Title of Invention

AN EXPRESSION VECTOR COMPRISING NUCLEIC ACID ENCODING TUMOR ANTIGENS

Abstract The present invention relates to an expression vector comprising the nucleic aid sequence encoding the amino acid sequence illustrated in sequence ID No.30 or sequence ID No.32.
Full Text

Tumor Antigens for Prevention and/or Treatment of Cancer
RELATED APPLICATIONS
This application claims priority to Ser. Nos. 60/471,119 filed May 16,2003 and 60/471,193 filed May 16,2003.
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 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 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 hybridiaation (ISH) and laser capture microscopy (Rosenberg, Immunity, 1999; Sgroi et al, 1999, Schena et al, 1995, Offiinga et al, 2000). The TAAs are antigens expressed or 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 can ehcit 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 cytokme or a co-stimulatory molecule has also been shown to be usefiil in developing effective therapeutic vaccines (Hodge et al, 95, Bronte et al, 1995, Chamberlain et al, 1996).
There is a need in the art for reagents and methodologies usefiil 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.
^ 2.

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 encoded by SEQ ID NO.: 29 or SEQ ID NO.: 31 or has the amino acid sequence of SEQ ID NO.: 30 or SEQ ID NO.: 32. 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 an immune stimulator, such as a co-stimulatory molecule or adjuvant.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. A. Nucleotide sequences of AAC2-1 and AAC2-2. B. Alignment of predicted amino acid sequence of AAC2-1 and AAC2-2, Missing nucleotides or amino acids are indicated by a "*". Differences between sequences are underlined.
Figure 2. A. Human lymphocytes differentiate into effector cells secreting IFN-y in response to peptides derived from the AAC2-2 protein. T cells were stimulated with the groups of peptides shown in Table III (groups 1-9). After three rounds of stimulation, the lymphocytes were analyzed for peptide-specific IFN-y production by ELISPOT. . B. The graph in the inset shows that activated cells stimulated by peptide Group #6 are capable of antigen-specific CTL activity killing peptide loaded T2 target cells. Peptide ECS elicits dominant activity in inducing both CTL activity and IFN-y secretion.
Figure 3. Murine T cells from HLA-A2-Kb transgenic mice recognize and secrete IFN-y in response to DNA immunization with a human AAC2-2-encoding DNA plasmid. Spleen cells from pEF6-hAAC2-2-immunized mice were re-stimulated with the different groups of peptides. After six days, the cells were harvested and tested for IFN-y secretion in response to each respective peptide group or a control HLA-A2-binding 9-mer HIV peptide. ELISPOT plates were incubated over- night and developed. Each group responded with high levels of IFN-y production (over 250 spots) in response to PMA and ionomycin used as a positive control. One of the higjily reactive peptides groups (group 6) is also recognized by human lymphocytes from the HLA-A-0201* donors tested so far.
Figure 4. DNA vaccination with a gene encoding human AAC2-2 completely abrogates the growth of implanted B16F10 melanoma cells. This effect is not due to a non-specific immune


response as shown by the inability of plasmid encoding flu-NP protein and the human flkl
(VEGFR-2) to prevent tumor growth.
Figure 5. Survival of mice after implantation of B16 F10 melanoma cells into C57BL/6 mice
showing the ability of DNA vaccination with a human AAC2-2 vector to completely protect
against the effects of tumor growth. This protective effect is antigen-specific and can not be
elicited through vaccination with other genes.
Figure 6. T lymphocytes from C57BL/6 mice exhibit effector cell activity and secrete IFN-y in
response to peptides of human AAC2-2 following DNA vaccination with the pEF6-hAAC2-2
expression plasmid. These peptides can exhibit cross-reactivity on B6 MHC class I. The
peptides in group 1 and group 5 induce strong reactivity by C57BL/6 T cells.
Figure?. BFA4cDNA sequence.
Figures. BFA4 amino acid sequence.
Figure 9. Immune response against BFA4 peptides.
Figure 10. BCYl nucleotide (A) and amino acid (B) sequences.
Figure 11. Immune response against specific BCYl peptides.
Figure 12. BFA5 cDNA sequence.
Figure 13. BFA5 amino acid sequence.
Figure 14. Immune response against BFA5-derived peptides.
Figure 15. BCZ4 cDNA and amino acid sequence.
Figure 16. Immune response against BCZ4-derived peptides.
Figure 17. BFY3 cDNA and amino acid sequence.
Figure 18. Immune response against BFY3-derived peptides.
DETAILED DESCRIPTION
The present, invention provides reagents and methodologies useful for treating and / or 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 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 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 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), 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.
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, gplOO); 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 that induces or enhances an aiti-tumor immune responso 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., /. Exp. Med., 180:347-352 (1994)), gp75 (TRP-1) (Wang et al, /. 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 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; Gaugler et at., Immunogenetics, 44:323-330 (1996); U.S. Pat. No. 5,939,526), N-acetylglucosaminyltransferase-V (Guilloux et at., J. Exp. Med., 183:1173-1183 (1996)), pl5


(Robbins et al., J. Immunol. 154:5944-5950 (1995)), 6-catenin (Robbins et al., J. Exp. Med., 183:1185-1192 (1996)), MUM-1 (Coulie et al., Proc. Natl. Acad. Sci. USA, 92:7976-7980 (1995)), cyclin dependent kinase-4 (CDK4) (Wolfel et al., Science, 269:1281-1284 (1995)), p21-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)), pl85 HER2/neu (erb-Bl; 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 antigens (CEA) (Kwong et al., J. Natl. 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. Immunol., 151:1654-1662 (1993)); EBNA gene products of EBV (i.e., EBNA-1; Rickinson et al., Cancer Surveys, 13:53-80 (1992)); E7, E6 proteins of human papillomavirus (Ressing et al., J. Immunol, 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. Immunol., 153:4775-4787 (1994)); KSA (U.S. Patent No. 5,348,887), kinesin 2 (Dietz, et al. Biochem Biophys Res Commun 2000 Sep 7;275(3):731-8), HlP-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-1 (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 Vaccines 2000, Cancer Research Institute, New York, NY), BFA4 (SEQ ID NOS.: 23 and 24), BCYl (SEQ ID NOS.: 25 and 26), BFA5 (SEQ ID NOS.: 27 and 28), BCZ4 (SEQ ID NOS.: 29 and 30), and BFY3 (SEQ ID NOS. 31 and 32), including "wild-type" (i.e., nonnally encoded by the genome, naturally-occurring), modified, and mutated versions as well as other firagments and derivatives thereof. Any of these TAs may be utilized alone or in combination with one another in a co-immunization protocol.
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 preferred that that the AA be found within or near blood vessels that supply a tumor.


Inunimization 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. / Urol., 2001, 166(4): 1275-9; Stames, et al. J. Thorac. Cardiovasc. Surg., 2001,122(3): 518-23; Dias, et al. Blood, 2002, 99: 2179-2184), the VEGF receptor (i.e., VEGF-R, flk-l/BODR; Stames, 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 (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 (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 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 synthase), collagen receptors, integrins the surface
proteolglycan NG2, AAC2-1 (SEQ ID N0.:1), or AAC2-2 (SEQ ID N0.: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 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 firagments or.derivatives thereof, such as that contained in a DNA insert in an ATCC Deposit. The term "nucleic acid sequence" or "nucleic acid molecule" refers to a DNA or RNA sequence. The term encompasses molecules formed fi-om any of the


known base analogs of DNA and RNA such as, but riot limited to 4-acetylcytosine, 8-hydroxy-N6-methyladenosine, aziridinyl-cytosine, pseudoisocytosiue, 5-(carboxyhydroxyhnethyI) uracil, 5-fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5-carboxy-methylaminomethyluracil, dihydrouracil, inosine, N6-iso-pentenyladenine, l-methyladenine, 1-methylpseudouracil, 1-methylguanine, 1-methylinosine, 2,2-dimethyl-g;uanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaniino-methyl-2-thiouracil, beta-D-mannosylqueosine, 5' -methoxycarbonyl-methyluracil, 5-methoxyuracil, 2-meihylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, oxybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, N-uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudoiuacil, queosine, 2-thiocytosine, and 2,6-diaininopurine, among others.
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 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 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 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 inathematical model or computer program (i.e., an 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. 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); Anderson et al, Nucleic Acid Hybridisation: A Practical Approach Ch. 4 (IRL Press Limited)). The terin "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 sodium citrate at 50-65°C or 0.015 M sodium chloride, 0.0015 M sodium citrate, and 20% formamide at 37-50°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¬pyrrolidone, 0.1% sodium pyrophosphate, 0.1% sodium dodecylsulfate, NabodS04, (SDS), ficoll, Denhardt's solution, sonicated sahnon 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; 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 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. Flanking sequences may be homologous (i.e., from the same species and / or strain as the host
/ 9

cell), heterologous (i.e., firom a species other than the host cell species or strain), hybrid (i.e., a combination of flanking sequences fi:om 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, 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 sequaice need not necessarily be contiguous with the coding sequence, so long as it fiinctions correctly. Thus, for 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 fi:om the coding sequence and affect transcription of the sequence.
In certain embodiments, it is preferred that the flankiiig sequence is a trascriptional regulatory region that drives high-level gene expression in the target cell. The transcriptional regidatory 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 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 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 gene promoters; the SV40 early promoter region (Bemoist 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 03SV-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 expression vectors such as the beta-lactamase promoter (Villa-Kamaroff et al, 1978, Proc. Natl
^ (O

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 \^ch is active in pancreatic acinar cells (Swift et al,
1984, Cell 38:639-46; Omitz et al, 1986, Cold Spring Harbor Symp. Quant. Biol 50:399-409 (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 mammary tumor virus control region in testicular, breast, lymphoid and mast cells (Leder et al,
1986, Cell 45:485-95); the albumin gene control region in Uver (Pinkert et al, 1987, Genes and Devel 1:268-76); the alpha-feto-protein gene control region in Hver (Krumlauf e/ al, 1985, Mol Cell Biol, 5:1639-48; Hammer et al, 1987, Science 235:53-58); the alpha 1-antitrypsin gene control region in Uver (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; KoUias et al, 1986, Ce// 46:89r94); 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 in the hypothalamus (Mason et al, 1986, Science 234:1372-78), and the tyrosinase promoter in melanoma cells (Hart, I. Semin Oncol 1996 Feb;23(l):154-8; Siders, et al. Cancer Gene Ther 1998 Sep-Oct;5(5):281-91), 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 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 available firom 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 sphced into the vector at a position 5' or 3' to nucleic acid coding sequence, it is typically located at a site 5' firom the promoter. Other suitable enhancers are known in the art, and would be applicable to the present invention.
^ II

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 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.
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 integratmg into a chromosome of the cell, may be maintained transiently as an episomal 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 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is naturally found when isolated firom 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 polypeptide is substantially fi-ee 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 are prepared. Further contemplated are related polypeptides such as, for example, Augments, 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 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 fragmrat of a nucleic acid or polypeptide comprises a truncation of the sequence (i.e., 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 iutemal deletions as compared to the parental sequence. In preferred embodiments, truncations 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 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 preferred embodiments, the variants have from 1 to 3, or from 1 to 5, or from 1 to 10, or from 1 to 15, or from 1 to 20, or from 1 to 25, or from 1 to 30, or from 1 to 40, or from 1 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 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 piolypeptide 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 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 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 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 fimctional and 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 decreased immunogenicity. Suitable conservative amino acid substitutions are shown in Table I.


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 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 from other species are known, one skilled in the art may compare tiie amino acid sequence of a polypeptide to such similar polypeptides. By performing such analyses, one can identify residues and portions of the 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. 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

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 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 vWll remove an existing N-linked carbohydrate chain. Also provided is a rearrangement of Nrlinked 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 residues are deleted or substituted with another amino acid {e.g., serine) as compared to the subject amino acid sequence set. Cysteine variants are useftil 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 firom unpaired cysteines.
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 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, 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 pq)tide, or other
f^ii.

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 firom 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 subsequentiy be removed fi-om 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 CXCIO (IP-10), CCL7 (MCP-3), or CCL5 (RANTES), for example:
A fusion motif may enhance transport of an immunogenic target to an MHC processing compartment, such as the endq)lasmic reticulum. These sequences, referred to as tranduction or transcytosis sequences, include sequences derived fit)m HTV 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-1 protein (hPERl; in particular, SRRHHCRSKAKRSRHH).
In addition, the polypeptide or variant thereof may be fused to a homologous polypeptide to form a homodimer or to a heterologous polypeptide to form a heterodimer. Heterologous p^tides 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 hgand or a portion thereoif which binds to a transmembrane receptor protein; an enzyme or portion thereof 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 immunoglobulin constant region; and a polypeptide which has a therapeutic activity different from the polypeptide or variaiit thereof
In certain embodiments, it may be advantageous to combine a nucleic acid sequence encoding an immunogenic target, polypeptide, or derivative thereof with one or more co-stimulatoiy 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 polypeptide, for example. Suitable co-stimulatory molecules include, for instance, polypeptides that bind members of the CD28 family (i.e., CD28, ICOS; Hutloff, et al. Nature 1999,397: 263-265; Peach, et al. J Exp. 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 (CD86; Ellis, et al. J. Immunol, 156(8): 2700-9);

polypeptides which bind members of the integrin family (i.e., LFA-1 (CDl la / CD18); Sedwick, et al. J Immunol 1999,162: 1367-1375; Wulfing, et al. Science 1998, 282: 2266-2269; Lub, et al. Immunol Today 1995,16:479-483) including members of the ICAM family (i.e., ICAM-1, -2 or -3); polypeptides which bind CD2 family members (i.e., CD2, signalling lymphocyte activation molecule (CDwl50 or "SLAM"; Aversa, et al. J Immunol 1997, 158: 4036-4044)) such as CD58 (LiFA-3; CD2 ligand; Davis, et al. Immunol Today 1996, 17: 177-187) or SLAM Ugands (Sayos, et al. Nature 1998, 395: 462-469); polypeptides which bind heat stable antigen (HSA or CD24; Zliou, et al. Eur J Immunol 1997, 27: 2524-2528); polypeptides which bind to members of the TNF receptor (TNFR) family (i.e., 4-lBB (CD137; Vinay, et al. Semin Immunol 1998,10: 481-489), OX40 (CD134; Weinberg, et al. Semin Immunol 1998, 10: 471-480; ffiggins, et al. J Immunol 1999, 162: 486-493), and CD27 (Lens, et al. Semin Immunol 1998, 10: 491-499)) such as 4-lBBL (4-lBB ligand; Vinay, et al. Semin Immunol 1998, 10: 481-48; DeBenedette, et al. /Immunol 1997, 158: 551-559), TNFR associated factor-1 (TRAF-1; 4-lBB ligand; Saoulli, et al. J Exp Med 1998, 187: 1849-1862, Arch, et al. Mol Cell Biol 1998,18: 558-565), TRAF-2 (4-lBB and OX40 Ugand; SaouUi, et al. J Exp Med 1998, 187: 1849-1862; Oshima, et al, Int Immunol 1998, 10: 517-526, Kawamata, et al. J Biol Chem 1998, 273: 5808-5814), TRAF-3 (4-lBB and OX40 ligand; Arch, et al. Mol Cell Biol 1998, 18: 558-565; Jang, et al. Biochem Biophys Res Commun 1998, 242: 613-620; Kawamata S, et al. J Biol Chem 1998, 273: 5808-5814), OX40L (OX40 ligand; 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 Chem 1998, 273: 5808-5814), and CD70 (CD27 ligand; Couderc, et al. Cancer Gene Ther., 5(3): 163-75). CD 154 (CD40 ligand or "CD40L"; Gurunathan, et al. J. Immunol,, 1998, 161: 4563-4571; Sine, et al. Hum. Gene Ther., 2001,12:1091-1102) may also be suitable.
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 Immimol. 1: 209-219). Suitable cytokines include, for example, interleukin-2 (IL-2) (Rosenberg, et al. Nature Med 4: 321-327 (1998)), IL-4, IL-7, IL-12 (reviewed by PardoU, 1992; Harries, et al. J. Gene Med. 2000 Jul-Aug;2(4):243-9; Rao, et al. J. Immunol. 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 (CSF (Disis, et al. Blood, 88: 202-210 (1996)), tumor necrosis factor-alpha (TNF-a), or

interferons such as IFN-a or ENF-y, Other cytokines may also be suitable for practicing the present invention, as is known in the art.
Oiemokines 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 immunity (Biragyn, et al. Nature Biotech. 1999, 17: 253-258). The chemokines CCL3 (MEP-la) 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 inunune mechanisms may be blocked, resulting in enhanced immune responses. For instance, treatment with anti-CTLA-4 (Shrikant, et al. Immunity, 1996,14:145-155; Sutinuller, et al. J. Exp. Med., 2001,194: 823-832), anti-CD25 (Sutinuller, 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 req)onses and would be suitable in practicing the present 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, ICAM-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, 158: 3947-3958 (1997); Iwasaki, et al. /. Immunol 158:4591-4601 (1997)), IL-12 + GM-CSF + TNF-a (Ahlers, et al. Int Immunol 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 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.
1999. Vaccine, 17: 3124-2135; Dubensky, et al. 2000. Mol Med. 6: 723-732; Leitiier, 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, et al. Ann. Rev. Immunol, 2000, 18: 927-974; Leitner, supra; Cho, et al. J. Immunol.
r n

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 (Gurunaflian, supra; Sullivan, et al. 2000. Nature, 408: 605-609; Hanke, et al. 1998. Vaccine, 16: 439-445; Amara, et al. 2001. Science, 292: 69-74), and the use of mucosal delivery vectors such as iSa/woweZ/fl parji, et al. 1997. Ce//, 91: 765-775; Woo, etal. 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. Oncogene 2000 Dec 27;19(56):6566-73). For example, m 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 + doxorubicin, for example. Olher 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 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 chemotherq)eutic regimens. For example, metastatic colorectal cancer is typically treated with Camptosar (irinotecan or CPT-11), 5-fluorouracil or leucovorin, alone or in combination with one another. Proteinase and integrin inhibitors such as as the MMP inhibitors marimastate (British Biotech), COL-3 (CoUagenex), Neovastat (Aetema), AG3340 (Agouron), BMS-275291 (Bristol Myers Squibb), CGS 27023A (Novartis) or the integrin inhibitors Vitaxin (Medimmune), or MED 1522 (Merck KgaA) may also be suitable for 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.
>^^

Many anti-angiogenic agents are known in the art and would be suitable tor 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-P)), cytokines (i.e., interferons such as IFN-a, -p, -y, platelet fector 4 (PF-4), PR-39), proteases (i.e., cleaved AT-III, collagen XVIII fragment (Endostatin)), HmwKallikrein-d5 plasmin fragment (Angiostatin), prothrombin-Fl-2, TSP-1), protease inhibitors (i.e., tissue inhibitor of metalloproteases such as TIMP-l, -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, HUI77, XLS 13; anti-VEGF; anti-integrin (i.e., Vitaxin, (Lxsys))), and gjycosidases (i.e., heparinase-I, -HI). "Chemical" or modified physiological agents known or believed to have anti-angiogenic potential mclude, 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.), fiavone acetic acid. Bay 12-9566 (Bayer Corp.), AG3340 (Agouron, Inc.), COS 27023A (Novartis), 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), 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 01/93897), cyclic-RGD peptide, accutin-disintegrin, benzodiazepenes, humanized anti-avb3 Ab, Rh-PAI-2, amiloride, p-amidobenzamidine, 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 practicmg the present invention.
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 novyt was modified to eliminate a toxin gene carried on a phage episome and administered to mice with


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 introducing a nucleic acid to a host include retrovirus, adenovirus, adeno-associated virus (AAV), herpes virus, and poxvirus, among others. It is imderstood 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 (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 avian retroviruses. Examples of suitable retroviral vectors include, for example, Moloney murine leukemia virus (MoMuLV), Harvey murine 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 ' can be provided by, for example, helper cell lines encoding retrovirus structural genes. Suitable helper cell lines include T2, PAS 17 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 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 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., et al, 1991, Science, 252 (5004): 431-4; Crystal, R., et al., 1994, Nat. Genet.. 8


(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 (Rosenfeld, M., et al, 1992, Cell, 68 (1): 143-55) injection into muscle (Quantin, B., et al, 1992, Proc. Natl Acad. Sci. U.SA., 89 (7): 2581-4), peripheral intravenous injection (Herz, J., and Gerard, R, 1993, Proc. Natl Acad. Sci U.SA., 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 specificity in integrating into the host cell genome (Hermonat, P., et al., 1984, Proc. Natl Acad. Sci. U.S.A., 81 (20): 6466-70). And Herpes Simplex Virus type-1 (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 Neuroscl, 14 (10): 428-32; Glorioso, et al, 1995, Mol Biotechnol, 4 (1): 87-99; Glorioso, et al, 1995, Annu. Rev. Microbiol, 49: 675-710).
Poxvirus is smother useM 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 Immunol., 158: 25-38; Moss, etal. 1991. Science, 252:1662-166T). 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 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+B14R); A type inclusion body region (ATI; A26L); hemagglutinin gene (HA; A56R); host range gene region (C7L-K1L); and, large subunit, ribonucleotide reductase (I4L). 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, VCP1433, placZH6H4Lreverse, pMPC6H6K3E3 and pC3H6IHVB 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 use in practicing fee present invention (see, for example, U.S. Pat. No. 5,756,103). ALVAC(2) is identical to ALVAC(l) except that ALVAC(2) genome comprises fee vaccinia E3L and K3L genes under fee control of vaccinia promoters (U.S. Pat. No. 6,130,066; Seattle et al., 1995a, 1995b, 1991; Chang et al., 1992; Davies et al., 1993). Bofe ALVAC(l) and ALVAC(2) have been demonstrated to be useful in expressing foreign DNA sequences, such as TAs (Tartaglia et al., 1993 a,b; U.S. Pat. No. 5,833,975). ALVAC was deposited under fee terms of fee Budapest Treaty wife fee American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, USA, ATCC accession number VR-2547.
Anofeer iiseful poxvirus vector is TROVAC. TROVAC refers to an attenuated fowlpox that was a plaque-cloned isolate derived from fee FP-1 vaccine strain of fowlpoxvirus which is licensed for vaccination of 1 day old chicks. TROVAC was likewise deposited under fee terms of fee Budapest Treaty wife fee ATCC, accession number 2553.
"Non-viral" plasmid vectors may also be suitable in practicing fee present invention. Preferred plasmid vectors are compatible wife bacterial, insect, and / or mammalian host cells. , Such vectors include, for example, PCR-II, pCR3, and pcDNA3.1 (Invitrogen, San Diego, CA), pBSn (Stratagene, La Jolla, CA), pET15 (Novagen, Madison, WI), pGEX (Pharmacia Biotech, Piscataway, NJ), pEGFP-N2 (Clontech, Palo Alto, CA), pETL (BlueBacH, Invitrogen), pDSR-alpha (PCT pub. No. WO 90/14363) and pFastBacDual (Gibco-BRL, Grand Island, NY) as well
(Si
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
products (e.g., TOPO™ TA cloning kit, PCR2.1 plasmid derivatives, Invitrogen, Carlsbad, CA). Bacterial vectors may also be used wife fee current invention. These vectors include, for example. Shigella, Salmonella, Vibrio cholerae, Lactobacillus, Bacille calmette guerin (BCG), and Streptococcus (see for example, WO 88/6626; WO 90/0594; WO 91/13157; WO 92/1796; and WO 92/21376). Many ofeer non-viral plasmid expression vectors and systems are known in fee art and could be used wife fee current invention.
Suitable nucleic acid delivery techniques include DNA-ligand complexes, adenovirus-ligand-DNA complexes, direct injection of DNA, CaP04 precipitation, gene gun techniques, electroporation, and colloidal dispersion systems, among ofeers. Colloidal dispersion systems 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 mtact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, R., et al, 1981, Trends Biochem. Sd., 6: 11). IThe 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 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 phosphatidylcholine, dipahnitoylphosphatidylcholine 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:



The immimogemc targets of the present invention may also be used to generate antibodies for use in screening assays or for immunotherE^y. Other uses would be apparent to one of skill in the art. The term "antibody" includes antibody fragments, as are known in the art, including Fab, Fab2, 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, Harlow, et al. Antibodies: A Laboratory Manual, Cold Spring EsrhoxLaboraiOTy, 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); Riechmann et al. Nature, 332:323-329 (1988); Presta (Cuir. 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., J. Mol. Biol., 222:581 (1991); Cole et al,. Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boemer et al., J. Immunol, 147(l):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); Fishwfld 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 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 therefiom may also be conjugated to therapeutic moieties such as cytotoxic drugs or toxins, or active fi'agments 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 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 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 correlated with base levels to determine whether a particular disease is present within the patient, the patient's 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 immunogenic 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 ttierapeutic modality may need to be utilized. In one embodiment, nucleic acid probes 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 e3q)ression 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 patient. The expression profiling technique may be combined with high throughput screening techniques to allow rapid identification of usefiil compounds and monitor the effectiveness of treatment with a drug candidate (see, for example, Zlokamik, et al.. Science 279, 84-8 (1998)). Drug candidates may be chemical compounds, nucleic acids, proteins, antibodies, or derivatives therefi-om, whether naturally occurring or synthetically derived. Drug candidates thus identified


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 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 containmg 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 oh the condition of the patient and other factors, but, once again, can be determined using routine methods.
The pharmaceutical composition may be administered orally, parentally, by inhalation spray, rectally, intranodally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. The term "phaimaceutically acceptable carrier" or "physiologically acceptable carrier" as used herein refers to one or more 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 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
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 mcluding saline, dextrose, or
water. The term parenteral as used herein includes subcutaneous, intravenous, intramuscular,
mtrastemal, infusion, or mtraperitoneal administration. Suppositories for rectal administration of
the drug can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa
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 freating 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

.-1^.
'9/

admiiiistration, and the particular compound employed For example, a poxyiral vector may be administered as a composition comprising 1x10* 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 a boosting step in which the targeted immunogen is administered m another form. The form of the targeted immunogen in the priming and boosting stq)s 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 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 administered as a combioation, 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 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 aiid 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% NaCl. In addition, sterile, fixed oils are conventioiully 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 intervening days during which no does is applied. Suitable compositions may comprise fix>m 0.001% to 10% w/w, for example, from 1% to 2% by weight of the formulation, although it may cornprise 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, 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 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 tban 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 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. 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 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 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), p97 (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.
Preferred embodiments of administratable compositions include, for example, nucleic acids or polypeptides m liquid preparations such as su^ensions, 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
^^30

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.
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 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.
EXAMPLES
Example 1
AAC2 Tumor Associated Antigen
A version of the AAC2 coding sequence (AAC2-1) was provided by a collaborator and
found to have high sequence similarity to a murine bcl-6-associated zdnc finger protein
("BAZF"). Based on this sequence information, PCR primers were designed as shown below:
CACCATGGGT TCCCCCGCCG CCCCGGA (forward primer, SEQ ID NO.: 6)
CTAGGGCCCC CCGAGAATGT GGTAGTGCAC TTT (reverse primer; iSEQ ID NO.: 7)
RNA was isolated from confluent HUVEC (BioWhittacker; Cat, No. CC2517, Lot No.
1F0141) cultures using Trizol"^ as indicated by the manufacturer (Life Technologies, Inc., Cat
No. 15596). High fidelity RT-PCR was then performed using the forward and reverse primers
(24 cycles at 94 degrees, 2 min.; 94 degrees, 30 sec; 56.8 degrees, 30 sec; 68 degrees, 1 mm 40
sec; cycle 25 is 68 degrees, 7 min) resulting in the isolation of a 1,447 base pair cDNA. The
cDNA was cloned into the pEF6-T0P0 eukaryotic expression plasmid and termed "pEF6-
hAAC2-2". The cDNA pEF6-hAAC2-2 was sequenced using four primers and aligned to the
sequence of AAC2-1 and murine BAZF (Fig. 1). As shown therein, AAC2-2 is missing the
serine residue (S) found at position 245 in AAC2-1. Secondly, a sfretch of 17 amino acids at
positions 298 to 316 (SEFFSCQNCEAVAGCSS) of AAC2-2 showed only 11.8% sequence
^S
identity with amino acids 298-316 of AAC2-1 (Fig. 1). Interestingly, the stretch of 17 amino acids between positions 298 and 316 is 100% identical with murine BAZF suggesting that this may be critical for transcription factor function along with the long stretch of serines (zinc finger). AAC2-2 was then cloned into the pcDNA3.1-zeo eukaryotic ejcpression plasmid ("pcDNA3.1-hAAC2-2").
Example 2 Human T-cell Reactivity Against AAC-2 Peptides
Using the AAC2-2 amino acid sequence, a library of 9-mer peptides predicted to bind to HLA-A-0201 was constructed (Table HI; "N" indicates the sequence is not found witiiin the mouse homolog, while "Y" indicates tiie sequence is found within the mouse homolog). Twenty-three of the peptides were dissolved in DMSO at 10 mg/ml (Table TV) and used in human PBMC cultures to test for their ability to elicit CDS and CD4 ap T-cell responses in vitro.
3^



Using GM-CSF and IL-4, dendritic cells (DC) were generated from peripheral blood monocytes of blood donors expressing HLA-A-0201. DC were pulsed with the different pools of 9-mer AAC2-2 peptides shown in Table IV.

These DC were used to stimulate autologous T-cell-enriched PBMC preparations. The T cells were re-stimulated witii autologous PBMC and then re-stimulated with CD40-hgand-activated autologous B cells. After the third and fourth round of stimulation with each peptide pool, ELISPOT analysis for IFN-y production indicated that the T cells responded most strongly

to one of the pools of AAC2-2 peptides (peptide group 6; Fig. 2A). Peptide group 6 includes the following peptides: ILTDVTLLV (aa 36-44), TLLVGGQPL (aa 41-49), and FMYTSRLRL (aa 95-103). Flow cytometric analysis (FACS) showed that the lymphocytes from this peptide-specific line consisted of >50% CDS T cells with a memory (CD45RO"*) phenotype. Very few cells ( Analysis of CTL activity from this peptide pool-specific T-cell line also demonstrated that the activated T cells were capable of killing peptide-loaded TAP-deficient T2 cells in an HLA-A-0201-restricted fashion (Fig. 2B). This analysis also revealed that ILTDVTLLV was a dominant peptide that stunulated the majority of the peptide-specific CTL activity. Thus, it was determined that AAC2-2 peptides are immunogenic in the human immune system.
EXAMPLES Immunogenicity ofAAC2-2 in vivo
Using DNA immunization into HLA-A2-Kb transgenic mice, it was found that the AAC2-2 protein is processed into immunogenic peptides and can elicit an HLA-A-0201-restricted T-cell response in vivo. Mice were immunized on day 1 by injection with pEF6-hAAC2-2 and boosted with the same plasmid at day 21. Lymphocytes were harvested from immunized mice 21 days after boosting and re-stimulated in vitro with the different groups of AAC2-2 peptides shown in Table IV. Peptide-specific effector T-cell fimction towards these peptides was found using IFN-y ELISPOT analysis (Fig. 3). It was found that the same pool of peptides (group 6) previously shown to be strongly immunogenic in human PBMC cultures also elicited significant reactivity by T cells after DNA vaccination (Fig. 3). Thus, the AAC2 gene product administered as a DNA-based vaccine is immunogenic in vivo and elicits a strong cell-mediated immune response characterized by the activation of CTL activity.
3^

EXAMPLE 4 TherapeuticAAC2-2 Vaccine
Therapeutic vaccination against the AAC2-2 gene product using the pEF6-hAAC2-2 DNA vaccine was found to completely block the growth of a solid tumor. Groups of eight C57BL/6 mice were subcutaneously challenged with 10'* B16F10 melanoma cells, a vigorous and relatively non-immunogenic tumor cell line. The mice were then immunized at weekly intervals starting at 6 days after tumor challenge. Control mice (eight per group) treated either with a plasmid encoding the flu-NP protein or saline alone all developed large tumors, hi contrast, all the mice (8/8) immunized with pEF6-hAAC2-2 had no detectable tumor over a 50-day period (Fig. 4). All mice remamed tumor-free through 80 days (data not shown). Fig. 5 plots the survival of mice treated with the different DNA vectors shown after melanoma implantation showing again the complete effectiveness of AAC2-2 vaccination in protecting mice against melanoma growth. No adverse health effects have been observed as a result of immunization with the human AAC2-2 gene-encoding vector (immunized mice were as active as control mice and showed no weight loss).
As shown in Figs. 4 and 5, vaccination with a plasmid encoding the human VEGFR-2 (pBLAST-hflkl) did not protect tumor-challenged mice. In fact, the tumors grew even more rapidly in these mice. Analysis of sera from mice vaccinated with the pBLAST-hflkl plasmid by ELISA found that IgG against the VEGFR-2 protein is induced in significant titres (data not shown). These results suggest that an antibody-based immune response directed against VEGFR-2 may not be not effective in preventing angiogenesis and solid tumor growth.
Inhibition of melanoma solid tumor growth in C57BL/6 mice immunized with pEF6-hAAC2-2 correlates with an immune response against the protein (Fig. 6). Immunization of C57BL/6 mice was performed as described above. Spleen cells from immunized mice were re-stimulated with the same peptide pools used in experiments with HLA-A2-Kb transgenic mice (Table III). A significant number of peptides cross-react on C57BL/6 class IMHC (Kb and Db molecules). Two pools of peptides in particular (group 1 and group 5) were found to ehcit strong effector cell activity in the IFN-y ELISPOT assays (Fig. 6). All of the peptides in these groups are also identical to the corresponding sequence in the murine BAZF protein. These results strongly suggest that immunization with the human AAC2-2 activates an immune response against its murine orthologue BAZF in mice and can inhibit tumor angiogenesis as a result These results are from a single experiment, and not all experiments showed these results.

F

5JL

Example 5 BFA4 Tumor Antigen
The BFA4 sequraice was found to be the "trichorbinophalangeal syndrome 1" (TRPS-1) gene (Genebank ID #6684533; Momeniet et al, Nature Genetics, 24(1), 71-74,2000), a known transcription factor with no function attributed previously in any form of cancer. The BFA4 cDNA sequence is shown in Fig. 7 (SEQ ID NO.: 23) and the deduced amino acid sequence is shown in Fig. 8 (SEQ ID NO.: 24).


The pcDNA3.2BFA4 (3.6 mg) was also used for DNA immunization to generate polyclonal sera in chickens.
B. Cloning of BFA4
Complete cDNA sequence for BFA4 is ~10kb and gene is e)q)ressed in BT474 ductal carcinoma cells. Primers 7717 (forward primer) and 7723 (reverse primer) were designed to amplify full-length BFA4 gene by amplification of 4kb, 7kb or lOkb products by RT-PCR.
Primer 7717: BFA4-BamHl/Fl (5'end forward) with Kozak:
5' CGGGATCCACCATGGTCCGGAAAAAGAACCCC 3'(BaniHI for DNA3.1, MP76)
Primer 7723: BFA4-BamHI /Rl (3' end reverse 4Icb):
5' CGGGATCCCTCTTTAGGTTTTCCATTTTTTTCCAC 3' (BamHI for DNA3.1, MP76)
Ten mg of total RNA isolated and firozen in different batches fix)m BT-474 cells using Trizol as indicated by the manufacturer (Gibco BRL) was used in RT-PCR to amplify the BFA4 gene. RT-PCR conditions were optimized using Taq Platinum High Fidelity enzyme, OPC (Oligo Purification Cartridge; Applied Biosystems) purified primers and purified total RNA/polyA mRNA (BT 474 cells). Optimization resulted in a 4.0kb fi-agment as a single band.
To re-amplify the BFA4 sequence, mRNA was treated with DNase per manufacturers' instmctions (Gibco BRL). The 4kb DNA was reamplified using PCR using primers 7717 and 7723 primers (lOpmole/microlitre) and Taq Platinum High Fidelity polymerase (GIBCO BRL) enzyme. Thermocycler conditions for both sets of reactions were as under: 94°C (2 min), followed by 30 cycles of 94°C (30 sec), 52°C (30sec), 67'C (4 min) and 67'C (5 min) and finally 40°C for 10 min. Three BFA4 clones were identified after pCR2.1/T0P0-TA cloning.
Several mutations were identified during analysis of the BFA4 sequence. To correct these sequences, the BamHI/XhoI fragment (5') of the BFA4 gene from clone JB-3552-1-2 (pCR2.1/T0P0/ BFA4) was exchanged with the XhoI/BamHI fiagment (3') of the BFA4 gene fi-om clone JB-3552-1-4 (pCR2.1/TOPO/BFA4). This recombined fiagment was then ligated into pMCS5 BamHI/CAP. Clone JB-3624-1-5 was generated and found to contain the correct sequence.
Nucleotide 344 of the isolated BFA4 clone was different from the reported sequence (C in BFA4, T in TRPS-1). The change resulted in a phe to ser amino acid change. To change this sequence to the reported sequence, the EcoRI/Bglll fragment (5') of the BFA4 gene from clone
^^r

JB-3552-1-2 (pCR2.1/TOPO/BFA4) was subcloned into. pUC8:2 to generate clone JB-3631-2. This clone was used as a template for Quickchange (Stratagene) mutagenesis to change amino acid 115 of the BFA4 protein from a serine to a phenylalanine as in the TRPSl protein. The selected clone was JB-3648-2-3. Mutagenesis was also repeated with pMCS5 BFA4 (BT474) as a template for Quickchange (Stratagene) mutagenesis to change amino acid 115 of the BFA4 protein from a serine to a phenylalanine as in the TRPSl protein. Several clones were found to be correct by DNA sequencing and one of the clones (JB-3685-1-18) was used for further subcloning.
JB-3685-1-18 was then used to subclone the BFA4 coding sequence into the BaniHI sites of four different expression vectors: 1) the poxviral (NYVAC) vector pSD554VC (C0PAK/H6; JB-3707-1-7); 2) pcDNA3.1/Zeo (+) (JB-3707-3-2); 3) pCAMycHis (JB-3707-5.1); and, 4) Semiliki Forest virus alphaviral repHcon vector pMP76 (JB-3735-1-23). The BFA4 coding sequence within JB-3707-1-7, JB-3707-5-1, and JB-3735-1-23 was confirmed by DNA sequencing.
A stop codon was introduced near the end of the cloned sequence in the pcDNA3.1/Zeo/BFA4 construct (JB-3707-3-2). A unique EcoRl site was opened and filled in to introduce a stop codon in-fi"ame with BFA4 coding sequence. Several putative clones were identified by the loss of EcoRl site, however three clones (JB-3756-1-2; JB-3756-3-1; and JB-3756-4-1) were sequenced. All three were found to be correct in the area of the fill-in. Clone JB-3756-3-1 identified as having the correct sequence and orientation.
Myc and myc/his tags (Evans et al, 1985) were introduced using ohgonucleotides, which were annealed and ligated into the pcDNA3.1/Zeo/BFA4 construct (JB-3707-3-2) at the EcoRI /EcoRV sites. Several clones were obtained for these constructs. Three clones having the correct sequences and orientations were obtained: 1) PcDNA3.1/Zeo/BFA4/myc-tag (JB-3773-1-2); 2) PcDNA3.1/Zeo/BFA4/mychis-tag (JB-3773-2-1); and, 3) PcDNA3.1/Zeo/BFA4/mychis-tag (JB-3773-2-2).
C. Expression of BFA4
1. Expression from poxviral vectors
The pSD554VC (C0PAK/H6; JB-3707-1-7) vector was used to generate ^fYVAC-BFA4 virus. In vitro recombination was performed with plasmid COPAK/H6/BFA4 and NYVAC in RK13/CEF cells. NYVAC-BFA4 (vP2033-NYVAC-RK13) was generated and amplified to P3 level after completion of three enrichments with final stock concentrations of 1.12 x 10^/ml
3"]

(10ml). Vero cells were infected with NYVAC-BFA4 at an M.O.L of 0.5 pfii/cell. Lysates and media were harvested 24h post-infection to confirm ejq)ression of BFA4 protein. One-twentieth of the concentrated media and 1/40 of the lysate were loaded onto a western blot and incubated with rabbit antisera against the BFA4 peptides CLP 2589, 2591, 2598 and 2594 (see above for peptide sequences and preparation of anti-BFA4 antiseara). An approximate 120kD band was detected in both the lysate and the concentrated media of NYVAC-BFA4-infected Vero cells which was not evident in either Vero control cells ("mock-infected"), Vero cells infected with the parental NYVAC virus, or concentrated media.
2. Expression from pcDNA3.1-based vectors
Transient transfection studies were performed to verify expression of BFA4 firom the pcDNA-based vectors and to analyze quality of polyclonal sera raised against BFA4 peptides. The following constructs were used to study expression of BFA4 gene: pcDNA 3.1 zeo^/BFA4, pMP76/BFA4, pcDNA 3.1 zeo^/ BFA4/Myc tag and pcDNA 3.1 zeo^/ BFA4/MycHis tag. BFA4 ejq)ression plasmids (5ng and 10 ^g) were co-transfected with pGL3 Luciferase (J Dg) (Promega) with the Gene porter reagent (Gene Therapy Systems) as the transfection reagent. At 48h post-transfection, whole cell extract was prepared by scraping cells in cell lysis reagent (200(xl) and 1 cycle of fireeze-thaw (-20°C freeze, 37°C thaw). Transfection efficiency was quantitated by analyzing expression of the luciferase reporter gene by measuring Relative Luciferase Units (RLU) in dupHcate. Similar RLU values were obtained in the samples c6-transfected with luciferase construct in the presence and absence of BFA4 expression vectors. There was no significant difference observed in toxicity or RLU values with differential amount (5ng and 10 fig) of BFA4 expression vectors. Preliminary western blot analysis using alkaline phosphatase system with the CHOKl cell extracts (pCDNA3.1 /zeo/ BFA4/MycHisTag) and an anti-BFA4 polyclonal antisera, revealed a band at e^proximately 120kDa band in extracts of BFA4 vector-transfected cells.
A stable transfection study was initiated to obtain stable clones of BFA4 expressing COS A2 cells. These cells are useful for in vitro stimulation assays. pcDNA 3.1 zeo^/BFAA (2.5\ig and 20 |ag), and pcDNA 3.1 zeo ^/ BFA4/MycHis tag (2.5|a.g) were used to study expression of BFA4). pGL3 Luciferase (2.5|j.g) was used as a conlrol vector to monitor transfection efficiency. The Gene porter reagent was used to faciUtate transfection of DNA vectors. After 48h post-transfection, whole cell extract were prepared by scraping cells in the cell lysis reagent (200jul)
^C^D

and 1 cycle of freeze-tiiaw at-20°C/37°C for first experiment Transfected cells obtained fi-om the second e^qieriment were trypsinized, fi-ozen stock established and cells were plated in increasing concentrations of Zeocin (0, 250, 500, 750 and 1000|ag/ml). Non-transfected CosA2cells survived at 60-80 % confluency for three weeks at 100|j,g/ml (Zeocin) and 10% confluency at 250ng/ml (Zeocin). However, after ttiree weeks, at higher drug concentration (500-lOOOjj.g/ml), live cells were not observed m the plates containing non-transfected cells and high Zeocin concentration (500-1 OOO^g/ml).
Several Zeocin-resistant clones growuig in differential drug concentrations (Zeocin-250, 500, 750 and lOOOjig/ml) were picked fi-om 10 cm plates after three weeks. These clones were fiMher expanded in a 3.5 cm plate(s) in the presence of Zeocin at 500, 750 and 1000 [ig/ml. Frozen lots of these clones were prepared and several clones fi-om each pool (pcDNA 3.1 zeo'^/BFA4, and pcDNA 3.1 TSO^I BFA4/MycHis tag) were expanded to T75 cm^ flasks in the presence of Zeocin at Img/ml. Five clones from each pool (pcDNA 3.1 zeo'^/BFA4, and pcDNA 3.1 zeoV BFA4/MycHis tag) were expanded to T75 cm^ flasks in the presence of Zeocin at Img/ml. Cells are maintained under Zeocin drug (Img/rol) selection. Six clones were used in BFA4 peptide-pulsed target experiment, and two clones were found to express BFA4 at a moderate level by immunological assays. The non-adherent cell lines K562A2 and EL4A2 were also transfected with these vectors to generate stable cell lines.
3. Prokaryotic expression vector
The BamHI -Xho-1 fi-agment (1.5 Kbp) fragment encoding N-terminal 54kDa BFDA4 from pCDNA3.1/BFA4 was cloned into pGEX4Tl-6His (Veritas) plasmid. This vector contains the tac promoter followed by the N-terminal glutathione S-transferase (GST~26kDa) and a hexahistidine tag to C terminus of the GST fiision protein.
The BFA4-N54 expression plasmid was transformed into BL21 cells and grown at 25°C in antibiotic selection medium (2L culture) to an OD (600nm) and thereafter induced with ImM IPTG. GST-BFA4-N54 was found to be soluble protein. Clarified extract of the soluble fraction was adsorbed batchwise to glutathione-Sepharose 4B and eluted with lOmM reduced glutathione. Fractions were analyzed after estimation of protein concentration and TCA precipitation. Specific polypeptide of Mr=85kDa in the eluate was confirmed by SDS-PAGE. The recombinant protein was purified by gluathione-Sepharose was absorbed on a NiNTA colunm for fijr&er purification. The bound protein was eluted with 0.25M imidazole. The protein was dialyzed versus TBS containing 40% Glycerol, resulting in 4.5 mg GST-BFA4-N54-6 His (N terminus
^t*(

BFA4 protein) protein. Expression of BFA4 was confirmed using the rabbit anti-BFA4 polyclonal antibody by western blot
p. Anti-BFA4 immune responses BFA4 peptides
In addition to genetic immunization vectors for BFA4, immunological reagents for BFA4 have been generated. A library of 100 nonamer peptides spanning the BFA4 gene product was synthesized. The peptides were chosen based on their potential ability to bind to HLA-A*0201. Table V lists 100 nonamer peptide epitopes for HLA-A*0201 fix>m the BFA4 protein tested (see below):
/^q2-





The peptide library was pooled into separate groups containing 7-10 different peptides for immunological testing as shown in Table VI (see below). In addition to a peptide library spanning BFA4, a recombinant protein spanning the N-terminal 300 amino acids (positions 1-300) has been synthesized and purified fixjm E. coli.
/M5'



2. Immune reactivity ofBFA4 peptides and generation of human effector T cells:
The BFA4 peptides were grouped into different pools of 7-10 peptides for immunological testing. Dissolved peptide pools were pulsed onto autologous HLA-A*0201 dendritic cells and used to activate autologous T-cell-enriched PBMC preparations. Activated T cells from each peptide-pool-stimulated culture were re-stimulated another 3 to 5 times using CD40L-activated autologous B-cells. IFN-y ELISPOT analysis and assays for CTL killing of peptide-pulsed target cells was performed to demonstrate the immunogenicity of these epitopes from BFA4.
Human T cells demonstrated effector cell activity against a number of pools of peptides from the BFA4 protein, as shown by their ability to secrete IFN-y in ELISPOT assays. These experiments were repeated after different rounds of APC stimulation resulting in the same reactive peptide groups. Peptide groups 1, 2, 4, 5, 6, 7, 8, 9, and 10 were found to be immunoreactive in these assays (Fig. 9A). Subsequently, these reactive peptide groups were de-convoluted in additional IFN-y ELISPOT assays in which single peptides from each group were tested separately. The individual peptides from BFA4 peptide groups 1, 5 6, 7, 8, 9, and 10 in ELISPOT assays (Fig. 9B). This analysis revealed a number of individual strongly reactive peptides from the BFA4 protein recognized by human T cells. It was also observed that many of these single peptides also induced CTL activity killing peptide-loaded human T2 lymphoma cell targets. These peptides are listed in Table VII:
r^-^


D. Immune responses against BFA4 after immunization in vivo:
The pcDNA3.1/Zeo-BFA4 plasmid was used to immunize transgenic mice expressing a hybrid HLA-A*0201 ala2 domain fused to a murine Kb a3 domain in C57BL/6 mice (A2-Kb mice). IFN-y ELISPOT analysis using the groups of pooled peptides after DNA immunization and removal of activated spleen cells revealed a number of reactive BFA4 peptide groups. Some of these groups (especially group 7 and 8) also reacted strongly in human T-cell cultures suggesting that overlapping groups of peptides are recognized by human T cells and are naturally processed and presented on HLA-A2 after vaccination.
Vaccination experiments were also performed with the NYVAC-BFA4 and the MP76-18-BFA4 vectors in A2-Kb mice. Mice were immunized subcutaneously with 10-2P^g of MP-76-18-BFA4 and 1-2 X 10^ pfo vP2033 (NYVAC-BFA4) and boosted 28 days later with the same amounts of each vector. Re-stimulation of spleen cells from the immunized mice with the pools of BFA4 peptides revealed induction of IFN-y production in response to BFA4 pqitide groups 2, 3, 4, 5, 7, 9, and 10 in ELISPOT assays. Thus, the BFA4 gene encoded in a CMV promoter

driven eukaryotic plasmid, KTVAC, or a Semliki replicase-based DNA plasmid, were all capable of inducing T-cell responses against the BFA4 protein in vivo.
Example 6 BCYl Tumor Antigen
The BCYl gene was detected as a partial open reading frame (ORF) homologous to a nematode gene called "posterior-expressed maternal gene-3" (PEM-3) playing a role in posterior to anterior patterning in Caenorhabtidis elegans embryos. No previous involvement of this gene in cancer has been documented.
A. BCYl and Amino Acid DNA Sequences
A partial DNA sequence was originally determined for BCYl. Primers, 9616SXC and 9617SXC, are derived from the BCY I partial DNA sequence and are designed to clone BCY I by RT-PCR from Calu 6 total RNA. The primers were designed such that the PCR product has BamHI sites at both ends and an ATG start codon and a Kozak sequence at the 5' end, as shown below:
9616SXC: 5' CAGTAGGGATCCACCATGGCCGAGCTGCGCCTGAAGGGC 3' 9617SXC: 5' CCACGAGGATCCTTAGGAGAATATTCGGATGGCTTGCG 3'
The 1.2 Kb expected amplicon was obtained using ThermoScript RT-PCR System (Invitrogen) under optimized conditions. The PCR products from three separate RT-PCR's were digested with BamHI and respectively inserted in pcDNA3.1/Zeo(+). The resulting clones were MC50A6, MC50A8 and MC50A19 from the first RT-PCR; MC54.21 from the second RT-PCR and MC55.29; and, MC55.32 from the third RT-PCR. The following primers were utilized in sequencing the clones:

9620MC 9621MC 9618MC 9619MC

5' TAATACGACTCACTATAGGG 3'
5' TAGAAGGCACAGTCGAGG 3'
5' GAAAACGACTTCCTGGCGGGGAG 3'
5' GCTCACCCAGGCGTGGGGCCTC 3'

DNA sequencing of all six clones indicated a consensus sequence (SEQ ID NO.: 25), as shown in Figs. lOA and lOB, having the following differences from the original partial BCYl sequence: a C to G substitution at position 1031 resulting in an amino acid change of Ala to Gly; a GC deletion at position 1032-1034 resulting in a Thr deletion; and, an A to G substitution at
M^

position 1177 resulting in an amino acid change of Thr to Ala. Clones MC50A8 and MC55.29 are identical to the consensus sequence. The amino acid sequence of BCYl is shown in Fig. lOB and(SEQroNO.:26).
B. Immunological reagents for BCYl breast cancer antigen:
A library of 100 nonamer peptides spanning the BCYl gene product was synthesized. The peptides were chosen based on their potential ability to bind to HLA-A*0201. Table Vin lists 100 nonamer peptide epitopes for HLA-A*0201 from the BCYl protein tested (see below):
^P







C. Immune reactivity of BCYl peptides and generation of human effector T cells
The library of 100 peptides from BCYl was separated into 10 groups of 7-10 peptides for immunological testing. Dissolved peptide pools Were pulsed onto autologous HLA-A*0201 dendritic cells and used to activate autologous T-cell-enriched PBMC preparations. Activated T cells from each peptide-pool-stimulated culture were re-stimulated another 3 to 5 times using CD40L-activated autologous B-cells. IFN-y ELISPOT analysis and assays for CTL killing of peptide-pulsed target cells was performed to demonstrate the immunogenicity of these epitopes from.BCYl.
Hunaan T cells demonstrated effector cell activity against a number of pools of peptides
from the BCYl protein, as shown by their abihty to secrete IFN-y in ELISPOT assays. These
experiments were repeated after different rounds of APC stimulation resulting in the same
reactive peptide groups. Peptide groups 1,2,3,4, 5,6, and 7 were found to be immxmoreactive in
•r these assays. Subsequentiy, these reactive peptide groups were de-convoluted in additional IFN-y
ELISPOT assays in which single peptides from each group were tested separately. This analysis
revealed a number of individual strongly reactive peptides from the BCYl protein recognized by
human T cells (Fig. 11). Many of these single peptides also induced CTL activity killing peptide-
loaded human T2 lymphoma cell targets. Table IX lists these pqitides.
Example 7 BFAS/NYBR-1 Breast Cancer Antigen A. Identification of BFA5
Microairay 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% 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: KIAA1074 (GenBank Accession No. X?vl_l 59732), aid, KIAA0565 (GenBank Accession No. ABOll 137) isolated fiom a number of fetal and adult brain 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 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). For the purposes of this apphcation, the sequence is referred to as BFA-5, the terms BFA-5 and NYBR-1 are interchangeable.
As shown previously by Jager, et al. and described in WO 01/47959, supra, BFA5 is specifically expressed in mannnary gland, being expressed in 12/19 breast tumors analyzed. The 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 rq)eats implying a role in protein-protein ititeractions; These ankyrin repeats may play a role in homo-dimerization of the protein. The BFA5 cDNA sequence is shown in FIG. 12 and SEQ ID NO.: 27. The BFA5 amino acid sequence is shown in FIG. 13 and SEQ ID NO.: 28.
B. ImmunoreactiyityofBFAS
i. Activation of human T cells and IFN-r secretion in ELISPOT.
A library of 100 peptides fi'om the BFA5/NYBR-1 coding sequence that are predicted to be medium to high binders to HLA-A*0201 were designed using Ranmiensee and Parker algorithms. The library was sub-divided into 10 pools often peptides (see Table XI), 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.
/J^"



ELISPOT analysis was performed on human T-cell cultures activated through four rounds of stimulation with each pool of BFA5 peptides. In Fig. 14A, the numbers under the X-axis indicate the number of each peptide pool (1-10). Reactivity against a CMV pp65 peptide and a Flu matrix peptide were used as positive controls for T-cell activation in the e3q)eriments. Each experiment was performed with PBMC and dendritic cells ifrom a single HLA-A*0201'*' donor 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 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 (Fig. 14A). Each reactive peptide pool was then separated into individual peptides and analyzed for immunogencity using ELISPOT analysis to isolate single reactive BFA5 peptides. As shown in Fig. 14B, BFA5 is highly immunogenic with several reactive single peptides than that of BFA4. Similar 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. Isolated T cells were tested in ^'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 individual peptides from the respective pool to the right. A hi^ 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 /^S'^



C. Immunological Reagents
Polyclonal antisera were generated against the following series of 22- to 23- mer peptides ofBFAS:

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 Hemocyanin (KLH)-coupled and administered as an emulsion with Freund's Incomplete Adjuvant FIA. The following results were observed:

Prebleed sample results exhibited IgG titers 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 ofBFAS protein is 153 kDa. A 220kD band was observed in the BT474 extract with CLP2980 antibody but not in the MDMB453 cell extracts however a ~130kD 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.
S^
^

EXAMPLES BCZ4 Tumor Antigen
A. BCZ4 Sequence
The BCZ4 sequence was detected as an over-e3q)ressed sequence in breast cancer samples. The nucleotide sequence and deduced amino acid sequence of BCZ4 are shown in Fig. 15, SEQ ID NO. 29 (BCZ4 cDNA), and SEQ ID NO. 30 (BCZ4 amino acid sequence).
B. Immunological reagents for BCZ4 breast cancer antigen:
A library of 100 nonamer peptides spanning the BCZ4 gene product was synthesized. The peptides were chosen based on their potential ability to bind to HLA-A*0201. Table XHI lists 100 nonamer peptide epitopes for HLA-A*0201 from the BCZ4 protein tested (see below):

/

b


C. Immune reactivity of BCZ4 peptides and generation of human effector T cells
Human PBMC from an HLA-A2.1 positive donor designated APIO were activated with autologous dendritic cells pulsed with different pools of 9-mer peptides from the. BCZ4 antigen (see Table XIII for list). The activated T cells were re-stimulated after 12 days with activated autologous CD40-ligand-activated B cells pulsed with the same respective peptide pools for another 8 to 10 days. This secondary activation was repeated more time for a total of 3 stimulations. The activated T cells were isolated after the 3rd stimulation and subjected to ELISPOT analysis for human IFN-y production against their respective BCZ4 peptide pools as shown (Fig. 16A). In Fig. 16A, the blue bars show reactivity against the BCZ4 peptide pools and the red bars are for an HLA-A2.1-binding HIV peptide as a negative control. Positive control HLA-A2.1-binding recall antigen peptides for CMV and flu were as used as positive control in the experiment Standard deviations are indicated. The experiment was repeatedon activated T cells after an additional round of peptide stimulation with the similar results.
The peptide pools were deconvoluted using IFN-y ELISPOT assays (Fig. 16 B). Human T cells from donor APIO were stimulated with the different pools of BCZ4 peptides shown in Table XIII. Stimulation was performed as described earlier for the other antigens described. After 4 and 5 rounds of stimulation, T cells were harvested and subjected to ELISPOT analysis for IFN-y production with each individual peptide in each pool. The bars shown represent individual peptide reactivity for each specific pool. Table XIII identifies each of the reactive peptides. This experiment was repeated with similar results following another round of stimulation of APIO donor T cells.
In addition to ELISPOT analysis, human T cells activated by BCZ4 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. Isolated T cells were tested in ^'Cr-release assays using peptide-loaded T2 cells. The % specific lysis at a 10:1 T-cell to target ratio is shown for T2 cells pulsed with individual BCZ4 peptides. A high level of cytotoxicity was observed for some peptides (Fig. 16C). CTL activity (percent specific lysis) induced by the control HIV pqitide was generally Table XFV lists the reactivity of the individual peptides:
^ ^ f



cycles of 94°C 30 seconds, 53°C 30 seconds, 67°C 2.5 minutes; and, 3) 6TC 7 minutes. PCR primers were designed to include EcoRI restriction sites and directly flank the ORF (i.e., no extraneous sequence). Primer sequences were as follows:
AS032F (forward primer) 5'
GGAATTC^4CATGGACATTGAAGCATATCTTGAAAGAATTG 3' AS034R (reverse primer) 5' GGAATTCCTGGTGAGCTGGATGACAAATAGAC AAGATTG 3'. A Kozak sequence was also included in the forward primer. pcDNA3.1/Zeo(+) was cut with EcoRI and treated with CEP to prevent self-ligation. The BCZ4 amplicon was then ligated into EcoRI digested pcDNA3.1/Zeo(+). Sequencing produced one clone (AS-579-5) which matched the expected BCZ4 sequence. BCZ4 protein was then ejqjressed fix)m this expression vector using standard techniques.
EXAMPLE 9 BFYS Tumor Antigen
A. BFY3 Sequence
The BFY3 sequence was detected as an over-expressed sequence in breast cancer samples. RT-PCR amplification of BFY3 w/EcoRI ends from HTB131 total RNA with AS007F (forward primer), 5' GGAATTG4CCATGCTTTGGAAATTGACGGAT 3' and ASOIOR (reverse primer) 5' GGAATTCCTCACTTTCTGTGCT TCTC CTCTTTGTCA 3' was performed. PCR product was digested with EcoRI and cloned into EcoRI.digested and CIP treated pcDNA3.1/Zeo(-i-) vector by ligation. Several positive clones were identified by restriction digestion and sequence results of AS-391-2 match expected BFY3 sequence. The nucleotide sequence and deduced amino acid sequence of BFYS are shown in Fig. 17, SEQ ID NO. 31 (BFY3 cDNA), and SEQ ID NO. 32 (BFY3 amino acid sequence).
B. Immunological reagents for BFYS breast cancer antigen
A library of 100 nonamer peptides spanning the BFY3 gene product was synthesized. The peptides were chosen based on their potential ability to bind to HLA-A*0201. Table XV lists 100 nonamer peptide epitopes for HLA-A*0201 from the BFYS protein tested (see below):




Human PBMC from an HLA-A2.1 positive donor designated AP31 were activated with autologous dendritic cells pulsed with different pools of 9-mer peptides from the BFY3 antigen (see Table 1 for list). The activated T cells were re-stimulated after 12 days with activated autologous CD40-ligand-activated B cells pulsed with the same respective peptide pools for another 8 to 10 days. This secondary activation was repeated 2 more time for a total of 4 stimulations. The activated T cells were isolated after the 4th stimulation and subjected to ELISPOT analysis for human IFN-y production against their respective BFY3 peptide pools as shown. The blue bars show reactivity against the BFY3 peptide pools and the red bars are for an HLA-A2.1-binding HTV peptide as a negative control. Standard deviations are indicated. The experiment was repeated 2 times on activated T cells from different rounds of peptide stimulation with the similar results (Fig. 18A).
The BFY3 peptide pools were deconvoluted and studied in IFN-y ELISPOT assays. Human T cells from donor APIO were stimulated with the different pools of BFY3 peptides shown in Table XV. Stimulation was performed as described earlier for the other antigens described. After 4 rounds of stimulation, the T cells from each culture were harvested and subjected to ELISPOT analysis for IFN-y production with each individual peptide in each pool. Fig. 18B illustrates individual peptide reactivity for each specific pool.
In addition to ELISPOT analysis, human T cells activated by BFY3 peptides were assayed for reactivity. Ten pools of peptides consisting of ten peptides per pool used to generate CTL. These 10 groups of effectors used to kill targets pulsed with corresponding peptide pools. Peptides from pools 1, 3, 5, 6, and 7 found to be recognized, indicating that peptides in those pools are capable of generating CTL (Fig. 18C). From these ten pools, peptides 3344, 3320, 3378, 2272, and 3387 were strongly recongized by CTL (Fig. 18D). "Moderately recognized" peptides include 3369, 3355, and 3362 (Fig. 18D). CosA2 cells transfected with BFY3 were killed by CTL generated from pools 1 and 3 indicating that processed and presented epitopes from these pools are immunologically relevant (Fig. 18E). The peptides responsible for this cytotoxicity are 3320 and 3344. Table XVT summarizes the properties of the BFY3 peptides.



C. BFY3 Expression Vectors
To construct a BFY3 expression vector, RT-PCR amplification of BFY3 w/EcoRI ends from HTB131 total RNA with AS007F (forward primer) 5' GGAATTC4CCATGCTTTGGAAATTGACGGAT 3' and ASOIOR (reverse, primer) 5' GGAATTCCTCACTTTCTGTGCTTCTCCTCTTTGTCA 3' was performed. PCR was performed using standard techniques. The amplified product was digested with EcoRI and cloned into CIP treated pcDNA3.1/Zeo(+) vector by ligation using standard techniques. Several positive ' clones were identified by restriction digestion and sequenced. Sequencing indicated that the sequence of clone AS-391-2 matched the expected BFY3 sequence. BFY3 protein was then expressed using firom the BFY3 expression vector using standard techniques.

EXAMPLE 10 Expression Vectors Encoding Multiple Tumor Antigens
In certain instances, it may be desirable to construct expression vectors encoding multiple tumor antigens. It has been determined that certain combinations of antigens, when combined into a single expression vector, encompasses the expression profiles of many patients in a single vector. For instance, one study of breast cancer samples from different patients indicated that the combination of BFA4 and BFA5 covered expression profiles of 74% of the samples; the combination of BCYl and BFA5 covered 65% of the samples; the combination of BCZ4 and BFA5 covered 69% of the samples; the combination of BFY3 and BFA5 covered 67% of the samples; the combination of BCYl, BFA4 and BFA5 covered 78% of the samples; the combination of BCZ4, BFA4 and BFA5 covered 81% of the samples; and, the combination of BFY3, BFA4, and BFA5 covered 74% of the samples. Accordingly, a multi-antigen expression construct may be built such that the most common expression profiles among breast cancer patients may be addressed using a single vector. Such a multiantigen expression vector is constructed using standard cloning techniques positioning nucleic acids encoding each of the tumor antigen sequences in proximity to a promoter or other transcriptional regulatory sequence. The expression vector may be engineered such that each nucleotide sequence encoding a tumor antigen is operably linked to a specific promoter, or the tumor antigens may collectively be operably linked to a single promoter and expressed as a single expression unit. Where a single expression unit is constructed, nucleotide sequences useful in separating the tumor antigen sequences following expression may be inserted between the tumor antigen sequences. Sequences useful for include IRES sequences, nucleotide sequences encoding amino acid sequences corresponding to protease cleavage sites, and the like. Suitable vectors for constructing such multiantigen expression vectors include, for example, poxviruses such as vaccinia, avipox, ALVAC and NYVAC.
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.



WE CLAIM :
1. An expression vector comprising the nucleic acid sequence encoding the amino
acid sequence illustrated in sequence ID No. 30 or sequence ID No. 32.
2. The expression vector as claimed in claim 1 wherein the nucleic acid sequence
is as illustrated in sequence ID No. 29 or sequence ID No. 31.
3. The expression vector as claimed in claim 1 wherein the vector is a plasmid or
a viral vector.
4. The expression vector as claimed in claim 3 wherein the viral vector is selected
from the group consisting of poxvirus, adenovirus, retrovirus, herpesvirus, and
adeno-associated virus.
5. The expression vector as claimed in claim 4 wherein the viral vector is a
poxvirus selected from the group consisting of vaccinia, NYVAC, avipox,
canarypox, ALVAC, ALVAC (2), fowlpox, and TROVAC.
6. The expression vector as claimed in claim 5 wherein the viral vector is a poxvirus selected from the group consisting of NYVAC, ALVAC, and ALVAC (2).
7. A composition comprising an expression vector as claimed in any of the preceding claims in a pharmaceutically acceptable carrier.

69

Documents:

3093-chenp-2005 abstract duplicate.pdf

3093-chenp-2005 abstract.pdf

3093-chenp-2005 claims duplicate.pdf

3093-chenp-2005 claims.pdf

3093-chenp-2005 correspondence-others.pdf

3093-chenp-2005 correspondence-po.pdf

3093-chenp-2005 descripition(completed) duplicate.pdf

3093-chenp-2005 descripition(completed).pdf

3093-chenp-2005 drawings duplicate.pdf

3093-chenp-2005 drawings.pdf

3093-chenp-2005 fomr-1.pdf

3093-chenp-2005 fomr-18.pdf

3093-chenp-2005 fomr-26.pdf

3093-chenp-2005 fomr-3.pdf

3093-chenp-2005 fomr-5.pdf

3093-chenp-2005 petition.pdf


Patent Number 232227
Indian Patent Application Number 3093/CHENP/2005
PG Journal Number 13/2009
Publication Date 27-Mar-2009
Grant Date 16-Mar-2009
Date of Filing 21-Nov-2005
Name of Patentee SANOFI PASTEUR, INC
Applicant Address Discovery Drive, Swiftwater, PA 18370,
Inventors:
# Inventor's Name Inventor's Address
1 RADVANYI, LASZLO 1755 Steeles Avenue, North York, Ontario M2R 3T4,
2 BERINSGTEIN NEIL 1755 Steeles Avenue, North York, Ontario M2R 3T4,
3 LOVITRT, COREY 1755 Steeles Avenue, North York, Ontario M2R 3T4,
4 PARRINGTON, MARK 1755 Steeles Avenue, North York, Ontario M2R 3T4,
5 SINGH-SANDHU 1755 Steeles Avenue, North York, Ontario M2R 3T4,
6 GALLICHAN, SCOTT 1755 Steeles Avenue, North York, Ontario M2R 3T4,
PCT International Classification Number C07K14/47
PCT International Application Number PCT/US04/15202
PCT International Filing date 2004-05-15
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/471,119 2003-05-16 U.S.A.
2 60/471,193 2003-05-16 U.S.A.