AU2011224126A1 - Tumor antigens BFA5 for prevention and/or treatment of cancer - Google Patents

Tumor antigens BFA5 for prevention and/or treatment of cancer Download PDF

Info

Publication number
AU2011224126A1
AU2011224126A1 AU2011224126A AU2011224126A AU2011224126A1 AU 2011224126 A1 AU2011224126 A1 AU 2011224126A1 AU 2011224126 A AU2011224126 A AU 2011224126A AU 2011224126 A AU2011224126 A AU 2011224126A AU 2011224126 A1 AU2011224126 A1 AU 2011224126A1
Authority
AU
Australia
Prior art keywords
expression vector
alvac
vector
nucleic acid
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2011224126A
Inventor
Neil Berinstein
Scott Gallichan
Corey Lovitt
Mark Parrington
Artur Pedyczak
Laszlo Radvanyi
Devender Singh-Sandhu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi Pasteur Ltd
Original Assignee
Sanofi Pasteur 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 Sanofi Pasteur Ltd filed Critical Sanofi Pasteur Ltd
Priority to AU2011224126A priority Critical patent/AU2011224126A1/en
Publication of AU2011224126A1 publication Critical patent/AU2011224126A1/en
Priority to AU2014201009A priority patent/AU2014201009A1/en
Abandoned legal-status Critical Current

Links

Abstract

TUMOR ANTIGENS BFA5 FOR PREVENTION AND/OR TREATMENT OF CANCER Abstract The present invention relates to a nucleic acid encoding a polypeptide and the use of the nucleic acid or polypeptide in preventing and/or treating cancer. In particular, the invention relates to improved vectors for the insertion and expression of foreign genes encoding tumor antigens for use in immunotherapeutic treatment of cancer.

Description

S&F Ref: 739027D1 AUSTRALIA PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT Name and Address Sanofi Pasteur Limited, of 1755 Steeles Avenue West, of Applicant: Toronto, Ontario, M2R 3T4, Canada Actual Inventor(s): Artur Pedyczak Corey Lovitt Devender Singh-Sandhu Scott Gallichan Mark Parrington Neil Berinstein Laszlo Radvanyi Address for Service: Spruson & Ferguson St Martins Tower Level 35 31 Market Street Sydney NSW 2000 (CCN 3710000177) Invention Title: Tumor antigens BFA5 for prevention and/or treatment of cancer The following statement is a full description of this invention, including the best method of performing it known to me/us: 5845c(5602127_1) Tumor Antigens BFA5 for Prevention and / or Treatment of Cancer RELATED APPLICATIONS This application claims priority to U.S. Provisional Application No. 60/462,945 filed 5 April 15, 2003. FIELD OF THE INVENTION The present invention relates to a nucleic acid encoding a polypeptide and the use of the nucleic acid or polypeptide in preventing and / or treating cancer. In particular, the invention 10 relates to improved vectors for the insertion and expression of foreign genes encoding tumor antigens for use in immunotherapeutic treatment of cancer. BACKGROUND OF THE INVENTION There has been tremendous increase in last few years in the development of cancer 15 vaccines with Tumour-associated antigens (TAAs) due to the great advances in identification of molecules based on the expression profiling on primary tumours and normal cells with the help of several techniques such as high density microarray, SEREX, immunohistochemistry (IHC), RT PCR, in-situ hybridization (ISH) and laser capture microscopy (Rosenberg, Immunity, 1999; Sgroi et al, 1999, Schena et al, 1995, Offringa et al, 2000). The TAAs are antigens expressed or 20 over-expressed by tumour cells and could be specific to one or several tumours for example CEA antigen is expressed in colorectal, breast and lung cancers. Sgroi et al (1999) identified several genes differentially expressed in invasive and metastatic carcinoma cells with combined use of laser capture microdissection and cDNA microarrays. Several delivery systems like DNA or viruses could be used for therapeutic vaccination against human cancers (Bonnet et al, 2000) and 25 can elicit immune responses and also break immune tolerance against TAAs. Tumour cells can be rendered more immunogenic by inserting transgenes encoding T cell co-stimulatory molecules such as B7.1 or cytokines such as IFN-y, IL2, or GM-CSF, among others. Co-expression of a TAA and a cytokine or a co-stimulatory molecule has also been shown to be useful in developing effective therapeutic vaccines (Hodge et al, 95, Bronte et al, 1995, Chamberlain et al, 1996). 30 There is a need in the art for reagents and methodologies useful in stimulating an immune response to prevent or treat cancers. The present invention provides such reagents and methodologies which overcome many of the difficulties encountered by others in attempting to treat cancer. 1 SUMMARY OF THE INVENTION The present invention provides an, immunogenic target for administration to a patient to prevent and / or treat cancer. In particular, the immunogenic target is a tumor antigen ("TA") and / or an angiogenesis-associated antigen ("AA"). In one embodiment, the immunogenic target is 5 encoded by SEQ ID NO.: 5 or has the amino acid sequence of SEQ ID NO.: 6. In certain embodiments, the TA and / or AA are administered to a patient as a nucleic acid contained within a plasmid or other delivery vector, such as a recombinant virus. The TA and / or AA may also be administered in combination with additional tumor antigens (i.e., SEQ ID NOS.: 1-4) and / or an immune stimulator, such as a co-stimulatory molecule or adjuvant. 10 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1. BFA4 cDNA sequence. Figure 2. BFA4 amino acid sequence. Figure 3. BCY1 nucleotide (A) and amino acid (B) sequences. 15 Figure 4. BFA5 cDNA sequence. Figure 5. BFA5 amino acid sequence. DETAILED DESCRIPTION The present invention provides reagents and methodologies useful for treating and / or 20 preventing cancer. All references cited within this application are incorporated by reference. In one embodiment, the present invention relates to the induction or enhancement of an immune response against one or more tumor antigens ("TA") to prevent and / or treat cancer. In certain embodiments, one or more TAs may be combined. In preferred embodiments, the immune response results from expression of a TA in a host cell following administration of a 25 nucleic acid vector encoding the tumor antigen or the tumor antigen itself in the form of a peptide or polypeptide, for example. As used herein, an "antigen" is a molecule such as a polypeptide or a portion thereof that produces an immune response in a host to whom the antigen has been administered. The immune response may include the production of antibodies that bind to at least one epitope of the antigen 30 and / or the generation of a cellular immune response against cells expressing an epitope of the antigen. The response may be an enhancement of a current immune response by, for example, causing increased antibody production, production of antibodies with increased affinity for the antigen, or an increased or more effective cellular response (i.e., increased T cells or T cells with 2 higher anti-tumor activity). An antigen that produces an immune response may. alternatively be referred to as being immunogenic or as an immunogen. In describing the present invention, a TA may be referred to as an "immunogenic target". TA includes both tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs), 5 where a cancerous cell is the source of the antigen. A TAA is an antigen that is expressed on the surface of a tumor cell in higher amounts than is observed on normal cells or an antigen that is expressed on normal cells during fetal development. A TSA is an antigen that is unique to tumor cells and is not expressed on normal cells. TA further includes TAAs or TSAs, antigenic fragments thereof, and modified versions that retain their antigenicity. 10 TAs are typically classified into five categories according to their expression pattern, function, or genetic origin: cancer-testis (CT) antigens (i.e., MAGE, NY-ESO-1); melanocyte differentiation antigens (i.e., Melan A/MART-1, tyrosinase, gp100); mutational antigens (i.e., MUM-1, p53, CDK-4); overexpressed 'self' antigens (i.e., HER-2/neu, p53); and, viral antigens (i.e., HPV, EBV). For the purposes of practicing the present invention, a suitable TA is any TA is that induces or enhances an anti-tumor immune response in a host in whom the TA is expressed. Suitable TAs include, for example, gplOO (Cox et al., Science, 264:716-719 (1994)), MART 1/Melan A (Kawakami et al., J. Exp. Med., 180:347-352 (1994)), gp75 (TRP-1) (Wang et aL, J. Exp. Med., 186:1131-1140 (1996)), tyrosinase (Wolfel et al., Eur. J. Immunol., 24:759-764 (1994); WO 200175117; WO 200175016; WO 200175007), NY-ESO-1 (WO 98/14464; WO 20 99/18206), melanoma proteoglycan (Hellstrom et al., J. Immunol., 130:1467-1472 (1983)), MAGE family antigens (i.e., MAGE-1, 2,3,4,6,12, 51; Van der Bruggen et al., Science, 254:1643 1647 (1991); U.S. Pat. Nos. 6,235,525; CN 1319611), BAGE family antigens (Boel et al., Immunity, 2:167-175 (1995)), GAGE family antigens (i.e., GAGE-1,2; Van den Eynde et al., J. Exp. Med., 182:689-698 (1995); U.S. Pat. No. 6,013,765), RAGE family antigens (i.e., RAGE-1; 25 Gaugler et at., Immunogenetics, 44:323-330 (1996); U.S. Pat. No. 5,939,526), N acetylglucosaninyltransferase-V (Guilloux et at., J. Exp. Med., 183:1173-1183 (1996)), p 15 (Robbins et al., J. Immunol. 154:5944-5950 (1995)), B-catenin (Robbins et al., J. Exp. Med., 183:1185-1192 (1996)), MUM-1 (Coulie et al., Proc. Nati. Acad. Sci. USA, 92:7976-7980 (1995)), cyclin dependent kinase-4 (CDK4) (Wolfel et al., Science, 269:1281-1284 (1995)), p21 30 ras (Fossum et at., Int. J. Cancer, 56:40-45 (1994)), BCR-abl (Bocchia et al., Blood, 85:2680 2684 (1995)), p53 (Theobald et al., Proc. Natl. Acad. Sci. USA, 92:11993-11997 (1995)), p 18 5 HER2/neu (erb-B1; Fisk et al., J. Exp. Med., 181:2109-2117 (1995)), epidermal growth factor receptor (EGFR) (Harris et al., Breast Cancer Res. Treat, 29:1-2 (1994)), carcinoembryonic 3 antigens (CEA) (Kwong et al., J. Nati. Cancer Inst., 85:982-990 (1995) U.S. Pat. Nos. 5,756,103; 5,274,087; 5,571,710; 6,071,716; 5,698.,530; 6,045,802; EP 263933; EP 346710; and, EP 784483); carcinoma-associated mutated mucins (i.e., MUC-1 gene products; Jerome et al., J. Inmunol., 151:1654-1662 (1993)); EBNA gene products of EBV (i.e., EBNA-1; Rickinson et al., 5 Cancer Survey's, 13:53-80 (1992)); E7, E6 proteins of human papillomavirus (Ressing et al., J. linmunol, 154:5934-5943 (1995)); prostate specific antigen (PSA; Xue et al., The Prostate, 30:73-78 (1997)); prostate specific membrane antigen (PSMA; Israeli, et al., Cancer Res., 54:1807-1811 (1994)); idiotypic epitopes or antigens, for example, immunoglobulin idiotypes or T cell receptor idiotypes (Chen et al., J. .nmunol., 153:4775-4787 (1994)); KSA (U.S. Patent No. 10 5,348,887), kinesin 2 (Dietz, et al. Biochem Biophys Res Commun 2000 Sep 7;275(3):731-8), HIP-55, TGFP-1 anti-apoptotic factor (Toomey, et al. Br J Biomed Sci 2001;58(3):177-83), tumor protein D52 (Bryne J.A., et al., Genomics, 35:523-532 (1996)), HIFT, NY-BR-i (WO 01/47959), NY-BR-62, NY-BR-75, NY-BR-85, NY-BR-87, NY-BR-96 (Scanlan, M. Serologic and Bioinformatic Approaches to the Identification of Human Tumor Antigens, in Cancer 15 Vaccines 2000, Cancer Research Institute, New York, NY), BFA4 (SEQ ID NOS.: 1 and 2), BCYI (SEQ ID NOS.: 3 and 4), and BFA5 (SEQ ID NOS.: 5 and 6) including "wild-type" (i.e., normally encoded by the genome, naturally-occurring), modified, and mutated versions as well as other fragments and derivatives thereof. Any of these TAs may be utilized alone or in combination with one another in a co-immunization protocol. 20 In certain cases, it may be beneficial to co-immunize patients with both TA and other antigens, such as angiogenesis-associated antigens ("AA"). An AA is an immunogenic molecule (i.e., peptide, polypeptide) associated with cells involved in the induction and / or continued development of blood vessels. For example, an AA may be expressed on an endothelial cell ("EC"), which is a primary structural component of blood vessels. For treatment of cancer, it is 25 preferred that that the AA be found within or near blood vessels that supply a tumor. Immunization of a patient against an AA preferably results in an anti-AA immune response whereby angiogenic processes that occur near or within tumors are prevented and /.or inhibited. Exemplary AAs include, for example, vascular endothelial growth factor (i.e., VEGF; Bemardini, et al. J. Urol., 2001, 166(4): 1275-9; Starnes, et al. J. Thorac. Cardiovasc. Surg., 30 2001, 122(3): 518-23; Dias, et al. Blood, 2002, 99: 2179-2184), the VEGF receptor (i.e., VEGF R, flk-1/KDR; Starnes, et al. J Thorac. Cardiovasc. Surg., 2001, 122(3): 518-23), EPH receptors (i.e., EPHA2; Gerety, et al. 1999, Cell, 4: 403-414), epidermal growth factor receptor (i.e., EGFR; Ciardeillo, et al. Clin. Cancer Res., 2001, 7(10): 2958-70), basic fibroblast growth factor 4 (i.e., bFGF; Davidson, et al. Clin. Exp. Metastasis 2000,18(6): 501-7; Poon, et al. Am J. Surg., 2001, 182(3):298-304), platelet-derived .cell growth factor (i.e., PDGF-B), platelet-derived endothelial cell growth factor (PD-ECGF; Hong, et al. J. Mol. Med., 2001, 8(2):141-8), transforming growth factors (i.e., TGF-a; Hong, et al. J. Mol. Med., 2001, 8(2):141-8), endoglin 5 (Balza, et al. Int. J. Cancer, 2001, 94: 579-585), Id proteins (Benezra, R. Trends Cardiovasc. Med., 2001, 11(6):237-41), proteases such as uPA, uPAR, and matrix metalloproteinases (MMP 2, MMP-9; Djonov, et al. J. Pathol., 2001, 195(2):147-55), nitric oxide synthase (Am. J. Ophthalmol., 2001, 132(4):551-6), aminopeptidase (Rouslhati, E. Nature Cancer, 2: 84-90, 2002), thrombospondins (i.e., TSP-1, TSP-2; Alvarez, et al. Gynecol. Oncol., 2001, 82(2):273-8; Seki, et 10 al. Int. J. Oncol., 2001, 19(2):305-10), k-ras (Zhang, et al. Cancer Res., 2001, 61(16):6050-4), Wnt (Zhang, et al. Cancer Res., 2001, 61(16):6050-4), cyclin-dependent kinases (CDKs; Drug Resist. Updat. 2000, 3(2):83-88), microtubules (Timar, et al. 2001. Path. Oncol. Res., 7(2): 85 94), heat shock proteins (i.e., HSP90 (Timar, supra)), heparin-binding factors (i.e., heparinase; Gohji, et al. Int. J. Cancer, 2001, 95(5):295-301), synthases (i.e., ATP synthase, thymidilate 15 synthase), collagen receptors, integrins (i.e., aup3, xujp5, alp1, a2p1, a5p1), the surface proteolglycan NG2, AAC2-1 (SEQ ID NO.:1), or AAC2-2 (SEQ ID NO.:2), among others, including "wild-type" (i.e., normally encoded by the genome, naturally-occurring), modified, mutated versions as well as other fragments and derivatives thereof. Any of these targets may be suitable in practicing the present invention, either alone or in combination with one another or 20 with other agents. In certain embodiments, a nucleic acid molecule encoding an immunogenic target is utilized. The nucleic acid molecule may comprise or consist of a nucleotide sequence encoding one or more immunogenic targets, or fragments or derivatives thereof, such as that contained in a DNA insert in an ATCC Deposit. The term "nucleic acid sequence" or "nucleic acid molecule" 25 refers to a DNA or 1RNA sequence. The term encompasses molecules forced from any of the known base analogs of DNA and RNA such as, but not limited to 4-acetylcytosine, 8-hydroxy N6-methyladenosine, aziridinyl-cytosine, pseudoisocytosine, 5-(carboxyhydroxylmethyl) uracil, 5-fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5-carboxy methylaminomethyluracil, dihydrouracil, inosine, N6-iso-pentenyladenine, 1-methyladenine, 1 30 methylpseudouracil, 1-methylguanine, 1-methylinosine, 2,2-dimethyl-guanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5 methylaminomethyluracil, 5-methoxyamino-methyl-2-thiouracil, beta-D-mannosylqueosine, 5' methoxycarbonyl-methyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5 5 oxyacetic acid methylester, uracil-5-oxyacetic acid, oxybutoxosine, pseudouracil, queosine, 2 thiocytosine, 5-methyl-2-thiouracil, 2-thiouraoil, 4-thiouracil, 5-methyluracil, N-uracil-5 oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine, among others. 5 An isolated nucleic acid molecule is one that: (1) is separated from at least about 50 percent of proteins, lipids, carbohydrates, or other materials with which it is naturally found when total nucleic acid is isolated from the source cells; (2) is not linked to all or a portion of a polynucleotide to which the nucleic acid molecule is linked in nature; (3) is operably linked to a polynucleotide which it is not linked to in nature; and / or, (4) does not occur in nature as part of a 10 larger polynucleotide sequence. Preferably, the isolated nucleic acid molecule of the present invention is substantially free from any other contaminating nucleic acid molecule(s) or other contaminants that are found in its natural environment that would interfere with its use in polypeptide production or its therapeutic, diagnostic,. prophylactic or research use. As used herein, the term "naturally occurring" or "native" or "naturally found" when used in connection 15 with biological materials such as nucleic acid molecules, polypeptides, host cells, and the like, refers to materials which are found in nature without manipulation by man. Similarly, "non naturally occurring" or "non-native" as used herein refers to a material that is not found in nature or that has been structurally modified or synthesized by man. The identity of two or more nucleic acid or polypeptide molecules is determined by 20 comparing the sequences. As known in the art, "identity" means the degree of sequence relatedness between nucleic acid molecules or polypeptides as determined by the match between the units making up the molecules (i.e., nucleotides or amino acid residues). Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., an 25 algorithm). Identity between nucleic acid sequences may also be determined by the ability of the related sequence to hybridize to the nucleic acid sequence or isolated nucleic acid molecule. In defining such sequences, the term "highly stringent conditions" and "moderately stringent conditions" refer to procedures that permit hybridization of nucleic acid strands whose sequences are complementary, and to exclude hybridization of significantly mismatched nucleic acids. 30 Examples of "highly stringent conditions" for hybridization and washing are 0.015 M sodium chloride, 0.0015 M sodium citrate at 65-68"C or 0.015 M sodium chloride, 0.0015 M sodium citrate, and 50% formamide at 42*C. (see, for example, Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual (2nd ed., Cold Spring Harbor Laboratory, 1989); 6 Anderson et al., Nucleic Acid Hybridisation: A Practical Approach Ch. 4 (IRL Press Limited)). The term "moderately stringent conditions" refers. to conditions under which a DNA duplex with a greater degree of base pair mismatching than could occur under "highly stringent conditions" is able to form. Exemplary moderately stringent conditions are 0.015 M sodium chloride, 0.0015 M 5 sodium citrate at 50-65*C or 0.015 M sodium chloride, 0.0015 M sodium citrate, and 20% formamide at 37-50 0 C. By way of example, moderately stringent conditions of 50'C in 0.015 M sodium ion will allow about a 21% mismatch. During hybridization, other agents may be included in the hybridization and washing buffers for the purpose of reducing non-specific and/or background hybridization. Examples are 0.1% bovine serum albumin, 0.1% polyvinyl 10 pyrrolidone, 0.1% sodium pyrophosphate, 0.1% sodium dodecylsulfate, NaDodSO4, (SDS), ficoll, Denhardt's solution, sonicated salmon sperm DNA (or another non-complementary DNA), and dextran sulfate, although other suitable agents can also be used. The concentration and types of these additives can be changed without substantially affecting the stringency of the hybridization conditions. Hybridization experiments are usually carried out at pH 6.8-7.4; 15 however, at typical ionic strength conditions, the rate of hybridization is nearly independent of pH. In certain embodiments of the present invention, vectors are used to transfer a nucleic acid sequence encoding a polypeptide to a cell. A vector is any molecule used to transfer a nucleic acid sequence to a host cell. In certain cases, an expression vector is utilized. An expression 20 vector is a nucleic acid molecule that is suitable for transformation of a host cell and contains nucleic acid sequences that direct and / or control the expression of the transferred nucleic acid sequences. Expression includes, but is not limited to, processes such as transcription, translation, and splicing, if introns are present. Expression vectors typically comprise one or more flanking sequences operably linked to a heterologous nucleic acid sequence encoding a polypeptide. 25 Flanking sequences may be homologous (i.e., from the same species and / or strain as the host cell), heterologous (i.e., from a species other than the host cell species or strain), hybrid (i.e., a combination of flanking sequences from more than one source), or synthetic, for example. A flanking sequence is preferably capable of effecting the replication, transcription and / or translation of the coding sequence and is operably linked to a coding sequence. As used herein, 30 the term operably linked refers to a linkage of polynucleotide elements in a functional relationship. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the coding sequence. However, a flanking sequence need not necessarily be contiguous with the coding sequence, so long as it functions correctly. Thus, for 7 example, intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence may still be considered operably linked to the coding sequence. Similarly, an enhancer sequence may be located upstream or downstream from the coding sequence and affect transcription of the sequence. 5 In certain embodiments, it is preferred that the flanking sequence is a transcriptional regulatory region that drives high-level gene expression in the target cell. The transcriptional regulatory region may comprise, for example, a promoter, enhancer, silencer, repressor element, or combinations thereof. The transcriptional regulatory region may be either constitutive, tissue specific, cell-type specific (i.e., the region is drives higher levels of transcription in a one type of 10 tissue or cell- as compared to another), or regulatable (i.e., responsive to interaction with a compound). The source of a transcriptional regulatory region may be any prokaryotic or eukaryotic organism, any vertebrate or invertebrate organism, or any plant, provided that the flanking sequence functions in a cell by causing transcription of a nucleic acid within that cell. A wide variety of transcriptional regulatory regions may be utilized in practicing the present 15 invention. Suitable transcriptional regulatory regions include, for example, the CMV promoter (i.e., the CMV-immediate early promoter); promoters fi-om eukaryotic genes (i.e., the estrogen inducible chicken ovalbumin gene, the interferon genes, the gluco-corticoid-inducible tyrosine aminotransferase gene, and the thymidine kinase gene); and the major early and late adenovirus 20 gene promoters; the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304 10); the promoter contained in the 3' long terminal repeat (LTR) of Rous sarcoma virus (RSV) (Yamamoto, et al., 1980, Cell 22:787-97); the herpes simplex virus thymidine kinase (HSV-TK) promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1444-45); the regulatory sequences of the metallothionine gene (Brinster et al., 1982, Nature 296:39-42); prokaryotic 25 expression vectors such as the beta-lactamase promoter (Villa-Kamaroff et al., 1978, Proc. NatL. Acad. Sci. U.S.A., 75:3727-31); or the tac promoter (DeBoer et al., 1983, Proc. NatL. Acad. Sci. U.S.A., 80:21-25). Tissue- and / or cell-type specific transcriptional control regions include, for example, the elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639-46; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol. 50:399-409 30 (1986); MacDonald, 1987, Hepatology 7:425-515); the insulin gene control region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-22); the immunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al., 1984, Cell 38:647-58; Adames et al., 1985, Nature 318:533-38; Alexander et al., 1987, Mol. Cell. Biol., 7:1436-44); the mouse 8 mammary tumor virus control region in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Cell 45:485-95); the albumin gene pontrolregion in liver (Pinkert et al., 1987, Genes and Devel. 1:268-76); the alpha-feto-protein gene control region in liver (Krumlauf et al., 1985, Mol. Cell. Biol., 5:1639-48; Hammer et al., 1987, Science 235:53-58); the alpha 1-antitrypsin gene 5 control region in liver (Kelsey et al., 1987, Genes and Devel. 1:161-71); the beta-globin gene control region in myeloid cells (Mogram et al., 1985, Nature 315:338-40; Kollias et al., 1986, Cell 46:89-94); the myelin basic protein gene control region in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-12); the myosin light chain-2 gene control region in skeletal muscle (Sani, 1985, Nature 314:283-86); the gonadotropic releasing hormone gene control region 10 in the hypothalamus (Mason et al., 1986, Science 234:1372-78), and the tyrosinase promoter in melanoma- cells (Hart, 1. Semin Oncol 1996 Feb;23(1):154-8; Siders, et al. Cancer Gene Ther 1998 Sep-Oct;5(5):2 8 1- 9 1), among others. Inducible promoters that are activated in the presence of a certain compound or condition such as light, heat, radiation, tetracycline, or heat shock proteins, for example, may also be utilized (see, for example, WO 00/10612). Other suitable 15 promoters are known in the art. As described above, enhancers may also be suitable flanking sequences. Enhancers are cis-acting elements of DNA, usually about 10-300 bp in length, that act on the promoter to increase transcription. Enhancers are typically orientation- and position-independent, having been identified both 5' and 3' to controlled coding sequences. Several enhancer sequences 20 available from mammalian genes are known (i.e., globin, elastase, albumin, alpha-feto-protein and insulin). Similarly, the SV40 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers are useful with eukaryotic promoter sequences. While an enhancer may be spliced into the vector at a position 5' or 3' to nucleic acid coding sequence, it is typically located at a site 5' from the promoter. Other suitable enhancers are 25 known in the art, and would be applicable to the present invention. While preparing reagents of the present invention, cells may need to be transfected or transformed. Transfection refers to the uptake of foreign or exogenous DNA by a'cell, and a cell has been transfected when the exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques are well known in the art (i.e., Graham et al., 1973, Virology 30 52:456; Sambrook et al., Molecular Cloning, A Laboratory Manual (Cold Spring Harbor Laboratories, 1989); Davis et al., Basic Methods in Molecular Biology (Elsevier, 1986); and Chu et al., 1981, Gene 13:197). Such techniques can be used to introduce one or more exogenous DNA moieties into suitable host cells. 9 In certain embodiments, it is preferred that transfection of a cell results in transformation of that cell. A cell is transformed when -there is a change in a characteristic of the cell, being transformed when it has been modified to contain a new nucleic acid. Following transfection, the transfected nucleic acid may recombine with that of the cell by physically integrating into a 5 chromosome of the cell, may be maintained transiently as an episornal element without being replicated, or may replicate independently as a plasmid. A cell is stably transformed when the nucleic acid is replicated with the division of the cell. The present invention further provides isolated immunogenic targets in polypeptide form. A polypeptide is considered isolated where it: (1) has been separated from at least about 50 10 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is naturally found when isolated from the source cell; (2) is not linked (by covalent or noncovalent interaction) to all or a portion of a polypeptide to which the "isolated polypeptide" is linked in nature; (3) is operably linked (by covalent or noncovalent interaction) to a polypeptide with which it is not linked in nature; or, (4) does not occur in nature. Preferably, the isolated 15 polypeptide is substantially free from any other contaminating polypeptides or other contaminants that are found in its natural environment that would interfere with its therapeutic, diagnostic, prophylactic or research use. Immunogenic target polypeptides may be mature polypeptides, as defined herein, and may or may not have an amino terminal methionine residue, depending on the method by which they 20 are prepared. Further contemplated are related polypeptides such as, for example, fragments, variants (i.e., allelic, splice), orthologs, homologues, and derivatives, for example, that possess at least one characteristic or activity (i.e., activity, antigenicity) of the immunogenic target. Also related are peptides, which refers to a series of contiguous amino acid residues having a sequence corresponding to at least a portion of the polypeptide from which its sequence is derived. In 25 preferred embodiments, the peptide comprises about 5-10 amino acids, 10-15 amino acids, 15-20 amino acids, 20-30 amino acids, or 30-50 amino acids. In a more preferred embodiment, a peptide comprises 9-12 amino acids, suitable for presentation upon Class I MHC molecules, for example. A fragment of a nucleic acid or polypeptide comprises a truncation of the sequence (i.e., 30 nucleic acid or polypeptide) at the amino terminus (with or without a leader sequence) and / or the carboxy terminus. Fragments may also include variants (i.e., allelic, splice), orthologs, homologues, and other variants having one or more amino acid additions or substitutions or internal deletions as compared to the parental sequence. In preferred embodiments, truncations 10 and/or deletions comprise about 10 amino acids, 20 amino acids, 30 amino acids, 40 amino acids, 50 amino acids, or more. The polypeptide fragments so produced will comprise about 10 amino acids, 25 amino acids, 30 amino acids, 40 amino acids, 50 amino acids, 60 amino acids, 70 amino acids, or more. Such polypeptide fragments may optionally comprise an amino terminal 5 methionine residue. It will be appreciated that such fragments can be used, for example, to generate antibodies or cellular immune responses to immunogenic target polypeptides. A variant is a sequence having one or more sequence substitutions, deletions, and/or additions as compared to the subject sequence. Variants may be naturally occurring or artificially constructed. Such variants may be prepared from the corresponding nucleic acid molecules. In 10 preferred embodiments, the variants have from I to 3, or from I to 5, or from I to 10, or from I to 15, or from 1 to 20, or from 1 to 25, or from 1 to 30, or from 1 to 40, or from I to 50, or more than 50 amino acid substitutions, insertions, additions and/or deletions. An allelic variant is one of several possible naturally-occurring alternate forms of a gene occupying a given locus on a chromosome of an organism or a population of organisms. A splice 15 variant is a polypeptide generated from one of several RNA transcript resulting from splicing of a primary transcript. An ortholog is a similar nucleic acid or polypeptide sequence from another species. For example, the mouse and human versions of an immunogenic target polypeptide may be considered orthologs of each other. A derivative of a sequence is one that is derived from a parental sequence those sequences having substitutions, additions, deletions, or chemically 20 modified variants. Variants may also include fusion proteins, which refers to the fusion of one or more first sequences (such as a peptide) at the amino or carboxy terminus of at least one other sequence (such as a heterologous peptide). "Similarity" is a concept related to identity, except that similarity refers to a measure of relatedness which includes both identical matches and conservative substitution matches. If two 25 polypeptide sequences have, for example, 10/20 identical amino acids, and the remainder are all non-conservative substitutions, then the percent identity and similarity would both be 50%. If in the same example, there are five more positions where there are conservative substitutions, then the percent identity remains 50%, but the percent similarity would be 75% (15/20). Therefore, in cases where there are conservative substitutions, the percent similarity between two polypeptides 30 will be higher than the percent identity between those two polypeptides. Substitutions may be conservative, or non-conservative, or any combination thereof. Conservative amino acid modifications to the sequence of a polypeptide (and the corresponding modifications to the encoding nucleotides) may produce polypeptides having functional and 11 chemical characteristics similar to those of a parental polypeptide. For example, a "conservative amino acid substitution" may involve a substitution of a native amino acid residue with a non native residue such that there is little or no effect on the size, polarity, charge, hydrophobicity, or hydrophilicity of the amino acid residue at that position and, in particlar, does not result in 5 decreased immunogenicity. Suitable conservative amino acid substitutions are shown in Table I. Table I Original Exemplary Substitutions Preferred Residues Substitutions Ala Val, Leu, Ile Val Arg Lys, Gln, Asn Lys Asn Gln Gin Asp Glu Glu Cys Ser, Ala Ser Gln Asn Asn Glu Asp Asp Gly Pro, Ala Ala His Asn, Gin, Lys, Arg Arg Ile Leu, Val, Met, Ala, Phe, Norleucine Leu Leu Norleucine, Ile, Val, Met, Ala, Phe Ile Lys Arg, 1,4 Diamino-butyric Acid, Gln, Asn Arg Met Leu, Phe, lie Leu Phe Leu, Val, Ile, Ala, Tyr Leu Pro Ala Gly Ser Thr, Ala, Cys Thr Thr Ser Ser Trp Tyr, Phe Tyr Tyr Trp, Phe, Thr, Ser Phe Val Ile, Met, Leu, Phe, Ala, Norleucine Leu A skilled artisan will be able to determine suitable variants of polypeptide using well known techniques. For identifying suitable areas of the molecule that may be changed without 10 destroying biological activity (i.e., MHC binding, immunogenicity), one skilled in the art may target areas not believed to be important for that activity. For example, when similar polypeptides with similar activities from the same species or fi-om other species are known, one skilled in the art may compare the amino acid sequence of a polypeptide to such similar polypeptides. By performing such analyses, one can identify residues and portions of the 15 molecules that are conserved among similar polypeptides. It will be appreciated that changes in areas of the molecule that are not conserved relative to such similar polypeptides would be less likely to adversely affect the biological activity and/or structure of a polypeptide. Similarly, the residues required for binding to MHC are known, and may be modified to improve binding. 12 However, modifications resulting in decreased binding to MHC will not be appropriate in most situations. One skilled in the art would also know that, even in relatively conserved regions, one may substitute chemically similar amino acids for the naturally occurring residues while retaining activity. Therefore, even areas that may be important for biological activity or for structure may 5 be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure. Other preferred polypeptide variants include glycosylation variants wherein the number and/or type of glycosylation sites have been altered compared to the subject amino acid sequence. In one embodiment, polypeptide variants comprise a greater or a lesser number of N-linked 10 glycosylation sites than the subject amino acid sequence. An N-linked glycosylation site is characterized by. the sequence Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X may be any amino acid residue except proline. The substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, substitutions that eliminate this sequence will remove an is existing N-linked carbohydrate chain. Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created. To affect 0-linked glycosylation of a polypeptide, one would modify serine and / or threonine residues. Additional preferred variants include cysteine variants, wherein one or more cysteine 20 residues are deleted or substituted with another amino acid (e.g., serine) as compared to the subject amino acid sequence set. Cysteine variants are useful when polypeptides must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines. 25 In other embodiments, the isolated polypeptides of the current invention include fusion polypeptide segments that assist in purification of the polypeptides. Fusions can be made either at the amino terminus or at the carboxy terminus of the subject polypeptide variant thereof. Fusions may be direct with no linker or adapter molecule or may be through a linker or adapter molecule. A linker or adapter molecule may be one or more amino acid residues, typically from about 20 to 30 about 50 amino acid residues. A linker or adapter molecule may also be designed with a cleavage site for a DNA restriction endonuclease or for a protease to allow for the separation of the fused moieties. It will be appreciated that once constructed, the fusion polypeptides can be derivatized according to the methods described herein. Suitable fusion segments include, among others, 13 metal binding domains (e.g., a poly-histidine segment), immunoglobulin binding domains (i.e., Protein A, Protein G, T cell, B cell, Fc receptor, or complement protein antibody-binding domains), sugar binding domains (e.g., a maltose binding domain), and/or a "tag" domain (i.e., at least a portion of a-galactosidase, a strep tag peptide, a T7 tag peptide, a FLAG peptide, or other 5 domains that can be purified using compounds that bind to the domain, such as monoclonal antibodies). This tag is typically fused to the polypeptide upon expression of the polypeptide, and can serve as a means for affinity purification of the sequence of interest polypeptide from the host cell. Affinity purification can be accomplished, for example, by column chromatography using antibodies against the tag as an affinity matrix. Optionally, the tag can subsequently be removed 10 from the purified sequence of interest polypeptide by various means such as using certain peptidases for cleavage. As described below, fusions may also be made between a TA and a co stimulatory components such as the chemokines CXC10 (IP-10), CCL7 (MCP-3), or CCL5 (RANTES), for example. A fusion motif may enhance transport of an immunogenic target to an MHC processing 15 compartment, such as the endoplasmic reticulum. These sequences, referred to as tranduction or transcytosis sequences, include sequences derived from HIV tat (see Kim et al. 1997 J. Immunol. 159:1666), Drosophila antennapedia (see Schutze-Redelmeier et al. 1996 J. Immunol. 157:650), or human period-I protein (hPERl; in particular, SRRHHCRSKAKRSRHH). In addition, the polypeptide or variant thereof may be fused to a homologous polypeptide 20 to form a homodimer or to a heterologous polypeptide to form a heterodimer. Heterologous peptides and polypeptides include, but are not limited to: an epitope to allow for the detection and/or isolation of a fusion polypeptide; a transmembrane receptor protein or a portion thereof, such as an extracellular domain or a transmembrane and intracellular domain; a ligand or a portion thereof which binds to a transmembrane receptor protein; an enzyme or portion thereof 25 which is catalytically active; a polypeptide or peptide which promotes oligomerization, such as a leucine zipper domain; a polypeptide or peptide which increases stability, such as an inimunoglobulin constant region; and a polypeptide which has a therapeutic activity different from the polypeptide or variant thereof. In certain embodiments, it may be advantageous to combine a nucleic acid sequence 30 encoding an immunogenic target, polypeptide, or derivative thereof with one or more nucleic acid sequences encoding one or more co-stimulatory component(s) such as cell surface proteins, cytokines or chemokines in a composition of the present invention. The co-stimulatory component may be included in the composition as a polypeptide or as a nucleic acid encoding the 14 polypeptide, for example. Suitable co-stimulatory molecules include, for instance, polypeptides that bind members of the CD28 family (i.e., CD28, ICOS; Hiutloff, et al. Nature 1999, 397: 263 265; Peach, et al. JExp Med 1994, 180: 2049-2058) such as the CD28 binding polypeptides B7.1 (CD80; Schwartz, 1992; Chen et al, 1992; Ellis, et al. J. Immunol., 156(8): 2700-9) and B7.2 5 (CD86; Ellis, et al. J. Immunol., 156(8): 2700-9); mutated and derivative B7 molecules (WO 00/66162); polypeptides which bind members of the integrin family (i.e., LFA-l (CD11a / CDI8); Sedwick, et al. J Inmunol 1999, 162: 1367-1375; W6lfing, et al. Science 1998, 282: 2266-2269; Lub, et al. Inmunol Today 1995, 16: 479-483) including members of the ICAM family (i.e., ]CAM-1, -2 or -3); polypeptides which bind CD2 family members (i.e., CD2, 10 signalling lymphocyte activation molecule (CDwl50 or "SLAM"; Aversa, et al. JImmunol 1997, 158: 4036-4044)) such as CD58 (LFA-3; CD2 ligand; Davis, et al. Inmunol Today 1996, 17: 177-187) or SLAM ligands (Sayos, et al. Nature 1998, 395: 462-469); polypeptides which bind heat stable antigen (HSA or CD24; Zhou, et al. Eur J mmnrunol 1997, 27: 2524-2528); polypeptides which bind to members of the TNF receptor (TNFR) family (i.e.,. 15 4-1BB (CD137; Vinay, et al. Semin Immunol 1998, 10: 481-489), OX40 (CD134; Weinberg, et al. Semin Immunol 1998, 10: 471-480; Higgins, et al. J inmunol 1999, 162: 486-493), and CD27 (Lens, et al. Senin Immunol 1998, 10: 491-499)) such as 4-IBBL (4-1BB ligand; Vinay, et al. Senin Iminunol 1998, 10: 481-48; DeBenedette, et al. JlImmunol 1997, 158: 551-559), TNFR associated factor-I (TRAF-1; 4-IBB ligand; Saoulli, et al. J Exp Med 1998, 187: 1849 20 1862, Arch, et al. Mol Cell Biol 1998, 18: 55 8-565), TRAF-2 (4-1 BB and OX40 ligand; Saoulli, et al. J Exp Med 1998, 187: 1849-1862; Oshina, et al. Int Immunol 1998, 10: 517-526, Kawamata, et al. JBiol Chem 1998, 273: 5808-5814), TRAF-3 (4-1BB and OX40 ligand; Arch, et al. Mol Cell Biol 1998, 18: 558-565; Jang, et al. Biochein Biophys Res Commun 1998, 242: 613-620; Kawamata S, et al. J Biol Chem 1998, 273: 5808-5814), OX40L (OX40 ligand; 25 Gramaglia, et al. J Immunol 1998, 161: 6510-6517), TRAF-5 (OX40 ligand; Arch, et al. Mol Cell Biol 1998, 18: 558-565; Kawamata, et al. J Biol Chen 1998, 273: 5808-5814), and CD70 (CD27 ligand; Couderc, et al. Cancer Gene Ther., 5(3): 163-75). CD154 (CD40 ligand or "CD4OL"; Gurunathan, et al. J. Imnunol., 1998, 161: 4563-4571; Sine, et al. Hum. Gene Ther., 2001, 12: 1091-1102) may also be suitable. 30 One or more cytokines may also be suitable co-stimulatory components or "adjuvants", either as polypeptides or being encoded by nucleic acids contained within the compositions of the present invention (Parmiani, et al. Immunol Lett 2000 Sep 15; 74(1): 41-4; Berzofsky, et al. Nature Immunol. 1: 209-219). Suitable cytokines include, for example, interleukin-2 (IL-2) 15 (Rosenberg, et al. Nature Med. 4: 321-327 (1998)), IL-4, IL-7, IL-12 (reviewed by Pardoll, 1992; Harries, et al. J. Gene Med. 2000 Jul-Aug;2(4):243-9; Rao, et al. J. Inmunol. 156: 3357-3365 (1996)), IL-15 (Xin, et al. Vaccine, 17:858-866, 1999), IL-16 (Cruikshank, et al. J. Leuk Biol. 67(6): 757-66, 2000), IL-18 (J. Cancer Res. Clin. Oncol. 2001. 127(12): 718-726), GM-CSF 5 (CSF (Disis, et al. Blood, 88: 202-210 (1996)), tumor necrosis factor-alpha (TNF-a), or interferons such as IFN-ax or rNF-y. Other cytokines may also be suitable for practicing the present invention, as is known in the art. Chemokines may also be utilized. For example, fusion proteins comprising CXCLIO (IP 10) and CCL7 (MCP-3) fused to a tumor self-antigen have been shown to induce anti-tumor 10 immunity (Biragyn, et al. Nature Biotech. 1999, 17: 253-258). The chemokines CCL3 (MIP l a) and CCL5 (RANTES) (Boyer, et al. Vaccine, 1999, 17 (Supp. 2): S53-S64) may also be of use in practicing the present invention. Other suitable chemokines are known in the art. It is also known in the art that suppressive or negative regulatory immune mechanisms may be blocked, resulting in enhanced immune responses. For instance, treatment with anti 15 CTLA-4 (Shrikant, et al. Immunity, 1996, 14: 145-155; Sutmuller, et al. J. Exp. Med., 2001, 194: 823-832), anti-CD25 (Sutmuller, supra), anti-CD4 (Matsui, et al. J. Immunol., 1999, 163: 184 193), the fusion protein IL13Ra2-Fc (Terabe, et al. Nature Immunol., 2000, 1: 515-520), and combinations thereof (i.e., anti-CTLA-4 and anti-CD25, Sutmuller, supra) have been shown to upregulate anti-tumor immune responses and would be suitable in practicing the present 20 invention. Any of these components may be used alone or in combination with other agents. For instance, it has been shown that a combination of CD80, 1CAM-1 and LFA-3 ("TRICOM") may potentiate anti-cancer immune responses (Hodge, et al. Cancer Res. 59: 5800-5807 (1999). Other effective combinations include, for example, IL-12 + GM-CSF (Ahlers, et al. J. Immunol., 25 158: 3947-3958 (1997); Iwasaki, et al. J. hImmunol. 158: 4591-4601 (1997)), IL-12 + GM-CSF + TNF-a (Ahlers, et al. Int. Inmunol. 13: 897-908 (2001)), CD80 + IL-12 (Fruend, et al. Int. J. Cancer, 85: 508-517 (2000); Rao, et al. supra), and CD86 + GM-CSF + IL-12 (Iwasaki, supra). One of skill in the art would be aware of additional combinations useful in carrying out the present invention.In addition, the skilled artisan would be aware of additional reagents or methods 30 that may be used to modulate such mechanisms. These reagents and methods, as well as others known by those of skill in the art, may be utilized in practicing the present invention. Additional strategies for improving the efficiency of nucleic acid-based immunization may also be used including, for example, the use of self-replicating viral replicons (Caley, et al. 16 1999. Vaccine, 17: 3124-2135; Dubensky, et al. 2000. Mol. Med. 6: 723-732; Leitner, et al. 2000. Cancer Res. 60: 51-55), codon optimization (Liu, et al. 2000. Mol. Ther., 1: 497-500; Dubensky, supra; Huang, et al. 2001. J. Virol. 75: 4947-4951), in vivo electroporation (Widera, et al. 2000. J. Immunol. 164: 4635-3640), incorporation of CpG stimulatory motifs (Gurunathan, 5 et al. Ann. Rev. Inmunol., 2000, 18: 927-974; Leitner, supra; Cho, et al. J. Immunol. 168(10):4907-13), sequences for targeting of the endocytic or ubiquitin-processing pathways (Thomson, et al. 1998. J. Virol. 72: 2246-2252; Velders, et al. 2001. J. Immunol. 166: 5366 5373), Marek's disease virus type 1 VP22 sequences (J. Virol. 76(6):2676-82, 2002), prime-boost regimens (Gurunathan, supra; Sullivan, et al. 2000. Nature, 408: 605-609; Hanke, et al. 1998. to Vaccine, 16: 439-445; Amara, et al. 2001. Science, 292: 69-74), and the use of mucosal delivery vectors such as Salmonella (Darji, et al. 1997. Cell, 91: 765-775; Woo, et al. 2001. Vaccine, 19: 2945-2954). Other methods are known in the art, some of which are described below. Chemotherapeutic agents, radiation, anti-angiogenic compounds, or other agents may also be utilized in treating and / or preventing cancer using immunogenic targets (Sebti, et al. 15 Oncogene 2000 Dec 27;19(56):6566-73). For example, in treating metastatic breast cancer, useful chemotherapeutic agents include cyclophosphamide, doxorubicin, paclitaxel, docetaxel, navelbine, capecitabine, and mitomycin C, among others. Combination chemotherapeutic regimens have also proven effective including cyclophosphamide + methotrexate + .5 fluorouracil; cyclophosphamide + doxorubicin + 5-fluorouracil; or, cyclophosphamide + 20 doxorubicin, for example. Other compounds such as prednisone, a taxane, navelbine, mitomycin C, or vinblastine have been utlized for various reasons. A majority of breast cancer patients have estrogen-receptor positive (ER+) tumors and in these patients, endocrine therapy (i.e., tamoxifen) is preferred over chemotherapy. For such patients, tamoxifen or, as a second line therapy, progestins (medroxyprogesterone acetate or megestrol acetate) are preferred. Aromatase 25 inhibitors (i.e., aminoglutethimide and analogs thereof such as letrozole) decrease the availability of estrogen needed to maintain tumor growth and may be used as second or third line endocrine therapy in certain patients. Other cancers may require different chemotherapeutic regimens. For example, metastatic colorectal cancer is typically treated with Camptosar (irinotecan or CPT-11), 5-fluorouracil or 30 leucovorin, alone or in combination with one another. Proteinase and integrin inhibitors such as as the MMP inhibitors marimastate (British Biotech), COL-3 (Collagenex), Neovastat (Aeterna), AG3340 (Agouron), BMS-275291 (Bristol Myers Squibb), CGS 27023A (Novartis) or the integrin inhibitors Vitaxin (Medimmune), or MED1522 (Merck KgaA) may also be suitable for 17 . use. As such, immunological targeting of immunogenic targets associated with colorectal cancer could be performed in combination with a treatment using those chemotherapeutic agents. Similarly, chemotherapeutic agents used to treat other types of cancers are well-known. in the art and may be combined with the immunogenic targets described herein. 5 Many anti-angiogenic agents are known in the art and would be suitable for co administration with the immunogenic target vaccines (see, for example, Timar, et al. 2001. Pathology Oncol. Res., 7(2): 85-94). Such agents include, for example, physiological agents such as growth factors (i.e., ANG-2, NK1,2,4 (HGF), transforming growth factor beta (TGF-D)), cytokines (i.e., interferons such as IFN-c., -s, -y, platelet factor 4 (PF-4), PR-39), proteases (i.e., 10 cleaved AT-III, collagen XVIII fragment (Endostatin)), HmwKallikrein-d5 plasmin fragment (Angiostatin), prothrombin-F1-2, TSP-1), protease inhibitors (i.e., tissue inhibitor of metalloproteases such as TIMP-1, -2, or -3; maspin; plasminogen activator-inhibitors such as PAI-1; pigment epithelium derived factor (PEDF)), Tumstatin (available through ILEX, Inc.), antibody products (i.e., the collagen-binding antibodies HUIV26, HU177, XL313; anti-VEGF; 15 anti-integrin (i.e., Vitaxin, (Lxsys))), and glycosidases (i.e., heparinase-I, -III). "Chemical" or modified physiological agents known or believed to have anti-angiogenic potential include, for example, vinblastine, taxol, ketoconazole, thalidomide, dolestatin, combrestatin A, rapamycin (Guba, et al. 2002, Nature Med., 8: 128-135), CEP-7055 (available from Cephalon, Inc.), flavone acetic acid, Bay 12-9566 (Bayer Corp.), AG3340 (Agouron, Inc.), CGS 27023A (Novartis), 20 tetracylcine derivatives (i.e., COL-3 (Collagenix, Inc.)), Neovastat (Aetema), BMS-275291 (Bristol-Myers Squibb), low dose 5-FU, low dose methotrexate (MTX), irsofladine, radicicol, cyclosporine, captopril, celecoxib, D45152-sulphated polysaccharide, cationic protein (Protamine), cationic peptide-VEGF, Suramin (polysulphonated napthyl urea), compounds that interfere with the function or production of VEGF (i.e., SU5416 or SU6668 (Sugen), 25 PTK787/ZK22584 (Novartis)), Distamycin A, Angiozyme (ribozyme), isoflavinoids, staurosporine derivatives, genistein, EMD121974 (Merck KcgaA), tyrphostins, isoquinolones, retinoic acid, carboxyamidotriazole, TNP-470, octreotide, 2-methoxyestradiol, aminosterols (i.e., squalamine), glutathione analogues (i.e., N-acteyl-L-cysteine), combretastatin A-4 (Oxigene), Eph receptor blocking agents (Nature, 414:933-938, 2001), Rh-Angiostatin, Rh-Endostatin (WO 30 01/93897), cyclic-RGD peptide, accutin-disintegrin, benzodiazepenes, humanized anti-avb3 Ab, Rh-PAI-2, amiloride, p-amidabenzamidine, anti-uPA ab, anti-uPAR Ab, L-phanylalanin-N methylamides (i.e., Batimistat, Marimastat), AG3340, and minocycline. Many other suitable agents are known in the art and would suffice in practicing the present invention. 18 The present invention may also be utilized in combination with "non-traditional" methods of treating cancer. For example, it has recently been demonstrated that administration of certain anaerobic bacteria may assist in slowing tumor growth. In one study, Clostridium noyi was modified to eliminate a toxin gene carried on a phage episome and administered to mice with 5 colorectal tumors (Dang, et al. P.N.A.S. USA, 98(26): 15155-15160, 2001). In combination with chemotherapy, the treatment was shown to cause tumor necrosis in the animals. The reagents and methodologies described in this application may be combined with such treatment methodologies. Nucleic acids encoding immunogenic targets may be administered to patients by any of several available techniques. Various viral vectors that have been successfully utilized for 10 introducing a nucleic acid to a host include retrovirus, adenovirus, adeno-associated virus (AAV), herpes virus, and poxvirus, among others. It is understood in the art that many such viral vectors are available in the art. The vectors of the present invention may be constructed using standard recombinant techniques widely available to one skilled in the art. Such techniques may be found in common molecular biology references such as Molecular Cloning: A Laboratory Manual 15 (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press), Gene Expression Technology (Methods in Enzymology, Vol. 185, edited by D. Goeddel, 1991. Academic Press, San Diego, CA), and PCR Protocols: A Guide to Methods and Applications (Innis, et al. 1990. Academic Press, San Diego, CA). Preferred retroviral vectors are derivatives of lentivirus as well as derivatives of murine or 20 avian retroviruses. Examples of suitable retroviral vectors include, for example, Moloney urine leukemia virus (MoMuLV), Harvey urine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), SIV, BIV, HIV and Rous Sarcoma Virus (RSV). A number of retroviral vectors can incorporate multiple exogenous nucleic acid sequences. As recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance 25 can be provided by, for example, helper cell lines encoding retrovirus structural genes. Suitable helper cell lines include T2, PA317 and PA12, among others. The vector virions produced using such cell lines may then be used to infect a tissue cell line, such as NIH 3T3 cells, to produce large quantities of chimeric retroviral virions. Retroviral vectors may be administered by traditional methods (i.e., injection) or by implantation of a "producer cell line" in proximity to the 30 target cell population (Culver, K., et al., 1994, Hum. Gene Ther., 5 (3): 343-79; Culver, K., et al., Cold Spring Harb. Symp. Quant. Biol., 59: 685-90); Oldfield, E., 1993, Hum. Gene Ther., 4 (1): 39-69). The producer cell line is engineered to produce a viral vector and releases viral particles in the vicinity of the target cell. A portion of the released viral particles contact the target cells 19 and infect those cells, thus delivering a nucleic acid of the present invention to the target cell. Following infection of the target cell, expression of the nucleic acid of the vector occurs. Adenoviral vectors have proven especially useful for gene transfer into eukaryotic cells (Rosenfeld, M., el al., 199.1, Science, 252 (5004): 431-4; Crystal, R., et al., 1994, Nat. Genet., 8 5 (1): 42-51), the study eukaryotic gene expression (Levrero, M., et al., 1991, Gene, 101 (2): 195 202), vaccine development (Graham, F. and Prevec, L., 1992, Biotechnology, 20: 363-90), and in animal models (Stratford-Perricaudet, L., et al., 1992, Bone Marrow Transplant., 9 (Suppl. 1): 151-2 ; Rich, D., et al., 1993, Hum. Gene Ther., 4 (4): 461-76). Experimental routes for administrating recombinant Ad to different tissues in vivo have included intratracheal instillation 10 (Rosenfeld, M., el al., 1992, Cell, 68 (1): 143-55) injection into muscle (Quantin, B., et al., 1992, Proc. Natl. Acad. Sci. US.A., 89 (7): 2581-4), peripheral intravenous injection (Herz, J., and Gerard, R., 1993, Proc. Natl. Acad. Sci. U.S.A., 90 (7): 2812-6) and stereotactic inoculation to brain (Le Gal La Salle, G., et al., 1993, Science, 259 (5097): 988-90), among others. Adeno-associated virus (AAV) demonstrates high-level infectivity, broad host range and 15 specificity in integrating into the host cell genome (Hermonat, P., et al., 1984, Proc. Natl. A cad. Sci. US.A., 81 (20): 6466-70). And Herpes Simplex Virus type-I (HSV-1) is yet another attractive vector system, especially for use in the nervous system because of its neurotropic property (Geller, A., et al., 1991, Trends Neurosci., 14 (10): 428-32; Glorioso, et al., 1995, Mol. Biotechnol., 4 (1): 87-99; Glorioso, et al., 1995, Annu. Rev. Microbiol., 49: 675-710). 26 Poxvirus is another useful expression vector (Smith, et al. 1983, Gene, 25 (1): 21-8; Moss, et al, 1992, Biotechnology, 20: 345-62; Moss, et al, 1992, Curr. Top. Microbiol. Inmunol., 158: 25-38; Moss, et al. 1991. Science, 252: 1662-1667). Poxviruses shown to be useful include vaccinia, NYVAC, avipox, fowlpox, canarypox, ALVAC, and ALVAC(2), among others. NYVAC (vP866) was derived from the Copenhagen vaccine strain of vaccinia virus by 25 deleting six nonessential regions of the genome encoding known or potential virulence factors (see, for example, U.S. Pat. Nos. 5,364,773 and 5,494,807). The deletion loci were also engineered as recipient loci for the insertion of foreign genes. The deleted regions are: thymidine kinase gene (TK; J2R); hemorrhagic region (u; B13R+BI4R); A type inclusion body region (ATI; A26L); hemagglutinin gene (HA; A56R); host range gene region (C7L-KlL); and, large 30 subunit, ribonucleotide reductase (14L). NYVAC is a genetically engineered vaccinia virus strain that was generated by the specific deletion of eighteen open reading frames encoding gene products associated with virulence and host range. NYVAC has been show to be useful for expressing TAs (see, for example, U.S. Pat. No. 6,265,189). NYVAC (vP866), vP994, vCP205, 20 vCP1433, placZH6H4Lreverse, pMPC6H6K3E3 and pC3H6FHVB were also deposited with the ATCC under the terms of the Budapest Treaty, accession numbers VR-2559, VR-2558, VR-2557, VR-2556, ATCC-97913, ATCC-97912, and ATCC-97914, respectively. ALVAC-based recombinant viruses (i.e., ALVAC-1 and ALVAC-2) are also suitable for 5 use in practicing the present invention (see, for example, U.S. Pat. No. 5,756,103). ALVAC(2) is identical to ALVAC(1) except that ALVAC(2) genome comprises the vaccinia E3L and K3L genes under the control of vaccinia promoters (U.S. Pat. No. 6,130,066; Beattie et al., 1995a, 1995b, 1991; Chang et al., 1992; Davies et al., 1993). Both ALVAC(1) and ALVAC(2) have been demonstrated to be useful in expressing foreign DNA sequences, such as TAs (Tartaglia et 10 al., 1993 a,b; U.S. Pat. No. 5,833,975). ALVAC was deposited under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, USA, ATCC accession number VR-2547. . Another useful poxvirus vector is TROVAC. TROVAC refers to an attenuated fowlpox that was a plaque-cloned isolate derived from the FP-1 vaccine strain of fowlpoxvirus which is is licensed for vaccination of I day old chicks. TROVAC was likewise deposited under the terms of the Budapest Treaty with the ATCC, accession number 2553. "Non-viral" plasmid vectors may also be suitable in practicing the present invention. Preferred plasmid vectors are compatible with bacterial, insect, and / or mammalian host cells. Such vectors include, for example, PCR-II, pCR3, and pcDNA3.1 (Invitrogen, San Diego, CA), 20 pBSII (Stratagene, La Jolla, CA), pET15 (Novagen, Madison, WI), pGEX (Pharmacia Biotech, Piscataway, NJ), pEGFP-N2 (Clontech, Palo Alto, CA), pETL (BlueBacI, Invitrogen), pDSR alpha (PCT pub. No. WO 90/14363) and pFastBacDual (Gibco-BRL, Grand Island, NY) as well as Bluescript plasmid derivatives (a high copy number COLEl-based phagemid, Stratagene Cloning Systems, La Jolla, CA), PCR cloning plasmids designed for cloning Taq-amplified PCR 25 products (e.g., TOPOTM TA cloning kit, PCR2.1 plasmid derivatives, Invitrogen, Carlsbad, CA). Bacterial vectors may also be used with the current invention. These vectors include, for example, Shigella, Salmonella, Vlibrio cholerae, Lactobacillus, Bacille calmette guarin (BCG), and Streptococcus (see for example, WO 88/6626; WO 90/0594; WO 91/13157; WO 92/1796; and WO 92/21376). Many other non-viral plasmid expression vectors and systems are known in 30 the art and could be used with the current invention. Suitable nucleic acid delivery techniques include DNA-ligand complexes, adenovirus ligand-DNA complexes, direct injection of DNA, CaPO 4 precipitation, gene gun techniques, electroporation, and colloidal dispersion systems, among others. Colloidal dispersion systems 21 include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. The preferred colloidal system of this invention is a liposome, which are artificial membrane vesicles useful as delivery vehicles in vitro and in vivo. RNA, DNA and intact virions can be encapsulated within 5 the aqueous interior and be delivered to cells in a biologically active form (Fraley, R., el al., 1981, Trends Biochem7. Sci., 6: 77). The composition of the liposome is usually a combination of phospholipids, particularly high-phase-transition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and the presence of 10 divalent cations. Examples of lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides. Particularly useful are diacylphosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated. Illustrative phospholipids include egg 15 phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine. An immunogenic target may also be administered in combination with one or more adjuvants to boost the immune response. Exemplary adjuvants are shown in Table 11 below: Table II Types of Immunologic Adjuvants 20 Type of Adjuvant General Examples Specific Examples/References Gel-type Aluminum hydroxide/phosphate ("alum (Aggerbeck and Heron, 1995) adjuvants") Calcium phosphate (Relyveld, 1986) Microbial Muramyl dipeptide (MDP) (Chedid et al., 1986) Bacterial exotoxins Cholera toxin (CT), E.coli labile toxin (LT)(Freytag and Clements, 1999) Endotoxin-based adjuvants Monophosphoryl lipid A (MPL) (Ulrich and Myers, 1995) Other bacterial CpG oligonucleotides (Corral and Petray, 2000), BCG sequences (Krieg, et al. Nature, 374:576), tetanus toxoid (Rice, et al. J. hnmunol. , 2001, 167: 1558-1565) Particulate Biodegradable (Gupta et al., 1998) Polymer microspheres immunostimulatory complexes (Morein and Bengtsson, 1999) (ISCOMs) Liposomes (Wassef et al., 1994) Oil-emulsion Freund's incomplete adjuvant (Jensen et al., 1998) and Microfluidized emulsions MF59 (Ott et al., 1995) 22 surfactant- SAF (Allison and Byars, 1992) based (Allison, 1999) adjuvants Saponins ' QS-21 (Kensil, 1996) Synthetic Muramyl peptide derivatives Murabutide (Lederer, 1986) Threony-MDP (Allison, 1997) Nonionic block copolymers L121 (Allison, 1999) Polyphosphazene (PCPP) (Payne et al., 1995) Synlhctic polynucleotides Poly A:U, Poly l:C (Johnson, 1994) Thalidomide derivatives CC-4047/ACTIMID (J. lrnmunol., 168(10):4914-9) The immunogenic targets of the present invention may also be used to generate antibodies for use in screening assays or for immunotherapy, which are another aspect of the present invention. Other uses would be apparent to one of skill in the art. The term "antibody" includes 5 antibody fragments, as are known in the art, including Fab, Fab 2 , single chain antibodies (Fv for example), humanized antibodies, chimeric antibodies, human antibodies, produced by several methods as are known in the art. Methods of preparing and utilizing various types of antibodies are well-known to those of skill in the art and would be suitable in practicing the present invention (see, for example, 10 Harlow, et al. Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; Harlow, et al. Using Antibodies: A Laboratory Manual, Portable Protocol No. 1, 1998; Kohler and Milstein, Nature, 256:495 (1975)); Jones et al. Nature, 321:522-525 (1986); Riechrnann et al. Nature, 332:323-329 (1988); Presta (Curr. Op. Struct. Biol., 2:593-596 (1992); Verhoeyen et al. (Science, 239:1534-1536 (1988); Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., 15 J. Mol. Biol., 222:581 (1991); Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boerner et al., J. Immunol., 147(1):86-95 (1991); Marks et al., Bio/Technology 10, 779-783 (1992); Lonberg et al., Nature 368 856-859 (1994); Morrison, Nature 368 812-13 (1994); Fishwild et al., Nature Biotechnology 14, 845-51 (1996); Neuberger, Nature Biotechnology 14, 826 (1996); Lonberg and Huszar, Intern. Rev. Immunol. 13 65-93 (1995); as 20 well as U.S. Pat. Nos. 4,816,567; 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and, 5,661,016). The antibodies or derivatives therefrom may also be conjugated to therapeutic moieties such as cytotoxic drugs or toxins, or active fragments thereof such as diptheria A chain, exotoxin A chain, ricin A chain, abrin A chain, curcin, crotin, phenomycin, enomycin, among others. Cytotoxic agents may also include radiochemicals. Antibodies and their derivatives may 25 be incorporated into compositions of the invention for use in vitro or in vivo. Nucleic acids, proteins, or derivatives thereof representing an immunogenic target may be used in assays to determine the presence of a disease state in a patient, to predict prognosis, or to 23 determine the effectiveness of a chemotherapeutic or other treatment regimen. Expression profiles, performed as is known in the' art, may be used to determine the relative level of expression of the immunogenic target. The level of expression may then be con-elated with base levels to determine whether a particular disease is present within the patient, the patient's 5 prognosis, or whether a particular treatment regimen is effective. For example, if the patient is being treated with a particular chemotherapeutic regimen, a decreased level of expression of an iminunogenic target in the patient's tissues (i.e., in peripheral blood) may indicate the regimen is decreasing the cancer load in that host. Similarly, if the level of expresssion is increasing, another therapeutic modality may need to be utilized. In one embodiment, nucleic acid probes 10 corresponding to a nucleic acid encoding an immunogenic target may be attached to a biochip, as is known in the art, for the detection and quantification of expression in the host. It is also possible to use nucleic acids, proteins, derivatives therefrom, or antibodies thereto as reagents in drug screening assays. The reagents may be used to ascertain the effect of a drug candidate on the expression of the immunogenic target in a cell line, or a cell or tissue of a 15 patient. The expression profiling technique may be combined with high throughput screening techniques to allow rapid identification of useful compounds and monitor the effectiveness of treatment with a drug candidate (see, for example, Zlokarnik, et al., Science 279, 84-8 (1998)). Drug candidates may be chemical compounds, nucleic acids, proteins, antibodies, or derivatives . therefrom, whether naturally occurring or synthetically derived. Drug candidates thus identified 20 may be utilized, among other uses, as pharmaceutical compositions for administration to patients or for use in further screening assays. Administration of a composition of the present invention to a host may be accomplished using any of a variety of techniques known to those of skill in the art. The composition(s) may be processed in accordance with conventional methods of pharmacy to produce medicinal agents for 25 administration to patients, including humans and other mammals (i.e., a "pharmaceutical composition"). The pharmaceutical composition is preferably made in the form of a dosage unit containing a given amount of DNA, viral vector particles, polypeptide or peptide, for example. A suitable daily dose for a human or. other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods. 30 The pharmaceutical composition may be administered orally, parentally, by inhalation spray, rectally, intranodally, or topically in dosage unit formulations containing conventional phanaceutically acceptable carriers, adjuvants, and vehicles. The term "pharmaceutically acceptable carrier" or "physiologically acceptable carrier" as used herein refers to one or more 24 formulation materials suitable for accomplishing or enhancing the delivery of a nucleic acid, polypeptide, or peptide as a pharmaceutical composition. A "pharmaceutical composition" is a composition comprising a therapeutically effective amount of a nucleic acid or polypeptide. The terms "effective amount" and "therapeutically effective amount" each refer to the amount of a 5 nucleic acid or polypeptide used to induce or enhance an effective immune response. It is preferred that compositions of the present invention provide for the induction or enhancement of an anti-tumor immune response in a host which protects the host from the development of a tumor and / or allows the host to eliminate an existing tumor from the body. For oral administration, the pharmaceutical composition may be of any of several forms 10 including, for example, a capsule, a tablet, a suspension, or liquid, among others. Liquids may be administered by injection as a composition with suitable carriers including saline, dextrose, or water. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular, intrasternal, infusion, or intraperitoneal administration. Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa is butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature. The dosage regimen for immunizing a host or otherwise treating a disorder or a disease with a composition of this invention is based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of 20 administration, and the particular compound employed. For example, a poxviral vector may be administered as a composition comprising 1 x 106 infectious particles per dose. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. A prime-boost regimen may also be utilized (see, for example, WO 01/30382 Al) in which the targeted immunogen is initially administered in a priming step in one form followed by 25 a boosting step in which the targeted immunogen is administered in another form. The form of the targeted immunogen in the priming and boosting steps are different. For instance, if the priming step utilized a nucleic acid, the boost may be administered as a peptide. Similarly, where a priming step utilized one type of recombinant virus (i.e., ALVAC), the boost step may utilize another type of virus (i.e., NYVAC). This prime-boost method of administration has been shown 30 to induce strong immunological responses. While the compositions of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more other compositions or agents (i.e., other immunogenic targets, co-stimulatory molecules, adjuvants). When 25 administered as a combination, the individual components can be formulated as separate compositions administered at the same' time or different times, or the components can be combined as a single composition. Injectable preparations, such as sterile injectable aqueous or oleaginous suspensions, may 5 be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Suitable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution, among others. For instance, a viral vector such as a poxvirus may be prepared in 0.4% 10 NaCl. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. For topical administration, a suitable topical dose of a composition may be administered one to four, and preferably two or three times daily. The dose may also be administered with 15 intervening days during which no does is applied. Suitable compositions may comprise from 0.001% to 10% w/w, for example, from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation. Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g.,. liniments, lotions, 20 ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose. The pharmaceutical compositions may also be prepared in a solid form (including granules, powders or suppositories). The pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional. adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Solid dosage 25 forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose, lactose, or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering 30 agents. Tablets and pills can additionally be prepared with enteric coatings. Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. 26 Such compositions may also comprise adjuvants, such as wetting sweetening, flavoring, and perfuming agents. Pharmaceutical compositions comprising a nucleic acid or polypeptide of the present invention may take any of several forms and may be administered by any of several routes. In 5 preferred embodiments, the compositions are administered via a parenteral route (intradermal, intramuscular or subcutaneous) to induce an immune response in the host. Alternatively, the composition may be administered directly into a lymph node (intranodal) or tumor mass (i.e., intratumoral administration). For example, the dose could be administered subcutaneously at days 0, 7, and 14. Suitable methods for immunization using compositions comprising TAs are 10 known in the art, as shown for p53 (Hollstein et al., 1991), p21-ras (Almoguera et al., 1988), HER-2 (Fendly et al., 1990), the melanoma-associated antigens (MAGE-1; MAGE-2) (van der Bruggen et al., 1991), p 9 7 (Hu et al., 1988), melanoma-associated antigen E (WO 99/30737) and carcinoembryonic antigen (CEA) (Kantor et al., 1993; Fishbein et al., 1992; Kaufman et al., 1991), among others. 15 Preferred embodiments of administratable compositions include, for example, nucleic acids or polypeptides in liquid preparations such as suspensions, syrups, or elixirs. Preferred injectable preparations include, for example, nucleic acids or polypeptides suitable for parental, subcutaneous, intradermal, intramuscular or intravenous administration such as sterile suspensions or emulsions. For example, a recombinant poxvirus may be in admixture with a 20 suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose or the like. The composition may also be provided in lyophilized form for reconstituting, for instance, in isotonic aqueous, saline buffer. In addition, the compositions can be co-administered or sequentially administered with other antineoplastic, anti-tumor or anti-cancer agents and/or with agents which reduce or alleviate ill effects of antineoplastic, anti-tumor or anti-cancer agents. 25 A kit comprising a composition of the present invention is also provided. The kit can include a separate container containing a suitable carrier, diluent or excipient. The kit can also include an additional anti-cancer, anti-tumor or antineoplastic agent and/or an agent that reduces or alleviates ill effects of antineoplastic, anti-tumor or anti-cancer agents for co- or sequential administration. Additionally, the kit can include instructions for mixing or combining ingredients 30 and/or administration. A better understanding of the present invention and of its many advantages will be had from the following examples, given by way of illustration. 27 -EXAMPLES Example 1 BFA 5 Breast Cancer A antigen 5 A. Identification of BFA5 Microarray profiling analysis indicated that BFA5 was expressed at low to high levels in 41 out of 54 breast tumor biopsy samples (76%) and at high levels in 31 out of 54 breast tumors (57%), as compared to a panel of 52 normal, non-tumor tissues. In situ hybridization (ISH) was performed using a series of BFA5 DNA probes and confirmed the microarray with at least 61% 10 of the tumors showing fairly strong signals. Further bioinformatics assessment confirmed the results of these gene expression analysis results. Sequence analysis of the BFA5 nucleotide sequence revealed a high degree of similarity to two unidentified human genes: KJAA1074 (GenBank Accession No. XM_159732); and, KIAA0565 (GenBank Accession No. AB011137) isolated from a number of fetal and adult brain 15 cDNA clones (Kikuno, et al. The complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro. DNA Res. 6: 197-205). These genes were found to contain putative Zn finger regions and a nuclear localization sequence. BFA5 was suggested by others to be a potential breast cancer antigen (Jager, et al. 2001. Identification of a tissue-specific putative transcription factor in breast tissue by serological screening of a breast cancer library. Cancer 20 Res. 61: 2055-2061 and WO 01/47959). In each of these publications, the nucleotide sequence BFA5 was designated NYBR-1 ("New York Breast Cancer-1"; GenBank Accession Nos. AF269087 (nucleotide) and AAK27325 (amino acid). As shown previously by Jager, et al. and described in WO 01/47959, supra, BFA5 is specifically expressed in mammary gland, being expressed in 12/19 breast tumors analyzed. The 25 structure of the BFA5/NYBR-1 gene has revealed that it encodes a 150-160 kD nuclear transcription factor with a bZIP site (DNA-binding domain followed by a leucine zipper motif). The gene also contains 5 tandem ankyrin repeats implying a role in protein-protein interactions. These ankyrin repeats may play a role in homo-dimerization of the protein.. The BFA5 cDNA sequence is shown in FIG. 4 and SEQ ID NO.: 5. The BFA5 amino acid sequence is shown in 30 FIG. 5 and SEQ ID NO.: 6. 28 B. Immunoreactivity of BFA5 1. Activation of human T cells and IFN-y secretion in ELISPOT. A library of 100 peptides from the BFA5/NYBR-1 coding sequence that are predicted to be medium to high binders to HLA-A*0201 were designed using Rammensee and Parker 5 algorithms. The library was sub-divided into 10 pools of ten peptides (Table 111), and each pool was used to activate 10 different T cell cultures after pulsing peptides on to mature autologous dendritic cells. Two experiments were performed with the library of BFA5/NYBR-1 peptides demonstrating immunoreactivity in HLA-A*0201 human T cells, as described below. 29 a _-<_j> C.,IJ II j-i
----------------------------
0. LO. CL nC CL~ I T > _j -J < _j- 1 1 L e J J(I Fy _jg : U) D f2X 0 JIo L.C w Rc j :u 6) ) 6 6 6)6) ) ) 6) 6) 6) a)6 )6 )6 0. - .
U- N CDa 30 01 (n w zD- W 0 W > 0 -> x w 0 I-~F aUW L~f w > > OW>>->> F - 0 ) W 0;7 . ! : * - j- )u -j -j 0 > a. m ~ ~ ~ L LOwL 0t wwWt t -r CD< CL U) P- ID L, 0) 0 - :E c- )I )0~ N C' )C -~ 5j- 0) w C )U) e e Y>
U)
0 'no E'no.1jjE .>.z Lo WD I-C > MCDU) or 0mC 4 m . or oC 0 C 0 D C C V- Tl NN NN "N c N31c ELISPOT analysis was performed on human T-cell cultures activated through four rounds of stimulation with each pool of BFA5 peptides. Reactivity against a CMV pp65 peptide and a Flu matrix peptide were used as positive controls for T-cell activation in the experiments. Each experiment was performed with PBMC and dendritic cells from a single HLA-A*0201+ donor 5 designated as "APIO". The results show that, although BFA4 is markedly reactive with high ELISPOT counts per 100,000 cells in the assay, BFA5 is even more reactive with 9/10 pools demonstrating ELISPOT reactivity. Similar results were obtained for both BFA4 and BFA5/NYBR-1 with a different HLA-A*0201. The bars reach a maximum at 600 spots because beyond that the ELISPOT reader does not give accurate counts. Cultures having a reading of 600 10 spots have more than this number of spots. . A large number of the BFA5 peptide pools of are reactive as shown by the high levels of IFN-y production. Each reactive peptide pool was then separated into individual peptides and analyzed for inmunogencity using ELISPOT analysis to isolate single reactive BFA5 peptides. BFA5 is highly immunogenic with several reactive single peptides than that of BFA4. Similar 15 results were obtained in two independent PBMC culture experiments. In addition to ELISPOT analysis, human T cells activated by BFA5 peptides were assayed to determine their ability to function as CTL. The cells were activated using peptide-pulsed dendritic cells followed by CD40 ligand-activated B cells (5 rounds of stimulation). The experiment shown was performed with isolated PBMC from HLA-A*0201+ donor AP31. 20 Isolated T cells were tested in 5 1 Cr-release assays using peptide-loaded T2 cells. The % specific lysis at a 10:1, 5:1, and 1:1 T-cell to target ratio is shown for T2 cells pulsed with either pools of BFA5/NYBR-1 peptides or with individual peptides. The graph shows CTL activity induced against targets loaded with a c non-specific HLA-A*0201 -binding HIV peptide (control) followed by the CTL activity against the peptide pool (Pool 1 etc.) and then the activity induced by 25 individual peptides from the respective pool to the right. A high level of cytotoxicity was observed for some peptides at a 1:1 E:T ratio. CTL activity (percent specific lysis) induced by the control HIV peptide was generally <10%. Similar results were obtained with another PBMC donor expressing HLA-A*0201 (AP1O). A large number of BFA5 peptides trigger T cell mediated cytotoxicity of BFA5 peptide-loaded target cells. Table IV lists those peptides having 30 immunogenic properties. Five peptides (LMDMQTFKA, ILIDSGADI, ILSVVAKLL, SQYSGQLKV, and ELCSVRLTL) were found to induce both IFN-y secretion and CTL activity in T cells from both donors. TABLE IV 32 Immunoreactive pept ides from BFA5 BFA5 peptides eliciting high BFA5 peptides inducing IFN-y release (>200 spotsll 00,000 cells) CTL lysis of pulsed cells Donor APIO Donor AP31 Donor APIO Donor AP31 LMDMOTFKA LMDMQTFKA LMDMQTFKA LMDMQTFKA I(VSIPTKAL KVSIPTKAL SIPTKALEL SIPTKALEL TVSQKDVCL SVPNKALEL YLLHENCML YLLHENCML YLLHENCML QLQSKNMWL QLQSKNMWL QLQSI(NMWL SLSIULDTV SLSKILDTV SLSKILDTV ILIDSGADI ILIDSGADI ILIDSGADI ILIDSGADI KVMEINREV AVYSEILSV ILSVVAKLL ILSVVAKLL ILSWVAKLL ILSWVAKLL SLTPLLLSI SLTPLLLSI SLTPLLLSI SQYSGQLKV SQYSGQLKV SQYsGQLKV SQYSGQLKV QIMEYIRKL QIMEYIRKL QIMEYIRKL SVPNKAFEL NLNYAGDAL NLNYAGDAL GVTAEHYAV KSQEPAFHI MLKLEIATL MLKLEIATL MLKLEIATL MLKKEIAML ALRIQDIEL VLKKKLSEA ELCSVRLTL ELCSVRLTL ELCSVRLTL ELCSVRLTL SLKINLNYA SLKINLNYA SLKINLNYA ATCGMKVSI ATCGMKVSI AELOMTLKL AELQMTLKL AELQMTLKL VFMADICGV ILKEKNAEL ILKEKNAEL NLVDVYGNM NLVDVYGNM KCTALMLAV 33 C. Immunological Reagents Polyclonal antisera were generated against the following series of 22- to 23- mer peptides of BFA5: 5 BFA5(1-23) KLH-MTKRKKTINLNIQDAQKRTALHW (CLP-2977) BFA5(312-334) KLH-TSEKFTWPAKG RPRKIAWEKKED (CLP-2978) BFA5(612-634) KLH-DEILPSESKQKDYEENSWDTESL (CLP-2979) BFA5(972-994) KLH-RLTLNQEEEKRRNADILNEKIRE (CLP-2980) BFA5(1117-1139) KLH-AENTMLTSKLKEKQDKEILEAEI (CLP-2981) 10 BFA5(1319-1341) KLH-NYNNHLKNRJYQYEKEKAETENS (CLP-2982) Prebleed samples from rabbits were processed and stored at -20*C. Rabbits were immunized as follows: 1) the peptides were administered as an emulsion with Freund's Complete Adjuvant (FCA); and, 2) two weeks later, the peptides were coupled with Keyhole-Limpet 15 Hemocyanin (KLH)-coupled and administered as an emulsion with Freund's Incomplete Adjuvant FIA. The following results were observed: TABLE V Peptide/protein IgG titer x 10, (after IgG titer x 10 (after first Immunization second Rbl/Rb2) Immunization Rbl/Rb2) CLP 2977 25/6 256/64 CLP 2978 25/25 64/256 CLP 2979 12/25 256/512 CLP 2980 25/12 1024/128 CLP 2981 814 256/64 CLP 2982 2/2 64/32 20 Prebleed sample results exhibited IgG titers <100 for all samples. To assess the quality of the polyclonal antisera, western blots were performed using sera against BFA5. Sera were separately screened against cell extracts obtained from the BT474, MDMB453, MCF-7, Calu-6, and CosA2 cells. The approximate expected MWr of BFA5 protein is 153 kDa. A 220kD band was observed in the BT474 extract with CLP2980 antibody but not in 25 the MDMB453 cell extracts however a -l30kD band was present in the MDMB453 extract. Both bands were found to be consistent with the polyclonal antibosera tested in this analysis. Neither of these bands is present in the negative control. Thus, it can be concluded that the polyclonal antisera are specific for BFA5. 34 While the present invention has been described in terms of the preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations that come within the scope of the invention as claimed. 5 35

Claims (63)

1. An expression vector comprising the nucleic acid sequence as illustrated in SEQ ID NO.: 5 or 5 Figure 4; a nucleic acid sequence encoding the amino acid sequence illustrated in SEQ ID NO.: 6 or Figure 5; or a fragment thereof
2. The expression vector of claim 1 wherein the vector is a plasmid or a viral vector.
3. The expression vector of claim 2 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus. 10
4. The expression vector of claim 3 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
5. The expression vector of claim 4 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC(2). 15
6. The expression vector of claim 1 further comprising at least one additional tumor-associated antigen.
7. The expression vector of claim 6 wherein the vector is a plasmid or a viral vector.
8. The expression vector of claim 7 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus. 20
9. The expression vector of claim 8 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
10. The expression vector of claim 9 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC(2). 25
11. The expression vector of claim 1 further comprising at least one nucleic sequence encoding an angiogenesis-associated antigen.
12. The expression vector of claim 11 wherein the vector is a plasmid or a viral vector.
13. The expression vector of claim 12 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus. 30
14. The expression vector of claim 13 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
15. The expression vector of claim 14 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC(2). 36
16. The expression vector of claim 6 further comprising at least one nucleic sequence encoding an angiogenesis-associated antigen.
17. The expression vector of claim 16 wherein the vector is a plasmid or a viral vector.
18. The expression vector of claim 17 wherein the viral vector is selected from the group 5 consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus.
19. The expression vector of claim 17 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
20. The expression vector of claim 18 wherein the viral vector is a poxvirus selected from the 10 group consisting of NYVAC, ALVAC, and ALVAC(2).
21. An expression vector selected from the group consisting of an expression vector of claim 1, an expression vector of claim 6, an expression vector of claim 11, and an expression vector of claim 16; further comprising a nucleic acid sequence encoding a co-stimulatory molecule.
22. The expression vector of claim 22 wherein the vector is a plasmid or a viral vector. 15
23. The expression vector of claim 23 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus.
24. The expression vector of claim 24 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC. 20
25. The expression vector of claim 18 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC(2).
26. A composition comprising an expression vector in a pharnaceutically acceptable carrier, said vector comprising the nucleic acid sequence shown in SEQ ID NO.:5 or Figure 4; a nucleic acid sequence encoding the amino acid sequence illustrated in SEQ ID NO.: 6 or Figure 5; or 25 a fragment thereof.
27. The composition of claim 26 wherein the vector is a plasmid or a viral vector.
28. The composition of claim 27 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus. .
29. The composition of claim 28 wherein the viral vector is a poxvirus selected from the group 30 consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
30. The composition of claim 29 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC(2). 37
31. A method for preventing or treating cancer comprising administering to a host an expression vector comprising the nucleic acid gquence illustrated in SEQ ID NO.: 5 or Figure 4; a nucleic acid encoding the amino acid sequence illustrated in SEQ ID NO.: 6 or Figure 5; or a fragment thereof. 5
32. The method of claim 31 wherein the vector is a plasmid or a viral vector.
33. The method of claim 32 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus.
34. The method of claim 33 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and 10 TROVAC.
35. The method of claim 34 wherein the viral vector is a poxvirus. selected from the group consisting of NYVAC, ALVAC, and ALVAC(2).
36. An isolated peptide derived from BFA5 as shown in Table X or XI.
37. A method for immunizing a host against the tumor antigen BFA5 comprising administering to 15 the patient a peptide shown in Table X or XI, either alone or in combination with another agent, where the individual components of the combination are administered simultaneously or separately from one another.
38. An isolated peptide derived from BFA5 as shown in Table X or XI.
39. A method for immunizing a host against thetumor antigen BFA5 comprising administering to 20 the patient a peptide shown in Table X or XI, either alone or in combination with another agent, where the individual components of the combination are administered simultaneously or separately from one another.
40. The expression vector of claim 6 wherein the additional tumor-associated antigen is encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NO.: 1; SEQ ID 25 NO.: 3; the nucleic acid sequence shown in Figure 1; the nucleic sequence illustrated in Figure 3A; a nucleic acid sequence encoding the amino acid sequence of SEQ ID NO.: 2; a nucleic acid sequence encoding the amino acid sequence of SEQ ID NO.: 4; the nucleic acid sequence encoding the amino acid sequence illustrated in Figure 2; a nucleic acid sequence encoding the amino acid sequence illustrated in Figure 3B; a nucleic acid hybridizable under 30 stringent conditions to any of the foregoing sequences; a fragment of any of the foregoing nucleic acid sequences; and, a derivative of any of the foregoing nucleic acid sequences.
41. The expression vector of claim 40 wherein the viral vector is selected from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and adeno-associated virus. 38
42. The expression vector of claim 41 wherein the viral vector is a poxvirus selected from the group consisting of vaccinia, NYVAC, avipox, canarypox, ALVAC, ALVAC(2), fowlpox, and TROVAC.
43. The expression vector of claim 42 wherein the viral vector is a poxvirus selected from the 5 group consisting of NYVAC, ALVAC, and ALVAC(2).
44. An expression vector selected from the group consisting of an expression vector of claim 40, an expression vector of claim 41, an expression vector of claim 42, and an expression vector of claim 42; further comprising a nucleic acid sequence encoding a co-stimulatory molecule.
45. An expression vector of claim 44 or claim 21 wherein the co-stimulatory molecule is human 10 B7.1 or a derivative thereof.
46. A composition comprising an expression vector of claim 40 in a pharmaceutically acceptable carrier.
47. A composition comprising an expression vector of claim 41 in a pharmaceutically acceptable carrier. 15
48. A composition comprising an expression vector of claim 42 in a pharmaceutically acceptable . carrier.
49. A composition comprising an expression vector of claim 43 in a pharmaceutically acceptable carrier.
50. A composition comprising an expression vector of claim 44 in a pharmaceutically acceptable 20 carrier.
51. A composition comprising an expression vector of claim 45 in a pharmaceutically acceptable carrier.
52. A method for preventing or treating cancer comprising administering to a host a composition of claim 46. 25
53. A method for preventing or treating cancer comprising administering to a host a composition of claim 47.
54. A method for preventing or treating cancer comprising administering to a host a composition of claim 48.
55. A method for preventing or treating cancer comprising administering to a host a composition 30 of claim 49.
56. A method for preventing or treating cancer comprising administering to a host a composition of claim 50. 39
57. A method for preventing or treating cancer comprising administering to a host a composition of claim 51.
58. An isolated DNA molecule comprising the nucleic acid of SEQ ID NO.:5 and at least one of the nucleic acid sequences of SEQ ID NO.: 3 or SEQ ID NO.: 5. 5
59. An expression vector comprising the isolated DNA molecule of claim 58.
60. An isolated DNA molecule comprising a nucleic acid encoding the amino acid sequence of SEQ ID NO. 6 and at least one of the amino acid sequences of SEQ ID NO.: 2 or SEQ ID NO.: 4.
61. An expression vector comprising the isolated DNA molecule of claim 60. 10
62. An isolated DNA molecule comprising the nucleic acid of SEQ ID NO.:5 and at least one of the nucleic acid sequences of SEQ ID NO.: 3 or SEQ ID NO.: 5; a nucleic acid hybridizable under stringent conditions to the nucleic acid sequences of SEQ ID NO.: 3 or SEQ ID NO.: 5; a fragment of the nucleic acid sequences of SEQ ID NO.: 3 or SEQ ID NO.: 5; and, a derivative of any of the nucleic acid sequences of SEQ ID NO.: 3 or SEQ ID NO.: 5. 15
63. An antibody having the ability to bind the amino acid sequence of SEQ ID NO.: 6 or a fragment the amino acid sequence of SEQ ID NO.: 6. Dated 19 September, 2011 Sanofi Pasteur Limited Patent Attorneys for the Applicant/Nominated Person SPRUSON & FERGUSON 40
AU2011224126A 2003-04-15 2011-09-19 Tumor antigens BFA5 for prevention and/or treatment of cancer Abandoned AU2011224126A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2011224126A AU2011224126A1 (en) 2003-04-15 2011-09-19 Tumor antigens BFA5 for prevention and/or treatment of cancer
AU2014201009A AU2014201009A1 (en) 2003-04-15 2014-02-26 Tumor antigens BFA5 for prevention and/or treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60/462,945 2003-04-15
AU2011224126A AU2011224126A1 (en) 2003-04-15 2011-09-19 Tumor antigens BFA5 for prevention and/or treatment of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2004230328A Division AU2004230328A1 (en) 2003-04-15 2004-04-15 Tumor antigens BFA5 for prevention and/or treatment of cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2014201009A Division AU2014201009A1 (en) 2003-04-15 2014-02-26 Tumor antigens BFA5 for prevention and/or treatment of cancer

Publications (1)

Publication Number Publication Date
AU2011224126A1 true AU2011224126A1 (en) 2011-10-13

Family

ID=45442036

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2011224126A Abandoned AU2011224126A1 (en) 2003-04-15 2011-09-19 Tumor antigens BFA5 for prevention and/or treatment of cancer
AU2014201009A Abandoned AU2014201009A1 (en) 2003-04-15 2014-02-26 Tumor antigens BFA5 for prevention and/or treatment of cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2014201009A Abandoned AU2014201009A1 (en) 2003-04-15 2014-02-26 Tumor antigens BFA5 for prevention and/or treatment of cancer

Country Status (1)

Country Link
AU (2) AU2011224126A1 (en)

Also Published As

Publication number Publication date
AU2014201009A1 (en) 2014-03-13

Similar Documents

Publication Publication Date Title
US20110311543A1 (en) Tumor Antigens BFA5 for Prevention and/or Treatment of Cancer
US20130011422A1 (en) Tumor Antigens for the Prevention and/or Treatment of Cancer
AU2011200127A1 (en) Multi-antigen vectors for melanoma
US8530442B2 (en) Modified CEA nucleic acid and expression vectors
US7786278B2 (en) Modified CEA nucleic acid and expression vectors
US20030113919A1 (en) Immunogenic targets for melanoma
US20030148973A1 (en) MAGE-A1 peptides for treating or preventing cancer
US8946174B2 (en) Tumor antigens BFA4 and BCY1 for prevention and / or treatment of cancer
AU2004280608B2 (en) Modified CEA/B7 vector
CA2481719C (en) Modified cea nucleic acid and expression vectors
AU2011224126A1 (en) Tumor antigens BFA5 for prevention and/or treatment of cancer
US20090156519A1 (en) Modified KSA and Uses Thereof

Legal Events

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