WO2009014382A2 - Cxcl11 adjuvant compositions and uses thereof - Google Patents

Cxcl11 adjuvant compositions and uses thereof Download PDF

Info

Publication number
WO2009014382A2
WO2009014382A2 PCT/KR2008/004317 KR2008004317W WO2009014382A2 WO 2009014382 A2 WO2009014382 A2 WO 2009014382A2 KR 2008004317 W KR2008004317 W KR 2008004317W WO 2009014382 A2 WO2009014382 A2 WO 2009014382A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
cxcll
composition
polynucleotide
immunogen
Prior art date
Application number
PCT/KR2008/004317
Other languages
French (fr)
Other versions
WO2009014382A3 (en
Inventor
Young Chul Sung
Sehwan Yang
Hong Namkoong
Bui Thanh Xuan
Se Jin Im
Original Assignee
Postech Academy-Industry Foundation
Genexine Co., 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
Application filed by Postech Academy-Industry Foundation, Genexine Co., Ltd. filed Critical Postech Academy-Industry Foundation
Publication of WO2009014382A2 publication Critical patent/WO2009014382A2/en
Publication of WO2009014382A3 publication Critical patent/WO2009014382A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/522Alpha-chemokines, e.g. NAP-2, ENA-78, GRO-alpha/MGSA/NAP-3, GRO-beta/MIP-2alpha, GRO-gamma/MIP-2beta, IP-10, GCP-2, MIG, PBSF, PF-4, KC
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • 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
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates generally to adjuvants, vacdne compositions, and methods useful for vaccination to modulate immune response.
  • the present invention provides use of a polynucleotide operably encoding a CXCLl 1 polypeptide or fragments, variant, or derivatives thereof or a CXCLl 1 polypeptide as an adjuvant.
  • Chemokines or chemoattractant cytokines are a family of small (8-10 kDa) proteins that play an important role in recruiting and activating leukocytes. See Luster et al. N. Engl. J. Med. 338: 436-445 (1998). Chemokines induce their biological effects by binding to seven trans-membrane-spanning G protein-coupled receptors. See id. Approximately 50 chemokines and about 16 different chemokine receptors interacting with the chemokines have been reported. Chemokines are subdivided into four main subclasses, designated as C, CC, CXC, and CX3C, according to the presence and position of the conserved cysteine motifs.
  • chemokines play important roles in various activities including maintaining homeostasis, angiogenesis/angiostasis, cellular differentiation and activation, lymphocyte homing, and influencing the overall Thl/Th2 balance of the immune responses. See D'Ambrosio et al., J. Immunol. Meth. 273: 3-13 (2003); Sallusto et al, J. Exp. Med. 187(6): 875-883 (1998).
  • Chemokine receptors often recognize more than one chemokine and, alternatively, several chemokines can bind to multiple receptors.
  • CXCR3 binds CXCL4 (PF4 or platelet factor 4), CXCL9 (monokine-induced by ⁇ -interferon (Mig)), CXCLlO (interferon-inducible protein 10 (IPlO)), and CXCLI l (interferon-inducible T cell- chemoattractant (1-TAC)).
  • Farber Biochem. Biophysic. Res. Comm. 192(1): 223-230 (1993); Loestcher et al. J. Exp. Med. 184:963-969 (1996); Murphy et al.
  • CXCL4 interacts selectively with a splice variant of the CXCR3 receptor, i.e., CXCR3-B while CXCL9, CXCLlO, and CXCLl 1 interact with both receptor variants.
  • CXCLl 1 is regulated by interferon and has potent chemoattractant activity for interleukin-2 (IL-2) activated T cells, but not for freshly isolated unstimulated T cells, neutrophils, or monocytes.
  • IL-2 interleukin-2
  • CXCL9, CXCLlO, and CXCLl 1 are three non-Glu-Leu-Arg CXC chemokines that are more closely related to each other, these ligands appear to mediate distinct biological phenomena in vivo. See Hancock et al. J. Exp. Med. 193:975-980 (2001); Khan et al. Immunity 12: 483-494 (2000); and Zhang et al. J. Immunol. 168: 3205-3212 (2002). This may be related to differential expression of these ligands as has been seen in cardiac and skin allograft rejection ( See Hancock et al. J. Exp. Med. 193:975-980 (2001) and Zhang et al. J.
  • the present invention provides a simple and safe yet effective adjuvant and methods to modulate an immune response using the adjuvant, i.e., a CXCLl 1 polypeptide or a polynucleotide encoding CXCLl 1.
  • the adjuvant i.e., a CXCLl 1 polypeptide or a polynucleotide encoding CXCLl 1.
  • the present invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant selected from the group consisting of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, an isolated CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO:2, and a combination of the polypeptide and polynucleotide, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
  • an adjuvant selected from the group consisting of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, an isolated CXCLl 1 polypeptide comprising at least 10 consecutive amino
  • the invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant, where the adjuvant comprises an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant; or a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant, where the adjuvant comprises an isolated CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
  • the CXCLl 1 polypeptide may comprise, e.g., at least 45 or at least 70 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO:2.
  • the CXCLl 1 polypeptide may comprise amino acids 27 to 89 of SEQ ID NO:2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO:2.
  • the instant invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant selected from the group consisting of an isolated polynucleotide which operably encodes a CXCLl 1 polypeptide comprising an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2, an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2, and a combination of the polypeptide and polynucleotide, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
  • an adjuvant selected from the group consisting of an isolated polynucleotide which operably encodes a CXCLl 1 polypeptide comprising an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2, an isolated CXCLl 1 poly
  • a method for modulating an immune response in an animal comprising administering to an animal in need thereof a composition comprising an immunogen, and an adjuvant which comprises an isolated polynucleotide operably encoding a CXCLl 1 polypeptide, which comprises an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2 , wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
  • the amino acid sequence may be, e.g., at least 90% or at least 95% identical to amino acids 27 to 89 of SEQ ID NO: 2.
  • the amino acid sequence may further be, e.g., identical to amino acids 27 to 89 of SEQ ID NO: 2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO: 2.
  • the polynucleotide encoding the CXCLl 1 polypeptide can be DNA or RNA such as messenger RNA (mRNA).
  • the polynucleotide may be incorporated into a vector such as a plasmid or a viral vector.
  • the vector can be incorporated into a cell. In a certain embodiment, the cell in which the vector is incorporated is not a tumor cell.
  • the method of modulating an immune response in an animal used in the present invention comprises administering a composition comprising an immunogen and an adjuvant comprising an isolated polynucleotide encoding the CXCLl 1 polypeptide, where the polynucleotide further comprises a heterologous nucleic acid.
  • the heterologous nucleic acid may encode a heterologous polypeptide fused to the CXCLl 1 polypeptide.
  • a CXCLl 1 polypeptide used as an adjuvant in the present invention may further comprise a heterologous polypeptide fused thereto.
  • the heterologous polypeptide may be, for example, but not by way of limitation, an antigen, an immunoglobulin Fc region, and a secretory signal peptide. In one embodiment, the heterologous polypeptide is not a chemokine.
  • an immunogen may be, but not by way of limitation, a polypeptide or a polynucletide encoding a polypeptide, or both.
  • the immunogen comprises an isolated polynucleotide which encodes an antigenic or immunogenic polypeptide or fragment, variant, or derivative thereof.
  • a polynucleotide immunogen and a polynucleotide which operably encodes the CXCLl 1 polypeptide are situated on the same vector.
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by two copies of different promoters or identical promoters on the same vector.
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by a single promoter as a bi ⁇ stronic transcript where the coding regions are separated by an internal ribosomal entry site (IRES).
  • IRS internal ribosomal entry site
  • polynucleotide immunogen and the polynucleotide which operably encodes the CXCLI l polypeptide are situated on separate vectors.
  • a polynucleotide immunogen encoding an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof and the polynucleotide operably encoding the CXCLI l polypeptide may be administered simultaneously.
  • the polynucleotide immunogen may be administered prior to the polynucleotide encoding the CXCLl 1 polypeptide or after the polynucleotide encoding the CXCLl 1 polypeptide.
  • the polynucleotide which encodes the CXCLI l polypeptide and a polynucleotide immunogen may be administered at a ratio of about 20: 1 to about 1:20, about 10:1 to about 1:10, or about 6:1.
  • the composition used in the method of the present invention may further comprise an additional adjuvant or a transfection facilitating agent.
  • an improved immune response elicited by the methods of the present invention may comprise an increased antibody response, for example, an IgG response, or an increased T-cell response such as a cytotoxic T lymphocyte (CTL) response.
  • the improved immune response may be an improved ThI response relative to administration of the immunogen alone.
  • the improved immune response may comprise increased serum levels of a cytokine or recruitment of cells selected from the group consisting of NK cells, CTLs, B lymphocytes, dendritic cells, macrophages, neutrophils, and a combination of two or more of the cells.
  • the improved immune response may comprise immunogen dose sparing. The dose sparing comprises achieving a comparable immune response to administration of the immunogen alone using reduced amount or reduced doses of the immunogen.
  • FIG. 1 depicts a schematic diagram of the schedule of administration of plasmids encoding OVA antigen with and without CXC chemokines including CXCLl 1. Also shown are the treatment conditions of control groups.
  • FIG. 2 depicts the level of total IgG in mice immunized with plasmids encoding antigen with or without chemokines at 3 weeks after priming.
  • FIGS. 3A, 3B and 3C depict the results of total IgG, IgG2a, and IgGl levels in mice immunized with plasmids encoding antigen with or without chemokines at 2 weeks after boosting.
  • FIG.4 depicts the effect of the chemokine adjuvants on CD4 + T cell responses.
  • FIG.5 depicts the effect of the chemokine adjuvants on CD8 + T cell response in term of IFN- ⁇ production.
  • FIG. 6 depicts FACS analysis of OVA-spe ⁇ fic CD8 + T cell responses, mediated by
  • FIGS. 7A and 7B depict FACS analysis of CD8 + T cell responses, mediated by CXC chemokines, assessed by staining for intracellular TNF- ⁇ and IL-2.
  • FiG. 8 depicts a schematic diagram of the schedule of administration of the trivalent influenza vaccine with or without the recombinant CXCLl 1-Fc protein.
  • FIG. 9 depicts the adjuvant effect of the recombinant CXCLl 1-Fc protein on CD8 + and CD4 + T cell responses in Balb/c mice model.
  • FIGS. 1OA and 1OB depict the results of end-point titration with anti-HA(H5Nl) antibodies to examine the adjuvant effect of the recombinant CXCLl 1-Fc protein for the induction of heterosubtypic immunity against influenza virus. Best Mode for Carrying Out the Invention
  • a or “an” entity refers to one or more of that entity; for example, “a polynucleotide”, is understood to represent one or more polynucleotides.
  • the terms “a” (or “an”), “one or more”, and “at least one” can be used interchangeably herein.
  • nucleic acid or “nucleic acid fragment” refers to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide or construct.
  • Two or more nucleic acids of the present invention can be present in a single polynucleotide construct, e.g., on a single plasmid, or in separate (non-identical) polynucleotide constructs, e.g., on separate plasmids.
  • any nucleic acid or nucleic acid fragment may encode a single polypeptide, e.g., a single antigen, cytokine, or regulatory polypeptide, or may encode more than one polypeptide, e.g., a nucleic acid may encode two or more polypeptides.
  • a nucleic acid may encode a regulatory element such as, e.g., a promoter or a transcription terminator, or may encode a specialized element or motif of a polypeptide or protein, such as a secretory signal peptide or a functional domain.
  • polynucleotide or “polynucleotides” is intended to encompass a single nucleic acid or nucleic acid fragment as well as plural nucleic acids or nucleic acid fragments, and refers to an isolated molecule or construct, e.g., a virus genome (e.g., a non-infectious viral genome), messenger RNA (mRNA), plasmid DNA (pDNA), or derivatives of pDNA (e.g., mini ⁇ rcles as described in (Darquet, A-M et al., Gene Therapy 4:1341-1349 (1997)) comprising a polynucleotide.
  • virus genome e.g., a non-infectious viral genome
  • mRNA messenger RNA
  • pDNA plasmid DNA
  • derivatives of pDNA e.g., mini ⁇ rcles as described in (Darquet, A-M et al., Gene Therapy 4:1341-1349 (1997) comprising a polynucleo
  • a polynucleotide may be provided in linear (e.g., mRNA), circular (e.g., plasmid), or branched form as well as double-stranded or single-stranded forms.
  • a linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease.
  • a polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g., an amide bond, such as found in peptide nucleic acids (PNA)).
  • polynucleotide or “polynucleotides” are used interchangeably with the terms “nucleotide,” “nucleotides,” “nucleic acid,” “nucleic acids,” “nucleotide sequence,” “polynucleotide sequence,” and “nucleic acid sequence.”
  • operably encoding refers to operable transcription of a given coding region to mRNA and translation of the mRNA to a polypeptide by a polynucleotide sequence comprising the coding region, transcriptional regulatory sequences, e.g., a promoter, andor translational regulatory sequences, e.g., a translation initiation factor, that are necessary for the transcription and translation of the coding region.
  • transcriptional regulatory sequences e.g., a promoter
  • translational regulatory sequences e.g., a translation initiation factor
  • the nucleic acid sequences may be obtained by chemical synthesis or by reverse-transcription of a messenger RNA (mRNA) corresponding to CXCLl 1 to a complementary DNA (cDNA) and converting the latter into a double stranded cDNA.
  • mRNA messenger RNA
  • cDNA complementary DNA
  • polypeptide is intended to encompass a singular
  • polypeptide as well as plural “polypeptides”, and comprises any chain or chains of two or more amino acids.
  • terms including, but not limited to “peptide”, “dipeptide”, “tripeptide”, “protein”, “amino acid chain”, or any other term used to refer to a chain or chains of two or more amino acids are included in the definition of a “polypeptide”, and the term “polypeptide” may be used instead of, or interchangeably with any of these terms.
  • polypeptides which have undergone post- translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
  • CXCLl 1 polypeptide encompasses full length CXCLl 1, mature CXCLl 1, or other variants of full length CXCLl 1, fragments of full length CXCLl 1, or allelic and other variants of fragments of full length CXCLl 1, derivatives of full-length CXCLl 1, derivatives of fragments of full-length CXCLl 1, analogues of full-length CXCLl 1, analogues of fragments of full-length CXCLl 1, and chimeric and fusion polypeptides comprising full length CXCLl 1 or one or more fragments of full length CXCLI l.
  • CXCLI l polynucleotide or "CXCLI l nucleic acid,” as used herein, encompass any nucleic acid sequence operably encoding CXCLI l polypeptides as described above.
  • CXCLl 1 polypeptides of the present invention include polypeptides which retain at least some of the activity of the CXCLl 1 polypeptide, e.g, modulating an immune response to an immunogen.
  • One example of the CXCLl 1 polypeptide activities is its binding affinity to CXCR3 to transduce its signals.
  • the fragment, analog, derivative, or variant of the CXCLl 1 polypeptide of the present invention may retain at least some affinity to CXCR3.
  • the affinity may be the same as, or lesser than, the full length CXCLI l polypeptide.
  • the fragment, analog, derivative, or variant of the CXCLl 1 polypeptides of the present invention retains, at least to a certain extent, the ability to attract CD4+ or CD8+ lymphocytes in the area.
  • the fragment, analog, derivative, or variant of the CXCLl 1 polypeptides of the present invention may induce an antibody response or induce cytokine secretion.
  • Fragments of the CXCLl 1 polypeptides of the present invention include proteolytic fragments, deletion fragments and in particular, fragments of CXCLl 1 polypeptides which exhibit increased solubility during expression, purification, and or administration to an animal.
  • Polypeptide fragments further include any portion of the polypeptide which comprises a portion of the native polypeptide that binds to CXCR3, including linear as well as three-dimensional portions.
  • An “epitopic fragment” of a polypeptide is a portion of the polypeptide that contains an epitope.
  • An “epitopic fragment” may, but need not, contain amino acid sequence in addition to one or more epitopes as defined below.
  • variant refers to a polypeptide that differs from the recited polypeptide due to amino acid substitutions, deletions, insertions, andor modifications. Variants may occur naturally, such as an allelic variant.
  • Non-naturally occurring variants may be produced using art-known mutagenesis techniques.
  • variant polypeptides differ from an identified sequence by substitution, deletion or addition of five amino acids or fewer.
  • Such variants may generally be identified by modifying a polypeptide sequence, and evaluating the antigenic properties of the modified polypeptide using, for example, the representative procedures described herein.
  • Polypeptide variants preferably exhibit at least about 60-70%, for example 75%,
  • variant polypeptides may comprise conservative or non- conservative amino acid substitutions, deletions or additions.
  • Derivatives of the CXCLl 1 polypeptides of the present invention are polypeptides which have been altered so as to exhibit additional features not found on the native polypeptide. Examples include fusion proteins.
  • An analog is another form of the CXCLl 1 polypeptide of the present invention.
  • An example is a proprotein which can be activated by cleavage of the proprotein to produce an active mature polypeptide.
  • Variants may also, or alternatively, contain other modifications, whereby, for example, a polypeptide or polynucleotide may be conjugated or coupled, e.g., fused to a heterologous polypeptide or polynucletoide, e.g., a signal (or leader) sequence at the N-terminal end of the protein which oo-translationally or post-translationally directs transfer of the protein.
  • a polypeptide or polynucleotide may be conjugated or coupled, e.g., fused to a heterologous polypeptide or polynucletoide, e.g., a signal (or leader) sequence at the N-terminal end of the protein which oo-translationally or post-translationally directs transfer of the protein.
  • polypeptide or polynucleotide may also be conjugated or produced coupled to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g., 6-His), or to enhance binding of the polypeptide to a solid support or to regulate the expression of the CXCLl 1 polypeptide.
  • a polypeptide or polynucleotide may be conjugated or coupled to an immunoglobulin Fc region or a nucleotide encoding the same.
  • the polypeptide may also be conjugated or coupled to a sequence that imparts or modulates the immune response to the polypeptide (e.g.
  • the sequence conjugated to the CXCLl 1 polypeptide may not be another cytokine or chemokine.
  • the polypeptide may also be conjugated or coupled to other polypeptides/epitopes from tumor, bacteria andor viruses to generate a hybrid immunogenic protein that alone or in combination with various adjuvants can elicit protective immunity to other pathogenic organisms.
  • epitope refers to portions of a polypeptide having antigenic or immunogenic activity in an animal, for example a mammal, for example, a human.
  • An "immunogenic epitope”, as used herein, is defined as a portion of a protein that elicits an immune response in an animal, as determined by any method known in the art.
  • antigenic epitope is defined as a portion of a protein to which an antibody or T-cell receptor can immunospe ⁇ fically bind as determined by any method well known in the art. Immunospe ⁇ fic binding excludes non-specific binding but does not necessarily exclude cross-reactivity with other antigens. Whereas all immunogenic epitopes are antigenic, antigenic epitopes need not be immunogenic.
  • immunogen encompass an antigenic or immunogenic polypeptide or a polynucleotide which operably encodes an antigenic or immunogenic polypeptide.
  • immunogens are described herein below.
  • an "antigenic polypeptide” or an “immunogenic polypeptide” is a polypeptide which, when introduced into an animal, reacts with the animal's immune system molecules, i.e., is antigenic, andor induces an immune response in the animal, i.e., is immunogenic. It is quite likely that an immunogenic polypeptide will also be antigenic, but an antigenic polypeptide, because of its size or conformation, may not necessarily be immunogenic.
  • Isolated antigenic and immunogenic polypeptides of the present invention in addition to the CXCLl 1 polypeptide may be provided as a recombinant protein, a purified subunit, a viral vector expressing the protein, or may be provided in the form of an inactivated whole cell vaccine, e.g., a live-attenuated virus vaccine, a heat-killed virus vaccine, etc.
  • a "coding region” is a portion of nucleic acid which consists of oodons translated into amino acids. Although a “stop oodon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, and the like, are outside the coding region.
  • oodon optimization is defined herein as modifying a nucleic acid sequence for enhanced expression in the cells of the host of interest by replacing at least one, more than one, or a significant number, of oodons of the native sequence with oodons that are more frequently or most frequently used in the genes of that host.
  • Various species exhibit particular bias for certain oodons of a particular amino acid. Methods of oodon optimization are well known to those of ordinary skill in the art.
  • isolated means that the polynucleotide or polypeptide or fragment, variant, or derivative thereof has been removed from other biological materials with which it is naturally associated.
  • An example of an isolated polynucleotide is a recombinant polynucleotide contained in a vector.
  • Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically.
  • a purified polypeptide of the present invention includes a polypeptide that is at least 70-100% pure, i.e., a polypeptide which is present in a composition wherein the polypeptide constitutes 70-100% by weight of the total composition.
  • the purified polypeptide of the present invention is 75%-99% by weight pure, 80%-99% by weight pure, 90-99% by weight pure, or 95% to 99% by weight pure.
  • An example of an isolated polynucleotide is a recombinant polynucleotide contained in a vector. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically. The relative degree of purity of a polynucleotide or polypeptide of the invention is easily determined by well-known methods.
  • compositions, and vaccines of the present invention are pharmaceutically acceptable.
  • an "effective amount” is the amount of a polypeptide, polynucleotide, composition or vaccine, when administered an individual either in a single dose or as part of a series, that is effective for treatment or prevention.
  • An amount is effective, for example, when its administration results in changes in an immune response, for example, humoral or cell-mediated. This amount varies depending upon the health and physical condition of the individual to be treated, the taxmomic group of individual to be treated (e.g. human, nonhuman primate, primate, etc.), the responsive capacity of the individual's immune system, the degree of protection desired, the formulation of the vaccine, a professional assessment of the medical situation, and other relevant factors. It is expected that the effective amount will fall in a relatively broad range that can be determined through routine trials.
  • a single dose of a polypeptide, polynucleotide, composition or vaccine is from about 10 ⁇ g to 10 mgAcg body weight.
  • peptide vaccine or “subunit vaccine” refers to a composition comprising one or more polypeptide immunogens, e.g., antigenic polypeptides or fragments thereof or antigen, which when administered to an animal are useful in stimulating an immune response.
  • DNA vaccine or “polynucleotide vaccine” refers to a composition comprising one or more polynucleotide immunogens, e.g., a polynucleotide encoding an antigenic polypeptide or fragment thereof or antigen.
  • the term "subject” is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, immunization, or therapy is desired.
  • Mammalian subjects include, but are not limited to, humans, domestic animals, farm animals, zoo animals such as bears, sport animals, pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, bears, cows; primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as oows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and so on.
  • the animal is a human subject.
  • mammals and comprises mammals and birds, as well as fish, reptiles, and amphibians.
  • the term animal also encompasses model animals, e.g., disease model animals.
  • the term animal includes valuable animals, either economically or otherwise, e.g., economically important breeding stock, racing animals, show animals, heirloom animals, rare or endangered animals, or companion animals.
  • the mammal can be a human subject, a food animal or a companion animal.
  • an "subject or animal in need thereof” refers to a subject or animal for whom it is desirable to treat, i.e., to prevent, cure, retard, or reduce the severity of disease symptoms, andor result in no worsening of the diseases over a specified period of time.
  • the present invention includes pharmaceutical, immunogenic, or vaccine compositions comprising an immunogen and an adjuvant which comprises an isolated polynucleotide operably encoding a CXCLI l polypeptide, fragment, derivative, analog or variant thereof, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a reference nucleotide sequence encoding amino acids 27 to 89 of SEQ ID NO: 2, wherein the polynucleotide sequence encodes a polypeptide, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the present invention is also directed to a pharmaceutical, immunogenic, or vaccine composition
  • an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence which encodes an amino acid sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acids 27 to 89 of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the present invention is directed to a pharmaceutical, im- munnogenic, or vaccine composition
  • an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence which operably encodes a CXCLI l polypeptide comprising an amino acid sequence least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acids X to Y of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • X can be any number selected from the group consisting of 22, 23, 24, 25, or 26 and Y can be any number selected from the group 89, 90, 91, 92, 93, or 94.
  • the present invention also includes a pharmaceutical, vaccine, or immunogenic composition
  • a pharmaceutical, vaccine, or immunogenic composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the full-length human CXCLl 1 polypeptide is 94 amino acids residues, including a secretory signal peptide sequence of about amino acids 1 to 21.
  • the CXCLl 1 polypeptide is also called small inducible cytokine BI l, Interferon-inducible T-cell ⁇ chemoattractant (I-TAC), Interferon- ⁇ inducible protein 9 (IP-9), H174, and Beta-Rl.
  • the gene encoding the CXCLl 1 polypeptide is called ITAC, SCYBl 1, and SCYB9B.
  • the gene encoding the CXCLl 1 polypeptide and its encoded amino acid sequence are described in PCT Publication No. WO 99/13082, incorporated herein by reference in its entirety.
  • the following polynucleotide sequence was reported as a nucleotide sequence encoding the human CXCLl 1 polypeptide sequence and has the accession number NM
  • Variants of human CXCLl 1 include, but are not limited to, the polypeptides with a mutation, e.g., V89A, an insertion, e.g., K78NRAS, or a deletion, or a combination thereof, e.g., NKGQ69-72IRK.
  • the following polynucleotide sequence was reported as a nucleotide sequence encoding the rhesus monkey CXCLl 1 polypeptide sequence and has the accession number AAK95956 in Genbank.
  • the beginning and ending residues of the domains listed below may vary depending upon the computer modeling program used or the method used for determining the domain.
  • a polynucleotide of the invention can also operably encode a derivative fusion protein, wherein two or more nucleic acid fragments, at least one of which encodes a CXCLl 1 polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide, e.g., immunoglobulin Fc fused to CXCLl 1.
  • a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment.
  • the heterologous nucleic acid or nucleotide sequence may be a regulatory element such as, as a way of non-limiting example, a promoter, enhancer, or terminator sequence.
  • the present invention includes a pharmaceutical, immunogenic, or vaccine composition
  • an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof, where the polynucleotide further comprises a heterologous nucleic acid or fragment thereof which encodes a heterologous polypeptide fused to the CXCLl 1 polypeptide, and where the heterologous polynucleotide comprises the immunogen which encodes an immunogenic or antigenic polypeptide.
  • Compositions may further comprise an additional heterologous polypeptide or polynucleotide immunogen.
  • the polynucleotide of the present invention fused to a heterologous nucleic acid or nucleotide sequence may induce an improved immune response relative to administration of the immunogen or fragment thereof in the absence of the polynucleotide operably encoding a CXCLl 1 polypeptide or fragment thereof.
  • a polynucleotide of the present invention operably encoding a CXCLl 1 polypeptide may be a circular or linearized plasmid or vector, or other linear DNA which may also be non-infectious and nonintegrating (i.e., does not integrate into the genome of vertebrate cells).
  • a linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease.
  • the present invention includes a pharmaceutical, immunological, or vaccine composition
  • a pharmaceutical, immunological, or vaccine composition comprising an immunogen, either polynucleotide or polypeptide, and an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or fragment, derivative, analog, or variant thereof, wherein the composition modulate, enhance, or improve an immune response, wherein the polynucleotide is contained in, or is, a vector.
  • Vectors of the present invention may also be used to produce an adjuvant comprising a CXCLI l polypeptide, fragment, derivative, or variant thereof for use in the compositions and methods of the invention.
  • the choice of vector and expression control sequences to which such nucleic acids are operably linked depends on the functional properties desired, e.g., protein expression in vivo, and in host cell to be expressed.
  • viral genomes e.g., DNA or RNA virus genes or RNA
  • a virus genome of the present invention is nonreplicative, noninfectious, and ⁇ r nonintegrating.
  • the virus genome may include viral vectors such as an adenoviral vector, an alphavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, an Epstein Barr viral vector, a papovaviral vector, a poxvirus vector, a vaccinia viral vector, or an adeno- asso ⁇ ated viral vector.
  • viral vectors such as an adenoviral vector, an alphavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, an Epstein Barr viral vector, a papovaviral vector, a poxvirus vector, a vaccinia viral vector, or an adeno-
  • a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA).
  • mRNA messenger RNA
  • the present invention incudes a method of modulating an immune response in an animal comprising administering an immunogen and an adjuvant as described above, where the adjuvant, and optionally the immunogen, is/are contained in a host cell.
  • the host cell may be a cell derived from the animal to which the composition is to be administered.
  • the host cell may be derived from immune related cells, for example, dendritic cells, macrophages, B- lymphocytes, microglia in the brain, or Kupffer cells in the liver.
  • a host cell is a non tumor cell.
  • a host cell may be an inactivated bacterial or parasitic cell used as a vaccine.
  • Nucleic acids and fragments thereof of the present invention can be altered from their native state in one or more ways.
  • a nucleic acid or fragment thereof which operably encodes a CXCLl 1 polypeptide can be a fragment which encodes only a portion of a full-length polypeptide, andor can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide.
  • the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell.
  • Vectors comprising a polynucleotide operably encoding CXCLl 1 polypeptides may also be used for the methods of the invention.
  • the choice of vector and expression control sequences to which such nucleic acids are operably linked depends on the functional properties desired, e.g., protein expression, and the host cell to be transformed.
  • Expression control elements useful for regulating the expression of an operably linked coding sequence are known in the art. Examples include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements. When an inducible promoter is used, it can be controlled, e.g., by a change in nutrient status, or a change in temperature, in the host cell medium.
  • inducible promoter When an inducible promoter is used, it can be controlled, e.g., by a change in nutrient status, or a change in temperature, in the host cell medium.
  • Any expression vector which is capable of eliciting expression in eukaryotic cells may be used in the present invention.
  • Suitable vectors include, but are not limited to plasmids pcDNA3, pHCMVEeo, pCR3.1, pEFl/His, pIND/GS, pRc/ HCMV2, pSV40£eo2, pTRACER-HCMV, pUB6/V5-His, pVAXl, and pZeoSV2 (available from Invitrogen, San Diego, CA), and plasmid pCI (available from Promega, Madison, WI).
  • Additional eukaryotic cell expression vectors are known in the art and are commercially available. Typically, such vectors contain convenient restriction sites for insertion of the desired DNA segment.
  • Exemplary vectors include pSVL and pKSV-10 (Pharmacia), pBPV-1, pml2d (International Biotechnologies), pTDTl (ATCC 31255), retroviral expression vector pMIG and pLL3.7, adenovirus shuttle vector pDC315, and AAV vectors.
  • Other exemplary vector systems are disclosed e.g., in U.S. Patent 6,413,777.
  • Frequently used regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and enhancers derived from retroviral LTRs, cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdmlP)), polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdmlP adenovirus major late promoter
  • polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters.
  • the recombinant expression vectors may carry sequences that regulate replication of the vector in host cells ⁇ e.g., origins of replication) andor selectable marker genes.
  • a selectable marker gene may not be used.
  • selectable marker genes are often used to facilitate selection of host cells into which the vector has been introduced (see, e.g., Axel, U.S Pat. Nos. 4,399,216; 4,634,665 and 5,179,017).
  • the selectable marker gene confers resistance to a drug, such as G418, hygromy ⁇ n or methotrexate, on a host cell into which the vector has been introduced.
  • a drug such as G418, hygromy ⁇ n or methotrexate
  • selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • vectors may include viral vectors such as an adenoviral vector, an alphavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, an Epstein Barr viral vector, a pa- povaviral vector, a poxvirus vector, a vaccinia viral vector, or an adeno- associated viral vector.
  • the viral vector can be a replication-defective viral vector.
  • Adenoviral vectors that have a deletion in its El gene or E3 gene may be used. When an adenoviral vector is used, the vector usually does not have a selectable marker gene.
  • the vector can include a prokaryotic replioon, i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extra-chromosomally in a bacterial host cell.
  • a prokaryotic replioon i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extra-chromosomally in a bacterial host cell.
  • a prokaryotic replioon may also include a gene whose expression confers a detectable marker such as a drug resistance. Examples of bacterial drug-resistance genes are those that confer resistance to ampi ⁇ llin or tetracycline.
  • Vectors that include a prokaryotic replicon can also include a prokaryotic or bacteriophage promoter for directing expression of the coding gene sequences in a bacterial host cell.
  • Promoter sequences compatible with bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment to be expressed. Examples of such plasmid vectors are pUC8, pUC9, pBR322 and pBR329 (BioRad), pPL and pKK223 (Pharmacia).
  • Any suitable prokaryotic host can be used to express a recombinant DNA molecule encoding a protein used in the methods of the invention.
  • R>r the purposes of this invention, numerous expression vector systems may be employed.
  • cells which have integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow selection of transfected host cells.
  • the marker may provide for pro- totrophy to an autotrophic host, bio ⁇ de resistance (e.g., antibiotics) or resistance to heavy metals such as copper.
  • the selectable marker gene can either be directly linked to the DNA sequences to be expressed, or introduced into the same cell by cotrans- formation.
  • neomycin phosphotransferase (neo) gene is an example of a selectable marker gene (Southern et al, J. MoI. Anal. Genet. 7:327-341 (1982)). Additional elements may also be needed for optimal synthesis of mRNA. These elements may include signal sequences, splice signals, as well as transcriptional promoters, enhancers, and termination signals.
  • Patent No. 5,580,859 the disclosure of which is incorporated herein by reference in its entirety.
  • a viral alphavector a non-infectious vector useful for administering RNA, may be used to introduce RNA into mammalian cells. Methods for the in vivo introduction of alphaviral vectors to mammalian tissues are described in Altman- Hamamdzic, S, et al., Gene Therapy 4, 815-822 (1997), the disclosure of which is incorporated herein by reference.
  • Viral replioons i.e., non-infectious RNA vectors packaged in a viral coat, e.g., a pioornavirus coat or an alphavirus coat, are also useful for efficient administration of RNA. See, e.g., US Patent No. 5,766,6CE, U.S. Patent No. 5,614,413, and PCT Publication No. WO 9507994, the disclosures of which are incorporated herein by reference.
  • Vectors operably encoding a CXCLl 1 polypeptide can be used for transformation or transfection of a suitable host cell, either for administration of the host cell to the subject to be treated, or for the in vitro production of CXCLl 1 polypeptide or polypeptide immunogens. Transformation or transfection can be by any suitable method. Methods for introduction of exogenous DNA into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide (s) in liposomes, and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors.
  • Transformation or transfection of host cells can be accomplished by conventional methods suited to the vector and host cell employed.
  • R>r transformation of prokaryotic host cells, electroporation and salt treatment methods can be employed (Cohen et al., Proc. Natl. Acad. ScL USA (59:2110-14 (1972)).
  • Ibr transformation or transfection of vertebrate cells, electroporation, cationic lipid or salt treatment methods can be employed. See, e.g., Graham et al., Virology 52:456-467 (1973); Wigler et al., Proc. Natl. Acad. ScL USA 7(5:1373-76 (1979).
  • the host cell line used for protein expression is most preferably of mammalian origin; those skilled in the art are credited with ability to preferentially determine particular host cell lines which are best suited for the desired gene product to be expressed therein.
  • Exemplary host cell lines include, but are not limited to NSO, SP2 cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells DG44 and DUXBI l (Chinese Hamster Ovary lines, DHFR minus), HELA (human cervical carcinoma), CVI (monkey kidney line), COS (a derivative of CVI with SV40 T antigen), R1610 (Chinese hamster fibroblast) BALBC/3T3 (mouse fibroblast), HAK (hamster kidney line), SP2O (mouse myeloma), P3x63-Ag3.653 (mouse myeloma), BFA-IcIBPT (
  • the present invention also provides pharmaceutical, immunogenic, or vaccine compositions and methods for modulating an immune response in an animal comprising administering the pharmaceutical, immunogenic, or vaccine composition which comprises an immunogen and a polynucleotide adjuvant as described above to an animal with optimal expression and safety conferred through oodon optimization and/ or other manipulations.
  • These compositions are prepared and administered in such a manner that the encoded gene products are optimally expressed in the animal of interest. As a result, these compositions and methods are useful in stimulating an immune response against the immunogen.
  • nucleic acid coding regions will encode the same polypeptide due to the redundancy of the genetic code. Deviations in the nucleotide sequence that comprise the oodons encoding the amino acids of any polypeptide chain allow for variations in the sequence coding for the gene. Since each oodon consists of three nucleotides, and the nucleotides comprising DNA are restricted to four specific bases, there are 64 possible combinations of nucleotides, 61 of which encode amino acids (the remaining three oodons encode signals ending translation). The "genetic code" which shows which oodons encode which amino acids is reproduced herein as Table 1.
  • amino acids are designated by more than one oodon.
  • R>r example the amino acids alanine and proline are coded for by four triplets, serine and arginine by six, whereas tryptophan and methionine are coded by just one triplet.
  • This degeneracy allows for DNA base composition to vary over a wide range without altering the amino acid sequence of the polypeptides encoded by the DNA.
  • Codon preference or oodon bias differences in oodon usage between organisms, is afforded by degeneracy of the genetic code, and is well documented among many organisms. Codon bias often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, inter alia, the properties of the oodons being translated and the availability of particular transfer RNA (tRNA) molecules.
  • mRNA messenger RNA
  • tRNA transfer RNA
  • the predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization.
  • the present invention relates to a polynucleotide adjuvant as described above comprising, consisting essentially of, or consisting of a coding optimized coding region which operably encodes a CXCLl 1 polypeptide disclosed herein.
  • the codon usage is adapted for optimized expression in the cells of a given prokaryote or eukaryote.
  • a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises a polynucleotide expression construct, vector, or host cell comprising a polynucleotide or fragment thereof which comprises all or partly codon-optimized coding regions, wherein the polynucleotide or fragment thereof operably encodes a CXCLl 1 polypeptide, or fragment, derivative, or variants thereof.
  • Codon usage tables are readily available, for example, at the "Codon Usage Database” available at www.kazusa.or.jp/codon/ (visited July 5, 2007), and these tables can be adapted in a number of ways. See Nakamura, Y., et al, "Codon usage tabulated from the international DNA sequence databases: status for the year 2000" Nucl. Acids Res. 28:292 (2000).
  • codon usage tables By utilizing the codon usage tables, one of ordinary skill in the art can apply the frequencies to any given polypeptide sequence, and produce a nucleic acid fragment comprising a codon-optimized coding region which encodes the polypeptide, but which uses codons optimal for a given species. Ibr example, in some embodiments of the present invention, the coding region is codon-optimized for expression in human.
  • Randomly assigning codons at an optimized frequency to encode a given polypeptide sequence can be done manually by calculating codon frequencies for each amino acid, and then assigning the codons to the polypeptide sequence randomly.
  • various algorithms and computer software programs are readily available to those of ordinary skill in the art. Ibr example, the "EditSeq" function in the Lasergene Package, available from DNAstar, Inc., Madison, WI, the backtranslation function in the VectorNTI Suite, available from InforMax, Inc., Bethesda, MD, and the "back- translate" function in the GCG- Wisconsin Package, available from Accelrys, Inc., San Diego, CA. Constructing a rudimentary algorithm to assign oodons based on a given frequency can also easily be accomplished with basic mathematical functions by one of ordinary skill.
  • Codon placement in a polynucleotide at an optimized frequency to encode a given polypeptide sequence can also be done in a directed manner.
  • a oodon may be assigned to a particular amino acid so as to create or destroy a restriction enzyme cleavage site. Creation or destruction of restriction enzyme sites may facilitate DNA manipulation by assisting with cloning or forming identifying markers.
  • a oodon may be assigned to a particular amino acid so as to achieve a desired secondary structure of the polynucleotide.
  • an entire coding region encoding a polypeptide sequence, or fragment, variant, or derivative thereof can be oodon optimized by any of the methods described herein or by other methods.
  • Various desired fragments, variants or derivatives may be designed, and each can then be oodon-optimized individually.
  • partially oodon-optimized coding regions of the present invention can be designed and constructed.
  • the invention includes a nucleic acid fragment of a oodon-optimized coding region encoding a polypeptide in which at least about 1%, 2%, 3,% 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% of the oodon positions have been oodon-optimized for a given species.
  • the partially oodon optimized coding regions contain a oodon that is preferentially used in the genes of a desired species, e.g., a mammalian species, e.g., humans, in place of a oodon that is normally used in the native nucleic acid sequence.
  • the present invention is also directed to a pharmaceutical, immunogenic, or vaccine composition
  • a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or fragment, variant or derivative thereof.
  • the adjuvant comprises, consists essentially of, or consists of a combination of a CXCLl 1 polypeptide or fragment, variant, or derivative thereof and a polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, variant, or derivative thereof.
  • the present invention is directed to a pharmaceutical, immunological, or vaccine composition
  • a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 27 to 89 of SEQ ID NO: 2 or a combination of the polypeptide and an isolated CXCLl 1 polynucleotide operably encoding the same.
  • the composition upon administration to an animal, may elicit an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the present invention is directed to a pharmaceutical, immunological, or vaccine composition
  • a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids X to Y of SEQ ID NO: 2 or a combination of the polypeptide and an isolated polynucleotide operably encoding the same, wherein X can be any number selected from the group consisting of 22, 23, 24, 25, 26, and 27 and Y can be any number selected from the group consisting of 89, 90, 91, 92, 93, and 94.
  • the composition upon administration to an animal, may modulate, enhance, or improve relative to administration of the immunogen in the absence of the adjuvant.
  • the amino acid sequence can be amino acids 29 to 89 of SEQ ID NO: 2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO: 2.
  • sequence identity between two polypeptides is determined by comparing the amino acid sequence of one polypeptide to the sequence of a second polypeptide.
  • sequence identity is determined by comparing the amino acid sequence of one polypeptide to the sequence of a second polypeptide.
  • whether any particular polypeptide is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to another polypeptide can be determined using methods and computer programs/ software known in the art such as, but not limited to, the BESTFIT program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711).
  • BESTFIT uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981), to find the best segment of homology between two sequences.
  • BESTFIT or any other sequence alignment program to determine whether a particular sequence is, for example, 95% identical to a reference sequence according to the present invention, the parameters are set such that the percentage of identity is calculated over the full length of the reference polypeptide sequence and that gaps in homology of up to 5% of the total number of amino acids in the reference sequence are allowed.
  • the present invention also includes a pharmaceutical, vaccine, or immunogenic com- position comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2 or a combination of the polypeptide and an isolated polynucleotide opearbly encoding the same.
  • an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20,
  • the CXCLl 1 polypeptide or fragment, derivative, analog, or variant thereof or a polypeptide immunogen comprising an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof described herein may have various alterations such as substitutions, insertions or deletions.
  • amino acids that can be substituted in the polypeptide include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleudne, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleudne) and aromatic side chains ( e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspart
  • a polypeptide in the present invention, can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids ( e.g. non-naturally occurring amino acids).
  • the polypeptides used in the present invention either a polypeptide mmunogen or a CXCLl 1 polypeptide, may be modified by either natural, such as posttranslational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
  • Modifications can occur anywhere in the polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, oovalent attachment of flavin, oovalent attachment of a heme moiety, oovalent attachment of a nucleotide or nucleotide derivative, oovalent attachment of a lipid or lipid derivative, oovalent attachment of phosphotidylinositol, cross-linking, cy- clization, disulfide bond formation, demethylation, formation of oovalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, ⁇ -carboxylation, gly- oosylation, GPI anchor formation, hydroxylation, iodination, methylation, myris- toylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino
  • Some embodiments of the invention involve the use of a CXCLl 1 polypeptide or fragment, derivative, or variant thereof that is fused to a heterologous polypeptide moiety to form a fusion protein.
  • fusion proteins can be used to accomplish various objectives, e.g., increased serum half-life, improved bioavailability, in vivo targeting, improved recombinant expression efficiency, improved host cell secretion, ease of purification, and higher avidity.
  • the heterologous moiety can be inert or biologically active. Also, it can be chosen to be stably fused to the CXCLl 1 polypeptide moiety or to be cleavable, in vitro or in vivo. Heterologous moieties to accomplish these other objectives are known in the art.
  • Pharmacologically active polypeptides such as the CXCLl 1 polypeptides may exhibit rapid in vivo clearance, necessitating large doses to achieve therapeutically effective concentrations in the body.
  • polypeptides smaller than about 60 kDa potentially undergo glomerular filtration, which sometimes leads to nephrotoxicity. Fusion or conjugation of relatively small polypeptides such CXCLI l polypeptides can be employed to reduce or avoid the risk of such nephrotoxicity.
  • Various heterologous amino acid sequences i.e., polypeptide moieties or "carriers," for increasing the in vivo stability, i.e., serum half-life, of therapeutic polypeptides are known. Examples include serum albumins such as, e.g., bovine serum albumin (BSA) or human serum albumin (HSA).
  • HSA human serum albumin
  • HSA can be used to form a fusion protein or polypeptide conjugate that displays pharmacological activity by virtue of the CXCLl 1 polypeptide moiety while displaying significantly increased in vivo stability, e.g., 10-fold to 100-fold higher.
  • the C-terminus of the HSA can be fused to the N-terminus of the CXCLl 1 polypeptide moiety.
  • HSA is a naturally secreted protein
  • the HSA signal sequence can be exploited to obtain secretion of the fusion protein in vivo in an animal, into the cell culture medium when the fusion protein is produced in a eukaryotic, e.g., mammalian, expression system.
  • a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant comprising an isolated polynucleotide which comprises a polynucleotide opearbly encoding the CXCLl 1 polypeptide and a heterologous nucleotide sequence, for example, a nucleotide sequence encoding HAS
  • the CXCLl 1 polypeptides, fragments, derivatives, or variants thereof for use in the methods of the present invention further comprise a targeting moiety.
  • Targeting moieties include a protein or a peptide which directs localization to a certain part of the body.
  • Some embodiments of the invention employ a CXCLl 1 polypeptide or fragment, derivative, or variant thereof fused to Fc region, i.e., the C-terminal portion of an Ig heavy chain constant region.
  • Fc region i.e., the C-terminal portion of an Ig heavy chain constant region.
  • Potential advantages of a CXCLl 1-polypeptide-Fc fusion include solubility, in vivo stability, and multivalency, e.g., dimerization.
  • the Fc region used can be an IgA, IgD, or IgG Fc region (hinge-CH2-CH3). Alternatively, it can be an IgE or IgM Fc region (hinge-CH2-CH3-CH4).
  • IgG Fc region is generally used, e.g., an IgGl Fc region or IgG4 Fc region.
  • Materials and methods for constructing and expressing DNA encoding Fc fusions are known in the art and can be applied to obtain fusions without undue experimentation.
  • Some embodiments of the invention employ a fusion protein such as those described in Capon et al., U.S Patent Nos. 5,428,130 and 5,565,335.
  • a native CXCLl 1 polypeptide, or fragment thereof occurs as a dimer with a CXCLl 1-polypeptide-Fc fusion.
  • the present invention provides an adjuvant comprising consisting essentially of, or consisting of an isolated polypeptide with a first polypeptide fragment and a second polypeptide fragment, where the first polypeptide fragment comprises an amino acid sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% similar to a CXCLI l reference amino acid sequence (SEQ ID NO.:2) and where the second polypeptide fragment comprises a fusion moiety.
  • leader sequence also referred to as a secretory signal peptide.
  • a leader sequence also referred to as a secretory signal peptide.
  • the native CXCLl 1 leader sequence is used, e.g., amino acids 1 to 21 of SEQ IDN O: 2, or a functional derivative of that sequence that retains the ability to direct the secretion of the peptide that is operably associated with it.
  • a heterologous mammalian leader sequence, or a functional derivative thereof may be used.
  • the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator or mouse ⁇ -glucuronidase.
  • the CXCLl 1 polypeptides or the polynucleotide encoding the CXCLl 1 polypeptides administered in the method of the invention can be fused to a polypeptide tag or a nucleotide sequence encoding the tag, respectively.
  • polypeptide tag is intended to mean any sequence of amino acids that can be attached to, connected to, or linked to the CXCLl 1 polypeptide and that can be used to identify, purify, concentrate or isolate the CXCLl 1 polypeptide.
  • the attachment of the polypeptide tag to the CXCLl 1 polypeptide may occur, e.g., by constructing a nucleic acid molecule that comprises: (a) a nucleic acid sequence that encodes the polypeptide tag, and (b) a nucleic acid sequence that encodes a CXCLl 1 polypeptide.
  • Exemplary polypeptide tags include, e.g., amino acid sequences that are capable of being post- translationally modified, e.g., amino acid sequences that are biotinylated.
  • Other Exemplary polypeptide tags include, e.g., amino acid sequences that are capable of being recognized andor bound by an antibody (or fragment thereof) or other specific binding reagent.
  • Polypeptide tags that are capable of being recognized by an antibody (or fragment thereof) or other specific binding reagent include, e.g., those that are known in the art as "epitope tags.”
  • An epitope tag may be a natural or an artificial epitope tag. Natural and artificial epitope tags are known in the art, including, e.g., artificial epitopes such as FLAG, Strep, or poly-histidine peptides.
  • FLAG peptides include the sequence Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys (SEQ ID NO: 9) or Asp- Tyr-Lys-Asp-Glu-Asp-Asp-Lys (SEQ ID NO: 10) (Einhauer, A. and Jungbauer, A., J. Biochem. Biophys. Methods 49:1-3:455-465 (2001)).
  • the Strep epitope has the sequence Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly (SEQ ID NO: 11).
  • the VSV-G epitope can also be used and has the sequence Tyr-
  • Thr-Asp-Ile-Glu-Met-Asn-Arg-Leu-Gly-Lys SEQ ID NO: 12
  • Another artificial epitope is a poly-His sequence having six histidine residues (His-His-His-His-His).
  • Naturally-occurring epitopes include the influenza virus hemagglutinin (HA) sequence Tyr-
  • Pro-Tyr-Asp-Val-Pro-Asp-Tyr-Ala-Ile-Glu-Gly-Arg (SEQ ID NO: 13) reoognized by the monoclonal antibody 12CA5 (Murray et al., Anal. Biochem. 229:170-179 (1995)) and the eleven amino acid sequence from human c-myc (Myc) reoqgnized by the monoclonal antibody 9E10 (Glu-Gln-Lys-Leu-Leu-Ser-Glu-Glu-Asp-Leu-Asn (SEQ ID NO: 14) (Manstein et al., Gene 162:129-134 (1995)).
  • Another useful epitope is the tripeptide Glu-Glu-Phe which is reoqgnized by the monoclonal antibody YL 122. (Stammers et al. FEBS Lett. 283:298-302(1991)).
  • a CXCLl 1 polypeptide and a polypeptide tag may be connected via a linking amino acid sequence.
  • a "linking amino acid sequence” may be an amino acid sequence that is capable of being reocgnized and ⁇ r cleaved by one or more proteases. Amino acid sequences that can be reocgnized and ⁇ r cleaved by one or more proteases are known in the art.
  • Exemplary amino acid sequences are those that are reocgnized by the following proteases: factor Vila, factor IXa, factor Xa, APC, t-PA, u-PA, trypsin, chymotrypsin, enterokinase, pepsin, cathepsin B,H,L,S,D, cathepsin G, renin, angiotensin oonverting enzyme, matrix met- alloproteases (oollagenases, stromelysins, gelatinases), macrophage elastase, Cir, and Cis.
  • the amino acid sequences that are reocgnized by the aforementioned proteases are known in the art.
  • Exemplary sequences reocgnized by certain proteases can be found, e.g., in U.S Patent No. 5,811,252. Also included is a polynucleotide enooding a CXCLl 1 polypeptide and a polynucleotide encoding a polypeptide tag which are connected via a nucleotide sequence enooding a linking amino acid sequence.
  • a CXCLl 1 polypeptide moiety By fusing a CXCLl 1 polypeptide moiety at the amino and ⁇ r carboxy termini of a suitable fusion partner, bivalent or tetravalent forms of a CXCLl 1 polypeptide or polypeptide fragment can be obtained for administration according to the method of the invention.
  • a CXCLl 1 polypeptide moiety can be fused to the amino and carboxy termini of an Ig moiety to produce a bivalent monomeric polypeptide containing two CXCLl 1 polypeptide moieties.
  • a polynucleotide operably enooding a CXCLl 1 polypeptide moiety that are fused to a polynucleotide sequence enooding the amino acid carboxy termini of an Ig moiety so that the expressed polypeptide is a bivalent monomeric polypeptide oontaining two CXCLl 1 polypeptide moieties. Upon dimerization of two of these monomers, by virtue of the Ig moiety, a tetravalent form of a CXCLl 1 polypeptide is obtained. Such multivalent forms can be used to achieve increased binding affinity for the target.
  • Multivalent forms of a CXCLl 1 polypeptide or polypeptide fragment of the invention or the polynucleotide operably encoding the same also can be obtained by placing CXCLl 1 polypeptide moieties in tandem to form ooncatamers, which can be employed alone or fused to a fusion partner such as Ig or HSA.
  • the heterologous polypeptide fused to the CXCLl 1 polypeptide is not a chemokine or a cytokine.
  • the heterologous polypeptide fused to the CXCLl 1 polypeptide is an immunogen comprising an antigenic or immunogenic polypeptide.
  • Some embodiments of the invention involve a CXCLl 1 polypeptide or fragment, analog, derivative, or variant thereof conjugated (covalently linked) to one or more polymers.
  • polymers suitable for such conjugation include polypeptides (discussed above), sugar polymers and polyalkylene glycol chains.
  • a polymer is conjugated to a CXCLl 1 polypeptide or fragment, analog, derivative, or variant thereof for the purpose of improving one or more of the following: solubility, stability, or bioavailability.
  • Conjugated polypeptides may be produced by methods well known for those of oridnary skill in the art.
  • the present invention is directed to a pharmaceutical, immunogenic, or vaccine composition
  • an adjuvant which comprises a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide and an immunogen.
  • the immunogen comprises a polynucleotide encoding an antigenic or immunogenic polypeptide.
  • the immunogen comprise a polypeptide such as an antigenic or immunogenic polypeptide.
  • the immunogen may comprise both an antigenic or immunogenic polypeptide and a polynucleotide encoding an antigenic or immunogenic polypeptide.
  • the polypeptide immunogen may be the same or different than the antigenic or immunogenic polypeptide encoded by the polynucleotide immunogen. Also included in the present invention is an immunogen which comprises an inactivated or attenuated whole cell or virus, e.g., a live- attenuated virus vaccine, a heat-killed virus vaccine, etc.
  • a polynucleotide encoding an antigenic or immunogenic polypeptide may be DNA or RNA, for example, in the form of messanger RNA (mRNA).
  • a polynucleotide immunogen may be, or be included in, a vector or may be in a host cell comprising the vector. Vectors of the invention are described above.
  • a polynucleotide immunogen of the invention can also encode a derivative fusion protein such as those described elsewhere herein, wherein two or more nucleic acid fragments, at least one of which encodes an antigenic or immunogenic polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide.
  • a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment.
  • the polynucleotide encodes an antigenic or immunogenic polypeptide comprising at least one immunogenic epitope, wherein the epitope elicits a B -cell (antibody) response, a T-cell (e.g., CTL) response, or both.
  • Polynucleotide immunogens used in the present invention can be altered from their native state in a manner similar to polynucleotides operably encoding a CXCLl 1 polypeptide such as described elsewhere herein.
  • all or part of the polynucleotide encoding an antigenic or immunogenic polypeptide may be oodon-optimized according to oodon usage in the animal in which the vaccine is to be delivered.
  • nucleic acid or fragment thereof which encodes an antigenic or immunogenic polypeptide can be a fragment which encodes only a portion of a full- length polypeptide, andor can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide.
  • the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell.
  • an immunogen may comprise an isolated polynucleotide encoding an antigenic or immunogenic polypeptide, wherein the antigenic or immunogenic polypeptide is expressed through operable association of the polynucleotide encoding the antigenic or immunogenic polypeptide with a promoter.
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and a polynucleotide operably encoding a CXCLl 1 polypeptide may be situated on the same vector.
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by two copies of different promoters or identical promoters on the the same vector.
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by a single promoter as a bi ⁇ stronic transcript where the coding regions are separated by an internal ribosomal entry site (IRES).
  • IRS internal ribosomal entry site
  • the polynucleotide encoding the antigenic or immunogenic polypeptide and a polynucleotide operably encoding a CXCLl 1 polypeptide may be situated on separate vectors.
  • the immunogen can also be a polypeptide produced in vitro. Such a polypeptide comprises an antigenic or immunogenic polypeptide.
  • an antigenic or immunogenic polypeptide included in the polypeptide immunogen or encoded by a polynucleotide immunogen comprises epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 8 to about 30 amino acids contained within the amino acid sequence of a polypeptide.
  • Certain polypeptides comprises immunogenic or antigenic epitopes of at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length.
  • Antigenic as well as immunogenic epitopes may be linear, i.e., be comprised of contiguous amino acids in a polypeptide, or may be three dimensional, i.e., where an epitope is comprised of non-contiguous amino acids which come together due to the secondary or tertiary structure of the polypeptide, thereby forming an epitope.
  • Polypeptide immunogens may have various modifications or alternations in a manner similar to a CXCLl 1 polypeptide described above.
  • the antigenic or immunogenic polypeptide used as an immunogen or encoded by a polynucleotide used as an immunogen in the present invention can be a viral polypeptide, a bacterial polypeptide, a fungal polypeptide, a parasite polypeptide, an allergenic polypeptide, a tumor- specific polypeptide, fragments, variants, derivatives of any of the polypeptides.
  • viral polypeptide may be derived from, but are not limited to, a virus selected from the group consisting of adenovirus, alphavirus, cali ⁇ virus (e.g., a cali ⁇ virus capsid peptide), coronavirus, distemper virus, Ebola virus, enterovirus, flavivirus, hepatitis virus (A-E)( e.g., a hepatitis B core or surface peptide), her- pesvirus( e.g., a herpes simplex virus or varicella zoster virus glycoprotein peptide), immunodeficiency virus ( e.g., a human immunodeficiency virus envelope or protease peptide), infectious peritonitis virus, influenza virus (e.g., an influenza A hemag- glutinin or neuraminidase peptide), leukemia virus, Marburg virus, onocgenic virus, orthomyxovirus, papilloma virus,
  • a virus
  • bacterial polypeptides may be derived from, but are not limited to, a member of a bacterial genus selected from the group consisting of Actinomyces, Bacillus, Bacteroides, Bordetella, Bartonella, Borrelia (e.g., a B. bergdorferi OspA peptide), Brucella, Campylobacter, Capnocytophaga, Chlamydia, Clostridium, Corynebacterium, Coxiella, Dermatophilus, Enterococcus, Ehrlichia, Escherichia, Fran ⁇ sella, Fusobacterium, Haemobartonella, Haemophilus (e.g., H.
  • a member of a bacterial genus selected from the group consisting of Actinomyces, Bacillus, Bacteroides, Bordetella, Bartonella, Borrelia (e.g., a B. bergdorferi OspA peptide), Bruce
  • influenzae type b outer membrane protein peptides Helicobacter, Klebsiella, L-form bacteria, Leptospira, Listeria, Mycobacteria, Mycoplasma, Neisseria, Neorickettsia, Nocardia, Pasteurella, Peptococcus, Peptostreptococcus, Pneumococcus, Proteus, Pseudomonas, Rickettsia, Rochalimaea, Salmonella, Shigella, Staphylococcus, Streptococcus ( e.g., S pyogenes M protein peptides), Treponema, and Yersinia (e.g., Y pestis Fl and V peptides).
  • S pyogenes M protein peptides e.g., S pyogenes M protein peptides
  • Treponema e.g., Y pestis Fl and V peptides.
  • fungal polypeptides may be derived from, but are not limited to, a member of a fungal genus selected from the group consisting of Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Coc ⁇ dioides, Conidiobolus, Cryptoooccus, Curvalaria, Epidermophyton, Exophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Muoor, Pae ⁇ lomyces, Peni ⁇ llium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhinosporidium, Rhizopus, Scolecobasidium, Sporothrix, Stemphylium, Trichophyton, Trichosporon, and Xylohypha.
  • Examples of protozoan parasite polypeptides may be derived from, but are not limited to, a member of a protozoan parasite genus selected from the group consisting of Babesia, Balantidium, Besnoitia, Cryptosporidium, Eimeri, Encephalitozoon, Entamoeba, Giardia, Hammondia, Hepatozoon, Isospora, Leishmania, Microsporidia, Neospora, Nosema, Pentatrichomonas, Plasmodium (e.g., P.
  • a member of a protozoan parasite genus selected from the group consisting of Babesia, Balantidium, Besnoitia, Cryptosporidium, Eimeri, Encephalitozoon, Entamoeba, Giardia, Hammondia, Hepatozoon, Isospora, Leishmania, Microsporidia, Neospora, Nosema, Pentatrichom
  • PfCSP falciparum ⁇ rcum- sporozoite
  • PfSSP2 sporozoite surface protein 2
  • PfLSA-I c-term carboxyl terminus of liver state peptide 1
  • PfExp-1 exported protein 1
  • helminth parasite polypeptides may be derived from, but are not limited to, a member of a helminth parasite selected from the group consisting of Acan- thocheilonema, Aelurostrongylus, Ancylostoma, Angiostrongylus, Ascaris, Brugia, Bunostomum, Capillaria, Chabertia, Cooperia, Crenosoma, Dictyocaulus, Dioctophyme, Dipetalonema, Diphyllobothrium, Diplydium, Dirofilaria, Dracunculus, Enterobius, Hlaroides, pep tides Haemonchus, Lagochilascaris, Loa, Mansonella, Muellerius, Nanophyetus, Necator, Nematodirus, Oesophagostomum, Onchocerca, Opisthorchis, Ostertagia, Parafilaria, Paragonimus, Parascaris,
  • ectoparasite polypeptides include, but are not limited to, peptides
  • ticks including hard ticks and soft ticks
  • flies such as midges, mosquitos, sand flies, black flies, horse flies, horn flies, deer flies, tsetse flies, stable flies, myiasis-causing flies and biting gnats
  • ants spiders, lice; mites; and true bugs, such as bed bugs and kissing bugs.
  • tumor- associated polypeptides include, but are not limited to, a tumor-specific immunoglobulin variable region, a GM2 peptide, a Tn peptide, an sTn peptide, a Thompson-Friedenreich peptide (TF), a Globo H peptide, an Le(y) peptide, a MUCl peptide, a MUC2 peptide, a MUC3 peptide, a MUC4 peptide, a MUC5AC peptide, a MUC5B peptide, a MUC7 peptide, a car ⁇ noembryonic peptide, a beta chain of human chorionic gonadotropin (hCG beta) peptide, a HERIneu peptide, a PSMA peptide, a EGFRvIII peptide, a KSA peptide, a PSA peptide, a PSCA peptide, a GPlOO peptide
  • fragments or variants of the antigenic or immunogenic polypeptide are also included in the present invention. Additional antigenic or immunogenic polypeptides may be found, for example in "Foundations in Microbiology,” Talaro, et al. , eds., McGraw-Hill Companies (Oct., 1998), Reids, et al. , “Virology,” 3d ed., Lippinoott- Raven (1996), “Biochemistry and Molecular Biology of Parasites", Marr, et al. , eds., Academic Press (1995), and Deacon, J., “Modern Mycology”, Blackwell Science Inc (1997), which are incorporated herein by reference.
  • the polypeptides of the present invention are isolated or purified. No particular level of purification is required. Once a polypeptide has been recombinantly expressed, it may be purified by any method known in the art for pu- rification of a polypeptide, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • the present invention provides a pharmaceutical, immunogenic, vaccine composition and methods for delivery of the composition, where the composition as described herein, comprises an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide or a combination thereof.
  • an immunogen can be either a polypeptide or a polynucleotide or a combination thereof.
  • the adjuvant of the present invention can modulate, enhance, or improve the immune response when administered in an animal.
  • Compositions of the present invention induce an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the invention provides a composition comprising an immunogen and combinatorial adjuvants in which a polynucleotide (e.g., a plasmid or a viral vector expressing a CXCLI l polypeptide) operably encoding a CXCLI l polypeptide, or fragment, derivative, or variant thereof and a CXCLl 1 polypeptide, fragment, derivative, or variant thereof are combined together.
  • a polynucleotide e.g., a plasmid or a viral vector expressing a CXCLI l polypeptide
  • the immunogen used in the present invention comprises both a polynucleotide immunogen and polypeptide immunogen.
  • a single formulation may comprise (i) an immunogen comprising a polynucleotide encoding an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof or a polypeptide comprising the antigenic or immunogenic polypeptide or both polynucleotide and polypeptide and (ii) an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof or a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof or both polypeptide and polynucleotide.
  • the polynucleotide or polypeptide immunogens and a polynucleotide operably encoding CXCLl 1 polypeptide or a CXCLl 1 polypeptide may exist in any form known in the art.
  • the pharmaceutical composition of the present invention includes one or more known additional adjuvants.
  • adjuvant refers to any material having the ability to (1) alter or increase the immune response to a particular antigen or (2) increase or aid an effect of a pharmacological agent. Accordingly, a CXCLl 1 polypeptide, fragment, derivative, or variant thereof or a polynucleotide operably encoding a CXCLI l polypeptide is considered an "adjuvant".
  • This embodiment refers to a pharmaceutical composition comprising one or more additional adjuvants.
  • Suitable adjuvants include, but are not limited to, cytokines, e.g., chemokines, and growth factors; alum, bentonite, latex or acrylic particles, pluronic block polymers, cationic lipids, squalene, depot formers, surface active materials, lysole ⁇ thin, retinal, Quil A, liposomes, pluronic polymer formulations; macrophage stimulators, alternate pathway complement activators, non-ionic surfactants, bacterial components, aluminum-based salts, calcium-based salts, silica, polynucleotides, toxoids, serum proteins, viruses and virally-derived materials, poisons, venoms, imida- zoquiniline compounds, poloxamers, toll-like receptor (TLR) agonists, mLT, CpG, MPL, cationic lipids, Qs21, pluronic polymer formulations, macrophage stimulators, alternate pathway complement activators, non-ionic surfact
  • an adjuvant to increase the immune response to an antigenic or immunogenic polypeptide is typically manifested by a significant increase in immune-mediated protection.
  • an increase in humoral immunity is typically manifested by a significant increase in the titer of antibodies raised to the antigen, and an increase in T-cell activity is typically manifested in increased cell proliferation, or cellular cytotoxicity.
  • An adjuvant may also alter an immune response, for example, by changing a primarily humoral or Th 2 response into a primarily cellular, or Th 1 response.
  • compositions used in the methods of the invention can also be incorporated into the compositions used in the methods of the invention.
  • the composition of the present invention comprising a polynucleotide operably encoding a CXCLI l polypeptide, fragment, variant, or derivative thereof or a CXCLl 1 polypeptide, fragment, variant, or derivative thereof combined with a polynucleotide encoding an antigenic or immunogenic polypeptide, or fragment, variant, or derivative thereof or a polypeptide encoded by the polynucleotide may be additionally formulated andor coadministered with one or more additional therapeutic agents effective to treat, ameliorate or prevent a disease.
  • an adjuvant for example, a polynucleotide sequence operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof andor a CXCLl 1 polypeptide, fragment, derivative, or variant thereof can be combined with, or be included in, a whole cell immunogen as a combination regimen that can elicit an enhanced or improved immune response.
  • the whole-cell immunogen is not a tumor cell.
  • the immunogenic composition of the present invention which includes polynucleotides can also include one or more transfection facilitating agents that facilitate delivery of polynucleotides to the interior of a cell, andor to a desired location within a cell.
  • transfection facilitating materials include, but are not limited to inorganic materials such as calcium phosphate, alum (aluminum sulfate), ampipathic peptides, cationic lipids, neutral lipids, anionic lipids, and gold particles (e.g., "powder” type delivery vehicles); peptides that are, for example, cationic, intercell targeting (for selective delivery to certain cell types), intracell targeting (for nucleor localization or endosomal escape), and ampipathic (helix forming or pore forming); proteins that are, for example, basic (e.g., positively charged) such as histones, targeting (e.g., asialoprotein), viral (e.g., Sendai virus coat protein
  • a transfection facilitating material can be used alone or in combination with one or more other transfection facilitating materials.
  • Two or more transfection facilitating materials can be combined by chemical bonding (e.g., CDvalent and ionic such as in lipidated polylysine, PEGylated polylysine) (Toncheva, et al. , Biochim. Biophys. Acta 1380(3):354-368 (1988)), mechical mixing (e.g., free moving materials in liquid or solid phase such as "polylysine + cationic lipids" (Gao and Huang, Biochemistry 35:1027-1036 (1996); Trubetskoy, et al. ⁇ iochem. Biophys.
  • hydrophobic and amphiphilic additives such as, for example, sterols, fatty acids, gangliosides, glycolipids, lipopeptides, liposaccharides, neobees, niosomes, prostaglandins and sphingolipids, may also be included in the immunogenic compositions of the present invention.
  • these additives may be included in an amount between about 0.1 mol % and about 99.9 mol % (relative to total lipid).
  • these additives comprise about 1-50 mol % and, most preferably, about 2-25 mol %.
  • Preferred additives include lipopeptides, liposaccharides and steroids.
  • the present invention may contain suitable pharmaceutically acceptable carriers comprising ex ⁇ pients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically for delivery to the site of action.
  • suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium car- boxymethyl cellulose, sorbitol and dextran.
  • the suspension may also contain stabilizers.
  • Liposomes can also be used to encapsulate the molecules of this invention for delivery into the cell.
  • Exemplary "pharmaceutically acceptable carriers" are any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • the composition comprises isotonic agents, for example, sugars, polyaloohols such as mannitol, sorbitol, or sodium chloride.
  • the compositions comprise pharmaceutically acceptable substances such as wetting or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the composition of the invention.
  • compositions of the present invention may include various salts, ex ⁇ pients, delivery vehicles andor auxiliary agents as are disclosed, e.g., in U. S Patent Application Publication No. 20020019358, published February 14, 20CE, which is incorporated herein by reference in its entirety.
  • compositions of the present invention can be formulated according to known methods, whereby the substance to be delivered is combined with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their preparation are described, for example, in Remington's Pharmaceutical Sciences. 16th Edition, A. Osol, ed., Mack Publishing Co., Easton, PA (1980), and Remington's Pharmaceutical Sciences. 19th Edition, A.R. Gennaro, ed., Mack Publishing Co., Easton, PA (1995).
  • the pharmaceutical composition can be formulated as an emulsion, gel, solution, suspension, lyophilized form, or any other form known in the art.
  • the pharmaceutical, immunological, or vaccine composition can also contain pharmaceutically acceptable additives including for example, diluents, binders, stabilizers, and preservatives.
  • salts of the nucleic acid molecule constructs described herein are preferred.
  • Such salts can be prepared from pharmaceutically acceptable non-toxic bases including organic bases and inorganic bases.
  • Salts derived from inorganic bases include sodium, potassium, lithium, ammonium, calcium, magnesium, and the like.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, basic amino acids, and the like.
  • R>r aqueous pharmaceutical compositions used in vivo use of sterile pyrogen-free water is preferred.
  • Such formulations will contain an effective amount of the immunogenic composition together with a suitable amount of vehicle in order to prepare pharmaceutically acceptable compositions suitable for administration to a vertebrate.
  • compositions of the invention may be in a variety of forms, including, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions e.g., dispersions or suspensions.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
  • the composition can be formulated as a solution, micro emulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze- drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic add, collagen, polyorthoesters, and polylactic add. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York (1978).
  • the combination of conventional vacdne compositions with the adjuvant of the present invention provides for therapeutically benefidal effects at dose sparing concentrations.
  • immunological responses suffident for a therapeutically benefidal effect in patients predetermined for an approved oommer ⁇ al product, such as for the conventional product described above can be attained by using less of the approved commerdal product when supplemented or enhanced with the appropriate amount of an adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLI l polypeptide.
  • dose sparing is contemplated by administration of conventional vacdnes administered in combination with a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide.
  • the dose of conventional vacdne may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with the adjuvant of the invention.
  • using a combination of conventional and the adjuvant of the present invetion may allow both materials to be used in lesser amounts while still affording the desired level of immune response arising from administration of either component alone in higher amounts (e.g. one may use less of either immunological product when they are used in combination). This may be manifest not only by using lower amounts of materials being delivered at any time, but also to redudng the number of administrations points in a vacdnation regime ( e.g. 2 versus 3 or 4 injections), andor to redudng the kinetics of the immunological response (e.g. desired response levels are attained in 3 weeks in stead of 6 after immunization).
  • the dose of the adjuvant of the present invention may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with an immunogen.
  • an immunogen that does not normally elidt an immune response upon administration to an animal i.e., non-immunogenic antigens
  • one advantage of the adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide used in the present invention is the ability to elidt or greatly enhance an immune response in an animal to a non-immunogenic antigen, for example, an antigen to which the animal is tolerant, when a polynucleotide encoding an antigenic or immunogenic polypeptide or a polypeptide comprising an antigenic or immunogenic polypeptide is GO- administered with the adjuvant of the present invention as described herein.
  • kits for use in immunotherapy comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide in a sterile environment.
  • kits for use in immunotherapy comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide as well as one or more polypeptides as an immunogen or polynucleotides encoding the polypeptides in a sterile environment.
  • an immunogen and an adjuvant used in the present invention may be contained together or separately. In one embodiment, the adjuvant is in the amount of 1 ng to 30 mg.
  • the immunogen and the adjuvant of the present invention may be contained in glass containers, plastic containers, or strips of plastic or paper.
  • the composition is contained in a syringe and administered through a plunger.
  • the composition is administered through a cathether.
  • the composition may further comprise a pharmaceutically acceptable carrier.
  • the kit can further comprise an instruction sheet for administration of the composition into an animal.
  • the components of the pharmaceutical composition are preferably provided as a liquid solution, such as a suspension, a solution, or an emulsion; or in lyophilized form as a dried powder or a cake.
  • the oompositin may be included in a suitable vehicle, such as sterile pyrogen-free water, for reoonstitution of the composition comprising an immunogen and a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide, or any buffer described herein, including PBS, normal saline, Tris buffer, and sodium phosphate vehicle.
  • a suitable vehicle such as sterile pyrogen-free water
  • the container in which the pharmaceutical composition is packaged prior to use can comprise a hermetically sealed container enclosing an amount of the lyophilized formulation or a solution containing the formulation suitable for a pharmaceutically effective dose thereof, or multiples of an effective dose.
  • the pharmaceutical composition is packaged in a sterile container, and the hermetically sealed container is designed to preserve sterility of the pharmaceutical formulation until use.
  • the container can be associated with administration means and or instruction for use.
  • Suitable additional agents that may be coformulated with andor coadministered with an immunogen andor an adjuvant of the present invention are described above in the composition section.
  • the present invention further provides a method for delivering a pharmaceutical, vaccine or immunogenic composition into an animal or a subject, comprising administering to an animal or subject in need of the compositions as described herein.
  • the composition comprising an immunogen and an adjuvant of the present invention induces an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
  • the present invention provides a method of enhancing or modulating an immune response in an animal in need of such an enhanced or modulated immune response, comprising administering to the animal a composition as described herein.
  • the composition contains an adjuvant comprising a polynucleotide encoding an immunogenic or antigenic polypeptide or the polypeptide encoded by the polynucleotide or a combination thereof.
  • the needed immunogenic or antigenic polypeptide is expressed in the animal, in a sufficient amount to induce andor modify a desired immune response in the vertebrate to prevent disease, cure disease, reduce the severity of disease symptoms, or prolong the life of the animal.
  • the present invention provides a method of enhancing or modulating an immune response in a healthy animal for large-scale antibody production, comprising administering to the animal a composition as described herein.
  • the composition contains a polynucleotide operably encoding a CXCLI l polypeptide, fragment, derivative, or variant thereof or a CXCLl 1 polypeptide.
  • the CXCLl 1 polypeptide is expressed or directly introduced in the animal, in a sufficient amount to produce a vigorous antibody response in the animal.
  • the antibodies thus produced are then recovered from the animal by, for example, the collection of serum, milk, or saliva.
  • Such antibodies may be useful for research or diagnostic purposes, or for additional therapies in animals in need of such therapies.
  • passive antibody treatment using antibodies produced by this method may prevent disease, cure disease, reduce the severity of disease symptoms, or prolong the life of an animal.
  • the polynucleotide encoding the CXCLl 1 polypeptide or the CXCLl 1 polypeptide of the present invention when used as an adjuvant, can induce an improved immune response, e.g., increase the B- and T-cell responses to poorly immunogenic polypeptides or polynucleotides encoding the same, influence the quality of the immune response (e.g., change the class of antibody produced or change a humoral response to a cellular response) to a specific immnogenic or antigenic polypeptide, and enable the eli ⁇ tation of an immune response to an immunogen, e.g., a tumor antigen, that would otherwise be impossible due to tolerance.
  • an immunogen e.g., a tumor antigen
  • the polynucleotide operably encoding the CXCLl 1 polypeptide or a CXCLl 1 polypeptide can further improve an immune response elicited by an immunogen, wherein the immune response is improved or enhanced relative to the immune response in the absence of the adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide.
  • the improved or enhanced immune response may be induction of an improved or enhanced humoral immune response.
  • the improved or enhanced humoral immune response may include an improved or enhanced antibody response, e.g., secretion of IgG, IgA, IgE, IgM, or IgD or a combination thereof than that would not have been secreted when a composition is administered to an animal in the absence of the adjuvant of the present invention.
  • the improved B cell response may further include secretion of larger amount of the antibodies than that would have been secreted when a composition is administered to an animal in the absence of the adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide.
  • a distinctive feature of the present invention is that a pharmaceutical, immunogenic, or vaccine composition
  • an immunogen and an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide
  • a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide
  • humoral immune responses i.e., antibody responses, as well as ThI responses
  • the immunogen with other chemokine adjuvants e.g., CXCL4, CXCL9, or CXCLlO.
  • the improved or enhanced immune response may not a cell mediated immune response.
  • the adjuvant when the composition of the present invention comprising an immunogen and an adjuvant is administered in an animal in need thereof, the adjuvant may improve or enhance natural killer T cell or cytotoxic T cell related immune responses.
  • administration of compositions of the present invention may shift a primarily humoral response into a primarily cellular response.
  • natural killer T cells may produce interferon- ⁇ granulocyte- macrophage colony- stimulating factor as well as multiple other cytokines and chemokines such as IL-2 and TNF- ⁇ in improved quantities relative to the administration of the composition in the absence of the adjuvant.
  • Cytotoxic T cell related immune response may include activation and differentiation of CD4+ or CD8+ cells or secretion of cytokines or chemokines, e.g., IL-2, by CD4+ andor CD8+ cells compared to the activation and differentiation of the cells when the composition of the present invention is administered in the absence of the adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide.
  • the improved immune response includes activation, development, recruitment, and/ ⁇ r differentiation of ThI cells, but not Th2 cells.
  • ThI cells may express CXCR3 on its surface to interact with the CXCLl 1 polypeptide.
  • the improved or enhanced immune response comprises recruitment of cells selected from the group consisting of natural killer (NK) cells, Cytotoxic T cells (CTLs), B lymphocytes, dendritic cells, mactrophages, neutrophils, and a combination of two or more of the cells.
  • NK natural killer
  • CTLs Cytotoxic T cells
  • B lymphocytes B lymphocytes
  • dendritic cells mactrophages
  • neutrophils and a combination of two or more of the cells.
  • the improved or enhanced immune response may include secretion of additional cytokines or chemokines or secretion of cytokines or chemokines in larger quantities than would have been secreted without the adjuvant of the present invention.
  • Non-limiting examples of the cytokines or chemokines are Interferons (type I, II and III) including but not limited to Interleukins (IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-I l, IL- 12, IL-13, IL- 14, IL-15, IL- 16, IL- 17, IL-18); interferons (INF- ⁇ , INF- ⁇ , INF- ⁇ ); tumor necrosis factor- ⁇ (TNF- ⁇ ); tumor necrosis factor- ⁇ (TNF- ⁇ ); tumor necrosis factor-c (TNF-c); growth hormone (somatotropin); prolactin; granulocyte-colony stimulating factor (G-CSF); myelomonocytic growth factor; leukemia inhibitory factor (LIF); onoostatin-M; ciliary neurotrophic factor (CNTF); cholinergic differentiation factor (CDF); macrophag
  • the improved or enhanced immune response may refer to reducing the level of certain cytokine or chemokine secretions.
  • the cytokines or chemokines which may be decreased may be IL-4, IL-IO, IL-5, or IL- 13.
  • the CXCLl 1 polypeptide may further elicit an improved immune response to an immunogen that would otherwise induce tolerance.
  • immunogen is tumor specific antigens described herein or viral antigenic or immunogenic polypeptides in patients with chronic infenction.
  • the present invention can also be applied in therapeutic andor prophylactic treatments of diseases, wherein an enhanced or improved immune response can trigger potent self-antigen recognition, such that the response can induce elimination of primary andor secondary attributes of a disease.
  • Increases in self-antigen recognition can be modulated by administration of a polynucleotide opearbly encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof andor a CXCLl 1 polypeptide, or fragment, variant, or derivative thereof, in combination with an immunogen, including but not limited to tumor specific antigens.
  • compositions may be delivered to an animal, e.g., a mammal.
  • the mammal can be a human.
  • compositions of the present invention can be accomplished according to any of various methods known in the art.
  • U.S Patent No. 5,676,954, incorporated herein by reference in its entirety reports on the injection of genetic material, complexed with cationic lipid carriers, into mice.
  • U.S Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and PCT international patent application PCT/US94/06069 (WO 9429469) the disclosures of which are incorporated herein by reference in their entireties, provide methods for delivering compositions comprising naked DNA, or DNA cationic lipid complexes to animals.
  • compositions of the present invention are utilized in a "prime boost” regimen.
  • An example of a “prime boost” regimen may be found in Yang, Z. et al.J. Virol. 77:799-803 (2002), which is incorporated herein by reference in its entirety.
  • one or more polynucleotide or polypeptide vaccine compositions of the present invention are delivered to an animal, thereby priming the immune response of the animal to an immunogen, and then a second composition is utilized as a boost vaccination.
  • compositions of the present invention are used to prime immunity, and then a second composition, e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated polypeptides or fragments, variants or derivatives thereof is used to boost an immune response.
  • a second composition e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated polypeptides or fragments, variants or derivatives thereof is used to boost an immune response.
  • a priming composition and a boosting composition are combined in a single composition or single formulation.
  • a single composition may comprise a polypeptide immunogen and an adjuvant comprising, consisting essentially of, or consisting of a CXCLl 1 polypeptide as the priming component and a polynucleotide immunogen encoding an antigenic and immunogenic polypeptide and an adjuvant comprising a polynucleotide operably encoding a CXCLI l polypeptide as the boosting component.
  • the compositions may be contained in a single vial where the priming component and boosting component are mixed together.
  • the polynucleotide component may provide a boost to the isolated protein component.
  • Compositions comprising both a priming component and a boosting component are referred to herein as "combinatorial vaccine compositions" or “single formulation heterologous prime -boost vacdne compositions.”
  • the priming composition may be administered before the boosting composition, or even after the boosting composition, if the boosting composition is expected to take longer to act.
  • the priming composition may be administered simultaneously with the boosting composition, but in separate formulations where the priming component and the boosting component are separated.
  • priming or “primary” and “boost” or “boosting” as used herein may refer to the initial and subsequent immunizations, respectively, i.e., in accordance with the definitions these terms normally have in immunology. However, in certain embodiments, e.g., where the priming component and boosting component are in a single formulation, initial and subsequent immunizations may not be necessary as both the “prime” and “boost” compositions are administered simultaneously.
  • compositions of the invention are useful for administration to any animal, preferably a mammal (such as apes, cows, horses, pigs, boars, sheep, rodents, goats, dogs, cats, chickens, monkeys, rabbits, ferrets, whales, and dolphins), and more preferably a human.
  • a mammal such as apes, cows, horses, pigs, boars, sheep, rodents, goats, dogs, cats, chickens, monkeys, rabbits, ferrets, whales, and dolphins
  • the immunogenic, pharmaceutical, or vaccine compositions of the present invention may be administered to any tissue of an animal, including, but not limited to, muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, tongue and connective tissue.
  • the immunogenic, pharmaceutical, or vaccine compositions of the present invention can be administered by orally, nasally, parenterally, topically, in- trabronchially, intravenously, subcutaneously, intramuscularly, intravenously, intra- tracheally, intranasally, transdermally, interdermally, subcutaneously, intraocularly, vaginally, rectally, intraperitoneally, intraintestinally, by inhalation, buccally, sub- lingually, vaginally, intraheptically, intracardiac, intrapancreatic, transmucosal (i.e., across a mucous membrane), intra-cavity ( e.g., oral, vaginal, or rectal), and intravenous (i.v.) transplantation, by inhalation or by an implanted pump.
  • the immunogenic compositions of the present invention are administered by intramuscular (i.m.) or subcutaneous (s.c.) routes.
  • Any mode of administration can be used so long as the mode results in the expression of the CXCLl 1 polypeptide and the immunogen in the composition in an amount sufficient to elicit a measurable immune response in an animal.
  • This includes needle injection, electroporation into skin or muscle, catheter infusion, biolistic injectors, particle accelerators (i.e., "gene guns” or pneumatic "needleless” injectors — for example, Med-E-Jet (Vahlsing, H., et al., J. Immunol.
  • EAPD Energy-assisted plasmid delivery
  • Parenteral injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Additionally, one approach for parenteral administration employs the implantation of a slow-release or sustained-released systems, which assures that a constant level of dosage is maintained, according to U.S. Pat. No. 3,710,795, incorporated herein by reference in its entirety.
  • a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide used in the methods of the invention may be directly infused into an organ or tissue.
  • the invention encompasses any suitable delivery method for a composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or a polynucleotide opearbly encoding a CXCLl 1 polypeptide to a selected target tissue, including bolus injection of an aqueous solution or implantation of a oontrolled-release system.
  • a oontrolled-release implant reduces the need for repeat injections.
  • compositions may also comprise an adjuvant comprising, consisting essentially of, or consisting of a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide and an immunogen dispersed in a biocompatible carrier material that functions as a suitable delivery or support system for the compounds.
  • sustained release carriers include semipermeable polymer matrices in the form of shaped articles such as suppositories or capsules.
  • Implantable or microcapsular sustained release matrices include polylactides (U.Si Patent No.
  • a polynucleotide or a polypeptide immunogen can be administered, either simultaneously or sequentially, with a polynucleotide operably encoding a CXCLl 1 polypeptide, or variant, derivative, or fragment thereof or a CXCLl 1 polypeptide, or variant, derivative, or fragment thereof as described herein.
  • a polynucleotide encoding multiple antigenic or immunogenic polypeptides each encoding for one or more antigens or multiple polypeptide im- munogens or antigens can be co-administered, either simultaneously or sequentially, with a polynucleotide operably encoding the CXCLl 1 polypeptide, or variant, derivative or fragment thereof or a CXCLl 1 polypeptide as described herein.
  • the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of viral diseases in human patients including, but not limited to, diseases caused by adenovirus, al- phavirus, cali ⁇ virus, coronavirus, distemper virus, Ebola virus, enterovirus, flavivirus, hepatitis virus (AE), herpesvirus, immunodeficiency virus, infectious peritonitis virus, influenza virus, leukemia virus, Marburg virus, orthomyx) virus, papilloma virus, parainfluenza virus, paramys) virus, parvovirus, pestivirus, picorna virus, pox virus, rabies virus, reovirus, retrovirus, rotavirus, as well as other cancer-causing or cancer- related virus.
  • the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of bacterial diseases in human patients including, but not limited to, diseases caused by Actinomyces, Bacillus, Bacteroides, Bordetella, Bartonella, Borrelia, Brucella, Campylobacter, Capnocytophaga, Chlamydia, Clostridium, Corynebacterium, Coxiella, Dermatophilus, Enterococcus, Ehrlichia, Escherichia, Fran ⁇ sella, Fu- sobacterium, Haemobartonella, Haemophilus, Helicobacter, Klebsiella, L-form bacteria, Leptospira, Listeria, Mycobacteria, Mycoplasma, Neisseria, Neorickettsia, Nocardia, Pasteurella, Peptococcus, Peptostreptococcus, Pneumococcus, Proteus, Pseu
  • the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of parasitic diseases in human patients including but not limited to, diseases caused by Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Coc ⁇ dioides, Conidiobolus, Cryptoooccus, Curvalaria, Epidermophyton, Ejophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Muoor, Pae ⁇ lomyces, Peni ⁇ llium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhi- nosporidium, Rhizopus, Soolecobasidium, Sporothri, Stemphylium, Trichophyton, Tri- chosporon, and X
  • the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of fungal diseases in human patients including, but not limited to, Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Coc ⁇ dioides, Conidiobolus, CryptoGoccus, Curvalaria, Epidermophyton, Ejophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Mu ⁇ or, Pae ⁇ lomyces, Peni ⁇ llium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhinosporidium, Rhizopus, Scolecobasidium, Sporothrix, Stemphylium, Trichophyton, Trichosporon, and Xylohypha.
  • the present invention can also be directed to methods of modulating an immune response in immunologically -based therapeutic andor prophylactic treatments of cancer including, but not limited to, cancers of oral cavity and pharynx (i.e., tongue, mouth, pharynx), digestive system (i.e., esophagus, stomach, small intestine, colon, rectum, anus, anal canal, anorectum, liver, gallbladder, pancreas), respiratory system (i.e., larynx, lung), bones, joints, soft tissues (including heart), skin, melanoma, breast, reproductive organs (i.e., cervix, endometirum, ovary, vulva, vagina, prostate, testis, penis), urinary system (i.e., urinary bladder, kidney, ureter, and other urinary organs), eye, brain, endocrine system (i.e., thyroid and other endocrine), lymphoma (i.e., B
  • Antigens to include in an immunogenic composition of the present invention are described, e.g., in Chamberlain, Drugs 57(3):309-25 (March, 1999), Nawrocki and Mackiewicz, Cancer Treat Rev 25(l):29-46 (Feb.,1999), Rosenberg, Immunity 10(3):281-7 (Mar., 1999), Rosenberg et al. , Adv Caner Res 70:145-77 (1996), Pandey et al. , Eur J Surg Oncol 25(2):209-14 (Apr., 1999), Herlyn et al. , Ann Med. 31(l):66-78 (Feg., 1999), and Rosenberg Immunol Today 18(4): 175-82 (Apr., 1997), which are incorporated herein by reference.
  • An additional embodiment of the present invention is directed to combining a composition of the present invention where the immunogen is a polynucleotide operably encoding, or a polypeptide comprising, a tumor antigen with one or more additional cancer therapies including, but not limited to bone marrow transplant, cord blood cell transplant, surgery, chemotherapy, radiation therapy, and immunotherapy.
  • additional cancer therapies including, but not limited to bone marrow transplant, cord blood cell transplant, surgery, chemotherapy, radiation therapy, and immunotherapy.
  • the polynucleotide, nucleic acid molecule construct(s), immunogenic composition or pharmaceutical composition of the present invention can be administered prior to the commencement of one or more of the additional cancer therapies, during the practice of one or more of the additional cancer therapies, and after the end of one or more of the additional cancer therapies.
  • Types of bone marrow transplant include, but are not limited to autologous bone marrow transplant and heterologous (i.e., from a donor) bone marrow transplant.
  • Types of surgery include, but are not limited to surgery for breast cancer, prostate cancer, colon cancer, brain cancer, and head and neck cancer.
  • Chemotherapeutic agents include, but are not limited to alkylating agents, including mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil, di- carbazine, streptazo ⁇ ne, carmustine, lomustine, semustine, chlorozoto ⁇ n, busulfan, triethylenemelamine, thiotepa, hexamethylmelamine; antimetabolites, including methotrexate; pyrimidine analogs, including fluoroura ⁇ l, 5-fluoroura ⁇ l, floxuridine (5'-fluoro-2'-deoxyuridine), idoxuridine, cytarabine, N-phosphonoacetyl-L-aspartate, 5-azacytidine, azaribine, 6-azauridine, pyrazofuran, 3-deazauridine, a ⁇ vi ⁇ n; purine analogs, including thioguanine, mercaptopurine, aza
  • An additional embodiment of the present invention is directed to combining any one of the viral disease treatment methods of the present invention with one or more ad- ditional viral therapies including, but not limited to anti- viral drugs, such as nucleoside analogs, immuloglobulin therapy and interferons.
  • anti-viral drugs include, but not limited to, Acyclovir, Gancyclovir, Ibscarnet, Zidovudine, Lamivudin [3TC], Ribavirin, and Amantadine, protease inhibitors, and a combination of one or more antiviral drugs.
  • An additional embodiment of the present invention is directed to combining any one of the bacterial infection treatment methods of the present invention with one or more additional bacterial infection treatments including, but not limited to, treatment with antibiotics, such as vancomycin, daptomy ⁇ n, cefotetan, chloramphenicol, penicillin, tetracyclin, trimethoprim, rifampin, clarithromycin, ciprofloxacin, erythromycin, azithromycin, metronidazole, trimethoprim, isoniazid, ciprofloxacin and clarithromycin.
  • antibiotics such as vancomycin, daptomy ⁇ n, cefotetan, chloramphenicol, penicillin, tetracyclin, trimethoprim, rifampin, clarithromycin, ciprofloxacin, erythromycin, azithromycin, metronidazole, trimethoprim, isoniazid, ciprofloxacin and clarithromycin.
  • An additional embodiment of the present invention is directed to combining any one of the fungal infection treatment methods of the present invention with one or more additional fungal infection treatments including, but not limited to, treatment with antifungal drugs, such as amphotericin B, caspofungin, fluconazole, flucytosine, intra- oonazole, ketoconazole and voriconazole.
  • antifungal drugs such as amphotericin B, caspofungin, fluconazole, flucytosine, intra- oonazole, ketoconazole and voriconazole.
  • An additional embodiment of the present invention is directed to combining any one of the parasitic infection treatment methods of the present invention with one or more additional parasitic infection treatments including, but not limited to, treatment with anti-parasitic drugs, chloroquine, doxycycline, a combination of atovaquone and proguanil or mefloquine, mefloquine and quinidine.
  • Determining an effective amount of an immunogenic composition depends upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the subject, the precise condition requiring treatment and its severity, and the route of administration. Based on the above factors, determining the precise amount, number of doses, and timing of doses are within the ordinary skill in the art and will be readily determined by the attending physician or veterinarian.
  • compositions of the invention may include a "therapeutically effective amount” or a “prophylactically effective amount” of a composition of the invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, e.g., modulating an immune response or elicits an improved immune response.
  • a therapeutically effective amount of the composition comprising an immunogen and an adjuvant of the present invention may vary according to factors such as the disease state, age, sex, and weight of the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • the pharmaceutical, immunologic, or vaccine composition of the present invention may be administered as a single dose, multiple doses or over an established period of time in an infusion. Dosage regimens also may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response).
  • a polynucleotide or vector operably encoding a CXCLl 1 polypeptide, fragment, derivative, or variant thereof or a CXCLl 1 polypeptide, fragment, derivative, or variant thereof may be administered to an animal once per day, one week out of the month, continuously ( e.g., by osmotic pump) or intermittently.
  • parenteral compositions in dosage unit form for ease of administration and uniformity of dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated, each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the immunogen and the adjuvant, i.e., the polynucleotide operably encoding the CXCLI l polypeptide and the CXCLl 1 polypeptide or CXCLl 1 protein and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an immunogen or an adjuvant of the present invention for the treatment of sensitivity in individuals.
  • a therapeutically effective dose range for an immunogen depends on the types or characteristics of the immunogen as well as manner of administration and types of an animal or subject administered thereto.
  • a therapeutically effective dose range for the CXCLl 1 polypeptide or a polynucleotide encoding the CXCLI l polypeptide is 0.001 - 10 mg/Kg per day.
  • a therapeutically effective dose range for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLI l polypeptide is 0.01 - 1 r ⁇ g/Kg per day.
  • a therapeutically effective dose range for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLI l polypeptide is 0.05-0.5 mg/Kg per day. In some embodiments a therapeutically effective dose range for for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLl 1 polypeptide is 0.05-0.2 mg/Kg per day. In some embodiments a therapeutically effective dose range for for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLl 1 polypeptide is 0.001-0.5 mg/Kg per day.
  • the dosage can range, e.g., from about 0.0001 to 100 mg/kg, from about 0.001 to 0.5 mg/kg, or 0.01 to 5 mg/kg ( e.g., 0.02 mgkg, 0.25 mgAcg, 0.5 mgAcg, 0.75 mgAcg, lmgAcg, 2 mg/kg, etc.), of the host body weight.
  • R>r example dosages can be 1 mgkg body weight or 10 mg/kg body weight or within the range of 1-10 mgAcg, preferably at least 1 mg/kg. Doses intermediate in the above ranges are also intended to be within the scope of the invention.
  • Subjects can be administered such doses daily, on alternative days, weekly or according to any other schedule determined by empirical analysis.
  • An exemplary treatment entails administration in multiple dosages over a prolonged period, for example, of at least six months. Additional exemplary treatment regimes entail administration once per every two weeks or once a month or once every 3 to 6 months.
  • Exemplary dosage schedules include 1-10 mg/kg or 15 mg/kg on consecutive days, 30 mgAcg on alternate days or 60 mgAcg weekly.
  • the immunogenic composition comprises an immunogen: adjuvant molar ratio.
  • the molar ratio ranges from about 100:1 to about 1:100; from about 50:1 to about 1:50; from about 30:1 to about 1:30; from about 25:1 to about 1:25, from about 20:1 to about 1:20; from about 19:1 to about 1:19; from about 18:1 to about 1:18; from about 17:1 to about 1:17; from about 16:1 to about 1:16; from about 15:1 to about 1:15; from about 14:1 to about 1:14; from about 13:1 to about 1:13; from about 12:1 to about 1:12; from about 11:1 to about 1:11; from about 10:1 to about 1:10; from about 9:1 to about 1:9; from about 8:1 to about 1:8; from about 7:1 to about 1:7; from about 6:1 to about 1:6; from about 5:1 to about 1:5; from about 4:1 to about 1:4;
  • CXCLl 1 SEQ ID NO:8
  • a polynucleotide encoding the murine CXCLl 1 was custom synthesized using TOP Gene Technologies ( www.topgenetech.corrO. and cloned into the Aspl 18 and Xbal sites of pShuttle-tet-10 mammalian expression vector.
  • the vector, pShuttle-tet-10 was constructed by inserting a custom synthesized (TOP Gene Technologies) tet operator sequence (5' TCC CTA TCA GTG ATA GAG ATC TCC CTA TCA GTG ATA GAG ATC GTC GAC GAG CTC-3' at the 3' end of the cytomegalovirus (CMV) promoter (SEQ ID NO.: 15) in the pShuttle 10 vector (Clontech).
  • TOP Gene Technologies TCC CTA TCA GTG ATA GAG ATC TCC CTA TCA GTG ATA GAG ATC GTC GAC GAG CTC-3' at the 3' end of the cytomegalovirus (CMV) promoter (SEQ ID NO.: 15) in the pShuttle 10 vector (Clontech).
  • mice were maintained at the animal facility of POSTECH under standard conditions according to the institutional Guidelines. All experiments were performed according to the guidelines of the institutional laboratory animal resources committee.
  • mice groups The immunization schedule of mice groups is shown in Hg. 1.
  • PBS phosphate-buffered saline
  • Electroporation was carried out using BTX electroporator (Mode: LV; voltage: 100V; P. length: 2OmS; No. of pulses: 6; interval between pulses: 1.0 sec; polarity: unipolar).
  • a booster immunization was performed the same way using the same doses of plasmid DNAs.
  • Serum samples from immunized mice were collected on the 21 st day and 35 th day by retroorbital bleeding and ELISA was performed to evaluate any modulatory effects of CXCLl 1 on the immune response to OVA.
  • Microtiter plates (Nalgene Nunc International, NY) were coated at 4 0 C for overnight with 1 ⁇ g/m# of OVA protein (purchased from Sigma- Aldrich Co.) in PBS Ibllowing morning, the plates were washed with PBS containing 0.05% tween and blocked at room temperature for 1 hour with 5% skim milk in 0.05% PBST. Serum samples collected from immunized mice were diluted in 5% milk in 0.05% PBST at 1:50 dilution added to the plates and incubated for 2 hours at 37 0 C. R>r endpoint dilution, individual serum samples were pooled per group and pooled serum was serially diluted before adding to the plate.
  • OVA protein purchased from Sigma- Aldrich Co.
  • 96- well filtration plates (Millpore, Bedford, MA) were coated with 3 ⁇ g/m# rat anti- mouse INF- ⁇ Ab (BD Pharmingen, San Diego, CA). After overnight incubation at room temperature, plates were washed twice with PBS and blocked with RPMI containing 10% FBS for 1 hour at 37 0 C.
  • Splenocyte cells (10 6 ) from 3-7 mice were harvested on the 21 st or 35 th day after prime or booster immunization (depending on the experiment), and re- suspended in complete medium (RPMI 1640 containing 10% FBS, 50 ⁇ m 2-ME, 2mM glutamine, IOOU of penicillin/ m-6, 100 ⁇ g of streptomy ⁇ n/m#), and applied into plates with or without OVA CD4 + and OVA CD8 + epitopes (these epitopes were synthesized by Peptron, Korea (www.peptron.CDm).
  • the fragments of OVA used for generation OVA::CD4 and OVA::CD8 fusions were OVA 323-339 and O VA 257 _ 264 respectively, and were previously described by Chang et ah, J. Immunol. 172:2818-2826 (2004)).
  • the plates were washed and 50 j ⁇ of 2 ⁇ g/m# biotinylated rat anti-mouse INF- ⁇ Ab was added (BD Pharmingen). After incubation at room temperature for 3 hours, the plates were washed, and 50 j ⁇ of 1:2000 diluted streptavidin-alkaline phosphatase (BD Pharmingen) was added. After 45 minutes of incubation, the plates were washed.
  • mice were co-immunized twice (as depicted schematically in Hg. 1) with plasmid pShuttle-tet-10-mCXCR3-ligands in combination with pGXIO-OVA, and 2 weeks after the final immunization (booster immunization) seven mice per group were sacrificed and their splenocytes were harvested.
  • Hg. 4 depicts that the level of IFN- ⁇ secreting CD4 + T cells, from mice GO- administered with plasmid DNAs expressing CXCL4 and OVA, was modestly decreased as compared to control mice, injected only with OVA expressing plasmid (Gl).
  • IFN- ⁇ secreting CD4 + T cells from the mice co-administered with plasmid DNAs expressing CXCL9, or CXCLlO, or CXCLl 1 and OVA significantly enhanced the production of the CD4 + T cells as compared to control mice, injected only with OVA expressing plasmid (Gl) (Hg. 4).
  • CXCLl 1 showed the highest capability for inducing INF-g secreting CD4 + T cells to the OVA antigen.
  • OVA-spe ⁇ fic CD8 + T cell responses were induced by each group (FIG. 1) administered with combination of OVA and a CXCR3 ligand expressing plasmids
  • OVA-spe ⁇ fic intracellular IFN- ⁇ staining and ELISPOT assays were performed using isolated splenocytes, 2 weeks after final (booster) immunization. At 2 weeks after booster immunization, seven mice per group were sacrificed and their splenocytes were harvested.
  • an IFN- ⁇ ELISPOT assay was performed in response to OVA CD8 + epitope stimulator.
  • IFN- ⁇ secreting cells ISCs
  • the data represent the average value and standard deviation of three independent experiments. As shown in Hg. 5, a co-injection of CXCLl 1 chemokine and OVA-enooded plasmids elicited a significant change in the production of IFN- ⁇ in response to OVA DNA vaccination.
  • the GO- administration of the plasmid DNA encoding CXCL4, or CXCL9, or CXCLlO, or CXCLl 1 with pGXIO-OVA also enhanced the level of IFN- ⁇ secreting CD8 + T cells.
  • Splenocytes were harvested from spleens of mice on the 35 th day after final (booster) immunization.
  • FACS analysis of splenocytes was performed. 2x10 6 cells in lm-6 of complete media/tube cells were stimulated by OVA CD8 + epitope and treated with BFA (100Ox) for 6 hours. The cells were then washed with FACS buffer [1% Fetal Bovine Serum (FBS), 0.09% sodium azide in PBS] and stained with Antigen presenting cell (APC)-oonjugated anti-CD8 mAb (BD Pharmingen, San Diego, CA).
  • FBS Fetal Bovine Serum
  • APC Antigen presenting cell
  • the cells were once again washed with FACS buffer and treated with 200 j ⁇ of lysing solution (BD Pharmingen, San Diego, CA) and incubated overnight at 4 0 C. Subsequently, the cells were washed with washing buffer (0.5% saponin in FACS buffer), and stained with PE-conjugated IFN- ⁇ mAb, CD8-APCs mAb, IL-2 mAb, and TNF- ⁇ mAb (BD Pharmingen, San Diego, CA), while incubating at 4 0 C for 30 minutes.
  • washing buffer (0.5% saponin in FACS buffer
  • splenocytes were isolated from seven mice per group, 2 weeks after boosting, and examined by FACS analysis. Total 3x10 6 splenocyte cells for each tube were stained with CD8-APCs mAb and PE-conjugated with TNF- ⁇ mAb (a) and IL-2 mAb (b), and were analyzed by FACS The data represent the average value and standard deviation of three independent experiments. As shown in Hg. 7a, co- administration of CXCLl 1 chemokine DNA with the OVA DNA vaccine could dramatically enhance TNF- ⁇ production, which is consistent with the results of INF- ⁇ staining described above.
  • CXCLl 1 a polynucleotide encoding the murine CXCLl 1 (mCXCLl 1) was custom synthesized using TOP Gene Technologies (www.topgenetech.com) and cloned into the Asp! IS and Xbal sites of pShuttle-tet-10 mammalian expression vector.
  • the vector, pShuttle-tet-10 was constructed by inserting a custom synthesized (TOP Gene Technologies) tet operator sequence (5'TCC CTA TCA GTG ATA GAG ATC TCC CTA TCA GTG ATA GAG ATC GTC GAC GAG CTC-3' at the 3' end of the cytomegalovirus (CMV) promoter in the pShuttle 10 vector (Clontech).
  • TOP Gene Technologies tet operator sequence
  • Recombinant replication-defective adenovirus expressing CXCLl 1 plus mouse Fc was generated according to the AdEasy Vector System (QBiogene, Carlsbad, CA). After recombination of pShuttle-tet- 10-CXCLl 1-Fc with the adenoviral backbone vector, pAdEsay in Escherichia coli BJ5183, the recombinant adenovirus expressing CXCLl 1-Fc were generated and expanded in 293 cells.
  • HiTrap recombinant protein A FF (Amersham bios ⁇ ences, Piscataway) columns were equilibrated with PBS (pH 7.0). The filtered supernatants were added to the columns and eluted with 0.1M sodium citrate (pH 3.0). The eluted proteins were finally obtained after dialysis with membrane (MWCO 12 14K, Spectrapor, Collinso Dominguez) more than three times. The concentration of a recombinant protein sample was determined by anti-mouse CXCLl 1/I-TAC antibody (R & D system, Minneapolis, cat#.
  • AF572 as a capture Ab
  • bioninylated anti-mouse CXCLl 1/I-TAC antibody R & D system, Minneapolis, cat#. BAF572
  • detection Ab R & D system, Minneapolis, cat#. BAF572
  • recombinant mouse CXCLl 1/I-TAC R & D system, Minneapolis, cat#. 572-MC
  • mice 5-6 weeks of age, were purchased from Japan SLC (Shizuoka, Japan). The mice were maintained at the animal facility of POSTECH under standard conditions according to the institutional Guidelines. All experiments were performed according to the guidelines of the institutional laboratory animal resources committee.
  • mice groups The immunization schedule of mice groups is shown in Hg. 8.
  • PBS phosphate-buffered saline
  • lour weeks after the 1 st immunization a booster immunization was performed the same way using the same doses of TIV and CXCLl 1-Fc.
  • Serum samples from immunized mice were collected on the 49 ⁇ day by retroorbital bleeding and ELISA was performed to evaluate any modulatory effects of CXCLl 1 on the immune response against influenza HA protein.
  • 96-well filtration plates (Millpore, Bedford, MA) were coated with 3 ⁇ g/m# rat anti- mouse INF- ⁇ Ab (BD Pharmingen, San Diego, CA). After overnight incubation at room temperature, plates were washed twice with PBS and blocked with RPMI containing 10% FBS for 1 hour at 37 0 C.
  • Splenocyte cells (10 6 ) from 6 mice in each group were harvested on the 49 th day after prime or booster immunization (depending on the experiment), and resuspended in complete medium (RPMI 1640 containing 10% FBS, 50 /M 2-ME, 2mM glutamine, IOOU of penicillinM, lOO ⁇ g of streptomy ⁇ n/m#), and applied into plates with or without HA CD4 + epitope (amino acids, SFERFEIFPKE) (see Haberman et al., J of Immunol.
  • mice were immunized twice (as depicted schematically in Hg. 8) with trivalent influenza vac ⁇ ne(TIV) in combination with or without recombinant CXCLl 1-Fc, and 3 weeks after the final immunization (booster immunization) all six mice per group were sacrificed and their splenocytes were harvested.
  • the effects of CD- administration of CXCLl 1 with TIV on the generation of HA spe ⁇ fic-CD4 + and CD8 + T cells were examined by ex vivo IFN- ⁇ ELISPOT assay.
  • Results were expressed as the number of IFN- ⁇ secreting cells (ISCs)/10 6 cells (Rg. 9). The data represent the average value and standard deviation.
  • Total IxIO 6 splenocytes cells were prepared, and each CD4 + epitope or CD8 + epitope of HA protein was used as a stimulator in ELISPOT assay.
  • Hg. 9 depicts that the significant IFN- ⁇ secreting CD4 + and CD8 + T cells were generated by oo-delivery of trivalent influenza vaccine (TIV) with CXCLl 1-Fc although TIV alone oould not induce antigen-specific T cell responses.
  • TIV trivalent influenza vaccine

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention is directed to modulating an immune response in an animal comprising administering a vaccine or pharmaceutical composition comprising an immunogen and an adjuvant which comprises a polynucleotide comprising one or more regions of nucleic acid operably encoding a CXCL11 polypeptide or fragment, variant, or derivative thereof. The present invention is further directed to modulating an immune response in an animal comprising administering a vaccine or pharmaceutical composition comprising an immunogen and an adjuvant which comprises a CXCL11 polypeptide or fragment, variant, or derivative thereof. The immune response elicited by administration of the composition of the present invention is improved relative to administration of the immunogen in the absence of the adjuvant:

Description

Description
CXCLIl ADJUVANT COMPOSITIONS AND USES THEREOF
Technical Field
[1] The present invention relates generally to adjuvants, vacdne compositions, and methods useful for vaccination to modulate immune response. In particular, the present invention provides use of a polynucleotide operably encoding a CXCLl 1 polypeptide or fragments, variant, or derivatives thereof or a CXCLl 1 polypeptide as an adjuvant.
[2]
Background Art
[3] Chemokines or chemoattractant cytokines are a family of small (8-10 kDa) proteins that play an important role in recruiting and activating leukocytes. See Luster et al. N. Engl. J. Med. 338: 436-445 (1998). Chemokines induce their biological effects by binding to seven trans-membrane-spanning G protein-coupled receptors. See id. Approximately 50 chemokines and about 16 different chemokine receptors interacting with the chemokines have been reported. Chemokines are subdivided into four main subclasses, designated as C, CC, CXC, and CX3C, according to the presence and position of the conserved cysteine motifs. Rossi, Annu Rev Immunol. 18: 217-42 (2000); D'Ambrosio et al., J. Immunol. Meth. 273: 3-13 (2003). In addition to chemotatic functions, chemokines play important roles in various activities including maintaining homeostasis, angiogenesis/angiostasis, cellular differentiation and activation, lymphocyte homing, and influencing the overall Thl/Th2 balance of the immune responses. See D'Ambrosio et al., J. Immunol. Meth. 273: 3-13 (2003); Sallusto et al, J. Exp. Med. 187(6): 875-883 (1998).
[4] Chemokine receptors often recognize more than one chemokine and, alternatively, several chemokines can bind to multiple receptors. R>r instance, CXCR3 binds CXCL4 (PF4 or platelet factor 4), CXCL9 (monokine-induced by γ-interferon (Mig)), CXCLlO (interferon-inducible protein 10 (IPlO)), and CXCLI l (interferon-inducible T cell- chemoattractant (1-TAC)). Farber, Biochem. Biophysic. Res. Comm. 192(1): 223-230 (1993); Loestcher et al. J. Exp. Med. 184:963-969 (1996); Murphy et al. Pharm. Rev. 52: 145-176 (2000). In particular, CXCL4 interacts selectively with a splice variant of the CXCR3 receptor, i.e., CXCR3-B while CXCL9, CXCLlO, and CXCLl 1 interact with both receptor variants. Lasagni et al. J. Exp. Med. 197(11): 1537-49 (2003). In particular, CXCLl 1 is regulated by interferon and has potent chemoattractant activity for interleukin-2 (IL-2) activated T cells, but not for freshly isolated unstimulated T cells, neutrophils, or monocytes. Cole et al, J. Exp. Med. 187: 2009-2(El (1998).
[5] While CXCL9, CXCLlO, and CXCLl 1 are three non-Glu-Leu-Arg CXC chemokines that are more closely related to each other, these ligands appear to mediate distinct biological phenomena in vivo. See Hancock et al. J. Exp. Med. 193:975-980 (2001); Khan et al. Immunity 12: 483-494 (2000); and Zhang et al. J. Immunol. 168: 3205-3212 (2002). This may be related to differential expression of these ligands as has been seen in cardiac and skin allograft rejection ( See Hancock et al. J. Exp. Med. 193:975-980 (2001) and Zhang et al. J. Immunol. 168: 3205-3212 (2002)), atherosclerosis (Mach et al., J. Clin. Investig. 104: 1041-1050 (1999)), host response to infection (Amichay et al., J. Immunol. 157: 4511-4520 (1996)), and inflammatory skin disease pier et al., J. Pathol. 194: 398-405 (2001)). Alternatively, it was reported that the ligands of CXCR3 can preferentially activate distinct internalization pathways. Colvin et al., J. Biol. Chem. 279: 30219-30227 (2004). CXCLlO and CXCL9 predominantly induce a carboxyl- terminal dependent pathway, whereas CXCLl 1 predominantly induces a carboxyl-terminal independent pathway. Id.
[6] While many chemokines share similar functions, only some chemokines are recognized as having an adjuvant effect. Pinto et al., Cell. Immunol. 224: 106-113 (2003). It is also known that augmentation of immune response by chemokines is clearly not linked to the receptor specificity of the chemokines. See id. Therefore, studies exploring the use of chemokines as adjuvants for vaccines have shown inconsistent results. See Yu et al., Clin. Exp. Immunol. 115: 335-341 (1999); Xin et al., Clin. Immunol. 92: 90-96 (1999); Kim et al, J. Clin. Invest. 102: 1112-1124 (1998); Sn et al, J. Virol 74: 11173-11180 (2000). R>r example, Yoon et al showed that while CCL5, CXCL8, and CXCLlO induce enhanced ThI -type or humoral immune responses, there was no change in the immune responses induced by a DNA vaccine with CCL4. Immunol. 120: 182-191 (2006). All of the above references are herein incorporated by reference in their entireties.
[7]
Disclosure of Invention Technical Problem
[8] There remains a need in the art for convenient, safe, and efficacious adjuvants to modulate immune responses in an animal. The present invention provides a simple and safe yet effective adjuvant and methods to modulate an immune response using the adjuvant, i.e., a CXCLl 1 polypeptide or a polynucleotide encoding CXCLl 1. [9]
Technical Solution
[10] The present invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant selected from the group consisting of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, an isolated CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO:2, and a combination of the polypeptide and polynucleotide, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
[11] In one embodiment, the invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant, where the adjuvant comprises an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant; or a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant, where the adjuvant comprises an isolated CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
[12] The CXCLl 1 polypeptide may comprise, e.g., at least 45 or at least 70 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO:2. In addition, the CXCLl 1 polypeptide may comprise amino acids 27 to 89 of SEQ ID NO:2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO:2.
[13] In another embodiment, the instant invention is directed to a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen and an adjuvant selected from the group consisting of an isolated polynucleotide which operably encodes a CXCLl 1 polypeptide comprising an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2, an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2, and a combination of the polypeptide and polynucleotide, wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant. Also included is a method for modulating an immune response in an animal, comprising administering to an animal in need thereof a composition comprising an immunogen, and an adjuvant which comprises an isolated polynucleotide operably encoding a CXCLl 1 polypeptide, which comprises an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: 2 , wherein the immune response elicited by administration of the composition is improved relative to administration of the immunogen in the absence of the adjuvant.
[14] The amino acid sequence may be, e.g., at least 90% or at least 95% identical to amino acids 27 to 89 of SEQ ID NO: 2. The amino acid sequence may further be, e.g., identical to amino acids 27 to 89 of SEQ ID NO: 2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO: 2.
[15] The polynucleotide encoding the CXCLl 1 polypeptide can be DNA or RNA such as messenger RNA (mRNA). The polynucleotide may be incorporated into a vector such as a plasmid or a viral vector. The vector can be incorporated into a cell. In a certain embodiment, the cell in which the vector is incorporated is not a tumor cell.
[16] In another embodiment, the method of modulating an immune response in an animal used in the present invention comprises administering a composition comprising an immunogen and an adjuvant comprising an isolated polynucleotide encoding the CXCLl 1 polypeptide, where the polynucleotide further comprises a heterologous nucleic acid. The heterologous nucleic acid may encode a heterologous polypeptide fused to the CXCLl 1 polypeptide. A CXCLl 1 polypeptide used as an adjuvant in the present invention may further comprise a heterologous polypeptide fused thereto. The heterologous polypeptide may be, for example, but not by way of limitation, an antigen, an immunoglobulin Fc region, and a secretory signal peptide. In one embodiment, the heterologous polypeptide is not a chemokine.
[17] In the method of the present invention, an immunogen may be, but not by way of limitation, a polypeptide or a polynucletide encoding a polypeptide, or both. In certain embodiments, the immunogen comprises an isolated polynucleotide which encodes an antigenic or immunogenic polypeptide or fragment, variant, or derivative thereof. In one aspect of the invention, a polynucleotide immunogen and a polynucleotide which operably encodes the CXCLl 1 polypeptide are situated on the same vector. In certain embodiments, the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by two copies of different promoters or identical promoters on the same vector. Alternatively, the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by a single promoter as a biάstronic transcript where the coding regions are separated by an internal ribosomal entry site (IRES).
[18] In other embodiments, the polynucleotide immunogen and the polynucleotide which operably encodes the CXCLI l polypeptide are situated on separate vectors.
[19] A polynucleotide immunogen encoding an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof and the polynucleotide operably encoding the CXCLI l polypeptide may be administered simultaneously. Alternatively, the polynucleotide immunogen may be administered prior to the polynucleotide encoding the CXCLl 1 polypeptide or after the polynucleotide encoding the CXCLl 1 polypeptide. In certain embodiments, the polynucleotide which encodes the CXCLI l polypeptide and a polynucleotide immunogen may be administered at a ratio of about 20: 1 to about 1:20, about 10:1 to about 1:10, or about 6:1. In a certain embodiment, the composition used in the method of the present invention may further comprise an additional adjuvant or a transfection facilitating agent.
[20] In certain embodiments, an improved immune response elicited by the methods of the present invention may comprise an increased antibody response, for example, an IgG response, or an increased T-cell response such as a cytotoxic T lymphocyte (CTL) response. The improved immune response may be an improved ThI response relative to administration of the immunogen alone. The improved immune response may comprise increased serum levels of a cytokine or recruitment of cells selected from the group consisting of NK cells, CTLs, B lymphocytes, dendritic cells, macrophages, neutrophils, and a combination of two or more of the cells. In certain embodiments, the improved immune response may comprise immunogen dose sparing. The dose sparing comprises achieving a comparable immune response to administration of the immunogen alone using reduced amount or reduced doses of the immunogen.
[21] Also included are a pharmaceutical, immunogenic, or vaccine compositions for use in the methods of the present invention. Brief Description of the Drawings
[22] FIG. 1 depicts a schematic diagram of the schedule of administration of plasmids encoding OVA antigen with and without CXC chemokines including CXCLl 1. Also shown are the treatment conditions of control groups. [23] FIG. 2 depicts the level of total IgG in mice immunized with plasmids encoding antigen with or without chemokines at 3 weeks after priming.
[24] FIGS. 3A, 3B and 3C depict the results of total IgG, IgG2a, and IgGl levels in mice immunized with plasmids encoding antigen with or without chemokines at 2 weeks after boosting.
[25] FIG.4 depicts the effect of the chemokine adjuvants on CD4+ T cell responses.
[26] FIG.5 depicts the effect of the chemokine adjuvants on CD8+ T cell response in term of IFN-γ production.
[27] FIG. 6 depicts FACS analysis of OVA-speάfic CD8 + T cell responses, mediated by
CXCR3 chemokines, assessed by staining for intracellular IFN-γ.
[28] FIGS. 7A and 7B depict FACS analysis of CD8 + T cell responses, mediated by CXC chemokines, assessed by staining for intracellular TNF-α and IL-2.
[29] FiG. 8 depicts a schematic diagram of the schedule of administration of the trivalent influenza vaccine with or without the recombinant CXCLl 1-Fc protein.
[30] FIG. 9 depicts the adjuvant effect of the recombinant CXCLl 1-Fc protein on CD8 + and CD4+ T cell responses in Balb/c mice model.
[31] FIGS. 1OA and 1OB depict the results of end-point titration with anti-HA(H5Nl) antibodies to examine the adjuvant effect of the recombinant CXCLl 1-Fc protein for the induction of heterosubtypic immunity against influenza virus. Best Mode for Carrying Out the Invention
[32] It will be apparent to one skilled in the art, in view of the following detailed description and the claims appended hereto, that various substitutions and modifications may be made to the present invention without departing from the scope of the invention as claimed.
[33]
[34] Definitions
[35] It is to be noted that the term "a" or "an" entity refers to one or more of that entity; for example, "a polynucleotide", is understood to represent one or more polynucleotides. As such, the terms "a" (or "an"), "one or more", and "at least one" can be used interchangeably herein.
[36] Throughout this specification and claims, the word "comprise," or variations such as
"comprises" or "comprising," indicate the inclusion of any recited elements or group of elements in the specified method, structure, or composition but not the exclusion of any other elements or group of elements.
[37] As used herein, the term "consists of," or variations such as "consist of" or "consisting of," as used throughout the specification and claims, indicate the inclusion of any recited elements or group of elements, but that no additional elements or group of elements may be added to the specified method, structure or composition.
[38] As used herein, the term "consists essentially of," or variations such as "consist essentially of" or "consisting essentially of," as used throughout the specification and claims, indicate the inclusion of any recited elements or group of elements, and the optional inclusion of any recited elements or group of elements that do not materially change the basic or novel properties of the specified method, structure or composition.
[39] The terms "nucleic acid" or "nucleic acid fragment" refers to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide or construct. Two or more nucleic acids of the present invention can be present in a single polynucleotide construct, e.g., on a single plasmid, or in separate (non-identical) polynucleotide constructs, e.g., on separate plasmids. Furthermore, any nucleic acid or nucleic acid fragment may encode a single polypeptide, e.g., a single antigen, cytokine, or regulatory polypeptide, or may encode more than one polypeptide, e.g., a nucleic acid may encode two or more polypeptides. In addition, a nucleic acid may encode a regulatory element such as, e.g., a promoter or a transcription terminator, or may encode a specialized element or motif of a polypeptide or protein, such as a secretory signal peptide or a functional domain.
[40] The term "polynucleotide" or "polynucleotides" is intended to encompass a single nucleic acid or nucleic acid fragment as well as plural nucleic acids or nucleic acid fragments, and refers to an isolated molecule or construct, e.g., a virus genome (e.g., a non-infectious viral genome), messenger RNA (mRNA), plasmid DNA (pDNA), or derivatives of pDNA (e.g., miniάrcles as described in (Darquet, A-M et al., Gene Therapy 4:1341-1349 (1997)) comprising a polynucleotide. A polynucleotide may be provided in linear (e.g., mRNA), circular ( e.g., plasmid), or branched form as well as double-stranded or single-stranded forms. A linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease. A polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond ( e.g., an amide bond, such as found in peptide nucleic acids (PNA)). The term "polynucleotide" or "polynucleotides" are used interchangeably with the terms "nucleotide," "nucleotides," "nucleic acid," "nucleic acids," "nucleotide sequence," "polynucleotide sequence," and "nucleic acid sequence."
[41] As used herein, the term "operably encoding" refers to operable transcription of a given coding region to mRNA and translation of the mRNA to a polypeptide by a polynucleotide sequence comprising the coding region, transcriptional regulatory sequences, e.g., a promoter, andor translational regulatory sequences, e.g., a translation initiation factor, that are necessary for the transcription and translation of the coding region. Non-limiting examples of the nucleic acid sequences operably encoding CXCLI l polypeptides are described herein, and additional sequences are known in the art. R>r example, the nucleic acid sequences may be obtained by chemical synthesis or by reverse-transcription of a messenger RNA (mRNA) corresponding to CXCLl 1 to a complementary DNA (cDNA) and converting the latter into a double stranded cDNA.
[42] As used herein, the term "polypeptide" is intended to encompass a singular
"polypeptide" as well as plural "polypeptides", and comprises any chain or chains of two or more amino acids. Thus, as used herein, terms including, but not limited to "peptide", "dipeptide", "tripeptide", "protein", "amino acid chain", or any other term used to refer to a chain or chains of two or more amino acids, are included in the definition of a "polypeptide", and the term "polypeptide" may be used instead of, or interchangeably with any of these terms. The term further includes polypeptides which have undergone post- translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
[43] The term "CXCLl 1 polypeptide" as used herein, encompasses full length CXCLl 1, mature CXCLl 1, or other variants of full length CXCLl 1, fragments of full length CXCLl 1, or allelic and other variants of fragments of full length CXCLl 1, derivatives of full-length CXCLl 1, derivatives of fragments of full-length CXCLl 1, analogues of full-length CXCLl 1, analogues of fragments of full-length CXCLl 1, and chimeric and fusion polypeptides comprising full length CXCLl 1 or one or more fragments of full length CXCLI l.
[44] The terms "CXCLI l polynucleotide" or "CXCLI l nucleic acid," as used herein, encompass any nucleic acid sequence operably encoding CXCLI l polypeptides as described above.
[45] The terms "fragment", "analog", "derivative", or "variant" when referring to the
CXCLl 1 polypeptides of the present invention include polypeptides which retain at least some of the activity of the CXCLl 1 polypeptide, e.g, modulating an immune response to an immunogen. One example of the CXCLl 1 polypeptide activities is its binding affinity to CXCR3 to transduce its signals. Hence, the fragment, analog, derivative, or variant of the CXCLl 1 polypeptide of the present invention may retain at least some affinity to CXCR3. The affinity may be the same as, or lesser than, the full length CXCLI l polypeptide. In another aspect, the fragment, analog, derivative, or variant of the CXCLl 1 polypeptides of the present invention retains, at least to a certain extent, the ability to attract CD4+ or CD8+ lymphocytes in the area. The fragment, analog, derivative, or variant of the CXCLl 1 polypeptides of the present invention may induce an antibody response or induce cytokine secretion. Fragments of the CXCLl 1 polypeptides of the present invention include proteolytic fragments, deletion fragments and in particular, fragments of CXCLl 1 polypeptides which exhibit increased solubility during expression, purification, and or administration to an animal. Polypeptide fragments further include any portion of the polypeptide which comprises a portion of the native polypeptide that binds to CXCR3, including linear as well as three-dimensional portions.
[46] An "epitopic fragment" of a polypeptide is a portion of the polypeptide that contains an epitope. An "epitopic fragment" may, but need not, contain amino acid sequence in addition to one or more epitopes as defined below.
[47] The term "variant", as used herein, refers to a polypeptide that differs from the recited polypeptide due to amino acid substitutions, deletions, insertions, andor modifications. Variants may occur naturally, such as an allelic variant.
[48] Non-naturally occurring variants may be produced using art-known mutagenesis techniques. In a preferred embodiment, variant polypeptides differ from an identified sequence by substitution, deletion or addition of five amino acids or fewer. Such variants may generally be identified by modifying a polypeptide sequence, and evaluating the antigenic properties of the modified polypeptide using, for example, the representative procedures described herein.
[49] Polypeptide variants preferably exhibit at least about 60-70%, for example 75%,
80%, 85%, 90%, 94%, 95%, 96%, 97%, 98%, 99% or 99.9% sequence identity with identified polypeptides. Variant polypeptides may comprise conservative or non- conservative amino acid substitutions, deletions or additions. Derivatives of the CXCLl 1 polypeptides of the present invention, are polypeptides which have been altered so as to exhibit additional features not found on the native polypeptide. Examples include fusion proteins. An analog is another form of the CXCLl 1 polypeptide of the present invention. An example is a proprotein which can be activated by cleavage of the proprotein to produce an active mature polypeptide.
[50] Variants may also, or alternatively, contain other modifications, whereby, for example, a polypeptide or polynucleotide may be conjugated or coupled, e.g., fused to a heterologous polypeptide or polynucletoide, e.g., a signal (or leader) sequence at the N-terminal end of the protein which oo-translationally or post-translationally directs transfer of the protein. The polypeptide or polynucleotide may also be conjugated or produced coupled to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g., 6-His), or to enhance binding of the polypeptide to a solid support or to regulate the expression of the CXCLl 1 polypeptide. R>r example, a polypeptide or polynucleotide may be conjugated or coupled to an immunoglobulin Fc region or a nucleotide encoding the same. The polypeptide may also be conjugated or coupled to a sequence that imparts or modulates the immune response to the polypeptide (e.g. a T-cell epitope or B-cell epitope) andor enhances uptake and/ or processing of the polypeptide by antigen presenting cells or other immune system cells. In one embodiment, the sequence conjugated to the CXCLl 1 polypeptide may not be another cytokine or chemokine. The polypeptide may also be conjugated or coupled to other polypeptides/epitopes from tumor, bacteria andor viruses to generate a hybrid immunogenic protein that alone or in combination with various adjuvants can elicit protective immunity to other pathogenic organisms.
[51] The terms "epitope" or "epitopies", as used herein, refers to portions of a polypeptide having antigenic or immunogenic activity in an animal, for example a mammal, for example, a human. An "immunogenic epitope", as used herein, is defined as a portion of a protein that elicits an immune response in an animal, as determined by any method known in the art. The term "antigenic epitope", as used herein, is defined as a portion of a protein to which an antibody or T-cell receptor can immunospeάfically bind as determined by any method well known in the art. Immunospeάfic binding excludes non-specific binding but does not necessarily exclude cross-reactivity with other antigens. Whereas all immunogenic epitopes are antigenic, antigenic epitopes need not be immunogenic.
[52] The terms "immunogen" or "immunogens," as used herein, encompass an antigenic or immunogenic polypeptide or a polynucleotide which operably encodes an antigenic or immunogenic polypeptide. Non-limiting examples of immunogens are described herein below.
[53] As used herein, an "antigenic polypeptide" or an "immunogenic polypeptide" is a polypeptide which, when introduced into an animal, reacts with the animal's immune system molecules, i.e., is antigenic, andor induces an immune response in the animal, i.e., is immunogenic. It is quite likely that an immunogenic polypeptide will also be antigenic, but an antigenic polypeptide, because of its size or conformation, may not necessarily be immunogenic. Isolated antigenic and immunogenic polypeptides of the present invention in addition to the CXCLl 1 polypeptide, may be provided as a recombinant protein, a purified subunit, a viral vector expressing the protein, or may be provided in the form of an inactivated whole cell vaccine, e.g., a live-attenuated virus vaccine, a heat-killed virus vaccine, etc.
[54] As used herein, a " coding region" is a portion of nucleic acid which consists of oodons translated into amino acids. Although a "stop oodon" (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, and the like, are outside the coding region.
[55] The term "oodon optimization" is defined herein as modifying a nucleic acid sequence for enhanced expression in the cells of the host of interest by replacing at least one, more than one, or a significant number, of oodons of the native sequence with oodons that are more frequently or most frequently used in the genes of that host. Various species exhibit particular bias for certain oodons of a particular amino acid. Methods of oodon optimization are well known to those of ordinary skill in the art.
[56] As used herein, the term "isolated" means that the polynucleotide or polypeptide or fragment, variant, or derivative thereof has been removed from other biological materials with which it is naturally associated. An example of an isolated polynucleotide is a recombinant polynucleotide contained in a vector. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically.
[57] As used herein, the term "purified" means that the polynucleotide or polypeptide or fragment, variant, or derivative thereof is substantially free of other biological material with which it is naturally associated, or free from other biological materials derived, e.g., from a recombinant host cell that has been genetically engineered to express the polypeptide of the invention. R>r example, a purified polypeptide of the present invention includes a polypeptide that is at least 70-100% pure, i.e., a polypeptide which is present in a composition wherein the polypeptide constitutes 70-100% by weight of the total composition. In some embodiments, the purified polypeptide of the present invention is 75%-99% by weight pure, 80%-99% by weight pure, 90-99% by weight pure, or 95% to 99% by weight pure. An example of an isolated polynucleotide is a recombinant polynucleotide contained in a vector. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically. The relative degree of purity of a polynucleotide or polypeptide of the invention is easily determined by well-known methods.
[58] The term "pharmaceutically acceptable" refers to components of a composition that are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity or other complications commensurate with a reasonable benefit/risk ratio. In some embodiments, the polypeptide, polynucleotides, compositions, and vaccines of the present invention are pharmaceutically acceptable.
[59] An "effective amount" is the amount of a polypeptide, polynucleotide, composition or vaccine, when administered an individual either in a single dose or as part of a series, that is effective for treatment or prevention. An amount is effective, for example, when its administration results in changes in an immune response, for example, humoral or cell-mediated. This amount varies depending upon the health and physical condition of the individual to be treated, the taxmomic group of individual to be treated (e.g. human, nonhuman primate, primate, etc.), the responsive capacity of the individual's immune system, the degree of protection desired, the formulation of the vaccine, a professional assessment of the medical situation, and other relevant factors. It is expected that the effective amount will fall in a relatively broad range that can be determined through routine trials. Typically a single dose of a polypeptide, polynucleotide, composition or vaccine is from about 10μg to 10 mgAcg body weight. The term "peptide vaccine" or "subunit vaccine" refers to a composition comprising one or more polypeptide immunogens, e.g., antigenic polypeptides or fragments thereof or antigen, which when administered to an animal are useful in stimulating an immune response. The term "DNA vaccine" or "polynucleotide vaccine" refers to a composition comprising one or more polynucleotide immunogens, e.g., a polynucleotide encoding an antigenic polypeptide or fragment thereof or antigen.
[60] The term "subject" is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, immunization, or therapy is desired. Mammalian subjects include, but are not limited to, humans, domestic animals, farm animals, zoo animals such as bears, sport animals, pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, bears, cows; primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as oows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and so on. In certain embodiments, the animal is a human subject.
[61] The term "animal" is intended to encompass a singular "animal" as well as plural
"animals" and comprises mammals and birds, as well as fish, reptiles, and amphibians. The term animal also encompasses model animals, e.g., disease model animals. In some embodiments, the term animal includes valuable animals, either economically or otherwise, e.g., economically important breeding stock, racing animals, show animals, heirloom animals, rare or endangered animals, or companion animals. In particular, the mammal can be a human subject, a food animal or a companion animal.
[62] As used herein, an "subject or animal in need thereof" refers to a subject or animal for whom it is desirable to treat, i.e., to prevent, cure, retard, or reduce the severity of disease symptoms, andor result in no worsening of the diseases over a specified period of time.
[63] The terms "priming" or "primary" and "boost" or "boosting" as used herein to refer to the initial and subsequent immunizations, respectively, i.e., in accordance with the definitions these terms normally have in immunology.
[64] Polynucleotides
[65] The present invention includes pharmaceutical, immunogenic, or vaccine compositions comprising an immunogen and an adjuvant which comprises an isolated polynucleotide operably encoding a CXCLI l polypeptide, fragment, derivative, analog or variant thereof, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[66] Included within the scope of the invention is a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to a reference nucleotide sequence encoding amino acids 27 to 89 of SEQ ID NO: 2, wherein the polynucleotide sequence encodes a polypeptide, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[67] The present invention is also directed to a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence which encodes an amino acid sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acids 27 to 89 of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[68] In certain embodiments, the present invention is directed to a pharmaceutical, im- munnogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide comprising a nucleotide sequence which operably encodes a CXCLI l polypeptide comprising an amino acid sequence least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to amino acids X to Y of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant. X can be any number selected from the group consisting of 22, 23, 24, 25, or 26 and Y can be any number selected from the group 89, 90, 91, 92, 93, or 94.
[69] The present invention also includes a pharmaceutical, vaccine, or immunogenic composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2, wherein the composition, upon administration to an animal, elicits an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[70] The full-length human CXCLl 1 polypeptide is 94 amino acids residues, including a secretory signal peptide sequence of about amino acids 1 to 21. The CXCLl 1 polypeptide is also called small inducible cytokine BI l, Interferon-inducible T-cell α chemoattractant (I-TAC), Interferon-γ inducible protein 9 (IP-9), H174, and Beta-Rl. The gene encoding the CXCLl 1 polypeptide is called ITAC, SCYBl 1, and SCYB9B. The gene encoding the CXCLl 1 polypeptide and its encoded amino acid sequence are described in PCT Publication No. WO 99/13082, incorporated herein by reference in its entirety. [71] The following polynucleotide sequence was reported as a nucleotide sequence encoding the human CXCLl 1 polypeptide sequence and has the accession number NM
005409 in Genbank.
[72] Full- Length Human CXCLl 1 Polynucleotides: 282nt (SEQ ID NO: 1):
[73] ATGAGTGTGA AGGGCATGGC TATAGCCTTG GCTGTGATAT
TGTGTGCTAC [74] AGTTGTTCAA GGCTTCCCCA TGTTCAAAAG AGGACGCTGT
CTTTGCATAG [75] GCCCTGGGGT AAAAGCAGTG AAAGTGGCAG ATATTGAGAA
AGCCTCCATA [76] ATGTACCCAA GTAACAACTG TGACAAAATA GAAGTGATTA
TTACCCTGAA [77] AGAAAATAAA GGACAACGAT GCCTAAATCC CAAATCGAAG
CAAGCAAGGC
[78] TTATAATCAA AAAAGTTGAA AGAAAGAATT TT
[79] [80] The following polypeptide sequence was reported as the human CXCLl 1 sequence and has the accession number NP_005400 in Genbank. [81] Full-Length Human CXCLl 1 Polypeptides: 94aa (SEQ ID NO:2):
[82] MSVKGMAIAL AVILCATVVQ GFPMFKRGRC LCIGPGVKAV KVADIEKASI
[83] MYPSNNCDKi EVIITLKENK GQRCLNPKSK QARLIIKKVE RKNF
[84] Variants of human CXCLl 1 include, but are not limited to, the polypeptides with a mutation, e.g., V89A, an insertion, e.g., K78NRAS, or a deletion, or a combination thereof, e.g., NKGQ69-72IRK. [85] The following polynucleotide sequence was reported as a nucleotide sequence encoding the rhesus monkey CXCLl 1 polypeptide sequence and has the accession number AAK95956 in Genbank.
[86] Full-length Monkey CXCLl 1 Polynucleotides: 285nts (SEQ ID NO: 3)
[87] atgagtgtga agggpatggp tatagpctta gptatgatat tgtgtactac
[88] ggttgttcaa ggtttcccta tgttcaaaag aggacgptgt ctttgpatag
[89] gpcctggagt aaaagpagtg aaagtggpag atattgagaa agpctccata
[90] atttacccaa gtaacaactg tgacaaaata gaagtgatta ttaccctgaa
[91] agaaaataaa ggacaacgat gpctaaatcc caaatcgaag caagpaagac [92] ttataatcaa aaaagttgaa agaaagaatt tttaa
[93]
[94] The following polypeptide sequence was reported as the rhesus monkey CXCLl 1 sequence and has the accession number NP_001028122 in Genbank. [95] Full-length Monkey CXCLl 1 Polypeptides: 94aa (SEQ ID NO: 4)
[96] MSVKGMAIAL AMILCTTVVQ GFPMFKRGRC LCIGPGVKAV KVADIEKASI
[97] iYPSNNCDKi EVIITLKENK GQRCLNPKSK QARLIIKKVE RKNF
[98]
[99] The following polynucleotide sequence was reported as a polynucleotide sequence encoding the chimpanzee CXCLI l polypeptide sequence and has the accession number XM_001151687.
[100] Full-length Chimpanzee CXCLl 1 Polynucleotides: 285nts (SEQ ID NO: 5) [101] atgagtgtga agggpatggp tatagpcttg gptgtgatat tgtgtgptac [102] agttgttcaa ggpttcccca tgttcaaaag aggacgptgt ctttgpatag [103] gpcctggggt aaaagpagtg aaagtggpag atattgagaa agpctccata [104] atgtacccaa gtaacaactg tgacaaaata gaagtgatta ttaccctgaa [105] agaaaataaa ggacaacgat gpctaaatcc caaatcgaag caagpaaggp [106] ttataatcaa aaaagttgaa agaaagaatt tttaa [107] [108] The following polypeptide sequence was reported as the chimpanzee CXCLl 1 sequence and has the accession number XP_001151744. [109] Full-length Chimpanzee CXCL Polypeptides: 94aa (SEQ ID NO: 6) [110] MSVKGMAIAL AVILCATVVQ GFPMFKRGRC LCIGPGVKAV KVADIEKASI [111] MYPSNNCDKI EVIITLKENK GQRCLNPKSK QARLIIKKVE RKNF [112] [113] The following polynucleotide sequence was reported as a polynucleotide sequence encoding the mouse CXCLl 1 polypeptide sequence and has the accession number NM
019494.
[114] Full-Length Mouse CXCLl 1 Polynucleotides: 300nt (SEQ ID NO: 7) [115] ATGAACAGGA AGGTCACAGC CATAGCCCTG GCTGCGATCA
TCTGGGCCAC [116] AGCTGCTCAA GGCTTCCTTA TGTTCAAACA GGGGCGCTGT
CTTTGCATCG [117] GCCCCGGGAT GAAAGCCGTC AAAATGGCAG AGATCGAGAA
AGCTTCTGTA [118] ATTTACCCGA GTAACGGCTG CGACAAAGTT GAAGTGATTG TTACTATGAA
[119] GGCTCATAAA CGACAAAGGT GCCTGGACCC CAGATCCAAG CAAGCTCGCC
[120] TCATAATGCA GGCAATAGAA AAAAAGAATT TTTTAAGGCG TCAAAACATG
[121]
[122] The following polypeptide sequence was reported as the mouse CXCLl 1 sequence and has the accession number NP_062367 in Genbank.
[123] Full-Length Mouse CXCLl 1 Polypeptides: lOOaa (SEQ ID NO:8):
[124] MNRKVTAIAL AAIIWATAAQ GFLMFKQGRC LCIGPGMKAV KMAEIEKASV
[125] IYPSNGCDKV EVIVTMKAHK RQRCLDPRSK QARLIMQAIE KKNFLRRQNM
[126]
[127] The domains for human CXCLl 1 are identified as follows: signal sequence (about amino acids 1 to 21), small cytokine CXC sequence (about amino acids 27 to 89), and C-terminal sequence (about amino acids 90 to 94). See smart.embl.de/smart/show motifs.pl?ID=O14625 (visited July 5, 2007). Alternatively, the domains for human CXCLl 1 may be identified as follows: transdomain sequence (about amino acids 5 to 22 of SEQ ID NO: 2) and small cytokine CXC sequence (about 23 to 89 of SEQ ID NO: 2). See smart.embl.de/smart/show motifs.pl?ID=O14625 (visited July 5, 2007). As one of skill in the art will appreciate, the beginning and ending residues of the domains listed below may vary depending upon the computer modeling program used or the method used for determining the domain.
[128] A polynucleotide of the invention can also operably encode a derivative fusion protein, wherein two or more nucleic acid fragments, at least one of which encodes a CXCLl 1 polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide, e.g., immunoglobulin Fc fused to CXCLl 1. Additionally, a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment. In one embodiment, the heterologous nucleic acid or nucleotide sequence may be a regulatory element such as, as a way of non-limiting example, a promoter, enhancer, or terminator sequence. In certain embodiments, the present invention includes a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof, where the polynucleotide further comprises a heterologous nucleic acid or fragment thereof which encodes a heterologous polypeptide fused to the CXCLl 1 polypeptide, and where the heterologous polynucleotide comprises the immunogen which encodes an immunogenic or antigenic polypeptide. Compositions may further comprise an additional heterologous polypeptide or polynucleotide immunogen. The polynucleotide of the present invention fused to a heterologous nucleic acid or nucleotide sequence may induce an improved immune response relative to administration of the immunogen or fragment thereof in the absence of the polynucleotide operably encoding a CXCLl 1 polypeptide or fragment thereof.
[129] A polynucleotide of the present invention operably encoding a CXCLl 1 polypeptide may be a circular or linearized plasmid or vector, or other linear DNA which may also be non-infectious and nonintegrating (i.e., does not integrate into the genome of vertebrate cells). A linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease.
[130] The present invention includes a pharmaceutical, immunological, or vaccine composition comprising an immunogen, either polynucleotide or polypeptide, and an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or fragment, derivative, analog, or variant thereof, wherein the composition modulate, enhance, or improve an immune response, wherein the polynucleotide is contained in, or is, a vector. Vectors of the present invention may also be used to produce an adjuvant comprising a CXCLI l polypeptide, fragment, derivative, or variant thereof for use in the compositions and methods of the invention. The choice of vector and expression control sequences to which such nucleic acids are operably linked depends on the functional properties desired, e.g., protein expression in vivo, and in host cell to be expressed.
[131] Alternatively, viral genomes, e.g., DNA or RNA virus genes or RNA, may be used to administer polynucleotides into vertebrate cells. In certain embodiments, a virus genome of the present invention is nonreplicative, noninfectious, andύr nonintegrating. In certain embodiments, the virus genome may include viral vectors such as an adenoviral vector, an alphavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, an Epstein Barr viral vector, a papovaviral vector, a poxvirus vector, a vaccinia viral vector, or an adeno- assoάated viral vector. References citing methods for the in vivo introduction of non- infectious virus genomes to vertebrate tissues are well known to those of ordinary skill in the art, and are cited supra.
[132] In other embodiments, a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA). Methods for introducing RNA sequences into vertebrate cells are described in, for example, U.Si Patent No. 5,580,859, the disclosure of which is incorporated herein by reference in its entirety.
[133] In certain embodiments, the present invention incudes a method of modulating an immune response in an animal comprising administering an immunogen and an adjuvant as described above, where the adjuvant, and optionally the immunogen, is/are contained in a host cell. The host cell may be a cell derived from the animal to which the composition is to be administered. In other embodiments, the host cell may be derived from immune related cells, for example, dendritic cells, macrophages, B- lymphocytes, microglia in the brain, or Kupffer cells in the liver. In certain embodiments, a host cell is a non tumor cell. Alternatively, a host cell may be an inactivated bacterial or parasitic cell used as a vaccine. Nucleic acids and fragments thereof of the present invention can be altered from their native state in one or more ways. R>r example, a nucleic acid or fragment thereof which operably encodes a CXCLl 1 polypeptide can be a fragment which encodes only a portion of a full-length polypeptide, andor can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide. R>r example, the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell.
[134]
[135] Vectors
[136] Vectors comprising a polynucleotide operably encoding CXCLl 1 polypeptides may also be used for the methods of the invention. The choice of vector and expression control sequences to which such nucleic acids are operably linked depends on the functional properties desired, e.g., protein expression, and the host cell to be transformed.
[137] Expression control elements useful for regulating the expression of an operably linked coding sequence are known in the art. Examples include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements. When an inducible promoter is used, it can be controlled, e.g., by a change in nutrient status, or a change in temperature, in the host cell medium. [138] Any expression vector which is capable of eliciting expression in eukaryotic cells may be used in the present invention. Examples of suitable vectors include, but are not limited to plasmids pcDNA3, pHCMVEeo, pCR3.1, pEFl/His, pIND/GS, pRc/ HCMV2, pSV40£eo2, pTRACER-HCMV, pUB6/V5-His, pVAXl, and pZeoSV2 (available from Invitrogen, San Diego, CA), and plasmid pCI (available from Promega, Madison, WI). Additional eukaryotic cell expression vectors are known in the art and are commercially available. Typically, such vectors contain convenient restriction sites for insertion of the desired DNA segment. Exemplary vectors include pSVL and pKSV-10 (Pharmacia), pBPV-1, pml2d (International Biotechnologies), pTDTl (ATCC 31255), retroviral expression vector pMIG and pLL3.7, adenovirus shuttle vector pDC315, and AAV vectors. Other exemplary vector systems are disclosed e.g., in U.S. Patent 6,413,777.
[139] Frequently used regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and enhancers derived from retroviral LTRs, cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdmlP)), polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters. For further description of viral regulatory elements, and sequences thereof, see e.g., Stinski, U.S Pat. No. 5,168,062; Bell, U.S Pat. No. 4,510,245, and Schaffner, U.S Pat. No. 4,968,615.
[140] The recombinant expression vectors may carry sequences that regulate replication of the vector in host cells {e.g., origins of replication) andor selectable marker genes. When the expression vector is directly administered in vivo to an animal, a selectable marker gene may not be used. When the expression vector is incorporated into a host cell in vitro, selectable marker genes are often used to facilitate selection of host cells into which the vector has been introduced (see, e.g., Axel, U.S Pat. Nos. 4,399,216; 4,634,665 and 5,179,017). For example, typically the selectable marker gene confers resistance to a drug, such as G418, hygromyάn or methotrexate, on a host cell into which the vector has been introduced. Frequently used selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
[141] In certain embodiments, vectors may include viral vectors such as an adenoviral vector, an alphavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, an Epstein Barr viral vector, a pa- povaviral vector, a poxvirus vector, a vaccinia viral vector, or an adeno- associated viral vector. The viral vector can be a replication-defective viral vector. Adenoviral vectors that have a deletion in its El gene or E3 gene may be used. When an adenoviral vector is used, the vector usually does not have a selectable marker gene.
[142] The vector can include a prokaryotic replioon, i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extra-chromosomally in a bacterial host cell. Such replioons are well known in the art. In addition, vectors that include a prokaryotic replioon may also include a gene whose expression confers a detectable marker such as a drug resistance. Examples of bacterial drug-resistance genes are those that confer resistance to ampiάllin or tetracycline.
[143] Vectors that include a prokaryotic replicon can also include a prokaryotic or bacteriophage promoter for directing expression of the coding gene sequences in a bacterial host cell. Promoter sequences compatible with bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment to be expressed. Examples of such plasmid vectors are pUC8, pUC9, pBR322 and pBR329 (BioRad), pPL and pKK223 (Pharmacia). Any suitable prokaryotic host can be used to express a recombinant DNA molecule encoding a protein used in the methods of the invention.
[144] R>r the purposes of this invention, numerous expression vector systems may be employed. R>r example, one class of vector involves the use of polyάstronic systems with internal ribosome binding sites. Additionally, cells which have integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow selection of transfected host cells. The marker may provide for pro- totrophy to an autotrophic host, bioάde resistance ( e.g., antibiotics) or resistance to heavy metals such as copper. The selectable marker gene can either be directly linked to the DNA sequences to be expressed, or introduced into the same cell by cotrans- formation. The neomycin phosphotransferase (neo) gene is an example of a selectable marker gene (Southern et al, J. MoI. Anal. Genet. 7:327-341 (1982)). Additional elements may also be needed for optimal synthesis of mRNA. These elements may include signal sequences, splice signals, as well as transcriptional promoters, enhancers, and termination signals.
[145] Methods for introducing RNA sequences into vertebrate cells are described in U.S
Patent No. 5,580,859, the disclosure of which is incorporated herein by reference in its entirety. A viral alphavector, a non-infectious vector useful for administering RNA, may be used to introduce RNA into mammalian cells. Methods for the in vivo introduction of alphaviral vectors to mammalian tissues are described in Altman- Hamamdzic, S, et al., Gene Therapy 4, 815-822 (1997), the disclosure of which is incorporated herein by reference. Viral replioons, i.e., non-infectious RNA vectors packaged in a viral coat, e.g., a pioornavirus coat or an alphavirus coat, are also useful for efficient administration of RNA. See, e.g., US Patent No. 5,766,6CE, U.S. Patent No. 5,614,413, and PCT Publication No. WO 9507994, the disclosures of which are incorporated herein by reference.
[146] Vectors operably encoding a CXCLl 1 polypeptide can be used for transformation or transfection of a suitable host cell, either for administration of the host cell to the subject to be treated, or for the in vitro production of CXCLl 1 polypeptide or polypeptide immunogens. Transformation or transfection can be by any suitable method. Methods for introduction of exogenous DNA into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide (s) in liposomes, and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors.
[147] Transformation or transfection of host cells can be accomplished by conventional methods suited to the vector and host cell employed. R>r transformation of prokaryotic host cells, electroporation and salt treatment methods can be employed (Cohen et al., Proc. Natl. Acad. ScL USA (59:2110-14 (1972)). Ibr transformation or transfection of vertebrate cells, electroporation, cationic lipid or salt treatment methods can be employed. See, e.g., Graham et al., Virology 52:456-467 (1973); Wigler et al., Proc. Natl. Acad. ScL USA 7(5:1373-76 (1979).
[148] The host cell line used for protein expression is most preferably of mammalian origin; those skilled in the art are credited with ability to preferentially determine particular host cell lines which are best suited for the desired gene product to be expressed therein. Exemplary host cell lines include, but are not limited to NSO, SP2 cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells DG44 and DUXBI l (Chinese Hamster Ovary lines, DHFR minus), HELA (human cervical carcinoma), CVI (monkey kidney line), COS (a derivative of CVI with SV40 T antigen), R1610 (Chinese hamster fibroblast) BALBC/3T3 (mouse fibroblast), HAK (hamster kidney line), SP2O (mouse myeloma), P3x63-Ag3.653 (mouse myeloma), BFA-IcIBPT (bovine endothelial cells), RAJI (human lymphocyte) and 293 (human kidney). Host cell lines are typically available from <x>mmerάal services, the American Tissue Culture Collection or from published literature.
[149]
[150] Codon Optimization
[151] The present invention also provides pharmaceutical, immunogenic, or vaccine compositions and methods for modulating an immune response in an animal comprising administering the pharmaceutical, immunogenic, or vaccine composition which comprises an immunogen and a polynucleotide adjuvant as described above to an animal with optimal expression and safety conferred through oodon optimization and/ or other manipulations. These compositions are prepared and administered in such a manner that the encoded gene products are optimally expressed in the animal of interest. As a result, these compositions and methods are useful in stimulating an immune response against the immunogen.
[152] As appreciated by one of ordinary skill in the art, various nucleic acid coding regions will encode the same polypeptide due to the redundancy of the genetic code. Deviations in the nucleotide sequence that comprise the oodons encoding the amino acids of any polypeptide chain allow for variations in the sequence coding for the gene. Since each oodon consists of three nucleotides, and the nucleotides comprising DNA are restricted to four specific bases, there are 64 possible combinations of nucleotides, 61 of which encode amino acids (the remaining three oodons encode signals ending translation). The "genetic code" which shows which oodons encode which amino acids is reproduced herein as Table 1. As a result, many amino acids are designated by more than one oodon. R>r example, the amino acids alanine and proline are coded for by four triplets, serine and arginine by six, whereas tryptophan and methionine are coded by just one triplet. This degeneracy allows for DNA base composition to vary over a wide range without altering the amino acid sequence of the polypeptides encoded by the DNA.
[153] Table 1 [Table 1]
[Table ]
TABLE 1: The Standard Genetic Code
Figure imgf000025_0001
[154] [155] It is to be appreciated that any polynucleotide that encodes a polypeptide in accordance with the invention falls within the scope of this invention, irregardless of the oodons used.
[156] Many organisms display a bias for use of particular oodons to code for insertion of a particular amino acid in a growing polypeptide chain. Codon preference or oodon bias, differences in oodon usage between organisms, is afforded by degeneracy of the genetic code, and is well documented among many organisms. Codon bias often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, inter alia, the properties of the oodons being translated and the availability of particular transfer RNA (tRNA) molecules. The predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization.
[157] In one embodiment, the present invention relates to a polynucleotide adjuvant as described above comprising, consisting essentially of, or consisting of a coding optimized coding region which operably encodes a CXCLl 1 polypeptide disclosed herein. The codon usage is adapted for optimized expression in the cells of a given prokaryote or eukaryote.
[158] Also provided is a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises a polynucleotide expression construct, vector, or host cell comprising a polynucleotide or fragment thereof which comprises all or partly codon-optimized coding regions, wherein the polynucleotide or fragment thereof operably encodes a CXCLl 1 polypeptide, or fragment, derivative, or variants thereof.
[159] Given the large number of gene sequences available for a wide variety of animal, plant and microbial species, it is possible to calculate the relative frequencies of codon usage. Codon usage tables are readily available, for example, at the "Codon Usage Database" available at www.kazusa.or.jp/codon/ (visited July 5, 2007), and these tables can be adapted in a number of ways. See Nakamura, Y., et al, "Codon usage tabulated from the international DNA sequence databases: status for the year 2000" Nucl. Acids Res. 28:292 (2000).
[160] By utilizing the codon usage tables, one of ordinary skill in the art can apply the frequencies to any given polypeptide sequence, and produce a nucleic acid fragment comprising a codon-optimized coding region which encodes the polypeptide, but which uses codons optimal for a given species. Ibr example, in some embodiments of the present invention, the coding region is codon-optimized for expression in human.
[161] Randomly assigning codons at an optimized frequency to encode a given polypeptide sequence, can be done manually by calculating codon frequencies for each amino acid, and then assigning the codons to the polypeptide sequence randomly. Additionally, various algorithms and computer software programs are readily available to those of ordinary skill in the art. Ibr example, the "EditSeq" function in the Lasergene Package, available from DNAstar, Inc., Madison, WI, the backtranslation function in the VectorNTI Suite, available from InforMax, Inc., Bethesda, MD, and the "back- translate" function in the GCG- Wisconsin Package, available from Accelrys, Inc., San Diego, CA. Constructing a rudimentary algorithm to assign oodons based on a given frequency can also easily be accomplished with basic mathematical functions by one of ordinary skill.
[162] Codon placement in a polynucleotide at an optimized frequency to encode a given polypeptide sequence can also be done in a directed manner. R>r example, a oodon may be assigned to a particular amino acid so as to create or destroy a restriction enzyme cleavage site. Creation or destruction of restriction enzyme sites may facilitate DNA manipulation by assisting with cloning or forming identifying markers. Alternatively, a oodon may be assigned to a particular amino acid so as to achieve a desired secondary structure of the polynucleotide.
[163] In certain embodiments, an entire coding region encoding a polypeptide sequence, or fragment, variant, or derivative thereof can be oodon optimized by any of the methods described herein or by other methods. Various desired fragments, variants or derivatives may be designed, and each can then be oodon-optimized individually. In addition, partially oodon-optimized coding regions of the present invention can be designed and constructed. R>r example, the invention includes a nucleic acid fragment of a oodon-optimized coding region encoding a polypeptide in which at least about 1%, 2%, 3,% 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% of the oodon positions have been oodon-optimized for a given species. The partially oodon optimized coding regions contain a oodon that is preferentially used in the genes of a desired species, e.g., a mammalian species, e.g., humans, in place of a oodon that is normally used in the native nucleic acid sequence.
[164] Polypeptides
[165] The present invention is also directed to a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or fragment, variant or derivative thereof. In one aspect, the adjuvant comprises, consists essentially of, or consists of a combination of a CXCLl 1 polypeptide or fragment, variant, or derivative thereof and a polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, variant, or derivative thereof.
[166] In one embodiment, the present invention is directed to a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 27 to 89 of SEQ ID NO: 2 or a combination of the polypeptide and an isolated CXCLl 1 polynucleotide operably encoding the same. The composition, upon administration to an animal, may elicit an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[167] In another embodiment, the present invention is directed to a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids X to Y of SEQ ID NO: 2 or a combination of the polypeptide and an isolated polynucleotide operably encoding the same, wherein X can be any number selected from the group consisting of 22, 23, 24, 25, 26, and 27 and Y can be any number selected from the group consisting of 89, 90, 91, 92, 93, and 94. In one aspect, the composition, upon administration to an animal, may modulate, enhance, or improve relative to administration of the immunogen in the absence of the adjuvant. In particular, the amino acid sequence can be amino acids 29 to 89 of SEQ ID NO: 2, amino acids 22 to 94 of SEQ ID NO: 2, or SEQ ID NO: 2.
[168] As known in the art, "sequence identity" between two polypeptides is determined by comparing the amino acid sequence of one polypeptide to the sequence of a second polypeptide. When discussed herein, whether any particular polypeptide is at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to another polypeptide can be determined using methods and computer programs/ software known in the art such as, but not limited to, the BESTFIT program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711). BESTFIT uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981), to find the best segment of homology between two sequences. When using BESTFIT or any other sequence alignment program to determine whether a particular sequence is, for example, 95% identical to a reference sequence according to the present invention, the parameters are set such that the percentage of identity is calculated over the full length of the reference polypeptide sequence and that gaps in homology of up to 5% of the total number of amino acids in the reference sequence are allowed.
[169] The present invention also includes a pharmaceutical, vaccine, or immunogenic com- position comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of an isolated CXCLl 1 polypeptide comprising an amino acid sequence at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, or at least 90 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO: 2 or a combination of the polypeptide and an isolated polynucleotide opearbly encoding the same.
[170] The CXCLl 1 polypeptide or fragment, derivative, analog, or variant thereof or a polypeptide immunogen comprising an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof described herein may have various alterations such as substitutions, insertions or deletions. Exemplary amino acids that can be substituted in the polypeptide include amino acids with basic side chains ( e.g., lysine, arginine, histidine), acidic side chains ( e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleudne, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleudne) and aromatic side chains ( e.g., tyrosine, phenylalanine, tryptophan, histidine).
[171] In the present invention, a polypeptide can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids ( e.g. non-naturally occurring amino acids). The polypeptides used in the present invention, either a polypeptide mmunogen or a CXCLl 1 polypeptide, may be modified by either natural, such as posttranslational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in the polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, oovalent attachment of flavin, oovalent attachment of a heme moiety, oovalent attachment of a nucleotide or nucleotide derivative, oovalent attachment of a lipid or lipid derivative, oovalent attachment of phosphotidylinositol, cross-linking, cy- clization, disulfide bond formation, demethylation, formation of oovalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, γ-carboxylation, gly- oosylation, GPI anchor formation, hydroxylation, iodination, methylation, myris- toylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. ( See, for instance, Proteins - Structure And Molecular Properties, 2nd Ed., T.E. Creighton, W.H. Freeman and Company, New York (1993); Posttranslational Covalent Modification of Proteins, B.C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol.182:626-646 (1990); Rattan et al., Ann. NY Acad. Sci.663:4&-62 (1992)).
[172] Some embodiments of the invention involve the use of a CXCLl 1 polypeptide or fragment, derivative, or variant thereof that is fused to a heterologous polypeptide moiety to form a fusion protein. Such fusion proteins can be used to accomplish various objectives, e.g., increased serum half-life, improved bioavailability, in vivo targeting, improved recombinant expression efficiency, improved host cell secretion, ease of purification, and higher avidity. Depending on the objective(s) to be achieved, the heterologous moiety can be inert or biologically active. Also, it can be chosen to be stably fused to the CXCLl 1 polypeptide moiety or to be cleavable, in vitro or in vivo. Heterologous moieties to accomplish these other objectives are known in the art.
[173] Pharmacologically active polypeptides such as the CXCLl 1 polypeptides may exhibit rapid in vivo clearance, necessitating large doses to achieve therapeutically effective concentrations in the body. In addition, polypeptides smaller than about 60 kDa potentially undergo glomerular filtration, which sometimes leads to nephrotoxicity. Fusion or conjugation of relatively small polypeptides such CXCLI l polypeptides can be employed to reduce or avoid the risk of such nephrotoxicity. Various heterologous amino acid sequences, i.e., polypeptide moieties or "carriers," for increasing the in vivo stability, i.e., serum half-life, of therapeutic polypeptides are known. Examples include serum albumins such as, e.g., bovine serum albumin (BSA) or human serum albumin (HSA).
[174] Due to its long half- life, wide distribution in mammalian tissues, and lack of enzymatic or immunological function, essentially full-length human serum albumin (HSA), or an HSA fragment, is commonly used as a heterologous moiety. Through ap- plication of methods and materials such as those taught in Yeh et al., Proc. Natl. Acad. ScL USA, 89:1904-08 (1992) and Syed et al., Blood 89:3243-52 (1997), HSA can be used to form a fusion protein or polypeptide conjugate that displays pharmacological activity by virtue of the CXCLl 1 polypeptide moiety while displaying significantly increased in vivo stability, e.g., 10-fold to 100-fold higher. The C-terminus of the HSA can be fused to the N-terminus of the CXCLl 1 polypeptide moiety. Since HSA is a naturally secreted protein, the HSA signal sequence can be exploited to obtain secretion of the fusion protein in vivo in an animal, into the cell culture medium when the fusion protein is produced in a eukaryotic, e.g., mammalian, expression system. Also included is a pharmaceutical, immunological, or vaccine composition comprising an immunogen and an adjuvant comprising an isolated polynucleotide which comprises a polynucleotide opearbly encoding the CXCLl 1 polypeptide and a heterologous nucleotide sequence, for example, a nucleotide sequence encoding HAS
[175] In certain embodiments, the CXCLl 1 polypeptides, fragments, derivatives, or variants thereof for use in the methods of the present invention further comprise a targeting moiety. Targeting moieties include a protein or a peptide which directs localization to a certain part of the body.
[176] Some embodiments of the invention employ a CXCLl 1 polypeptide or fragment, derivative, or variant thereof fused to Fc region, i.e., the C-terminal portion of an Ig heavy chain constant region. Potential advantages of a CXCLl 1-polypeptide-Fc fusion ("immunofusin") include solubility, in vivo stability, and multivalency, e.g., dimerization. The Fc region used can be an IgA, IgD, or IgG Fc region (hinge-CH2-CH3). Alternatively, it can be an IgE or IgM Fc region (hinge-CH2-CH3-CH4). An IgG Fc region is generally used, e.g., an IgGl Fc region or IgG4 Fc region. Materials and methods for constructing and expressing DNA encoding Fc fusions are known in the art and can be applied to obtain fusions without undue experimentation. Some embodiments of the invention employ a fusion protein such as those described in Capon et al., U.S Patent Nos. 5,428,130 and 5,565,335. In certain embodiments of the invention a native CXCLl 1 polypeptide, or fragment thereof occurs as a dimer with a CXCLl 1-polypeptide-Fc fusion.
[177] In another embodiment, the present invention provides an adjuvant comprising consisting essentially of, or consisting of an isolated polypeptide with a first polypeptide fragment and a second polypeptide fragment, where the first polypeptide fragment comprises an amino acid sequence at least 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% similar to a CXCLI l reference amino acid sequence (SEQ ID NO.:2) and where the second polypeptide fragment comprises a fusion moiety.
[178] Secretion of a peptide from a cell can be facilitated by a leader sequence, also referred to as a secretory signal peptide. In a preferred embodiment, either the native CXCLl 1 leader sequence is used, e.g., amino acids 1 to 21 of SEQ IDN O: 2, or a functional derivative of that sequence that retains the ability to direct the secretion of the peptide that is operably associated with it. Alternatively, a heterologous mammalian leader sequence, or a functional derivative thereof, may be used. For example, the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator or mouse β-glucuronidase.
[179] The CXCLl 1 polypeptides or the polynucleotide encoding the CXCLl 1 polypeptides administered in the method of the invention can be fused to a polypeptide tag or a nucleotide sequence encoding the tag, respectively. The term "polypeptide tag", as used herein, is intended to mean any sequence of amino acids that can be attached to, connected to, or linked to the CXCLl 1 polypeptide and that can be used to identify, purify, concentrate or isolate the CXCLl 1 polypeptide. The attachment of the polypeptide tag to the CXCLl 1 polypeptide may occur, e.g., by constructing a nucleic acid molecule that comprises: (a) a nucleic acid sequence that encodes the polypeptide tag, and (b) a nucleic acid sequence that encodes a CXCLl 1 polypeptide. Exemplary polypeptide tags include, e.g., amino acid sequences that are capable of being post- translationally modified, e.g., amino acid sequences that are biotinylated. Other Exemplary polypeptide tags include, e.g., amino acid sequences that are capable of being recognized andor bound by an antibody (or fragment thereof) or other specific binding reagent. Polypeptide tags that are capable of being recognized by an antibody (or fragment thereof) or other specific binding reagent include, e.g., those that are known in the art as "epitope tags." An epitope tag may be a natural or an artificial epitope tag. Natural and artificial epitope tags are known in the art, including, e.g., artificial epitopes such as FLAG, Strep, or poly-histidine peptides. FLAG peptides include the sequence Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys (SEQ ID NO: 9) or Asp- Tyr-Lys-Asp-Glu-Asp-Asp-Lys (SEQ ID NO: 10) (Einhauer, A. and Jungbauer, A., J. Biochem. Biophys. Methods 49:1-3:455-465 (2001)). The Strep epitope has the sequence Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly (SEQ ID NO: 11). The VSV-G epitope can also be used and has the sequence Tyr-
Thr-Asp-Ile-Glu-Met-Asn-Arg-Leu-Gly-Lys (SEQ ID NO: 12). Another artificial epitope is a poly-His sequence having six histidine residues (His-His-His-His-His-His). Naturally-occurring epitopes include the influenza virus hemagglutinin (HA) sequence Tyr-
Pro-Tyr-Asp-Val-Pro-Asp-Tyr-Ala-Ile-Glu-Gly-Arg (SEQ ID NO: 13) reoognized by the monoclonal antibody 12CA5 (Murray et al., Anal. Biochem. 229:170-179 (1995)) and the eleven amino acid sequence from human c-myc (Myc) reoqgnized by the monoclonal antibody 9E10 (Glu-Gln-Lys-Leu-Leu-Ser-Glu-Glu-Asp-Leu-Asn (SEQ ID NO: 14) (Manstein et al., Gene 162:129-134 (1995)). Another useful epitope is the tripeptide Glu-Glu-Phe which is reoqgnized by the monoclonal antibody YL 122. (Stammers et al. FEBS Lett. 283:298-302(1991)).
[180] In certain embodiments, a CXCLl 1 polypeptide and a polypeptide tag may be connected via a linking amino acid sequence. As used herein, a "linking amino acid sequence" may be an amino acid sequence that is capable of being reocgnized andύr cleaved by one or more proteases. Amino acid sequences that can be reocgnized andύr cleaved by one or more proteases are known in the art. Exemplary amino acid sequences are those that are reocgnized by the following proteases: factor Vila, factor IXa, factor Xa, APC, t-PA, u-PA, trypsin, chymotrypsin, enterokinase, pepsin, cathepsin B,H,L,S,D, cathepsin G, renin, angiotensin oonverting enzyme, matrix met- alloproteases (oollagenases, stromelysins, gelatinases), macrophage elastase, Cir, and Cis. The amino acid sequences that are reocgnized by the aforementioned proteases are known in the art. Exemplary sequences reocgnized by certain proteases can be found, e.g., in U.S Patent No. 5,811,252. Also included is a polynucleotide enooding a CXCLl 1 polypeptide and a polynucleotide encoding a polypeptide tag which are connected via a nucleotide sequence enooding a linking amino acid sequence.
[181] By fusing a CXCLl 1 polypeptide moiety at the amino andύr carboxy termini of a suitable fusion partner, bivalent or tetravalent forms of a CXCLl 1 polypeptide or polypeptide fragment can be obtained for administration according to the method of the invention. R>r example, a CXCLl 1 polypeptide moiety can be fused to the amino and carboxy termini of an Ig moiety to produce a bivalent monomeric polypeptide containing two CXCLl 1 polypeptide moieties. Also included is a polynucleotide operably enooding a CXCLl 1 polypeptide moiety that are fused to a polynucleotide sequence enooding the amino acid carboxy termini of an Ig moiety so that the expressed polypeptide is a bivalent monomeric polypeptide oontaining two CXCLl 1 polypeptide moieties. Upon dimerization of two of these monomers, by virtue of the Ig moiety, a tetravalent form of a CXCLl 1 polypeptide is obtained. Such multivalent forms can be used to achieve increased binding affinity for the target. Multivalent forms of a CXCLl 1 polypeptide or polypeptide fragment of the invention or the polynucleotide operably encoding the same also can be obtained by placing CXCLl 1 polypeptide moieties in tandem to form ooncatamers, which can be employed alone or fused to a fusion partner such as Ig or HSA. In one embodiment of the invention, the heterologous polypeptide fused to the CXCLl 1 polypeptide is not a chemokine or a cytokine. In another embodiment, the heterologous polypeptide fused to the CXCLl 1 polypeptide is an immunogen comprising an antigenic or immunogenic polypeptide.
[182] Some embodiments of the invention involve a CXCLl 1 polypeptide or fragment, analog, derivative, or variant thereof conjugated (covalently linked) to one or more polymers. Examples of polymers suitable for such conjugation include polypeptides (discussed above), sugar polymers and polyalkylene glycol chains. Typically, but not necessarily, a polymer is conjugated to a CXCLl 1 polypeptide or fragment, analog, derivative, or variant thereof for the purpose of improving one or more of the following: solubility, stability, or bioavailability. Conjugated polypeptides may be produced by methods well known for those of oridnary skill in the art.
[183] Immunogens
[184] The present invention is directed to a pharmaceutical, immunogenic, or vaccine composition comprising an adjuvant which comprises a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide and an immunogen. In one embodiment, the immunogen comprises a polynucleotide encoding an antigenic or immunogenic polypeptide. In another embodiment, the immunogen comprise a polypeptide such as an antigenic or immunogenic polypeptide. In other embodiments, the immunogen may comprise both an antigenic or immunogenic polypeptide and a polynucleotide encoding an antigenic or immunogenic polypeptide. The polypeptide immunogen may be the same or different than the antigenic or immunogenic polypeptide encoded by the polynucleotide immunogen. Also included in the present invention is an immunogen which comprises an inactivated or attenuated whole cell or virus, e.g., a live- attenuated virus vaccine, a heat-killed virus vaccine, etc.
[185] In one embodiment, a polynucleotide encoding an antigenic or immunogenic polypeptide may be DNA or RNA, for example, in the form of messanger RNA (mRNA). In certain embodiments, a polynucleotide immunogen may be, or be included in, a vector or may be in a host cell comprising the vector. Vectors of the invention are described above.
[186] A polynucleotide immunogen of the invention can also encode a derivative fusion protein such as those described elsewhere herein, wherein two or more nucleic acid fragments, at least one of which encodes an antigenic or immunogenic polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide. Additionally, a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment. Preferably, the polynucleotide encodes an antigenic or immunogenic polypeptide comprising at least one immunogenic epitope, wherein the epitope elicits a B -cell (antibody) response, a T-cell (e.g., CTL) response, or both.
[187] Polynucleotide immunogens used in the present invention can be altered from their native state in a manner similar to polynucleotides operably encoding a CXCLl 1 polypeptide such as described elsewhere herein. R>r example, all or part of the polynucleotide encoding an antigenic or immunogenic polypeptide may be oodon-optimized according to oodon usage in the animal in which the vaccine is to be delivered. In addition, a nucleic acid or fragment thereof which encodes an antigenic or immunogenic polypeptide can be a fragment which encodes only a portion of a full- length polypeptide, andor can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide. R>r example, the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell.
[188] In one embodiment, an immunogen may comprise an isolated polynucleotide encoding an antigenic or immunogenic polypeptide, wherein the antigenic or immunogenic polypeptide is expressed through operable association of the polynucleotide encoding the antigenic or immunogenic polypeptide with a promoter. In certain embodiments, the polynucleotide encoding the antigenic or immunogenic polypeptide and a polynucleotide operably encoding a CXCLl 1 polypeptide may be situated on the same vector. In certain embodiments, the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by two copies of different promoters or identical promoters on the the same vector. Alternatively, the polynucleotide encoding the antigenic or immunogenic polypeptide and the polynucleotide operably encoding the CXCLl 1 polypeptide are driven by a single promoter as a biάstronic transcript where the coding regions are separated by an internal ribosomal entry site (IRES).
[189] Alternatively, the polynucleotide encoding the antigenic or immunogenic polypeptide and a polynucleotide operably encoding a CXCLl 1 polypeptide may be situated on separate vectors. [190] In the compositions or methods of the present invention, the immunogen can also be a polypeptide produced in vitro. Such a polypeptide comprises an antigenic or immunogenic polypeptide.
[191] In the instant invention, an antigenic or immunogenic polypeptide included in the polypeptide immunogen or encoded by a polynucleotide immunogen comprises epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 8 to about 30 amino acids contained within the amino acid sequence of a polypeptide. Certain polypeptides comprises immunogenic or antigenic epitopes of at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length. Antigenic as well as immunogenic epitopes may be linear, i.e., be comprised of contiguous amino acids in a polypeptide, or may be three dimensional, i.e., where an epitope is comprised of non-contiguous amino acids which come together due to the secondary or tertiary structure of the polypeptide, thereby forming an epitope.
[192] As to the selection of peptides or polypeptides bearing an antigenic or immunogenic epitope (e.g., that contain a region of a protein molecule to which an antibody or T cell receptor can bind), it is well known in that art that relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, e.g., Sutcliffe, J. G., et al, Science 219:660-666 (1983).
[193] Polypeptide immunogens may have various modifications or alternations in a manner similar to a CXCLl 1 polypeptide described above.
[194] In certain embodiments, the antigenic or immunogenic polypeptide used as an immunogen or encoded by a polynucleotide used as an immunogen in the present invention can be a viral polypeptide, a bacterial polypeptide, a fungal polypeptide, a parasite polypeptide, an allergenic polypeptide, a tumor- specific polypeptide, fragments, variants, derivatives of any of the polypeptides.
[195] Examples of viral polypeptide may be derived from, but are not limited to, a virus selected from the group consisting of adenovirus, alphavirus, caliάvirus ( e.g., a caliά virus capsid peptide), coronavirus, distemper virus, Ebola virus, enterovirus, flavivirus, hepatitis virus (A-E)( e.g., a hepatitis B core or surface peptide), her- pesvirus( e.g., a herpes simplex virus or varicella zoster virus glycoprotein peptide), immunodeficiency virus ( e.g., a human immunodeficiency virus envelope or protease peptide), infectious peritonitis virus, influenza virus (e.g., an influenza A hemag- glutinin or neuraminidase peptide), leukemia virus, Marburg virus, onocgenic virus, orthomyxovirus, papilloma virus, parainfluenza virus ( e.g., hemagglutinin/neuraminidase peptides), paramyxovirus, parvovirus, pestivirus, pioorna virus ( e.g., a po- liovirus capsid peptide), pox virus (e.g., a vacdnia virus peptide), rabies virus ( e.g., a rabies virus glycoprotein G peptide), reovirus, retrovirus, rotavirus, as well as other cancer-causing or cancer-related virus.
[196] Examples of bacterial polypeptides may be derived from, but are not limited to, a member of a bacterial genus selected from the group consisting of Actinomyces, Bacillus, Bacteroides, Bordetella, Bartonella, Borrelia ( e.g., a B. bergdorferi OspA peptide), Brucella, Campylobacter, Capnocytophaga, Chlamydia, Clostridium, Corynebacterium, Coxiella, Dermatophilus, Enterococcus, Ehrlichia, Escherichia, Franάsella, Fusobacterium, Haemobartonella, Haemophilus ( e.g., H. influenzae type b outer membrane protein peptides), Helicobacter, Klebsiella, L-form bacteria, Leptospira, Listeria, Mycobacteria, Mycoplasma, Neisseria, Neorickettsia, Nocardia, Pasteurella, Peptococcus, Peptostreptococcus, Pneumococcus, Proteus, Pseudomonas, Rickettsia, Rochalimaea, Salmonella, Shigella, Staphylococcus, Streptococcus ( e.g., S pyogenes M protein peptides), Treponema, and Yersinia (e.g., Y pestis Fl and V peptides).
[197] Examples of fungal polypeptides may be derived from, but are not limited to, a member of a fungal genus selected from the group consisting of Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Cocάdioides, Conidiobolus, Cryptoooccus, Curvalaria, Epidermophyton, Exophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Muoor, Paeάlomyces, Peniάllium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhinosporidium, Rhizopus, Scolecobasidium, Sporothrix, Stemphylium, Trichophyton, Trichosporon, and Xylohypha.
[198] Examples of protozoan parasite polypeptides may be derived from, but are not limited to, a member of a protozoan parasite genus selected from the group consisting of Babesia, Balantidium, Besnoitia, Cryptosporidium, Eimeri, Encephalitozoon, Entamoeba, Giardia, Hammondia, Hepatozoon, Isospora, Leishmania, Microsporidia, Neospora, Nosema, Pentatrichomonas, Plasmodium (e.g., P. falciparum άrcum- sporozoite (PfCSP), sporozoite surface protein 2 (PfSSP2), carboxyl terminus of liver state peptide 1 (PfLSA-I c-term), and exported protein 1 (PfExp-1)), Pneumocystis, SarcDcystis, Schistosoma, Theileria, Toxoplasma, and Trypanosoma.
[199] Examples of helminth parasite polypeptides may be derived from, but are not limited to, a member of a helminth parasite selected from the group consisting of Acan- thocheilonema, Aelurostrongylus, Ancylostoma, Angiostrongylus, Ascaris, Brugia, Bunostomum, Capillaria, Chabertia, Cooperia, Crenosoma, Dictyocaulus, Dioctophyme, Dipetalonema, Diphyllobothrium, Diplydium, Dirofilaria, Dracunculus, Enterobius, Hlaroides, pep tides Haemonchus, Lagochilascaris, Loa, Mansonella, Muellerius, Nanophyetus, Necator, Nematodirus, Oesophagostomum, Onchocerca, Opisthorchis, Ostertagia, Parafilaria, Paragonimus, Parascaris, Physaloptera, Proto- strongylus, Setaria, Spirocerca, Spirometra, Stephanofilaria, Strongyloides, Strongylus, Thelazia, Toxascaris, Tojocara, Trichinella, Trichostrongylus, Trichuris, Uncinaria, and Wuchereria peptides.
[200] Examples of ectoparasite polypeptides include, but are not limited to, peptides
(including protective peptides as well as allergens) from fleas; ticks, including hard ticks and soft ticks; flies, such as midges, mosquitos, sand flies, black flies, horse flies, horn flies, deer flies, tsetse flies, stable flies, myiasis-causing flies and biting gnats; ants; spiders, lice; mites; and true bugs, such as bed bugs and kissing bugs.
[201] Examples of tumor- associated polypeptides include, but are not limited to, a tumor- specific immunoglobulin variable region, a GM2 peptide, a Tn peptide, an sTn peptide, a Thompson-Friedenreich peptide (TF), a Globo H peptide, an Le(y) peptide, a MUCl peptide, a MUC2 peptide, a MUC3 peptide, a MUC4 peptide, a MUC5AC peptide, a MUC5B peptide, a MUC7 peptide, a carάnoembryonic peptide, a beta chain of human chorionic gonadotropin (hCG beta) peptide, a HERIneu peptide, a PSMA peptide, a EGFRvIII peptide, a KSA peptide, a PSA peptide, a PSCA peptide, a GPlOO peptide, a MAGE 1 peptide, a MAGE 2 peptide, a TRP 1 peptide, a TRP 2 peptide, a tyrosinase peptide, a MART-I peptide, a PAP peptide, a CEA peptide, A BAGE peptide, a MAGE peptide and a RAGE peptide.
[202] Also included in the present invention are fragments or variants of the antigenic or immunogenic polypeptide, and any combination of the polypeptides. Additional antigenic or immunogenic polypeptides may be found, for example in "Foundations in Microbiology," Talaro, et al. , eds., McGraw-Hill Companies (Oct., 1998), Reids, et al. , "Virology," 3d ed., Lippinoott- Raven (1996), "Biochemistry and Molecular Biology of Parasites", Marr, et al. , eds., Academic Press (1995), and Deacon, J., "Modern Mycology", Blackwell Science Inc (1997), which are incorporated herein by reference.
[203] In some embodiments, the polypeptides of the present invention are isolated or purified. No particular level of purification is required. Once a polypeptide has been recombinantly expressed, it may be purified by any method known in the art for pu- rification of a polypeptide, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
[204]
[205] Compositions
[206] The present invention provides a pharmaceutical, immunogenic, vaccine composition and methods for delivery of the composition, where the composition as described herein, comprises an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide or a combination thereof. As described above, an immunogen can be either a polypeptide or a polynucleotide or a combination thereof. These compositions and methods are useful in stimulating an immune response when administered to an animal or subject in need thereof. The adjuvant of the present invention can modulate, enhance, or improve the immune response when administered in an animal. Compositions of the present invention induce an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[207] R>r example, the invention provides a composition comprising an immunogen and combinatorial adjuvants in which a polynucleotide ( e.g., a plasmid or a viral vector expressing a CXCLI l polypeptide) operably encoding a CXCLI l polypeptide, or fragment, derivative, or variant thereof and a CXCLl 1 polypeptide, fragment, derivative, or variant thereof are combined together. In another embodiment, the immunogen used in the present invention comprises both a polynucleotide immunogen and polypeptide immunogen. A single formulation may comprise (i) an immunogen comprising a polynucleotide encoding an antigenic or immunogenic polypeptide, or fragment, derivative, or variant thereof or a polypeptide comprising the antigenic or immunogenic polypeptide or both polynucleotide and polypeptide and (ii) an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof or a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof or both polypeptide and polynucleotide. The polynucleotide or polypeptide immunogens and a polynucleotide operably encoding CXCLl 1 polypeptide or a CXCLl 1 polypeptide may exist in any form known in the art.
[208] In one embodiment, the pharmaceutical composition of the present invention includes one or more known additional adjuvants. The term "adjuvant" refers to any material having the ability to (1) alter or increase the immune response to a particular antigen or (2) increase or aid an effect of a pharmacological agent. Accordingly, a CXCLl 1 polypeptide, fragment, derivative, or variant thereof or a polynucleotide operably encoding a CXCLI l polypeptide is considered an "adjuvant". This embodiment refers to a pharmaceutical composition comprising one or more additional adjuvants. Suitable adjuvants include, but are not limited to, cytokines, e.g., chemokines, and growth factors; alum, bentonite, latex or acrylic particles, pluronic block polymers, cationic lipids, squalene, depot formers, surface active materials, lysoleάthin, retinal, Quil A, liposomes, pluronic polymer formulations; macrophage stimulators, alternate pathway complement activators, non-ionic surfactants, bacterial components, aluminum-based salts, calcium-based salts, silica, polynucleotides, toxoids, serum proteins, viruses and virally-derived materials, poisons, venoms, imida- zoquiniline compounds, poloxamers, toll-like receptor (TLR) agonists, mLT, CpG, MPL, cationic lipids, Qs21, pluronic polymer formulations, macrophage stimulators, alternate pathway complement activators, non-ionic surfactants and a combination of two or more of the adjuvants. The ability of an adjuvant to increase the immune response to an antigenic or immunogenic polypeptide is typically manifested by a significant increase in immune-mediated protection. Ibr example, an increase in humoral immunity is typically manifested by a significant increase in the titer of antibodies raised to the antigen, and an increase in T-cell activity is typically manifested in increased cell proliferation, or cellular cytotoxicity. An adjuvant may also alter an immune response, for example, by changing a primarily humoral or Th2 response into a primarily cellular, or Th1 response.
[209] Supplementary active compounds can also be incorporated into the compositions used in the methods of the invention. Ibr example, the composition of the present invention comprising a polynucleotide operably encoding a CXCLI l polypeptide, fragment, variant, or derivative thereof or a CXCLl 1 polypeptide, fragment, variant, or derivative thereof combined with a polynucleotide encoding an antigenic or immunogenic polypeptide, or fragment, variant, or derivative thereof or a polypeptide encoded by the polynucleotide may be additionally formulated andor coadministered with one or more additional therapeutic agents effective to treat, ameliorate or prevent a disease.
[210] In certain embodiments, an adjuvant, for example, a polynucleotide sequence operably encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof andor a CXCLl 1 polypeptide, fragment, derivative, or variant thereof can be combined with, or be included in, a whole cell immunogen as a combination regimen that can elicit an enhanced or improved immune response. In one aspect of the present invention, the whole-cell immunogen is not a tumor cell.
[211] The immunogenic composition of the present invention which includes polynucleotides can also include one or more transfection facilitating agents that facilitate delivery of polynucleotides to the interior of a cell, andor to a desired location within a cell. Examples of the transfection facilitating materials include, but are not limited to inorganic materials such as calcium phosphate, alum (aluminum sulfate), ampipathic peptides, cationic lipids, neutral lipids, anionic lipids, and gold particles (e.g., "powder" type delivery vehicles); peptides that are, for example, cationic, intercell targeting (for selective delivery to certain cell types), intracell targeting (for nucleor localization or endosomal escape), and ampipathic (helix forming or pore forming); proteins that are, for example, basic (e.g., positively charged) such as histones, targeting (e.g., asialoprotein), viral (e.g., Sendai virus coat protein), and pore-forming; lipids that are, for example, cationic (e.g., DMRIE, DOSPA, DC-Choi), basic (e.g., steryl amine), neutral (e.g., cholesterol), anionic (e.g., phosphatidyl serine), and zwit- terionic (e.g., DOPE, DOPC); and polymers such as dendrimers, star-polymers, "homogenous" poly-amino acids ( e.g., poly-lysine, poly-arginine), "heterogenous" poly- amino acids ( e.g., mixtures of lysine & glycine), oo-polymers, polyvinylpyrrolidinone (PVP), and polyethylene glycol (PEG). A transfection facilitating material can be used alone or in combination with one or more other transfection facilitating materials. Two or more transfection facilitating materials can be combined by chemical bonding ( e.g., CDvalent and ionic such as in lipidated polylysine, PEGylated polylysine) (Toncheva, et al. , Biochim. Biophys. Acta 1380(3):354-368 (1988)), mechical mixing ( e.g., free moving materials in liquid or solid phase such as "polylysine + cationic lipids") (Gao and Huang, Biochemistry 35:1027-1036 (1996); Trubetskoy, et al.βiochem. Biophys. Acta 1131:311-313 (1992)), and aggregation ( e.g., co-precipitation, gel forming such as in cationic lipids + poly-lactide co-galactide, and polylysine + gelatin).
[212] Other hydrophobic and amphiphilic additives, such as, for example, sterols, fatty acids, gangliosides, glycolipids, lipopeptides, liposaccharides, neobees, niosomes, prostaglandins and sphingolipids, may also be included in the immunogenic compositions of the present invention. In such compositions, these additives may be included in an amount between about 0.1 mol % and about 99.9 mol % (relative to total lipid). Preferably, these additives comprise about 1-50 mol % and, most preferably, about 2-25 mol %. Preferred additives include lipopeptides, liposaccharides and steroids. [213] In some embodiments, the present invention may contain suitable pharmaceutically acceptable carriers comprising exάpients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically for delivery to the site of action. Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium car- boxymethyl cellulose, sorbitol and dextran. Optionally, the suspension may also contain stabilizers. Liposomes can also be used to encapsulate the molecules of this invention for delivery into the cell. Exemplary "pharmaceutically acceptable carriers" are any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In some embodiments, the composition comprises isotonic agents, for example, sugars, polyaloohols such as mannitol, sorbitol, or sodium chloride. In some embodiments, the compositions comprise pharmaceutically acceptable substances such as wetting or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the composition of the invention.
[214] Compositions of the present invention may include various salts, exάpients, delivery vehicles andor auxiliary agents as are disclosed, e.g., in U. S Patent Application Publication No. 20020019358, published February 14, 20CE, which is incorporated herein by reference in its entirety.
[215] Compositions of the present invention can be formulated according to known methods, whereby the substance to be delivered is combined with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their preparation are described, for example, in Remington's Pharmaceutical Sciences. 16th Edition, A. Osol, ed., Mack Publishing Co., Easton, PA (1980), and Remington's Pharmaceutical Sciences. 19th Edition, A.R. Gennaro, ed., Mack Publishing Co., Easton, PA (1995). The pharmaceutical composition can be formulated as an emulsion, gel, solution, suspension, lyophilized form, or any other form known in the art. In addition, the pharmaceutical, immunological, or vaccine composition can also contain pharmaceutically acceptable additives including for example, diluents, binders, stabilizers, and preservatives.
[216] Administration of pharmaceutically acceptable salts of the nucleic acid molecule constructs described herein is preferred. Such salts can be prepared from pharmaceutically acceptable non-toxic bases including organic bases and inorganic bases. Salts derived from inorganic bases include sodium, potassium, lithium, ammonium, calcium, magnesium, and the like. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, basic amino acids, and the like.
[217] R>r aqueous pharmaceutical compositions used in vivo, use of sterile pyrogen-free water is preferred. Such formulations will contain an effective amount of the immunogenic composition together with a suitable amount of vehicle in order to prepare pharmaceutically acceptable compositions suitable for administration to a vertebrate.
[218] Compositions of the invention may be in a variety of forms, including, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions ( e.g., injectable and infusible solutions), dispersions or suspensions. The preferred form depends on the intended mode of administration and therapeutic application. In one embodiment, compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies.
[219] The composition can be formulated as a solution, micro emulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze- drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
[220] In some embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic add, collagen, polyorthoesters, and polylactic add. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York (1978).
[221] In the instant invention, the combination of conventional vacdne compositions with the adjuvant of the present invention provides for therapeutically benefidal effects at dose sparing concentrations. R>r example, immunological responses suffident for a therapeutically benefidal effect in patients predetermined for an approved oommerάal product, such as for the conventional product described above, can be attained by using less of the approved commerdal product when supplemented or enhanced with the appropriate amount of an adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLI l polypeptide. Thus, dose sparing is contemplated by administration of conventional vacdnes administered in combination with a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide.
[222] In particular, the dose of conventional vacdne may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with the adjuvant of the invention.
[223] Further, using a combination of conventional and the adjuvant of the present invetion may allow both materials to be used in lesser amounts while still affording the desired level of immune response arising from administration of either component alone in higher amounts (e.g. one may use less of either immunological product when they are used in combination). This may be manifest not only by using lower amounts of materials being delivered at any time, but also to redudng the number of administrations points in a vacdnation regime ( e.g. 2 versus 3 or 4 injections), andor to redudng the kinetics of the immunological response ( e.g. desired response levels are attained in 3 weeks in stead of 6 after immunization).
[224] In particular, the dose of the adjuvant of the present invention, may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with an immunogen.
[225] In some embodiments, an immunogen that does not normally elidt an immune response upon administration to an animal, i.e., non-immunogenic antigens, are also within the scope of the present invention. Indeed, one advantage of the adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide used in the present invention is the ability to elidt or greatly enhance an immune response in an animal to a non-immunogenic antigen, for example, an antigen to which the animal is tolerant, when a polynucleotide encoding an antigenic or immunogenic polypeptide or a polypeptide comprising an antigenic or immunogenic polypeptide is GO- administered with the adjuvant of the present invention as described herein.
[226] Determining the precise amounts of the adjuvant and an immunogen is based on a number of factors as described above, and is readily determined by one of ordinary skill in the art.
[227] Methods of making various nucleic acid molecule constructs comprising antigen- encoding polynucleotides or methods of preparing a polypeptide antigen or immunogen are well known to those skilled in the art of molecular biology, and disclosed, for example, in U.S. Patent Nos. 4,713,339 and 4,965,196, the disclosures of which are incorporated herein by reference in their entireties.
[228]
[229] Pharmaceutical kits
[230] The present invention also provides kits for use in immunotherapy comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide in a sterile environment. Also provided are kits for use in immunotherapy comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide as well as one or more polypeptides as an immunogen or polynucleotides encoding the polypeptides in a sterile environment. R>r example, an immunogen and an adjuvant used in the present invention may be contained together or separately. In one embodiment, the adjuvant is in the amount of 1 ng to 30 mg.
[231] The immunogen and the adjuvant of the present invention may be contained in glass containers, plastic containers, or strips of plastic or paper. In one embodiment, the composition is contained in a syringe and administered through a plunger. In another embodiment, the composition is administered through a cathether. The composition may further comprise a pharmaceutically acceptable carrier.
[232] The kit can further comprise an instruction sheet for administration of the composition into an animal. The components of the pharmaceutical composition are preferably provided as a liquid solution, such as a suspension, a solution, or an emulsion; or in lyophilized form as a dried powder or a cake. If the composition comprising an immunogen and adjuvant are provided in lyophilized form, the oompositin may be included in a suitable vehicle, such as sterile pyrogen-free water, for reoonstitution of the composition comprising an immunogen and a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide, or any buffer described herein, including PBS, normal saline, Tris buffer, and sodium phosphate vehicle.
[233] The container in which the pharmaceutical composition is packaged prior to use can comprise a hermetically sealed container enclosing an amount of the lyophilized formulation or a solution containing the formulation suitable for a pharmaceutically effective dose thereof, or multiples of an effective dose. The pharmaceutical composition is packaged in a sterile container, and the hermetically sealed container is designed to preserve sterility of the pharmaceutical formulation until use. Optionally, the container can be associated with administration means and or instruction for use.
[234] Suitable additional agents that may be coformulated with andor coadministered with an immunogen andor an adjuvant of the present invention are described above in the composition section.
[235]
[236] Methods of administration
[237] The present invention further provides a method for delivering a pharmaceutical, vaccine or immunogenic composition into an animal or a subject, comprising administering to an animal or subject in need of the compositions as described herein. Upon administration according to this method, the composition comprising an immunogen and an adjuvant of the present invention induces an improved immune response relative to administration of the immunogen in the absence of the adjuvant.
[238] Similarly, the present invention provides a method of enhancing or modulating an immune response in an animal in need of such an enhanced or modulated immune response, comprising administering to the animal a composition as described herein. In this method, the composition contains an adjuvant comprising a polynucleotide encoding an immunogenic or antigenic polypeptide or the polypeptide encoded by the polynucleotide or a combination thereof. Upon administration of the composition according to this method, the needed immunogenic or antigenic polypeptide is expressed in the animal, in a sufficient amount to induce andor modify a desired immune response in the vertebrate to prevent disease, cure disease, reduce the severity of disease symptoms, or prolong the life of the animal.
[239] Also, the present invention provides a method of enhancing or modulating an immune response in a healthy animal for large-scale antibody production, comprising administering to the animal a composition as described herein. In this method, the composition contains a polynucleotide operably encoding a CXCLI l polypeptide, fragment, derivative, or variant thereof or a CXCLl 1 polypeptide. Upon administration of the composition according to this method, the CXCLl 1 polypeptide is expressed or directly introduced in the animal, in a sufficient amount to produce a vigorous antibody response in the animal. The antibodies thus produced are then recovered from the animal by, for example, the collection of serum, milk, or saliva. Such antibodies may be useful for research or diagnostic purposes, or for additional therapies in animals in need of such therapies. Ibr example, passive antibody treatment using antibodies produced by this method may prevent disease, cure disease, reduce the severity of disease symptoms, or prolong the life of an animal.
[240] The polynucleotide encoding the CXCLl 1 polypeptide or the CXCLl 1 polypeptide of the present invention, when used as an adjuvant, can induce an improved immune response, e.g., increase the B- and T-cell responses to poorly immunogenic polypeptides or polynucleotides encoding the same, influence the quality of the immune response (e.g., change the class of antibody produced or change a humoral response to a cellular response) to a specific immnogenic or antigenic polypeptide, and enable the eliάtation of an immune response to an immunogen, e.g., a tumor antigen, that would otherwise be impossible due to tolerance. The polynucleotide operably encoding the CXCLl 1 polypeptide or a CXCLl 1 polypeptide can further improve an immune response elicited by an immunogen, wherein the immune response is improved or enhanced relative to the immune response in the absence of the adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide.
[241] The improved or enhanced immune response may be induction of an improved or enhanced humoral immune response. The improved or enhanced humoral immune response may include an improved or enhanced antibody response, e.g., secretion of IgG, IgA, IgE, IgM, or IgD or a combination thereof than that would not have been secreted when a composition is administered to an animal in the absence of the adjuvant of the present invention. The improved B cell response may further include secretion of larger amount of the antibodies than that would have been secreted when a composition is administered to an animal in the absence of the adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide. A distinctive feature of the present invention is that a pharmaceutical, immunogenic, or vaccine composition comprising an immunogen and an adjuvant comprising a polynucleotide operably encoding a CXCLl 1 polypeptide or a CXCLl 1 polypeptide can enhance both humoral immune responses, i.e., antibody responses, as well as ThI responses, relative to the administration of the immunogen in the absence of the adjuvant, or relative to the administration of the immunogen with other chemokine adjuvants, e.g., CXCL4, CXCL9, or CXCLlO.
[242] Alternatively, the improved or enhanced immune response may not a cell mediated immune response. R>r example, when the composition of the present invention comprising an immunogen and an adjuvant is administered in an animal in need thereof, the adjuvant may improve or enhance natural killer T cell or cytotoxic T cell related immune responses. Furthermore, administration of compositions of the present invention may shift a primarily humoral response into a primarily cellular response. As a non-limiting example, natural killer T cells may produce interferon-γ granulocyte- macrophage colony- stimulating factor as well as multiple other cytokines and chemokines such as IL-2 and TNF- α in improved quantities relative to the administration of the composition in the absence of the adjuvant. Cytotoxic T cell related immune response may include activation and differentiation of CD4+ or CD8+ cells or secretion of cytokines or chemokines, e.g., IL-2, by CD4+ andor CD8+ cells compared to the activation and differentiation of the cells when the composition of the present invention is administered in the absence of the adjuvant comprising a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide. In certain embodiments, the improved immune response includes activation, development, recruitment, and/όr differentiation of ThI cells, but not Th2 cells. ThI cells may express CXCR3 on its surface to interact with the CXCLl 1 polypeptide.
[243] In one embodiment, the improved or enhanced immune response comprises recruitment of cells selected from the group consisting of natural killer (NK) cells, Cytotoxic T cells (CTLs), B lymphocytes, dendritic cells, mactrophages, neutrophils, and a combination of two or more of the cells.
[244] In certain embodiment, the improved or enhanced immune response may include secretion of additional cytokines or chemokines or secretion of cytokines or chemokines in larger quantities than would have been secreted without the adjuvant of the present invention. Non-limiting examples of the cytokines or chemokines are Interferons (type I, II and III) including but not limited to Interleukins (IL-I, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-I l, IL- 12, IL-13, IL- 14, IL-15, IL- 16, IL- 17, IL-18); interferons (INF-α, INF-β, INF-γ); tumor necrosis factor-α (TNF-α); tumor necrosis factor-β (TNF-β); tumor necrosis factor-c (TNF-c); growth hormone (somatotropin); prolactin; granulocyte-colony stimulating factor (G-CSF); myelomonocytic growth factor; leukemia inhibitory factor (LIF); onoostatin-M; ciliary neurotrophic factor (CNTF); cholinergic differentiation factor (CDF); macrophage colony-stimulating factor (M-CSF); transforming growth factor α(TGF-α); transforming growth factor beta2 (TGF- β2); nerve growth factor (NGF); platelet- derived growth factor (PDGF); gonadotropin; melanoma growth stimulating activity (MGSA); macrophage inflammatory protein 1 beta (MIP-I beta); epidemal growth factor (EGF) family: heparin-binding EGF-like growth factor (HB-EGF); am- phiregulin; epiregulin; epigen; betacellulin; neuregulin-1; neuregulin-2; neuregulin-3; neuregulin-4; fibroblast growth factor (FGF); erythropoietin; insulin-like growth factor I (IGF-I); insulin-Like growth factor II (IGF-II), and a combination of two or more of the cytokines. In some embodiments, the improved immune response comprises increased serum levels of a cytokine selected from the group consisting of IFN-γ TNF- α IL-2, and a combination of two or more of the cytokines.
[245] Furthermore, the improved or enhanced immune response may refer to reducing the level of certain cytokine or chemokine secretions. Examples of the cytokines or chemokines which may be decreased may be IL-4, IL-IO, IL-5, or IL- 13.
[246] The CXCLl 1 polypeptide may further elicit an improved immune response to an immunogen that would otherwise induce tolerance. Examples of such immunogen is tumor specific antigens described herein or viral antigenic or immunogenic polypeptides in patients with chronic infenction.
[247] The present invention can also be applied in therapeutic andor prophylactic treatments of diseases, wherein an enhanced or improved immune response can trigger potent self-antigen recognition, such that the response can induce elimination of primary andor secondary attributes of a disease. Increases in self-antigen recognition can be modulated by administration of a polynucleotide opearbly encoding a CXCLl 1 polypeptide, or fragment, derivative, or variant thereof andor a CXCLl 1 polypeptide, or fragment, variant, or derivative thereof, in combination with an immunogen, including but not limited to tumor specific antigens. Other methods in this invention include use of autologous or allogenic genetically modified normal cells expressing a CXCLI l polypeptide (for e.g., fibroblasts); gene modification which expresses a CXCLl 1 polypeptide for antigen processing and presentation by dendritic cells (DCs), application of CXCLl 1 -targeted effector cells like lymphocytes or natural killer cells (NKs); transfer of genes encoding antibodies to known tumor antigens in vivo; vaccination with DNA-encoding cancer antigens. Diseases that can respond to a treatment effected by enhanced self antigen recognition are cellular proliferation disorders, for example, cancer. [248] In any of the methods disclosed herein, the composition may be delivered to an animal, e.g., a mammal. In the instant invention, the mammal can be a human.
[249] Administration of the compositions of the present invention according to any of the above methods can be accomplished according to any of various methods known in the art. R>r example, U.S Patent No. 5,676,954, incorporated herein by reference in its entirety, reports on the injection of genetic material, complexed with cationic lipid carriers, into mice. Also, U.S Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and PCT international patent application PCT/US94/06069 (WO 9429469), the disclosures of which are incorporated herein by reference in their entireties, provide methods for delivering compositions comprising naked DNA, or DNA cationic lipid complexes to animals.
[250] One or more compositions of the present invention are utilized in a "prime boost" regimen. An example of a "prime boost" regimen may be found in Yang, Z. et al.J. Virol. 77:799-803 (2002), which is incorporated herein by reference in its entirety. In these embodiments, one or more polynucleotide or polypeptide vaccine compositions of the present invention are delivered to an animal, thereby priming the immune response of the animal to an immunogen, and then a second composition is utilized as a boost vaccination. One or more compositions of the present invention are used to prime immunity, and then a second composition, e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated polypeptides or fragments, variants or derivatives thereof is used to boost an immune response.
[251] In one embodiment, a priming composition and a boosting composition are combined in a single composition or single formulation. R>r example, a single composition may comprise a polypeptide immunogen and an adjuvant comprising, consisting essentially of, or consisting of a CXCLl 1 polypeptide as the priming component and a polynucleotide immunogen encoding an antigenic and immunogenic polypeptide and an adjuvant comprising a polynucleotide operably encoding a CXCLI l polypeptide as the boosting component. In this embodiment, the compositions may be contained in a single vial where the priming component and boosting component are mixed together. In general, because the peak levels of expression of protein from the polynucleotide does not occur until later ( e.g., 7-10 days) after administration, the polynucleotide component may provide a boost to the isolated protein component. Compositions comprising both a priming component and a boosting component are referred to herein as "combinatorial vaccine compositions" or "single formulation heterologous prime -boost vacdne compositions." In addition, the priming composition may be administered before the boosting composition, or even after the boosting composition, if the boosting composition is expected to take longer to act.
[252] In another embodiment, the priming composition may be administered simultaneously with the boosting composition, but in separate formulations where the priming component and the boosting component are separated.
[253] The terms "priming" or "primary" and "boost" or "boosting" as used herein may refer to the initial and subsequent immunizations, respectively, i.e., in accordance with the definitions these terms normally have in immunology. However, in certain embodiments, e.g., where the priming component and boosting component are in a single formulation, initial and subsequent immunizations may not be necessary as both the "prime" and "boost" compositions are administered simultaneously.
[254] Compositions of the invention are useful for administration to any animal, preferably a mammal (such as apes, cows, horses, pigs, boars, sheep, rodents, goats, dogs, cats, chickens, monkeys, rabbits, ferrets, whales, and dolphins), and more preferably a human.
[255] The immunogenic, pharmaceutical, or vaccine compositions of the present invention may be administered to any tissue of an animal, including, but not limited to, muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, tongue and connective tissue.
[256] In one embodiment, the immunogenic, pharmaceutical, or vaccine compositions of the present invention can be administered by orally, nasally, parenterally, topically, in- trabronchially, intravenously, subcutaneously, intramuscularly, intravenously, intra- tracheally, intranasally, transdermally, interdermally, subcutaneously, intraocularly, vaginally, rectally, intraperitoneally, intraintestinally, by inhalation, buccally, sub- lingually, vaginally, intraheptically, intracardiac, intrapancreatic, transmucosal (i.e., across a mucous membrane), intra-cavity ( e.g., oral, vaginal, or rectal), and intravenous (i.v.) transplantation, by inhalation or by an implanted pump. R>r example, the immunogenic compositions of the present invention are administered by intramuscular (i.m.) or subcutaneous (s.c.) routes.
[257] Any mode of administration can be used so long as the mode results in the expression of the CXCLl 1 polypeptide and the immunogen in the composition in an amount sufficient to elicit a measurable immune response in an animal. This includes needle injection, electroporation into skin or muscle, catheter infusion, biolistic injectors, particle accelerators (i.e., "gene guns" or pneumatic "needleless" injectors — for example, Med-E-Jet (Vahlsing, H., et al., J. Immunol. Methods 171,11-22 (1994)), Pigjet (Schrijver, R., et al, Vaccine 15, 1908-1916 (1997)), Biojector (Davis, H., et al, Vaccine 12, 1503-1509 (1994); Gramzinski, R., et al, MoI Med. 4, 109-118 (1998)), gelfoam sponge depots, other commercially available depot materials, osmotic pumps ( e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery. Certain modes of administration are intramuscular needle-based injection and pulmonary application via catheter infusion. Energy-assisted plasmid delivery (EAPD) methods may also be employed to administer the compositions of the invention. One such method involves the application of brief electrical pulses to injected tissues, a procedure commonly known as electroporation. See generally Mir, L.M. et al, Proc. Natl. Acad. Sci USA 96 :4262-7 (1999); Hartikka, J. et al, MoI. Ther. 4:407-15 (2001); Mathiesen, L, Gene Ther. 6:508-14(1999); Rizzuto G. et al, Hum. Gen. Ther. 77:1891-900 (2000). Each of the references cited in this paragraph is incorporated herein by reference in its entirety.
[258] Parenteral injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Additionally, one approach for parenteral administration employs the implantation of a slow-release or sustained-released systems, which assures that a constant level of dosage is maintained, according to U.S. Pat. No. 3,710,795, incorporated herein by reference in its entirety.
[259] In addition to systemic administration, a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide used in the methods of the invention may be directly infused into an organ or tissue.
[260] The invention encompasses any suitable delivery method for a composition comprising an immunogen and an adjuvant which comprises, consists essentially of, or consists of a CXCLl 1 polypeptide or a polynucleotide opearbly encoding a CXCLl 1 polypeptide to a selected target tissue, including bolus injection of an aqueous solution or implantation of a oontrolled-release system. Use of a oontrolled-release implant reduces the need for repeat injections.
[261] The compositions may also comprise an adjuvant comprising, consisting essentially of, or consisting of a CXCLl 1 polypeptide or a polynucleotide operably encoding a CXCLl 1 polypeptide and an immunogen dispersed in a biocompatible carrier material that functions as a suitable delivery or support system for the compounds. Suitable examples of sustained release carriers include semipermeable polymer matrices in the form of shaped articles such as suppositories or capsules. Implantable or microcapsular sustained release matrices include polylactides (U.Si Patent No. 3,773,319; EP 58,481), copolymers of L-glutamic acid and γ-ethyl-L-glutamate (Sdman et al., Biopolymers 22:547-56 (1985)); poly (2-hydroxyethyl-methacrylate), ethylene vinyl acetate (Langer et al., J. Biomed. Mater. Res. 15:167-277 (1981); Langer, Chem. Tech. 12:98-105 (1982)) or poly-D-(-)-3hydroxybutyric acid (EP 133,988).
[262] In certain embodiments, a polynucleotide or a polypeptide immunogen can be administered, either simultaneously or sequentially, with a polynucleotide operably encoding a CXCLl 1 polypeptide, or variant, derivative, or fragment thereof or a CXCLl 1 polypeptide, or variant, derivative, or fragment thereof as described herein. Alternatively, a polynucleotide encoding multiple antigenic or immunogenic polypeptides each encoding for one or more antigens or multiple polypeptide im- munogens or antigens can be co-administered, either simultaneously or sequentially, with a polynucleotide operably encoding the CXCLl 1 polypeptide, or variant, derivative or fragment thereof or a CXCLl 1 polypeptide as described herein.
[263] In one embodiment, the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of viral diseases in human patients including, but not limited to, diseases caused by adenovirus, al- phavirus, caliάvirus, coronavirus, distemper virus, Ebola virus, enterovirus, flavivirus, hepatitis virus (AE), herpesvirus, immunodeficiency virus, infectious peritonitis virus, influenza virus, leukemia virus, Marburg virus, orthomyx) virus, papilloma virus, parainfluenza virus, paramys) virus, parvovirus, pestivirus, picorna virus, pox virus, rabies virus, reovirus, retrovirus, rotavirus, as well as other cancer-causing or cancer- related virus.
[264] In another embodiment, the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of bacterial diseases in human patients including, but not limited to, diseases caused by Actinomyces, Bacillus, Bacteroides, Bordetella, Bartonella, Borrelia, Brucella, Campylobacter, Capnocytophaga, Chlamydia, Clostridium, Corynebacterium, Coxiella, Dermatophilus, Enterococcus, Ehrlichia, Escherichia, Franάsella, Fu- sobacterium, Haemobartonella, Haemophilus, Helicobacter, Klebsiella, L-form bacteria, Leptospira, Listeria, Mycobacteria, Mycoplasma, Neisseria, Neorickettsia, Nocardia, Pasteurella, Peptococcus, Peptostreptococcus, Pneumococcus, Proteus, Pseudomonas, Rickettsia, Rochalimaea, Salmonella, Shigella, Staphylococcus, Streptococcus, Treponema, and Yersinia .
[265] In still another embodiment, the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of parasitic diseases in human patients including but not limited to, diseases caused by Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Cocάdioides, Conidiobolus, Cryptoooccus, Curvalaria, Epidermophyton, Ejophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Muoor, Paeάlomyces, Peniάllium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhi- nosporidium, Rhizopus, Soolecobasidium, Sporothri, Stemphylium, Trichophyton, Tri- chosporon, and Xylohypha.
[266] In certain embodiments, the present invention may be directed to methods of modulating an immune response in a prophylactic andor therapeutic treatment of fungal diseases in human patients including, but not limited to, Absidia, Acremonium, Alternaria, Aspergillus, Basidiobolus, Bipolaris, Blastomyces, Candida, Cocάdioides, Conidiobolus, CryptoGoccus, Curvalaria, Epidermophyton, Ejophiala, Geotrichum, Histoplasma, Madurella, Malassezia, Microsporum, Moniliella, Mortierella, Muαor, Paeάlomyces, Peniάllium, Phialemonium, Phialophora, Prototheca, Pseudallescheria, Pseudomicrodochium, Pythium, Rhinosporidium, Rhizopus, Scolecobasidium, Sporothrix, Stemphylium, Trichophyton, Trichosporon, and Xylohypha.
[267] The present invention can also be directed to methods of modulating an immune response in immunologically -based therapeutic andor prophylactic treatments of cancer including, but not limited to, cancers of oral cavity and pharynx (i.e., tongue, mouth, pharynx), digestive system (i.e., esophagus, stomach, small intestine, colon, rectum, anus, anal canal, anorectum, liver, gallbladder, pancreas), respiratory system (i.e., larynx, lung), bones, joints, soft tissues (including heart), skin, melanoma, breast, reproductive organs (i.e., cervix, endometirum, ovary, vulva, vagina, prostate, testis, penis), urinary system (i.e., urinary bladder, kidney, ureter, and other urinary organs), eye, brain, endocrine system (i.e., thyroid and other endocrine), lymphoma (i.e., B-cell lymphoma including non-hodgkin's lymphoma , hodgkin's disease), multiple myeloma, leukemia (i.e., acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, chronic myeloid leukemia). Antigens to include in an immunogenic composition of the present invention are described, e.g., in Chamberlain, Drugs 57(3):309-25 (March, 1999), Nawrocki and Mackiewicz, Cancer Treat Rev 25(l):29-46 (Feb.,1999), Rosenberg, Immunity 10(3):281-7 (Mar., 1999), Rosenberg et al. , Adv Caner Res 70:145-77 (1996), Pandey et al. , Eur J Surg Oncol 25(2):209-14 (Apr., 1999), Herlyn et al. , Ann Med. 31(l):66-78 (Feg., 1999), and Rosenberg Immunol Today 18(4): 175-82 (Apr., 1997), which are incorporated herein by reference.
[268] An additional embodiment of the present invention is directed to combining a composition of the present invention where the immunogen is a polynucleotide operably encoding, or a polypeptide comprising, a tumor antigen with one or more additional cancer therapies including, but not limited to bone marrow transplant, cord blood cell transplant, surgery, chemotherapy, radiation therapy, and immunotherapy. The polynucleotide, nucleic acid molecule construct(s), immunogenic composition or pharmaceutical composition of the present invention can be administered prior to the commencement of one or more of the additional cancer therapies, during the practice of one or more of the additional cancer therapies, and after the end of one or more of the additional cancer therapies. Types of bone marrow transplant include, but are not limited to autologous bone marrow transplant and heterologous (i.e., from a donor) bone marrow transplant. Types of surgery include, but are not limited to surgery for breast cancer, prostate cancer, colon cancer, brain cancer, and head and neck cancer.
[269] Chemotherapeutic agents include, but are not limited to alkylating agents, including mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil, di- carbazine, streptazoάne, carmustine, lomustine, semustine, chlorozotoάn, busulfan, triethylenemelamine, thiotepa, hexamethylmelamine; antimetabolites, including methotrexate; pyrimidine analogs, including fluorouraάl, 5-fluorouraάl, floxuridine (5'-fluoro-2'-deoxyuridine), idoxuridine, cytarabine, N-phosphonoacetyl-L-aspartate, 5-azacytidine, azaribine, 6-azauridine, pyrazofuran, 3-deazauridine, aάviάn; purine analogs, including thioguanine, mercaptopurine, azathioprine, pentostatin, erythrohy- droxynonyladenine; vinca alkaloids, including vincristine and vinblastine; epipodophyllotoxins, including etoposide and teniposide; antibiotics, including dactinomyάn, daunorubiάn, doxorubicin, bleomycin sulfate, plicamyάn, mitomycin; enzymes, including L-asparaginase; platinum coordination complexes, including άsplatin, carboplatin; hydroxyurea, procarbazine, mitotane; and hormones or related agents, including adrenocorticosteroids such as prednisone and prednisolone; aminog- lutethimide; progestins such as hydroxyprogesterone caproate, medroxyprogesterone acetate, megesterol acetate, estrogens and androgens such as diethylstilbestrol, flu- oxymesterone, ethynyl estradiol, antiestrogens such as tamoxifen, and gonadotropin- releasing hormone analogs such as leuprolide.
[270] An additional embodiment of the present invention is directed to combining any one of the viral disease treatment methods of the present invention with one or more ad- ditional viral therapies including, but not limited to anti- viral drugs, such as nucleoside analogs, immuloglobulin therapy and interferons. Examples of anti-viral drugs include, but not limited to, Acyclovir, Gancyclovir, Ibscarnet, Zidovudine, Lamivudin [3TC], Ribavirin, and Amantadine, protease inhibitors, and a combination of one or more antiviral drugs.
[271] An additional embodiment of the present invention is directed to combining any one of the bacterial infection treatment methods of the present invention with one or more additional bacterial infection treatments including, but not limited to, treatment with antibiotics, such as vancomycin, daptomyάn, cefotetan, chloramphenicol, penicillin, tetracyclin, trimethoprim, rifampin, clarithromycin, ciprofloxacin, erythromycin, azithromycin, metronidazole, trimethoprim, isoniazid, ciprofloxacin and clarithromycin.
[272] An additional embodiment of the present invention is directed to combining any one of the fungal infection treatment methods of the present invention with one or more additional fungal infection treatments including, but not limited to, treatment with antifungal drugs, such as amphotericin B, caspofungin, fluconazole, flucytosine, intra- oonazole, ketoconazole and voriconazole.
[273] An additional embodiment of the present invention is directed to combining any one of the parasitic infection treatment methods of the present invention with one or more additional parasitic infection treatments including, but not limited to, treatment with anti-parasitic drugs, chloroquine, doxycycline, a combination of atovaquone and proguanil or mefloquine, mefloquine and quinidine.
[274] Determining an effective amount of an immunogenic composition depends upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the subject, the precise condition requiring treatment and its severity, and the route of administration. Based on the above factors, determining the precise amount, number of doses, and timing of doses are within the ordinary skill in the art and will be readily determined by the attending physician or veterinarian.
[275] The pharmaceutical compositions of the invention may include a "therapeutically effective amount" or a "prophylactically effective amount" of a composition of the invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, e.g., modulating an immune response or elicits an improved immune response. A therapeutically effective amount of the composition comprising an immunogen and an adjuvant of the present invention may vary according to factors such as the disease state, age, sex, and weight of the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
[276] The pharmaceutical, immunologic, or vaccine composition of the present invention may be administered as a single dose, multiple doses or over an established period of time in an infusion. Dosage regimens also may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). R>r example, a polynucleotide or vector operably encoding a CXCLl 1 polypeptide, fragment, derivative, or variant thereof or a CXCLl 1 polypeptide, fragment, derivative, or variant thereof may be administered to an animal once per day, one week out of the month, continuously ( e.g., by osmotic pump) or intermittently. In addition, over time, the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated, each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the immunogen and the adjuvant, i.e., the polynucleotide operably encoding the CXCLI l polypeptide and the CXCLl 1 polypeptide or CXCLl 1 protein and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an immunogen or an adjuvant of the present invention for the treatment of sensitivity in individuals.
[277] A therapeutically effective dose range for an immunogen depends on the types or characteristics of the immunogen as well as manner of administration and types of an animal or subject administered thereto. In some embodiments a therapeutically effective dose range for the CXCLl 1 polypeptide or a polynucleotide encoding the CXCLI l polypeptide is 0.001 - 10 mg/Kg per day. In some embodiments a therapeutically effective dose range for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLI l polypeptide is 0.01 - 1 rηg/Kg per day. In some embodiments a therapeutically effective dose range for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLI l polypeptide is 0.05-0.5 mg/Kg per day. In some embodiments a therapeutically effective dose range for for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLl 1 polypeptide is 0.05-0.2 mg/Kg per day. In some embodiments a therapeutically effective dose range for for the CXCLl 1 polypeptide or the polynucleotide operably encoding the CXCLl 1 polypeptide is 0.001-0.5 mg/Kg per day.
[278] R>r treatment with an immunogen and a polynucleotide operably encoding a
CXCLl 1 polypeptide or a CXCLl 1 polypeptide, the dosage can range, e.g., from about 0.0001 to 100 mg/kg, from about 0.001 to 0.5 mg/kg, or 0.01 to 5 mg/kg ( e.g., 0.02 mgkg, 0.25 mgAcg, 0.5 mgAcg, 0.75 mgAcg, lmgAcg, 2 mg/kg, etc.), of the host body weight. R>r example dosages can be 1 mgkg body weight or 10 mg/kg body weight or within the range of 1-10 mgAcg, preferably at least 1 mg/kg. Doses intermediate in the above ranges are also intended to be within the scope of the invention. Subjects can be administered such doses daily, on alternative days, weekly or according to any other schedule determined by empirical analysis. An exemplary treatment entails administration in multiple dosages over a prolonged period, for example, of at least six months. Additional exemplary treatment regimes entail administration once per every two weeks or once a month or once every 3 to 6 months. Exemplary dosage schedules include 1-10 mg/kg or 15 mg/kg on consecutive days, 30 mgAcg on alternate days or 60 mgAcg weekly.
[279] In certain embodiments, the immunogenic composition comprises an immunogen: adjuvant molar ratio. In one aspect of the invention, the molar ratio ranges from about 100:1 to about 1:100; from about 50:1 to about 1:50; from about 30:1 to about 1:30; from about 25:1 to about 1:25, from about 20:1 to about 1:20; from about 19:1 to about 1:19; from about 18:1 to about 1:18; from about 17:1 to about 1:17; from about 16:1 to about 1:16; from about 15:1 to about 1:15; from about 14:1 to about 1:14; from about 13:1 to about 1:13; from about 12:1 to about 1:12; from about 11:1 to about 1:11; from about 10:1 to about 1:10; from about 9:1 to about 1:9; from about 8:1 to about 1:8; from about 7:1 to about 1:7; from about 6:1 to about 1:6; from about 5:1 to about 1:5; from about 4:1 to about 1:4; from about 3:1 to about 1:3; from about 2:1 to about 1:2 and, still preferably, from about 1:1 to about 1:1. In certain embodiments, a person of ordinary skill in the art can compare expression levels of the immunogen andor adjuvant of the present invention by standard methods to arrive at a proper ratio.
[280] The following examples are included for purposes of illustration only and are not intended to limit the scope of the present invention, which is defined by the appended claims. All references cited in the Examples are incorporated herein by reference in their entireties.
[281] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., Sambrook et al., ed., Cold Spring Harbor Laboratory Press: (1989); Molecular Cloning:A Laboratory Manual, Sambrook et al., ed., Cold Springs Harbor Laboratory, New York (1992), DNA Cloning, D. N. Glover ed., Volumes I and II (1985); Oligonucleotide Synthesis, M. J. Gait ed., (1984); Mullis et al. U.S Pat. No: 4,683,195, Nucleic Acid Hybridization, B. D. Hames & S J. Higgins eds. (1984); Transcription And Translation, B. D. Hames & S J. Higgins eds. (1984); Culture Of Animal Cells, R. I. Freshney, Alan R. Liss, Inc., (1987); Immobilized Cells And Enzymes, IRL Press, (1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology, Academic Press, Inc., N.Y.; Gene Transfer Vectors For Mammalian Cells, J. H. Miller and M. P. Calos eds., Cold Spring Harbor Laboratory (1987); Methods In Enzymology, VoIs. 154 and 155 (Wu et al. eds.); Immunochemical Methods In Cell And Molecular Biology, Mayer and Walker, eds., Academic Press, London (1987); Handbook Of Experimental Immunology, Volumes I-IV, D. M. Weir and C. C. Blackwell, eds., (1986); Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y., (1986); and in Ausubel et al., CurrentProtocols in Molecular Biology, John Wiley and Sons, Baltimore, Maryland (1989).
[282] Standard reference works setting forth general principles of immunology include Current Protocols in Immunology, John Wiley & Sons, New York; Klein, J., Immunology : The Science of Self-Nonself Discrimination, John Wiley & Sons, New York (1982); Kennett, R., et al., eds., Monoclonal Antibodies, Hybridoma: A New Dimension in Biological Analyses , Plenum Press, New York (1980); Campbell, A., "Monoclonal Antibody Technology" in Burden, R., et al., eds., Laboratory Techniques in Biochemistry and Molecular Biology, Vol. 13, Elsevere, Amsterdam (1984), Kuby Immunnology 4th ed. Ed. Richard A. Goldsby, Thomas J. Kindt and Barbara A. Osborne, H. Freemand & Co. (2000); Roitt, L, Brostoff, J. and Male D., Immunology 6 th ed. London: Mosby (2001); Abbas A., Abul, A. and Lichtman, A., Cellular and Molecular Immunology Ed. 5, Elsevier Health Sciences Division (2005); Kontermann and Dubel, Antibody Engineering, Springer Verlan (2001); Sambrook and Russell, Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Press (2001); Lewin, Genes VIII, Prentice Hall (2003); Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Press (1988); Dieffenbach and Dveksler, PCR Primer Cold Spring Harbor Press (2003).
[283]
Mode for the Invention
[284] EXAMPLEl:
[285] Plasmid DNA preparation and purification
[286] To construct a mammalian vector for efficient expression of members of the CXC family of chemokines, CXCLl 1 (SEQ ID NO:8), a polynucleotide encoding the murine CXCLl 1 (SEQ ID NO:7) was custom synthesized using TOP Gene Technologies ( www.topgenetech.corrO. and cloned into the Aspl 18 and Xbal sites of pShuttle-tet-10 mammalian expression vector. The vector, pShuttle-tet-10, was constructed by inserting a custom synthesized (TOP Gene Technologies) tet operator sequence (5' TCC CTA TCA GTG ATA GAG ATC TCC CTA TCA GTG ATA GAG ATC GTC GAC GAG CTC-3' at the 3' end of the cytomegalovirus (CMV) promoter (SEQ ID NO.: 15) in the pShuttle 10 vector (Clontech).
[287] To construct the pGXIO-OVA expression plasmid, the OVA gene from pCMV-OVA plasmid (Cho et ah, J. Immunol. 167:5549-5557; (2001)) was digested with EcoRI and Xbal and inserted into pGXIO vector that was previously reported (Ha et al., Gene Therapy, 12:634-638 (2005)). The plasmid DNA was isolatedby a widely available Qiagen plasmid purification kit. The quality ofDNA was assessed by standard gel electrophoresis on 1% agarose gel. Western blot analysis confirmed efficient expression of the CXCLl 1 from the plasmid pShuttle-tet-10-mCXCLl 1 (data not shown).
[288] Animals
[289] C57BL/6 mice, 5-6 weeks of age, were purchased from Japan SLC (Shizuoka,
Japan). The mice were maintained at the animal facility of POSTECH under standard conditions according to the institutional Guidelines. All experiments were performed according to the guidelines of the institutional laboratory animal resources committee.
[290] Immunization and sample collection
[291] The immunization schedule of mice groups is shown in Hg. 1. The first stage immunization was performed on groups of 5- to 6-week-old male and female mice (n=7) by intramuscular injection (i.m.) with electroporation with a combination of plasmids, lOμg pGXIO-OVA plasmid DNA encoding OVA immunogen and 60μg of of pShuttle- tet-10-mCXCLl 1 plasmid DNA encoding the chemokine CXCLl 1 (G5), formulated in phosphate-buffered saline (PBS) (pH7.2). Electroporation was carried out using BTX electroporator (Mode: LV; voltage: 100V; P. length: 2OmS; No. of pulses: 6; interval between pulses: 1.0 sec; polarity: unipolar). Three weeks after the 1st immunization, a booster immunization was performed the same way using the same doses of plasmid DNAs. Serum samples from immunized mice were collected on the 21 st day and 35th day by retroorbital bleeding and ELISA was performed to evaluate any modulatory effects of CXCLl 1 on the immune response to OVA.
[292] ELISA
[293] The immune-modulatory function of the CXCl 1, as a genetic adjuvant, was evaluated by co-immunizing the C57BL/6 mice intramuscularly (i.m) on two occasions (at zero week and 3 week) with pShuttle-tet-10-mCXCLl l in combination with the DNA vaccine encoding OVA (pGXIO-OVA). Subsequently, the OVA- speάfic IgG levels in the sera were determined by ELISA on the 21 st day and the 35th day, as schematically shown in FIG. 1. Microtiter plates (Nalgene Nunc International, NY) were coated at 40C for overnight with 1 μg/m# of OVA protein (purchased from Sigma- Aldrich Co.) in PBS Ibllowing morning, the plates were washed with PBS containing 0.05% tween and blocked at room temperature for 1 hour with 5% skim milk in 0.05% PBST. Serum samples collected from immunized mice were diluted in 5% milk in 0.05% PBST at 1:50 dilution added to the plates and incubated for 2 hours at 370C. R>r endpoint dilution, individual serum samples were pooled per group and pooled serum was serially diluted before adding to the plate. After incubation, plates were washed and were incubated with 1:3000 dilutions of goat anti-mouse total IgG, IgGl and IgG2a antibodies conjugated to horseradish peroxidase (Southern Biotechnology Associates, Birmingham, AL). After incubation at room temperature for 1 hour, the plates were washed and assayed for color development by the addition of 5O/i6 of 1:1 mixtures of TMB peroxidase substrate (purchased from KPL Inc.) and peroxidase substrate solution B (purchased from KPL Inc.). When color was developed sufficiently, the reaction was stopped with 5O/i2 of 2N H 2SO4. Plates were read at 450nm with an ELISA microplate reader.
[294] Modulation of systemic antibody responses by CXCR3 ligands as adjuvants
[295] The OVA-speάfic IgG levels in the sera obtained from three mice per group (as diagramed in Hg. 1), were determined by ELISA on the 21 st day after priming immu- nization. No detectable OVA-speάfic IgG response was induced by the control naive group (Naive). In contrast, the plasmid pGXIO-OVA (Gl) showed detectable IgG response after first administration (Rg. 2). Co-injection of CXCL4 (G2), CXCL9 (G3), CXCLlO (G4), and CXCLI l (G5) all induced moderate increases in the OVA-speάfic IgG responses in sera compared to pGXIO-OVA alone injection group (Gl). Antibody responses were expressed as absorbance at 450nm.
[296] To determine any modulatory effects consequent to booster immunization, C57BL/6 mice, that were prime immunized on day zero, were once again immunized intramuscularly with 10μg of OVA antigen +/- different chemokines (60μg) on the 21 st day. At 2 weeks after booster immunization (i.e., 35th day from priming), sera from five mice per group were obtained. Pooled sera from of each group were made with same volume of each individual sera. Serum anti-OVA total IgG, IgG2a, and IgGl end-point dilution titer using sera pool of each group were determined by ELISA. Antibody responses were expressed as absorbance at 450nm (FIG.3)
[297] Co-injection of pShuttle-tet-10-mCXCLl 1 with pGXIO-OVA was found to show a differential pattern of IgG production upon boosting on the 21st day (Rg.3a). The groups (G1-G5) denoted in Rg.3 refers to the immunization groups depicted schematically in Rg.l. Co-injection of pShuttle-tet-10-mCXCL9 and pGXIO-OVA (G3) slightly increased the levels of total IgG, IgG2a, and IgGl compared to injection of pGXIO-OVA alone (Gl) (Rg.3a-c). However, there was no significant change in the generation of the IgG response in the CXCL4 (G2) or CXCLlO (G4) co-injection groups. In contrast, a higher level of IgG production was observed when the plasmid expressing CXCLI l (G5) was co-injected with pGXIO-OVA, compared to CXC4, or CXC9, or CXClO co-injection groups (G2-G4). Moreover, the strongest anti-OVA antibody responses including IgG2a and IgGl were also observed in CXCLl 1 co- injection group (G5). This suggested that co-injection of CXCLl 1 expressing plasmid with the OVA DNA vaccine, in contrast to co-injection with plasmids encoding other chemokines, can modulate the production of OVA-speάfic IgG induced in response to OVA DNA vaccination in the absence of adjuvant.
[298] ELISPOT assay
[299] 96- well filtration plates (Millpore, Bedford, MA) were coated with 3 μg/m# rat anti- mouse INF-γ Ab (BD Pharmingen, San Diego, CA). After overnight incubation at room temperature, plates were washed twice with PBS and blocked with RPMI containing 10% FBS for 1 hour at 370C. Splenocyte cells (106) from 3-7 mice (the number of mice varied depending on the experiment) were harvested on the 21st or 35th day after prime or booster immunization (depending on the experiment), and re- suspended in complete medium (RPMI 1640 containing 10% FBS, 50 μm 2-ME, 2mM glutamine, IOOU of penicillin/ m-6, 100 βg of streptomyάn/m#), and applied into plates with or without OVA CD4+ and OVA CD8+ epitopes (these epitopes were synthesized by Peptron, Korea (www.peptron.CDm). The fragments of OVA used for generation OVA::CD4 and OVA::CD8 fusions were OVA323-339 and O VA257_264 respectively, and were previously described by Chang et ah, J. Immunol. 172:2818-2826 (2004)). After 20 hours, the plates were washed and 50 jΛ of 2 μg/m# biotinylated rat anti-mouse INF-γ Ab was added (BD Pharmingen). After incubation at room temperature for 3 hours, the plates were washed, and 50 jΛ of 1:2000 diluted streptavidin-alkaline phosphatase (BD Pharmingen) was added. After 45 minutes of incubation, the plates were washed. For spot development, 50 μJl of 5-bromo-4-chloro-3-indolyl phosphate/ nitroblue tetrazolium solution (66 ml of NBT (nitroblue tetrazolium solution) and 33 jΛ of BCIP (5-bromo-4-chloro-3-indolyl phosphate) added to 10 ml of alkaline phosphate buffer (100 mM Nacl, 5 mM MgCl2, 100 mM Tris-Cl, pH 9.5); purchased from Promega] was added as substrate. When spot color was sufficiently developed, the reaction was stopped by washing the plates with water. The number of spots were counted using AID ELISPOT Reader System (Autoimmun Diagnostika, Strassber, Germany).
[300] Induction of CD4-T cell responses by CXCR3 ligands as adjuvants [301] To determine any effects after booster immunization on the CD4+ T cell responses, groups of mice were co-immunized twice (as depicted schematically in Hg. 1) with plasmid pShuttle-tet-10-mCXCR3-ligands in combination with pGXIO-OVA, and 2 weeks after the final immunization (booster immunization) seven mice per group were sacrificed and their splenocytes were harvested. The effects of co-administration of plasmid DNAs encoding various CXCR ligand chemokines with pGXIO-OVA on the generation of OVA speάfic-CD4+ T cells were examined. Results were expressed as the number of IFN-γ secreting cells (ISCs)/ 106 cells (Rg 4). The data represent the average value and standard deviation of three independent experiments. [302] Total 1x106 splenocytes cells were prepared, and OVA CD4+ epitope was used as a stimulator in ELISPOT assay. T cells obtained from the spleens of these mice were stimulated with the syngeneic APC pulsed with OVA protein antigen. To examine antigen- specific T cell responses, an IFN-γ ELISPOT assay was performed in response to the OVA CD4+ epitope stimulator. Hg. 4 depicts that the level of IFN-γ secreting CD4+ T cells, from mice GO- administered with plasmid DNAs expressing CXCL4 and OVA, was modestly decreased as compared to control mice, injected only with OVA expressing plasmid (Gl). Hg. 4 further shows that the IFN-γ secreting CD4 + T cells, from the mice co-administered with plasmid DNAs expressing CXCL9, or CXCLlO, or CXCLl 1 and OVA significantly enhanced the production of the CD4 +T cells as compared to control mice, injected only with OVA expressing plasmid (Gl) (Hg. 4). Interestingly, among the four CXCR3 ligands tested, CXCLl 1 showed the highest capability for inducing INF-g secreting CD4 +T cells to the OVA antigen.
[303] Induction of CD8-T cell responses by CXCR3 ligands as adjuvants
[304] To analyze if OVA-speάfic CD8 + T cell responses were induced by each group (FIG. 1) administered with combination of OVA and a CXCR3 ligand expressing plasmids, OVA-speάfic intracellular IFN-γ staining and ELISPOT assays were performed using isolated splenocytes, 2 weeks after final (booster) immunization. At 2 weeks after booster immunization, seven mice per group were sacrificed and their splenocytes were harvested. To examine antigen- specific T cell responses, an IFN-γ ELISPOT assay was performed in response to OVA CD8+ epitope stimulator. A total IxIO6 splenocytes cells were prepared, and OVA CD8+ epitope was used as a stimulator in ELISPOT assay. Results were expressed as the number of IFN-γ secreting cells (ISCs)/ 106 cells. The data represent the average value and standard deviation of three independent experiments. As shown in Hg. 5, a co-injection of CXCLl 1 chemokine and OVA-enooded plasmids elicited a significant change in the production of IFN-γ in response to OVA DNA vaccination. The GO- administration of the plasmid DNA encoding CXCL4, or CXCL9, or CXCLlO, or CXCLl 1 with pGXIO-OVA also enhanced the level of IFN-γ secreting CD8+ T cells.
[305] FACS analysis
[306] Splenocytes were harvested from spleens of mice on the 35th day after final (booster) immunization. To confirm antigen- specific T cell responses, in term of IFN-γ production with the ELISPOT assays described above, FACS analysis of splenocytes was performed. 2x106 cells in lm-6 of complete media/tube cells were stimulated by OVA CD8+ epitope and treated with BFA (100Ox) for 6 hours. The cells were then washed with FACS buffer [1% Fetal Bovine Serum (FBS), 0.09% sodium azide in PBS] and stained with Antigen presenting cell (APC)-oonjugated anti-CD8 mAb (BD Pharmingen, San Diego, CA). After incubating 30 minutes at 40C in the dark, the cells were once again washed with FACS buffer and treated with 200 jΛ of lysing solution (BD Pharmingen, San Diego, CA) and incubated overnight at 40C. Subsequently, the cells were washed with washing buffer (0.5% saponin in FACS buffer), and stained with PE-conjugated IFN-γ mAb, CD8-APCs mAb, IL-2 mAb, and TNF-α mAb (BD Pharmingen, San Diego, CA), while incubating at 40C for 30 minutes. Hnally, 50,000-100,000 cells were collected using FACSCalibur (BD Biosάences, San Jose, CA) and analyzed with CellQuest software (BD Biosάences, San Jose, CA). The data represent the average value and standard deviation of three independent experiments.
[307]
[308] The adjuvant effect of the CXCLl 1, in terms of CD8+ T cell response, was confirmed in the OVA-speάfic IFN-γ intracellular staining assay using FACS Two weeks after boosting, splenocytes were prepared from mice, and stimulated with CD8 + specific epitope. Levels of OVA-speάfic CD8 + T cells in splenocytes were assayed with CD8-APC mAb, and PE-conjugated with IFN-γ mAb. The results from intracellular IFN-γ staining were similar to those of from IFN-γ ELISPOT assay. As shown in Hg. 6, the CXCL4 and CXCLl 1 induced about 2.5-fold more OVA-speάfic IFN-γ secreting CD8+ T cells than OVA alone.
[309] Collectively, these results indicate that the CXCLl 1 can efficiently induce antigen- specific CD 8 + T cell responses as well as CD4+ T cell responses, in terms of IFN-γ production, in the OVA DNA vaccine mouse model. In addition, there were some effects of other CXCR3 chemokine ligand, such as CXCL9 and CXCLlO, on both CD4+ and CD8+T cells responses.
[310] The effects of the DNA oprablv encoding CXCR3 ligands on CD8 ±-T cells in term of producing other cytokines
[311] To investigate other potential effects of the chemokines DNA ligands on CD8 + T cells, splenocytes were isolated from seven mice per group, 2 weeks after boosting, and examined by FACS analysis. Total 3x106 splenocyte cells for each tube were stained with CD8-APCs mAb and PE-conjugated with TNF-α mAb (a) and IL-2 mAb (b), and were analyzed by FACS The data represent the average value and standard deviation of three independent experiments. As shown in Hg. 7a, co- administration of CXCLl 1 chemokine DNA with the OVA DNA vaccine could dramatically enhance TNF-α production, which is consistent with the results of INF-γ staining described above. Similarly, enhanced secretion of IL-2 was also observed when mice were co- injected with CXCLl 1 and OVA encoding plasmid DNAs (Rg. 7b). The data demonstrate that CXCLl 1 had a stronger effect, compared to other CXCR3 chemokine ligands, on the production of IL-2 from CD8 + T cells (Rg. 7b).
[312]
[313] Statistical Analysis [314] All analysis was performed using Microsoft Excel program. Data were expressed as mean and standard deviation. The tests of significance were performed by t-test (α=0.05).
[315]
[316] EXAMPLE 2
[317] Construction of recombinant adenoviral vector expressing CXCLI l fused to mouse Fc
[318] To construct an adenoviral vector for efficient expression of members of the CXC family of chemokines, CXCLl 1, a polynucleotide encoding the murine CXCLl 1 (mCXCLl 1) was custom synthesized using TOP Gene Technologies (www.topgenetech.com) and cloned into the Asp! IS and Xbal sites of pShuttle-tet-10 mammalian expression vector. The vector, pShuttle-tet-10, was constructed by inserting a custom synthesized (TOP Gene Technologies) tet operator sequence (5'TCC CTA TCA GTG ATA GAG ATC TCC CTA TCA GTG ATA GAG ATC GTC GAC GAG CTC-3' at the 3' end of the cytomegalovirus (CMV) promoter in the pShuttle 10 vector (Clontech). And codon-optimized Fc encoding mouse IgG2a Fc (SEQ ID NO: 16) was custom synthesized using TOP Gene Technologies, and cloned into pShuttle-tet-10-CXCLl 1 to make easily purify recombinant murine CXCLl 1 fused with mouse Fc.
[319] Recombinant replication-defective adenovirus expressing CXCLl 1 plus mouse Fc was generated according to the AdEasy Vector System (QBiogene, Carlsbad, CA). After recombination of pShuttle-tet- 10-CXCLl 1-Fc with the adenoviral backbone vector, pAdEsay in Escherichia coli BJ5183, the recombinant adenovirus expressing CXCLl 1-Fc were generated and expanded in 293 cells.
[320] Production and purification of CXCLl 1-Fc
[321] To produce CXCLl 1-Fc, recombinant adenovirus expressing CXCLl 1-Fc was expanded in 293 cells. During amplification, 293 cells infected with rAd-CXCLl l-Fc were cultured with DMEM (Cambrex Bio Science Walkers ville), 10% fetal bovine serum, and antibiotics (Invitrogen, Carlsbad). At final step, 293 cells were cultured with serum free media (JRH Biosάences, Kansas). At 48hrs after rAd-CXCLl 1-Fc infection, the serum- free supernatants including CXCLI l-Fc protein were harvest and used for its purification. For purification, HiTrap recombinant protein A FF (Amersham biosάences, Piscataway) columns were equilibrated with PBS (pH 7.0). The filtered supernatants were added to the columns and eluted with 0.1M sodium citrate (pH 3.0). The eluted proteins were finally obtained after dialysis with membrane (MWCO 12 14K, Spectrapor, Rancho Dominguez) more than three times. The concentration of a recombinant protein sample was determined by anti-mouse CXCLl 1/I-TAC antibody (R & D system, Minneapolis, cat#. AF572) as a capture Ab, bioninylated anti-mouse CXCLl 1/I-TAC antibody (R & D system, Minneapolis, cat#. BAF572) as a detection Ab, and recombinant mouse CXCLl 1/I-TAC (R & D system, Minneapolis, cat#. 572-MC) as a standard protein for the measurement of CXCLl 1.
[322] Animals
[323] Balb/c mice, 5-6 weeks of age, were purchased from Japan SLC (Shizuoka, Japan). The mice were maintained at the animal facility of POSTECH under standard conditions according to the institutional Guidelines. All experiments were performed according to the guidelines of the institutional laboratory animal resources committee.
[324] Immunization and sample collection
[325] The immunization schedule of mice groups is shown in Hg. 8. The first stage immunization was performed on two groups of 5- to 6- week-old female mice (n=6) by intramuscular injection (i.m.) with 3μg trivalent influenza vaccine (TIV, Green Cross Co., Korea) with or without 3μg recombinant CXCLl 1-Fc protein, which are formulated in phosphate-buffered saline (PBS) (pH7.2). lour weeks after the 1 st immunization, a booster immunization was performed the same way using the same doses of TIV and CXCLl 1-Fc. Serum samples from immunized mice were collected on the 49 ^ day by retroorbital bleeding and ELISA was performed to evaluate any modulatory effects of CXCLl 1 on the immune response against influenza HA protein.
[326] ELISA
[327] The immune-modulatory function of the CXCl 1, as a protein adjuvant, was evaluated by co-immunizing the Balb/c mice intramuscularly (i.m) on two occasions (at zero week and 4 week) with recombinant CXCLl 1-Fc in combination with trivalent influenza vaccine (TIV). To determine the induction of heterosubtypic antibody responses, H5N1 HA-speάfic were determined by ELISA on 49 1^ day, as schematically shown in FIG. 8. Microtiter plates (Nalgene Nunc International, NY) were coated at 40C for overnight with 0.5μg/m# of H5N1 HA protein (produced by Prof. Hwang IH's Lab in POSTECH) in PBS Following morning, the plates were washed with PBS containing 0.05% tween and blocked at room temperature for 1 hour with 5% skim milk in 0.05% PBST. For endpoint dilution, individual serum samples were pooled per group and pooled serum was serially diluted before adding to the plate. After incubation, plates were washed and were incubated with 1:3000 dilutions of goat anti-mouse total IgG, IgGl and IgG2a antibodies conjugated to horseradish peroxidase (Southern Biotechnology Associates, Birmingham, AL). After incubation at room temperature for 1 hour, the plates were washed and assayed for color development by the addition of 5O/i6 of 1 : 1 mixtures of TMB peroxidase substrate (purchased from KPL Inc.) and peroxidase substrate solution B (purchased from KPL Inc.). When color was developed sufficiently, the reaction was stopped with 5O/i6 of 2N H2SO4. Plates were read at 450nm with an ELISA microplate reader.
[328] ELISPOT assay
[329] 96-well filtration plates (Millpore, Bedford, MA) were coated with 3 μg/m# rat anti- mouse INF-γ Ab (BD Pharmingen, San Diego, CA). After overnight incubation at room temperature, plates were washed twice with PBS and blocked with RPMI containing 10% FBS for 1 hour at 370C. Splenocyte cells (106) from 6 mice in each group were harvested on the 49th day after prime or booster immunization (depending on the experiment), and resuspended in complete medium (RPMI 1640 containing 10% FBS, 50 /M 2-ME, 2mM glutamine, IOOU of penicillinM, lOOμg of streptomyάn/m#), and applied into plates with or without HA CD4 + epitope (amino acids, SFERFEIFPKE) (see Haberman et al., J of Immunol. 1990, 145: 3087-3094) and HA CD8+ epitope (amino acids, IYSTVASSL) (see Fraser et al., Immunology, 2006, 119: 126-133) that were synthesized by Peptron, Korea (www.peptron.com). After 20 hours, the plates were washed, and 5O/i6 of 2μg/m# biotinylated rat anti-mouse INF-γ Ab was added (BD Pharmingen). After incubation at room temperature for 3 hours, the plates were washed, and 5O/i2 of 1:2000 diluted strep tavidin- alkaline phosphatase (BD Pharmingen) was added. After 45 minutes of incubation, the plates were washed. For spot development, 5O/i6 of 5-bromo-4-chloro-3-indolyl phosphate/ nitroblue tetrazolium solution (66 ml of NBT (nitroblue tetrazolium solution) and 33 ml of BCIP (5-bromo-4-chloro-3-indolyl phosphate) added to 10 mi of alkaline phosphate buffer (100 mM NaCl, 5 mM MgCl2, 100 mM Tris-Cl, pH 9.5); purchased from Promega] was added as substrate. When spot color was sufficiently developed, the reaction was stopped by washing the plates with water. The number of spots were counted using AID ELISPOT Reader System (Autoimmun Diagnostika, Strassber, Germany).
[330] Induction of CD4^and CD8+ T cell responses by co-delivery with CXCLl 1
[331] To determine the effect of CXCLl 1-Fc on the CD4+ and CD8+ T cell responses, groups of mice were immunized twice (as depicted schematically in Hg. 8) with trivalent influenza vacάne(TIV) in combination with or without recombinant CXCLl 1-Fc, and 3 weeks after the final immunization (booster immunization) all six mice per group were sacrificed and their splenocytes were harvested. The effects of CD- administration of CXCLl 1 with TIV on the generation of HA speάfic-CD4 + and CD8+ T cells were examined by ex vivo IFN-γ ELISPOT assay. Results were expressed as the number of IFN-γ secreting cells (ISCs)/106 cells (Rg. 9). The data represent the average value and standard deviation. Total IxIO6 splenocytes cells were prepared, and each CD4+ epitope or CD8+ epitope of HA protein was used as a stimulator in ELISPOT assay. Hg. 9 depicts that the significant IFN-γ secreting CD4 + and CD8+ T cells were generated by oo-delivery of trivalent influenza vaccine (TIV) with CXCLl 1-Fc although TIV alone oould not induce antigen-specific T cell responses. [332] Induction of heterosubtypic antibody responses by cp-delivery of CXCLI l [333] The H5N1 HA-speάfic IgG levels in the sera obtained from six mice per group (as diagramed in Hg. 8), were determined by ELISA on the 21 stday after boost immunization. No detectable OVA-speάfic IgG response was induced by the control naive group (data not shown). In contrast, trivalent influenza vaccine (TIV, Gl) induced detectable IgG response after twice administration (Rg. 10). Co-injection of CXCLl 1 (G2) induced moderate increases in the OVA-speάfic total IgG responses and decreased to 1/4 of IgGl responses in sera compared to TIV alone vaccinated group (Gl). However, CXCLI l co-delivered with TIV generated 32-folds increased IgG2a responses. When IgG2a/IgGl was calculated, more than 100-folds higher IgG2a/IgGl ratio was induced by co-administration of CXCLl 1-Fc. Antibody responses were expressed as absorbance at 450nm. (FIG .10) [334]

Claims

Claims
[1] An adjuvant composition comprising:
(i) an isolated polynucleotide operably encoding a CXCLl 1 polypeptide;
(ii) an isolated CXCLI l polypeptide; or
(iii) a combination of (i) and (ii) ; wherein said composition, upon administration to an animal or subject with an immunogen, elicits an improved immune response to the immunogen relative to administration of said immunogen in the absence of said adjuvant.
[2] An adjuvant composition comprising:
(i) an isolated polynucleotide operably encoding a CXCLl 1 polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ
ID NO: 2;
(ii) an isolated CXCLI l polypeptide comprising at least 10 consecutive amino acids of amino acids 22 to 94 of SEQ ID NO:2; or
(iii) a combination of (i) and (ii) ; wherein said composition, upon administration to an animal or subject with an immunogen, elicits an improved immune response to the immunogen relative to administration of said immunogen in the absence of said adjuvant.
[3] The composition of claim 2, wherein said CXCLl 1 polypeptide comprises SEQ
ID NO:2.
[4] An adjuvant composition comprising:
(i) an isolated polynucleotide which operably encodes a CXCLl 1 polypeptide comprising an amino acids 27 to 89 of SEQ ID NO: 2 or an amino acid sequence at least 85% identical to amino acids 27 to 89 of SEQ ID NO: %
(ii) an isolated CXCLl 1 polypeptide comprising an amino acids 27 to 89 of SEQ
ID NO: 2 or an amino acid sequence at least 85% identical to amino acids 27 to
89 of SEQ ID NO: % or
(iii) a combination of (i) and (ii); wherein said composition, upon administration to an animal or subject with an immunogen, elicits an improved immune response to the immunogen relative to administration of said immunogen in the absence of said adjuvant.
[5] The composition of any one of claims 1 to 4, wherein said polynucleotide is incorporated into a vector.
[6] The composition of claim 5, wherein said vector is a plasmid or a viral vector.
[7] The composition of claim 6, wherein said viral vector is selected from the group consisting of an adenovirus vector, and adeno-assoάated virus vector, an al- phavirus vector, an enterovirus vector, a pestivirus vector, a lentiviral vector, a baculoviral vector, a herpesvirus vector, a papovaviral vector, a poxvirus vector, and a parvovirus vector.
[8] The composition of any one of claims 1 to 5, wherein said polynucleotide encoding said CXCLl 1 polypeptide further comprises a heterologous nucleic acid.
[9] The composition of claim 8, wherein said heterologous nucleic acid encodes a heterologous polypeptide fused to said CXCLl 1 polypeptide.
[10] The composition of claim 9, wherein said heterologous polypeptide is selected from the group consisting of an antigen, an immunoglobulin Fc region, and a secretory signal peptide.
[11] A vaccine composition comprising an immunogen and the adjuvant composition of any one of claims 1 to 10.
[12] The composition of claim 11, wherein said immunogen comprises an isolated polynucleotide which operably encodes an antigenic or immunogenic polypeptide or fragment, variant, or derivative thereof, or an encoded polypeptide thereof.
[13] The composition of claim 12, wherein said polynucleotide is incorporated into a vector.
[14] The composition of claim 13, wherein the polynucleotide operably encoding said antigenic or immunogenic polypeptide and a polynucleotide which encodes said
CXCLl 1 polypeptide are situated on the same vector.
[15] The method of claim 14, wherein the polynucleotide encoding said antigenic or immunogenic polypeptide and the polynucleotide operably encoding said
CXCLl 1 polypeptide are driven by two copies of different promoters or identical promoters on the said same vector or by a single promoter as a biάstronic transcript where the coding regions are separated by an internal ribosomal entry site (IRES).
[16] The composition of claim 13, wherein the polynucleotide operably encoding said antigenic or immunogenic polypeptide and a polynucleotide which encodes said
CXCLI l polypeptide are situated on separate vectors.
[17] The composition of claim 16, wherein the polynucleotide operably encoding said antigenic or immunogenic polypeptide is administered prior to the polynu- cleotide encoding said CXCLl 1 polypeptide, or after the polynucleotide encoding said CXCLl 1 polypeptide, or simultaneously with the polynucleotide encoding said CXCLl 1 polypeptide.
[18] The composition of claim 12, wherein a polynucleotide which operably encodes said CXCLl 1 polypeptide and the polynucleotide encoding said antigenic or immunogenic polypeptide are administered at a ratio of about 20:1 to about 1:20.
[19] The composition of claim 12, wherein said polynucleotide operably encoding said polypeptide antigen further comprises a heterologous nucleic acid.
[20] The composition of claim 19, wherein said heterologous nucleic acid encodes a heterologous polypeptide fused to said antigenic or immunogenic polypeptide.
[21] The composition of claim 20, wherein said heterologous polypeptide is selected from the group consisting of an antigen, an immunoglobulin Fc region, and a secretory signal peptide.
[22] The composition of claim 12, wherein said antigenic or immunogenic polypeptide is selected from the group consisting of a viral polypeptide, a bacterial polypeptide, a fungal polypeptide, a parasite polypeptide, an allergenic polypeptide, a tumor-specific polypeptide, fragments, variants, derivatives of any of said polypeptides, and a combination of two or more of said polypeptides.
[23] The composition of claim 11, wherein said composition further comprises an additional adjuvant selected from the group consisting of alum, bentonite, latex or acrylic particles, pluronic block polymers, cationic lipids, squalene, depot formers, surface active materials, lysoleάthin, retinal, Quil A, liposomes, pluronic polymer formulations; macrophage stimulators, alternate pathway complement activators, non-ionic surfactants, bacterial components, aluminum- based salts, calcium-based salts, silica, polynucleotides, toxoids, serum proteins, viruses and virally-derived materials, poisons, venoms, imidazoquiniline compounds, poloxamers, toll-like receptor (TLR) agonists, mLT, CpG, MPL, cationic lipids, Qs21, and a combination of two or more of said adjuvants.
[24] The composition of claim 11, wherein said composition further comprises phamaceutical acceptable carrier.
[25] The composition of claim 11, wherein said composition further comprises a transfection facilitating agent selected from the group consisting of calcium phosphate, alum, gold particles, cationic peptides, ampipathic peptides, histones, targeting molecules, cationic lipids, neutral lipids, anionic lipids, and zwit- terionic lipids, dendrimers, star-polymers, poly-amino acids, co-polymers, polyvinylpyrrolidone (PVP), and polyethylene glycol (PEG). [26] The method for modulating an immune response in an animal or subject, comprising administering to an animal or subject in need thereof a composition according to any one of claims 11 to 25, wherein the immune response elicited by administration of said composition is improved relative to administration of said immunogen in the absence of said adjuvant.
PCT/KR2008/004317 2007-07-23 2008-07-23 Cxcl11 adjuvant compositions and uses thereof WO2009014382A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US93501407P 2007-07-23 2007-07-23
US60/935,014 2007-07-23

Publications (2)

Publication Number Publication Date
WO2009014382A2 true WO2009014382A2 (en) 2009-01-29
WO2009014382A3 WO2009014382A3 (en) 2009-04-02

Family

ID=40281982

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2008/004317 WO2009014382A2 (en) 2007-07-23 2008-07-23 Cxcl11 adjuvant compositions and uses thereof

Country Status (1)

Country Link
WO (1) WO2009014382A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102011001090A1 (en) 2011-03-04 2012-09-06 Geräte- und Pumpenbau GmbH Dr. Eugen Schmidt Merbelsrod Controllable cooling system for a motor vehicle, coolant pump therefor, usable in the coolant pump impeller and method for controlling a flow of coolant in such a cooling system
JP2013052202A (en) * 2011-09-01 2013-03-21 Kosumedei Seiyaku Kk Dna vaccine microneedle
US20160067324A1 (en) * 2013-04-24 2016-03-10 Medizinische Universität Wien Novel vaccine formulation for ocular immunization

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070087986A1 (en) * 2004-01-26 2007-04-19 Brett Premack Compositions and methods for enhancing immunity by chemoattractant adjuvants
WO2007062844A1 (en) * 2005-12-02 2007-06-07 Lmu Muenchen Chemokine-mucin fusions linked to glycosylphosphatidylinositol (gpi)-anchors in tissue regeneration and as tumour immune adjuvants
US20070160571A1 (en) * 2006-01-11 2007-07-12 Mitchell Krathwohl Composition and method for inducing protective vaccine response

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070087986A1 (en) * 2004-01-26 2007-04-19 Brett Premack Compositions and methods for enhancing immunity by chemoattractant adjuvants
WO2007062844A1 (en) * 2005-12-02 2007-06-07 Lmu Muenchen Chemokine-mucin fusions linked to glycosylphosphatidylinositol (gpi)-anchors in tissue regeneration and as tumour immune adjuvants
US20070160571A1 (en) * 2006-01-11 2007-07-12 Mitchell Krathwohl Composition and method for inducing protective vaccine response

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
COLE, K.E. ET AL.: 'Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3' J. EXP. MED. vol. 187, no. 12, 15 June 1998, pages 2009 - 2021 *
YOON, H.A. ET AL.: 'Differential polarization of immune responses by genetic cotransfer of chemokines changes the protective immunity of DNA vaccine against pseudorabies virus' IMMUNOLOGY. vol. 120, no. 2, February 2007, pages 182 - 191 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102011001090A1 (en) 2011-03-04 2012-09-06 Geräte- und Pumpenbau GmbH Dr. Eugen Schmidt Merbelsrod Controllable cooling system for a motor vehicle, coolant pump therefor, usable in the coolant pump impeller and method for controlling a flow of coolant in such a cooling system
WO2012119622A2 (en) 2011-03-04 2012-09-13 Geräte- und Pumpenbau GmbH Dr. Eugen Schmidt Controllable cooling system for a motor vehicle, coolant pump therefor, impeller for use in the coolant pump, and method for controlling a coolant flow in such a cooling system
JP2013052202A (en) * 2011-09-01 2013-03-21 Kosumedei Seiyaku Kk Dna vaccine microneedle
US20160067324A1 (en) * 2013-04-24 2016-03-10 Medizinische Universität Wien Novel vaccine formulation for ocular immunization
US10709778B2 (en) * 2013-04-24 2020-07-14 Medizinische Universität Wien Vaccine formulation for ocular immunization

Also Published As

Publication number Publication date
WO2009014382A3 (en) 2009-04-02

Similar Documents

Publication Publication Date Title
JP6549194B2 (en) Novel vaccine against multiple subtypes of influenza virus
WO2003055439A2 (en) Her2/neu target antigen and use of same to stimulate an immune response
CZ304884B6 (en) Composition for inducing enhanced immune response against peptide or protein antigen in a mammal and use thereof in the preparation of a vaccine
CN111533812B (en) DNA vaccine for SARS-COV-2 virus and its use
WO2017044709A1 (en) Targeting mda-5 activation for cancer immunotherapy
JP2023510112A (en) Vaccines against African swine fever virus and methods of using them
KR20090092271A (en) Dna composition for eliciting an immune response against tumor-associated macrophages
WO2009014382A2 (en) Cxcl11 adjuvant compositions and uses thereof
US10835589B2 (en) Vaccine directed to induction of immune response to protein and glycolipid antigens of bacterial cells through interaction of CD40L/CD40 receptor axis with complex of glycolipid/CD1d receptor in NKT cells and in dendritic cells
US20230293664A1 (en) Marburgvirus consensus antigens, nucleic acid constructs and vaccines made therefrom, and methods of using same
WO2020092880A1 (en) A novel dna vaccine against crimean-congo hemorrhagic fever virus (cchfv)
WO2003004053A1 (en) Methods for inducing an immune response with an elevated th1/th2 ratio, by intracellular induction of nfkappab
KR20180088458A (en) Vaccination of the MICA / B alpha 3 domain for the treatment of cancer
Yurina DNA Vaccine: Mechanism of Action and factors which increase its efficacy
AU2014232335B2 (en) Synthetic immunogens for prophylaxis or treatment of tuberculosis
US9889188B1 (en) Ebola virus composition/vaccine
AU2014268164B2 (en) Novel vaccines against multiple subtypes of influenza virus
WO2022024156A2 (en) Consensus sequence of the antigen telomerase and the use thereof in preventive and therapeutic vaccination
WO2017189722A1 (en) Vaccine directed to induction of immune response to protein and glycolipid antigens of bacterial cells through interaction of cd40l/cd40 receptor axis with complex of glycolipid/cd1d receptor in nkt cells and in dendritic cells
AU2002314374A1 (en) Methods for inducing an immune response with an elevated TH1/TH2 ratio by intracellular induction of NFKAPPAB

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: 08792870

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08792870

Country of ref document: EP

Kind code of ref document: A2