitor.
[00164] Certain embodiments of the present disclosure provide a method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to a nucleic acid vaccine as described herein.
[00165] In certain embodiments, the method comprises exposing the subject to one or more adjuvants.
[00166] Adjuvants are as described herein and include aluminum hydroxide, aluminum potassium sulfate, other mineral salts, oil emulsions, particulate adjuvants, and microbial derivatives. Examples of additional adjuvants include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS- 21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E. coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N-acetyl- muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L-alanyl-D- isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L-alanyl-D- isoglutaminyl-L-alanine-2-( -2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)- ethylamine (CGP 19835 A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80 emulsion.
[00167] Certain embodiments of the present disclosure provide a combination product comprising the following components:
a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to vaccinate a subject.
[00168] In certain embodiments, the selected antigen is present in a vaccine composition, an immunogenic composition, a therapeutic formulation, or a medicament.
[00169] In certain embodiments, the selected antigen is prepared into a composition.
[00170] In certain embodiments, the selected antigen is mixed with one or more excipients which are pharmaceutically acceptable and compatible with the selected antigen to form a composition. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof. In addition, a composition may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, and/or pH buffering agents.
[00171] In certain embodiments, the selected antigen is mixed with one or more adjuvants. Examples of additional adjuvants which may be effective include for example complete Freunds adjuvant (CFA), Incomplete Freunds adjuvant (IV A), Saponin, a purified extract fraction of Saporin such as Quil A, a derivative of Saporin such as QS-21, lipid particles based on Saponin such as ISCOM/ISCOMATIX, E. coli heat labile toxin (LT) mutants such as LTK63 and/or LTK72, aluminium hydroxide, N- acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nor-muramyl-L- alanyl-D-isoglutamine (CGP 11637, referred to as nor-MDP), N-acetylmuramyl-L- alanyl-D-isoglutaminyl-L-alanine-2-(l'-2'-dipalmitoyl-sn-glycero-3-hydroxyphosphoryl oxy)-ethylamine (CGP 19835 A, referred to as MTP-PE), and RIBI, which contains three components extracted from bacteria, monophosphoryl lipid A, trehalose dimycolate and cell wall skeleton (MPL+TDM+CWS) in a 2% squalene/Tween 80 emulsion.
[00172] In certain embodiments, a chemokine receptor CCX-CKR inhibitor may itself be used as an adjuvant, based on the fact that inhibition of the expression of the atypical chemokine receptor CCX-CKR enhances the immune response to an antigen.
[00173] Certain embodiments of the present disclosure provide use of a chemokine receptor CCX-CKR inhibitor as an adjuvant.
[00174] In certain embodiments, the methods described herein are used to produce an antibody to the selected antigen in an animal or a human subject. Antibodies are as described herein.
[00175] In certain embodiments, the methods described herein are used to produce a CTL response.
[00176] Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in a subject, the method comprising exposing the 1 subject to the selected antigen, wherein the subject comprises a reduced chemokine receptor CCX-CKR activity.
[00177] In certain embodiments, the subject is an animal subject.
[00178] Certain embodiments of the present disclosure provide a method of producing an antibody to a selected antigen in an animal subject, the method comprising exposing the animal subject to the selected antigen, wherein the animal subject comprises a reduced chemokine receptor CCX-CKR activity.
[00179] Animal subjects are as described herein. In certain embodiments, the animal subject comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene.
[00180] Certain embodiments of the present disclosure provide the use of an animal subject with reduced chemokine receptor CCX-CKR activity to produce an antibody. For example, CCX-CKR" " mice may be used to produce monoclonal antibodies to a selected antigen.
[00181] Certain embodiments of the present disclosure provide a method of producing a hybridoma cell.
[00182] Certain embodiments of the present disclosure provide a method of producing a hybridoma cell, the method comprising fusing a B cell with reduced chemokine receptor CCX-CKR activity with an immortalised cell. Methods for producing hybridoma cells are known.
[00183] Methods for reducing a chemokine receptor CCX-CKR activity are described herein.
[00184] In certain embodiments, the B cell comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene. In certain embodiments, the B cell comprises a homozygous knock out in a chemokine receptor gene. Methods for creating gene knock out or gene knock down in cells are known.
[00185] Certain embodiments of the present disclosure provide a hybridoma produced by a method as described herein.
[00186] A typical protocol is as follows: Animals (e.g. mice) are first exposed to the selected antigen. Usually this is done by a series of injections of the antigen, over the course of several weeks. Once splenocytes are isolated from the mammal's spleen, the B cells may be fused with immortalised myeloma cells. The myeloma cells are generally selected to ensure they are not secreting antibody themselves and that they lack the hypoxanthine-guanine phosphoribosyltransferase (HGPRT) gene, making them sensitive to HAT medium. The fusion may be accomplished, for example, using polyethylene glycol or Sendai virus.
[00187] Fused cells are incubated in HAT medium for roughly 10 to 14 days. Aminopterin blocks the pathway that allows for nucleotide synthesis and unfused myeloma cells die, as they cannot produce nucleotides by the de novo or salvage pathways because they lack HGPRT. Removal of the unfused myeloma cells is necessary because they have the potential to outgrow other cells, especially weakly established hybridomas. Unfused B cells die as they have a short life span. In this way, only the B cell-myeloma hybrids survive, since the HGPRT gene coming from the B cells is functional. These cells produce antibodies and are immortal. The incubated medium is then diluted into multi-well plates to such an extent that each well contains only one cell. Since the antibodies in a well are produced by the same B cell, they will be directed towards the same epitope, and are thus monoclonal antibodies.
[00188] The next stage is a rapid primary screening process, which identifies and selects only those hybridomas that produce antibodies of appropriate specificity. The hybridoma culture supernatant, secondary enzyme labeled conjugate, and chromogenic or fluorescent substrate, are then incubated, and the formation of a colored product indicates a positive hybridoma. Alternatively, immunocytochemical screening or flow cytometry can also be used.
[00189] The B cell that produces the desired antibodies can be cloned to produce many identical daughter clones. Supplemental media containing interleukin-6 are essential for this step. Once a hybridoma colony is established, it will continually grow in culture medium like RPMI-1640 (with antibiotics and fetal bovine serum) and produce antibodies.
[00190] Multiwell plates are used initially to grow the hybridomas, and after selection, are changed to larger tissue culture flasks. This maintains the well-being of the hybridomas and provides enough cells for cryopreservation and supernatant for subsequent investigations. The culture supernatant can yield 1 to 60 μg/ml of monoclonal antibody, which is maintained at -20 °C or lower until required.
[00191] By using culture supernatant or a purified immunoglobulin preparation, further analysis of a potential monoclonal antibody producing hybridoma can be made in terms of reactivity, specificity, and cross-reactivity.
[00192] Certain embodiments of the present disclosure provide a modified hybridoma.
[00193] Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising reduced chemokine receptor CCX-CKR activity. Methods for reducing chemokine receptor activity are as described herein.
[00194] Certain embodiments of the present disclosure provide a hybridoma of a B-cell and an immortalised cell, the B-cell component of the hybridoma comprising a knock out or a knock down of a chemokine receptor CCX-CKR gene.
[00195] Certain embodiments of the present disclosure provide a hybridoma comprising reduced chemokine receptor CCX-CKR activity. Methods for reducing chemokine receptor CCX-CKR activity are described herein.
[00196] Certain embodiments of the present disclosure provide an antibody produced from a hybridoma cell as described herein. Methods for producing antibodies from hybridoma cells are known.
[00197] Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody from a hybridoma cell as described herein.
[00198] Certain embodiments of the present disclosure provide a method of producing a monoclonal antibody, the method comprising producing an antibody from a hybridoma comprising a B-cell and an immortalised cell, wherein the B cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity. [00199] Standard techniques may be used for recombinant DNA technology, oligonucleotide synthesis, antibody production, peptide synthesis, tissue culture and transfection. Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures may be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), herein incorporated by reference.
EXAMPLE 1 - Deletion of CCX-CKR enhances the antibody response
[00200] Wild-type or CCX-CKR" " mice were immunised with ovalbumin in Alum-OH gel and peripheral blood was taken at the indicate time post-immunisation. Immunoglobulin-specific ELISAs were conducted to assess the effect on anti-OVA antibody production. Knock-out mice were produced as described in Comerford I.et al (2010) Blood 116(20): 4130-40.
[00201] OVA immunisation and ELISA: 8-10 week old female CCX-CKR+ + and CCX- CKR" " mice were immunised with 100 μg ovalbumin (OVA) in aluminum hydroxide gel (100 μί/π αιβε) intraperitoneally. Serum was harvested on days 0 (pre-bleed prior to immunisation), 14 and 21 post-immunisation. Direct ELISAs were used to quantify serum titers of anti-OVA antibodies. Briefly, 96-well plates were coated with 10 μg/mL OVA protein in 0.05M carbonate buffer (Na2C03 + NaHC03; pH9.6) overnight and subsequently blocked with 2% skim milk/PBS. Serum was serially diluted onto the 96- well plates in PBS and incubated for 2 hours at room temperature. After washing, antigen-specific antibodies were detected using rat anti-mouse -IgG, -IgGl or IgG2c antibodies conjugated to HRP. Bound antibody was detected using TMB substrate and plates read at 450/620nm. All washes were performed using 0.01% Tween20/PBS. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results. [00202] The results are shown in Figure 1. The data shows that deletion of CCX-CKR enhances the antibody response.
EXAMPLE 2- Deletion of CCX-CKR enhances the high-affinity antibody response
[00203] Wild-type or CCX-CKR" " mice were immunised with P-KLH in Alum and peripheral blood was taken at the indicate time post-immunisation. Immunoglobulin- specific ELISAs were conducted to assess the effect on high ( P(5)) and low (NP(16)) affinity and anti- P antibody production.
[00204] NP-KLH immunisation and ELISA: 8-10 week old female CCX-CKR+ + and CCX-CKR " mice were immunised with 100 μg NP24-KLH precipitated in alum (100 uL/mouse) intraperitoneally. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 14 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of high-affinity and global-affinity anti-NP antibodies. Briefly, 96-well plates were coated with 10 μg/mL of either NP5-BSA (high-affinity) or NP16-BSA (global affinity) in 0.05M carbonate buffer (Na2C03 + NaHC03; pH9.6) overnight and subsequently blocked with 3% BSA/PBS. Serum was serially diluted onto the 96-well plates in PBS and incubated for 2 hours at room temperature. After washing, antigen- specific antibodies were detected using rat anti-mouse IgG conjugated to HRP. Bound antibody was detected using TMB substrate and plates read at 450/620nm. All washes were performed using 0.01% Tween20/PBS. All washes were performed using 0.01% Tween20/PBS. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results.
[00205] The results are shown in Figure 2. The data shows that deletion of CCX-CKR enhances the high-affinity antibody response.
EXAMPLE 3 - Deletion of CCX-CKR enhances the generation of protective anti-SFV antibodies
[00206] Wild-type or CCX-CKR" " mice were immunised with Semliki Forrest Virus (γ- SFV via the intravenous route) and peripheral blood was collected on the indicated days post-immunisation. Anti-SFV-specific ELISAs were conducted to quantify anti-SFV antibody production (top and middle panels) and the ability of the antibodies to neutralise SFV by preventing its ability to infect Vero cells was determined (bottom panel).
[00207] 8-10 week old female CCX-CKR+ + and CCX-CKR" " mice were immunised with two doses known to not induce protective antibody responses in wild-type B6 mice: 2.5 x 108 and 2.5 x 107 PFU-equivalent of gamma (y)-irradiated (50KGy) semliki forest virus (SFV) (y-SFV) intravenously. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 15 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of anti-SFV antibodies. Briefly, 96-well plates were coated with boric acid-inactivated SFV in 0.05M carbonate buffer (Na2C03 + NaHC03; pH9.6) overnight and subsequently blocked with 3% BSA/PBS. Serum was serially diluted onto the 96-well plates in PBS and incubated for 2 hours at room temperature. After washing, antigen-specific antibodies were detected using rat anti-mouse IgG conjugated to HRP. Bound antibody was detected using TMB substrate and plates read at 450/620nm. All washes were performed using 0.01% Tween20/PBS. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results.
[00208] SFV neutralization assays were utilized to determine the virus neutralizing capability of serum anti-SFV antibodies. To do this, 100 PFU live SFV was co-cultured with the indicated serum dilutions (harvested 21 days post-immunisation) for 1 hour and then subjected to a plaque forming assay whereby the SFV/sera dilutions were added to Vero cell monolayers, incubated for 3 days and plaques counted using microscopy. Percentage of SFV neutralization was calculated by subtracting the number of plaques formed in test wells from the number of plaques formed from wells which received live SFV co-cultured with serum from naive mice. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results.
[00209] The results are shown in Figure 3. The data shows that deletion of CCX-CKR enhances the generation of protective anti-SFV antibodies. EXAMPLE 4 - Deletion of CCX-CKR enhances the generation of protective anti-SFV antibodies
[00210] Wild-type or CCX-CKR" " mice were immunised with Semliki Forrest and peripheral blood was collected on the indicated days post-immunisation. Anti-SFV- specific ELISAs were conducted to quantify anti-SFV antibody production (top panel) and the ability of the antibodies to neutralise SFV by preventing its ability to infect cells was determined (bottom panel).
[00211] 8-10 week old female CCX-CKR+ + and CCX-CKR"'" mice were immunised with 2.5 x 109 PFU-equivalent of gamma-irradiated (γ) (50KGy) semliki forest virus (SFV) (γ-SFV) intravenously. Serum was harvested on days 0 (pre-bleed prior to immunisation), 7, 15 and 21 post-immunisation. Direct ELISAs were used to quantify serum titres of anti-SFV antibodies. Briefly, 96-well plates were coated with boric acid- inactivated SFV in 0.05M carbonate buffer (Na2C03 + NaHC03; pH9.6) overnight and subsequently blocked with 3% BSA/PBS. Serum was serially diluted onto the 96-well plates in PBS and incubated for 2 hours at room temperature. After washing, antigen- specific antibodies were detected using rat anti-mouse IgG conjugated to HRP. Bound antibody was detected using TMB substrate and plates read at 450/620nm. All washes were performed using 0.01% Tween20/PBS. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results.
[00212] SFV neutralization assays were utilized to determine the virus neutralizing capability of serum anti-SFV antibodies. To do this, 100 PFU live SFV was co-cultured with the indicated serum dilutions (harvested 21 days post-immunisation) for 1 hour and then subjected to a plaque forming assay whereby the SFV/sera dilutions were added to vero cell monolayers, incubated for 3 days and plaques counted using microscopy. Percentage of SFV neutralization was calculated by subtracting the number of plaques formed in test wells from the number of plaques formed from wells which received live SFV co-cultured with serum from naive mice. Data represent mean + SEM (n=4/group) from one of two independent experiments, which obtained similar results
[00213] The results are shown in Figure 4. The data shows that deletion of CCX-CKR enhances the generation of protective anti-SFV antibodies. EXAMPLE 5 - Production of hybridoma cells from mice deficient in CCX-CKR activity
[00214] Hybridoma cells from mice deficient in CCX-CKR activity may be produced using the protocol set out below.
[00215] (i) Materials:
Gerbu Adjuvant MM, #3001A, 6x1 ml, Accurate Chemical & Scientific Corp.
Hybridoma-SFM, #12045-076, 1000ml, Invitrogen
HAT, H0262-10VL, 10 vials, Sigma
HT, H0137-10VL, 10 vials, Sigma
Recombinant human IL6, RIL61, 20ug, Endogen/Pierce
PEG 1500, #783641, 10x4ml, Roche Diagnostics
[00216] (ii) HAT Fusion Medium (500ml):
50ml FCS
10ml HAT (1 vial of Sigma H0262-10VL)
5 ml Pen/Strep
500ul (25mg) Gentamicin
300 U humIL6 (optional)
in Hyb-SFM
[00217] (iii) HT Fusion Medium (500ml)
50ml FCS
10ml HT ( 1 vial of Sigma HO 137- 1 OVL)
5 ml Pen/Strep
500ul (25mg) Gentamicin
in Hyb-SFM
[00218] (iv) CCX-CKR"'" mice
[00219] Gene knock out in the CCX-CKR gene may be produced as described in Comerford l et al (2010) Blood 116(20): 4130-40. [00220] (iv) Immunization
1. Use BALB/c CCX-CKR" " mice female mice, 8-10 weeks of age
2. Immunize mouse three times (dO, dl4, d21) i.p. with selected antigen and 100ml Gerbu adjuvant (total volume of 200ml). Use 1-lOOug of antigen depending on purity, concentration, and availability. The amount for the first immunization is twice the amount of all subsequent immunizations.
3. Bleed mouse before each immunization for serum collection (~100ul).
4. Perform mouse serum ELISA after d21 sampling. The titer from day 0 to day 21 should increase by 100-1000 fold.
5. When the titer is sufficient (usually after three immunizations), boost with antigen (no adjuvant, add 100ml lxPBS instead) three times hree days in a row (d28, d29, d30)
6. Perform fusion on d31
[00221] (v) Fusion Protocol
Prewarm all media and PEG 1500 to 37°C.
Typically need:
1-1.5L HAT medium (assuming 3-5 x 108 splenocytes)
-100ml HYB-SFM + 10% FBS
~25ml HYB-SFM, no FBS
Myeloma cells
-Assess and count X63-Ag8.653 myeloma cells. Leave in Hyb+10%FCS in incubator throughout spleen cell recovery.
Spleen cell collection
1. Euthanize mouse via cervical dislocation.
2. Place entire mouse in a beaker containing ~200ml of 80%Betadine, 20% 70% ethanol.
3. Remove spleen using aseptic techniques
4. Transfer spleen to Petri dish containing 5ml Hyb SFM + 10%FCS
5. In hood, move spleen to new Petri dish in 1ml Hyb-SFM+10%FCS.
6. Trim fat and connective tissue, then cut spleen into small pieces.
7. Using two sterile watchmaker's forceps, secure a piece of spleen with one forceps while "milking" cells from the piece into the medium. 8. Transfer cells to a 50ml tube, through several (~4) washes with 2-3ml medium (Also wash capsule sections. Wash "main" spleen cell collection 4 or 5 times, and capsule collection 2 or 3 times. Transfer all material, including chunks and capsules, to 50ml tube)
9. Pipette cell suspension a few times, then let sit (~lmin) until larger tissue pieces have fallen to the bottom of the tube.
10. Collect upper cell suspension and place in new 50ml tube. Centrifuge at 900-lOOOrpm for 5min.
11. Pour off supernatant.
12. Resuspend spleen cells in 20ml HYB-SFM + 10% FBS
13. Count spleen cells (both undiluted and 1 : 10)
14. Calculate required number of X63s (i.e., ½ number of spleen cells). Combine X63s directly with spleen cells.
Fusion
1. Centrifuge mixed cells 900-1000rpm 5 min.
2. Wash cells with 25ml Hyb-SFM medium (no additives) and centrifuge
3. Loosen pellet by finger- flicking. Aim for a slurry of cells; do not want chunks or pelleted cells.
4. Slowly add 1.5ml PEG per 3x108 mixed cells.
Dispense PEG along sides of tube. Does not need to be lml/min slow, just slow enough to keep from blasting cells. Do not touch cell/PEG slurry with pipette. After dispensing PEG, mix by swirling tube.
5. Incubate for 1 minute at 37C
6. Add very slowly (slower than cells over Ficoll) a total of 20ml Hyb-SFM. (use two 10ml pipettes).
lml in first minute
3 ml in second minute
16ml in third minute
7. Centrifuge
8. Calculate total amount of HAT Fusion medium necessary for plating. Use tissue-culture treated 24 well plates, 106 total cells/well.
For each well, calculate 2ml of medium
9. Subtract 200ml from total HAT medium volume and divide into workable volumes (<500ml) in wide-mouth Erlenmeyer flasks.
10. With the remaining 200ml HAT medium, slowly add 25ml at a time to the mixed cell pellet and gently pour even amounts into each HAT bulk volume (first 25ml into one flask, second 25ml into next flask, etc in an attempt to finish with 103 cells/ml/flask)
11. Put all but one flask into incubator to maintain temperature while plating.
12. Gently swirl flask to keep cells evenly distributed
13. Using 25ml pipette, plate 2ml/well. Gently swirl flask on a regular basis throughout plating (ex. after each 1-2 plates)
14. Wrap plates in Saran Wrap, three to a stack, place in incubator (37°C, 5% C02)
Clone Testing
1. After 10-14 days when clones are visible by eye and medium in some wells just begins to turn yellowish, test all wells by ELISA.
2. Pick individual clones from ELISA positive wells
Transfer to round bottom, cell-treated 96 well plates, 1 clone/well
150ul medium/well
Use 30ul to pick clones
3. When 96-well clones are just visible by eye (usually 3 days later), test by ELISA.
Test supernatants for IgG (or other fusion protein tag) and the protein of interest in parallel.
4. For clones that recognize the protein of interest and not the IgG or fusion protein tag, transfer to 24 well plates - 1 ml/well
5. Transfer to 6well plates - 3-4ml/well. Collect the supernatants for additional testing depending on the protein: e.g. fusions for CD markers or surface proteins should be tested by flow cytometry. Monoclonal antibodies to an IgG isotype should be tested against various isotypes, etc to identify clones of highest interest.
6. When well-grown, fix an aliquot of cells for clonality testing by flow cytometry, freeze an aliquot, and collect supernatant for mouse isotype ELISA 7. After an additional 2 weeks in HAT (4 weeks total), wean cells into HT medium.
8. After 2 weeks in HT medium, wean cells into Hyb+10%FCS
9. Wean to Hyb-SFM for bulk culture for purification.
EXAMPLE 5 - Immunogenic compositions
[00222] An immunogenic composition may be produced as follows:
One ml adult dose, as a sterile suspension in isotonic saline:
Selected Antigen: 5 -250 μg
Neutralising CCX-CKR Antibody: 10-500 μg
The selected antigen may be adsorbed on 0.5 mg aluminium as aluminium hydroxide. The composition may be administered by intramuscular administration.
[00223] Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. Where a specific range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is included therein. All smaller sub ranges are also included. The upper and lower limits of these smaller ranges are also included therein, subject to any specifically excluded limit in the stated range.
[00224] Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
[00225] As used herein, the singular forms "a", "an" and "the" include plural aspects unless the context already dictates otherwise. [00226] All methods described herein can be performed in any suitable order unless indicated otherwise herein or clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the example embodiments and does not pose a limitation on the scope of the claimed invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential.
[00227] The description provided herein is in relation to several embodiments which may share common characteristics and features. It is to be understood that one or more features of one embodiment may be combinable with one or more features of the other embodiments. In addition, a single feature or combination of features of the embodiments may constitute additional embodiments.
[00228] The subject headings used herein are included only for the ease of reference of the reader and should not be used to limit the subject matter found throughout the disclosure or the claims. The subject headings should not be used in construing the scope of the claims or the claim limitations.
[00229] Future patent applications may be filed on the basis of the present application, for example by claiming priority from the present application, by claiming a divisional status and/or by claiming a continuation status. It is to be understood that the following claims are provided by way of example only, and are not intended to limit the scope of what may be claimed in any such future application. Nor should the claims be considered to limit the understanding of (or exclude other understandings of) the present disclosure. Features may be added to or omitted from the example claims at a later date.
[00230] Although the present disclosure has been described with reference to particular examples, it will be appreciated by those skilled in the art that the disclosure may be embodied in many other forms.

Claims

1. A method of inducing an immune response to a selected antigen in a subject, the method comprising:
inhibiting activity of a chemokine receptor CCX-CKR in the subject; and exposing the subject to the selected antigen.
2. The method according to claim 1, wherein the immune response is a humoral response.
3. The method according to claims 1 or 2, wherein the immune response comprises induction of IgM and/or IgG antibodies to the selected antigen.
4. The method according to any one of claims 1 to 3, wherein the immune response comprises the induction of high affinity antibodies to the selected antigen.
5. The method according to any one of claims 1 to 4, wherein the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
6. The method according to any one of claims 1 to 5, wherein the inhibiting of activity of a chemokine CCX-CKR receptor comprises a gene knock out and/or a gene knock down.
7. The method according to claim 6, wherein the gene knock out and/or the gene knock down comprises a knock out and/or a knock down of a chemokine receptor CCX- CKR gene.
8. The method according to any one of claims 1 to 5, wherein the inhibiting of activity of a chemokine CCX-CKR receptor comprises exposing the subject to a chemokine receptor CCX-CKR inhibitor.
9. The method according to claim 8, wherein the exposing the subject to a chemokine receptor inhibitor comprises administering a chemokine receptor CCX-CKR inhibitor.
10. The method according to claims 8 or 9, wherein the chemokine receptor inhibitor comprises one or more of a chemokine receptor CCX-CKR antagonist, a small interfering RNA, a microRNA, an antisense RNA, a ligand for a chemokine receptor CCX-CKR, or an antibody and/or an antigen binding part thereof.
11. The method according to any one of claims 8 to 10, wherein the chemokine receptor CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof.
12. The method according to claim 11, wherein the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
13. The method according to any one of claims 1 to 12, wherein the method comprises exposing the subject to an adjuvant.
14. The method according to any one of claims 1 to 13, wherein the method is used to produce an antibody to the selected antigen in an animal or a human subject.
15. The method according to any one of claims 1 to 13, wherein the method is used for one or more of the following: to improve an immune response to the selected antigen in the subject; to vaccine the subject against the selected antigen; to improve protective immunity in the subject against an infectious agent comprising the selected antigen or a related antigen; to improve efficacy of a vaccine comprising the selected antigen; to improve high affinity antibody to the selected antigen production in the subject; and to reduce the amount of the selected antigen exposed to the subject to obtain a desired immunological response.
16. A method of inducing an immune response to a selected antigen in a subject, the method comprising administering to the subject the selected antigen and a chemokine receptor CCX-CKR inhibitor.
17. An immunogenic composition comprising a selected antigen and a chemokine receptor CCX-CKR inhibitor.
18. The immunogenic composition according to claim 17, wherein the selected antigen comprises one or more of a bacterial antigen, a viral antigen, a fungal antigen, a protozoan antigen, a metazoan antigen, a cancer antigen, a tumour antigen, an antigen derived from an allergen, an autoantigen and a cell surface molecule.
19. The immunogenic composition according to claims 17 or 18, wherein the chemokine receptor CCX-CKR inhibitor comprises one or more of a chemokine receptor CCX-CKR antagonist, a small interfering RNA, a microRNA, an antisense RNA, a ligand for a chemokine receptor CCX-CKR, an antibody and/or an antigen binding part thereof.
20. The immunogenic composition according to any one of claims 17 to 19, wherein the CCX-CKR inhibitor comprises an antibody and/or an antigen binding fragment thereof.
21. The immunogenic composition according to claim 20, wherein the antibody and/or the antigen binding fragment thereof binds to a chemokine receptor CCX-CKR.
22. The immunogenic composition according to any one of claims 17 to 21, wherein the immunogenic composition comprises an adjuvant.
23. Use of a chemokine receptor CCX-CKR inhibitor in the preparation of an immunogenic composition.
24. A method of inducing an immune response to a selected antigen in a subject, the method comprising exposing the subject to an immunogenic composition according to any one of claims 17 to 23.
25. A method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to the selected antigen and a chemokine receptor CCX- CKR inhibitor.
26. A vaccine comprising an antigen and a chemokine receptor CCX-CKR inhibitor.
27. Use of a chemokine receptor CCX-CKR inhibitor in the preparation of a vaccine.
28. A nucleic acid vaccine comprising (i) a nucleic acid encoding an antigen and a nucleic acid encoding a chemokine receptor CCX-CKR inhibitor; and/or (ii) a nucleic acid encoding an antigen and a chemokine receptor CCX-CKR inhibitor.
29. A method of vaccinating a subject against a selected antigen, the method comprising exposing the subject to a nucleic acid vaccine according to claim 28.
30. A method of improving an immune response to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
31. A method of reducing the amount of a selected antigen exposed to a subject to obtain a desired immunological response, the method comprising exposing the subject to a chemokine receptor CCX-CKR inhibitor.
32. Use of a chemokine receptor CCX-CKR inhibitor as an adjuvant.
33. A composition comprising:
(i) a chemokine receptor CCX-CKR inhibitor;
(ii) one or more selected antigens;
(iii) an excipient; and
optionally one or more adjuvants.
34. A combination product comprising the following components: a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to induce an immune response to the selected antigen in a subject.
35. A combination product comprising the following components:
a selected antigen; and
a chemokine receptor CCX-CKR inhibitor;
wherein the components are provided in a form for separate or coadministration to vaccinate a subject.
36. A method of producing an antibody to a selected antigen in an animal subject, the method comprising exposing the animal subject to the selected antigen, wherein the animal subject comprises a reduced chemokine receptor CCX-CKR activity.
37. The method according to claim 36, wherein the animal subject comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene.
38. An isolated or purified antibody produced according to claims 36 or 37.
39. A method of producing a hybridoma cell, the method comprising fusing a B cell with reduced chemokine receptor CCX-CKR activity with an immortalised cell.
40. The method according to claim 39, wherein the B cell comprises a knock out and/or a knock down in a chemokine receptor CCX-CKR gene.
41. A hybridoma produced according to claims 39 or 40.
42. An antibody produced from a hybridoma cell according to claim 41.
43. A method of producing a monoclonal antibody, the method comprising producing an antibody from a hybridoma comprising a B-cell and an immortalised cell, wherein the B cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity.
44. A hybridoma of a B-cell and an immortalised cell, wherein the B-cell component of the hybridoma comprises a reduced chemokine receptor CCX-CKR activity.
45. A hybridoma of a B-cell and an immortalised cell, wherein the B-cell component of the hybridoma comprises a knock out or a knock down of a chemokine receptor CCX-CKR gene.
46. A method of improving protective immunity against an infectious agent in a subject, the method comprising exposing the subject to a selected antigen associated with the infectious agent and a chemokine receptor CCX-CKR inhibitor.
47. A method of improving efficacy of a vaccine comprising a selected antigen, the method comprising exposing a subject to be vaccinated against the selected antigen to a chemokine receptor CCX-CKR inhibitor.
48. A method of improving high affinity antibody production to a selected antigen in a subject, the method comprising exposing the subject to a chemokine receptor CCX- CKR inhibitor.
PCT/AU2014/000390 2013-04-10 2014-04-10 Methods and compositions for inducing an immune response WO2014165916A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2013901227 2013-04-10
AU2013901227A AU2013901227A0 (en) 2013-04-10 Methods and compositions for inducing an immune response

Publications (1)

Publication Number Publication Date
WO2014165916A1 true WO2014165916A1 (en) 2014-10-16

Family

ID=51688731

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2014/000390 WO2014165916A1 (en) 2013-04-10 2014-04-10 Methods and compositions for inducing an immune response

Country Status (1)

Country Link
WO (1) WO2014165916A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040146926A1 (en) * 1999-10-12 2004-07-29 Chemocentryx, Inc. Chemokine receptor
WO2014036608A1 (en) * 2012-09-06 2014-03-13 Adelaide Research & Innovation Pty Ltd Methods and products for preventing and/or treating metastatic cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040146926A1 (en) * 1999-10-12 2004-07-29 Chemocentryx, Inc. Chemokine receptor
WO2014036608A1 (en) * 2012-09-06 2014-03-13 Adelaide Research & Innovation Pty Ltd Methods and products for preventing and/or treating metastatic cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
COMERFORD, I. ET AL.: "The atypical chemokine receptor CCX-CKR scavenges homeostatic chemokines in circulation and tissues and suppresses Th1 7 responses.", BLOOD, vol. 116, no. 20, 18 November 2010 (2010-11-18), pages 4130 - 4140 *
TAKATSUKA, S. ET AL.: "Generation of a panel of monoclonal antibodies against atypical chemokine receptor CCX-CKR by DNA immunization.'';", JOURNAL OF PHARMACOLOGICAL AND TOXICOLOGICAL METHODS, vol. 63, 2011, pages 250 - 257 *

Similar Documents

Publication Publication Date Title
EP1597280B1 (en) Monoclonal antibody production by ebv transformation of b cells
AU2009275226B2 (en) Neutralizing anti-influenza a virus antibodies and uses thereof
AU2009305113B2 (en) Dengue virus neutralizing antibodies and uses thereof
DK1597280T4 (en) Preparation of monoclonal antibodies by EBV transformation of B cells
US11773155B2 (en) Bispecific antibody against rabies virus, and application thereof
KR20170047192A (en) Ebola monoclonal antibodies
KR20110055517A (en) Human cytomegalovirus neutralising antibodies and use thereof
WO2014043215A1 (en) Bi-specifc diabodies for masking and targeting vaccines
US11773181B2 (en) Anti-survivin antibodies for cancer therapy
WO2014165916A1 (en) Methods and compositions for inducing an immune response
WO2022023292A9 (en) Corona virus spike protein-targeting antibodies and use thereof
US20220185911A1 (en) Therapeutic antibodies for treating lung cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14782126

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14782126

Country of ref document: EP

Kind code of ref document: A1