Combinations of tumor-associated antigens in compositions for various types of cancers

Abstract
Disclosed herein are methods and compositions for inducing an immune response against various combinations of tumor-associated antigens, which can promote effective immunologic intervention in pathogenic processes. Embodiments of the invention disclosed herein are directed to the use of effective combinations of TuAAs for the immunotherapy of patients with various types of cancer. Both immunogenic compositions for inducing an immune response to these combinations of antigens and methods for their use are disclosed.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


Disclosed herein are methods and compositions for inducing an immune response against various combinations of tumor-associated antigens, which can promote effective immunologic intervention in pathogenic processes.


2. Description of the Related Art


The American Cancer Society has estimated that over one million people get cancer each year, and that approximately one out of every two American men and one out of every three American women will have some type of cancer at some point during their lifetime.


Cancer generally develops when cells in a part of the body begin to grow out of control. Although there are many kinds of cancer, they usually start because of out-of-control growth of abnormal cells.


Normal body cells grow, divide, and die in an orderly fashion. Cancer cells are different in that they continue to grow and divide. Instead of dying, they outlive normal cells and continue to form new abnormal cells.


Usual treatment options for cancer include surgery, radiation therapy, and chemotherapy. A fourth branch of treatment is developing, which is referred to as immunotherapy. Immunotherapies attempt to help the immune system recognize cancer cells, and/or to strengthen a response against cancer cells in order to destroy the cancer. Immunotherapies include active and passive immunotherapies. Active immunotherapies attempt to stimulate the body's own immune system to fight the disease. Passive immunotherapies generally do not rely on the body to attack the disease; instead, they use immune system components (such as antibodies) created outside of the body.


Despite the various types of treatments, a continuing need exists for additional treatment options.


SUMMARY OF THE INVENTION

Embodiments of the invention disclosed herein are directed to the use of effective combinations of TuAAs for the immunotherapy of patients with various types of cancer. Both immunogenic compositions for inducing an immune response to these combinations of antigens and methods for their use are disclosed.


Some embodiments relate to methods of treating neoplastic diseases. The methods can include the step of immunizing a patient against, for example, PRAME and at least one other tumor associated antigen. To immunize a patient against an antigen such as, for example, PRAME, means to administer to the patient some portion of the antigen, or some other immunogenic agent, that is capable of inducing a specific immune response directed to the antigen. Accordingly, in some embodiments, immunizing against PRAME can include administering a complete and intact PRAME antigen to the patient, while in other embodiments it can include administering one or more epitopes, one or more epitope clusters, one or more fragments, and the like, of PRAME, and/or administering, for example, a nucleic acid encoding any of the foregoing epitope(s), cluster(s), fragment(s), and the like. Although PRAME is used in an exemplary way for convenience in this discussion, the principle is applicable to any antigen against which a patient may be immunized.


One embodiment relates to methods of treating ovarian or colorectal cancer. The methods can include the step of immunizing a patient against, for example, PRAME and at least one other tumor associated antigen. In some embodiments the tumor associated antigen can include, for example, SSX-2, NY-ESO-1, PSMA, and the like. Preferably, the methods can include immunizing against PRAME, NY-ESO-1 and SSX-2, for example. More preferably, the methods can include immunization against PRAME, NY-ESO-1, SSX-2, and PSMA, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2. In a preferred embodiment, the methods can induce a cytolytic T cell response.


In one embodiment, a method of inducing a cytolytic T-cell response in the treatment of ovarian or colorectal cancer is disclosed. The method can include, for example, the step of immunizing a pateint against PRAME and at least one of the tumor-associated antigens. In some embodiments, the tumor-associated antigen can be, for example, SSX-2, NY-ESO-1, PSMA, and the like. For example, the method can include immunization against PRAME, NY-ESO-1, and SSX-2. The method can further include immunization against PSMA. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2.


Other embodiments relate to methods of treating pancreatic cancer. The methods can include the step of immunizing against, for example, PSMA and at least one other tumor-associated antigen. The tumor-associated antigen can be, for example, SSX-2 and NY-ESO-1. Preferably, the methods can include immunizing against PSMA, NY-ESO-1, and SSX-2. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the methods can induce a cytolytic T cell response.


Further, in one embodiment, a method of inducing a cytolytic T-cell response in the treatment of pancreatic cancer is disclosed. The method can include, for example, the step of immunizing a patient against PSMA and at least one of the tumor-associated antigens. In some embodiments, the tumor-associated antigen can be, for example, SSX-2, NY-ESO-1, and the like. For example, the method can include immunization against PSMA, NY-ESO-1, and SSX-2. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Still other embodiments relate to methods of treating non-small cell lung cancer. The methods can include the step of immunizing a patient against, for example, PSMA and at least one other tumor associated antigen. For example, the tumor-associated antigen can be MAGE-3, SSX-2, NY-ESO-1, and the like. Preferably, the methods can include immunizing against PSMA, NY-ESO-1 and SSX-2. More preferably, the methods can include immunization against PSMA, NY-ESO-1, SSX-2, and MAGE-3, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the methods can induce a cytolytic T cell response.


In a further embodiment, a method of inducing a cytolytic T-cell response in the treatment of non-small cell lung cancer is disclosed. The method can include, for example, the step of immunizing a patient against PSMA and at least one of tumor-associated antigen. The tumor-associated antigen can be, for example, MAGE-3, SSX-2, NY-ESO-1, and the like. For example, the method can include immunization against PSMA, NY-ESO-1, and SSX-2. The method can further include immunization against PSMA, NY-ESO-1, SSX-2, and MAGE-3. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Alternative embodiments relate to methods of treating renal cell carcinoma. The methods can include the step of immunizing a patient against, for example, PSMA and at least one other tumor associated antigen. The tumor-associated antigen can be, for example, PRAME, SSX-2, and the like. Preferably, the methods can include immunizing against PSMA, PRAME, and SSX-2, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the methods can induce a cytolytic T cell response.


Further, in one embodiment, a method of inducing a cytolytic T-cell response in the treatment of renal cell carcinoma is disclosed. The method can include, for example, the step of immunizing a patient against PSMA and at least one other tumor-associated antigen. In some embodiments, the tumor-associated antigen can be PRAME, SSX-2, and the like. For example, the method can include immunization against PSMA, PRAME, and SSX-2. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Some embodiments relate to methods of treating melanoma. The methods can include the step of immunizing a patient against, for example, at least one tumor-associated antigen selected from each of two groups. The first group can include, for example, tyrosinase, Melan-A, and the like. The second group can include, for example, SSX-2, NY-ESO-1, and the like. Preferably, the method can include immunizing against Melan-A, SSX-2, and NY-ESO. More preferably, the method can include immunization against Melan-A, SSX-2, and tyrosinase, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2. In a preferred embodiment, the methods can induce a cytolytic T cell response.


In one embodiment, a method of inducing a cytolytic T-cell response in the treatment of melanoma is disclosed. The method can include, for example, the step of immunizing a patient against at least one tumor-associated antigen selected from each of two groups. The first group can include, for example, tyrosinase, Melan-A, and the like. The second group can include, for example, SSX-2, NY-ESO-1, and the like. For example, the method can include immunization against Melan-A, SSX-2, and NY-ESO. Alternatively, the method can include, for example, immunization against Melan-A, SSX-2, and tyrosinase. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2.


Some embodiments relate to the use of a composition comprising, for example, PRAME and at least one other tumor associated antigen, in the preparation of a medicament for the treatment of treating ovarian or colorectal cancer. In some embodiments the tumor associated antigen can include, for example, SSX-2, NY-ESO-1, PSMA, and the like. Preferably, the tumor-associated antigens can be NY-ESO-1 and SSX-2, for example. More preferably, the tumor-associated antigens can be NY-ESO-1, SSX-2, and PSMA, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2. In a preferred embodiment, the medicament can induce a cytolytic T cell response.


Other embodiments relate to the use of a composition comprising, for example, PSMA and at least one other tumor-associated antigen in the preparation of medicament for the treatment of pancreatic cancer. The tumor-associated antigen can be, for example, SSX-2 and NY-ESO-1. Preferably, the medicament comprises PSMA, NY-ESO-1, and SSX-2. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the medicament can induce a cytolytic T cell response.


Still other embodiments relate to the use of a composition comprising PSMA and at least one other tumor associated antigen in the preparation of a medicament for the treatment of non-small cell lung cancer. In some embodiments, the tumor-associated antigen can be MAGE-3, SSX-2, NY-ESO-1, and the like. Preferably, the composition comprises PSMA, NY-ESO-1 and SSX-2. More preferably, the composition comprises PSMA, NY-ESO-1, SSX-2, and MAGE-3, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the medicament can induce a cytolytic T cell response.


Alternative embodiments relate to the use of a composition comprising PSMA and at least one other tumor associated antigen in the preparation of a medicament for the treatment of renal cell carcinoma. The tumor-associated antigen can be, for example, PRAME, SSX-2, and the like. Preferably, the composition comprises PSMA, PRAME, and SSX-2, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2. In a preferred embodiment, the medicament can induce a cytolytic T cell response.


Still other embodiments relate to the use of a composition comprising at least one tumor-associated antigen selected from each of two groups in the preparation of a medicament for the treatment of melanoma. The first group can include, for example, tyrosinase, Melan-A, and the like. The second group can include, for example, SSX-2, NY-ESO-1, and the like. Preferably, the composition comprises Melan-A, SSX-2, and NY-ESO. More preferably, the composition comprises Melan-A, SSX-2, and tyrosinase, for example. The method can further include the step of immunizing against at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2, and Tie-2. In a preferred embodiment, the medicament can induce a cytolytic T cell response.


Other embodiments relate to immunogenic compositions for the treatment of or for inducing a T cell response against ovarian or colorectal cancer, for example. The compositions can include, for example, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, 5) nucleic acids encoding any of 1 to 4, and the like; wherein the antigens can include PRAME and a tumor-associated antigen, including, for example, SSX-2, NY-ESO-1, PSMA, and the like. The compositions can further include at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2 and Tie-2.


Still other embodiments relate to immunogenic compositions for the treatment of or for inducing a T cell response against pancreatic cancer. The compositions can include, for example, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens can include PSMA and at least one tumor-associated antigen. In a preferred embodiment, the tumor-associated antigen can be selected from SSX-2 and NY-ESO-1, for example. The compositions can further include at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Alternative embodiments relate to immunogenic compositions for the treatment of or for inducing a T cell response against non-small cell lung cancer. The compositions can include, for example, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens can include PSMA and at least one tumor-associated antigen, including, for example, MAGE-3, SSX-2 and NY-ESO-1. The compositions can further include at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Some embodiments relate to immunogenic compositions for the treatment of or for inducing a T cell response against renal cell carcinoma. The compositions can include, for example, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens can include PSMA and at least one tumor-associated antigen. In a preferred embodiment the tumor associated antigen can be PRAME or SSX-2. The compositions can further include at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, VEGFR2 and Tie-2.


Still other embodiments relate to an immunogenic composition for the treatment of or for inducing a T cell response against melanoma. The composition can include, for example, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens can be selected from each of two groups; wherein the first group includes, for example, tyrosinase and melan-A; and wherein the second group includes SSX-2 and NY-ESO-1, and the like. The compositions can further include at least one antigen associated with tumor neovasculature. The antigen associated with tumor neovasculature can be, for example, PSMA, VEGFR2 and Tie-2.


Further embodiments relate to compositions and methods for inducing a CTL response using combinations of tumor-associated antigens, including fragments of tumor-associated antigens, clusters and epitopes. The compositons can include nucleic acid constructs, for example, a single construct that encodes all of the desired angitens. In other embodiments a single construct encodes a single antigen, while in other embodiments one construct can have a combination of epitopes with similar immunogenicity and another construct can have epitopes with similar immunogenicity.


Still other embodiments relate to methods of designing and preparing immunogenic compositions, which methods can include the steps of determining the presence of one or more antigens on a tumor type, and obtaining the one or more antigens for inclusion in a composition that induces CTL.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the schedule of immunization with two plasmid (pCBP expressing SSX2 41-49 and pSEM expressing Melan A).



FIG. 2 is a bar graph that shows CTL activity obtained using the protocol in FIG. 1.



FIG. 3 shows the schedule of immunization of an entrain-and-amplify immunization protocol using plasmids and peptides representing two epitopes.



FIG. 4 is a table showing in vivo clearance of epitope-pulsed cells in mice immunized according to the protocol of FIG. 3.



FIG. 5 shows the preferred immunization protocols for inducing multivalent responses.




DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT

The frequency of expression of many tumor-associated antigens (TuAAs) in various types of cancers is known. However, the frequency of appearance of some antigens, and especially certain combinations of TuAAs in various types of cancers has not been reported. Accurate measurement of the presence of TuAAs in tumor tissues aids in determining which TuAAs will be useful for the treatment of a particular type of cancer.


Many attempts to develop active immunotherapies for cancer have utilized a single antigen. This can be problematic for two distinct reasons. Firstly, the expression of any particular TuAA in cancer can be mosaic with the antigen expression ranging from high in some cells within a tumor mass to completely absent in others. Moreover, the TuAA may be expressed in some lesions but not others. By directing an immune response against more than a single antigen, if properly selected, the number of tumor cells that can be recognized can be maximized. Secondly, it has been observed in some cases that tumors lose expression of a TuAA following immunization, giving rise to a resistant population. If the immune response is directed against more than one TuAA it becomes much more difficult for a resistant tumor to arise because it must then simultaneously lose expression of each of the antigens in order to escape. Thus, in treating cancer with immunotherapy, it can be advantageous to use a combination of TuAAs both due to more complete coverage of the population of tumor cells, and because there will be less chance of tumor escape through antigen loss of expression of the TuAAs, provided the tumor is positive for at least two of the TuAAs used.


Definitions


Unless otherwise clear from the context of the use of a term herein, the following listed terms shall generally have the indicated meanings for purposes of this description.


PROFESSIONAL ANTIGEN-PRESENTING CELL (pAPC)—a cell that possesses T cell costimulatory molecules and is able to induce a T cell response. Well characterized pAPCs include dendritic cells, B cells, and macrophages.


PERIPHERAL CELL—a cell that is not a pAPC.


HOUSEKEEPING PROTEASOME—a proteasome normally active in peripheral cells, and generally not present or not strongly active in pAPCs.


IMMUNOPROTEASOME—a proteasome normally active in pAPCs; the immunoproteasome is also active in some peripheral cells in infected tissues or following exposure to interferon.


EPITOPE —a molecule or substance capable of stimulating an immune response. In preferred embodiments, epitopes according to this definition include but are not necessarily limited to a polypeptide and a nucleic acid encoding a polypeptide, wherein the polypeptide is capable of stimulating an immune response. In other preferred embodiments, epitopes according to this definition include but are not necessarily limited to peptides presented on the surface of cells, the peptides being non-covalently bound to the binding cleft of class I MHC, such that they can interact with T cell receptors (TCR). Epitopes presented by class I MHC may be in immature or mature form. “Mature” refers to an MHC epitope in distinction to any precursor (“immature”) that may include or consist essentially of a housekeeping epitope, but also includes other sequences in a primary translation product that are removed by processing, including without limitation, alone or in any combination, proteasomal digestion, N-terminal trimming, or the action of exogenous enzymatic activities. Thus, a mature epitope may be provided embedded in a somewhat longer polypeptide, the immunological potential of which is due, at least in part, to the embedded epitope; likewise, the mature epitope can be provided in its ultimate form that can bind in the MHC binding cleft to be recognized by TCR.


MHC EPITOPE—a polypeptide having a known or predicted binding affinity for a mammalian class I or class II major histocompatibility complex (MHC) molecule.


HOUSEKEEPING EPITOPE—In a preferred embodiment, a housekeeping epitope is defined as a polypeptide fragment that is an MHC epitope, and that is displayed on a cell in which housekeeping proteasomes are predominantly active. In another preferred embodiment, a housekeeping epitope is defined as a polypeptide containing a housekeeping epitope according to the foregoing definition, that is flanked by one to several additional amino acids. In another preferred embodiment, a housekeeping epitope is defined as a nucleic acid that encodes a housekeeping epitope according to the foregoing definitions. Exemplary housekeeping epitopes are provided in U.S. application Ser. Nos. 10/117,937, filed on Apr. 4, 2002 (Pub. No. 20030220239 A1), and Ser. No. 10/657,022, and in PCT Application No. PCT/US2003/027706 (Pub. No. WO04022709A2), filed Sep. 5, 2003; and U.S. Provisional Application No. 60/282,211, filed on Apr. 6, 2001; 60/337,017, filed on Nov. 7, 2001; 60/363,210 filed Mar. 7, 2002; and 60/409,123, filed on Sep. 5, 2002. Each of the listed applications is entitled EPITOPE SEQUENCES. Each of the applications mentioned in this paragraph is incorporated herein by reference in its entirety.


IMMUNE EPITOPE—In a preferred embodiment, an immune epitope is defined as a polypeptide fragment that is an MHC epitope, and that is displayed on a cell in which immunoproteasomes are predominantly active. In another preferred embodiment, an immune epitope is defined as a polypeptide containing an immune epitope according to the foregoing definition, that is flanked by one to several additional amino acids. In another preferred embodiment, an immune epitope is defined as a polypeptide including an epitope cluster sequence, having at least two polypeptide sequences having a known or predicted affinity for a class I MHC. In yet another preferred embodiment, an immune epitope is defined as a nucleic acid that encodes an immune epitope according to any of the foregoing definitions.


TARGET CELL—In a preferred embodiment, a target cells is a cell associated with a pathogenic condition that can be acted upon by the components of the immune system, for example, a cell infected with a virus or other intracellular parasite, or a neoplastic cell. In another embodiment, a target cell is a cell to be targeted by the vaccines and methods of the invention. Examples of target cells according to this definition include but are not necessarily limited to: a neoplastic cell and a cell harboring an intracellular parasite, such as, for example, a virus, a bacterium, or a protozoan. Target cells can also include cells that are targeted by CTL as a part of an assay to determine or confirm proper epitope liberation and processing by a cell expressing immunoproteasome, to determine T cell specificity or immunogenicity for a desired epitope. Such cells can be transformed to express the liberation sequence, or the cells can simply be pulsed with peptide/epitope.


TARGET-ASSOCIATED ANTIGEN (TAA)—a protein or polypeptide present in a target cell.


TUMOR-ASSOCIATED ANTIGEN (TuAA)—a TAA, wherein the target cell is a neoplastic cell.


HLA EPITOPE—a polypeptide having a known or predicted binding affinity for a human class I or class II HLA complex molecule.


ANTIBODY—a natural immunoglobulin (Ig), poly- or monoclonal, or any molecule composed in whole or in part of an Ig binding domain, whether derived biochemically, or by use of recombinant DNA, or by any other means. Examples include inter alia, F(ab), single chain Fv, and Ig variable region-phage coat protein fusions.


SUBSTANTIAL SIMILARITY—this term is used to refer to sequences that differ from a reference sequence in an inconsequential way as judged by examination of the sequence. Nucleic acid sequences encoding the same amino acid sequence are substantially similar despite differences in degenerate positions or minor differences in length or composition of any non-coding regions. Amino acid sequences differing only by conservative substitution or minor length variations are substantially similar. Additionally, amino acid sequences comprising housekeeping epitopes that differ in the number of N-terminal flanking residues, or immune epitopes and epitope clusters that differ in the number of flanking residues at either terminus, are substantially similar. Nucleic acids that encode substantially similar amino acid sequences are themselves also substantially similar.


FUNCTIONAL SIMILARITY—this term is used to refer to sequences that differ from a reference sequence in an inconsequential way as judged by examination of a biological or biochemical property, although the sequences may not be substantially similar. For example, two nucleic acids can be useful as hybridization probes for the same sequence but encode differing amino acid sequences. Two peptides that induce cross-reactive CTL responses are functionally similar even if they differ by non-conservative amino acid substitutions (and thus may not be within the substantial similarity definition). Pairs of antibodies, or TCRs, that recognize the same epitope can be functionally similar to each other despite whatever structural differences exist. Testing for functional similarity of immunogenicity can be conducted by immunizing with the “altered” antigen and testing the ability of an elicited response, including but not limited to an antibody response, a CTL response, cytokine production, and the like, to recognize the target antigen. Accordingly, two sequences may be designed to differ in certain respects while retaining the same function. Such designed sequence variants of disclosed or claimed sequences are among the embodiments of the present invention.


EXPRESSION CASSETTE—a polynucleotide sequence encoding a polypeptide, operably linked to a promoter and other transcription and translation control elements, including but not limited to enhancers, termination codons, internal ribosome entry sites, and polyadenylation sites. The cassette can also include sequences that facilitate moving it from one host molecule to another.


EMBEDDED EPITOPE—in some embodiments, an embedded epitope is an epitope that is wholly contained within a longer polypeptide; in other embodiments, the term also can include an epitope in which only the N-terminus or the C-terminus is embedded such that the epitope is not wholly in an interior position with respect to the longer polypeptide.


MATURE EPITOPE—a peptide with no additional sequence beyond that present when the epitope is bound in the MHC peptide-binding cleft.


EPITOPE CLUSTER—a polypeptide, or a nucleic acid sequence encoding it, that is a segment of a protein sequence, including a native protein sequence, comprising two or more known or predicted epitopes with binding affinity for a shared MHC restriction element. In preferred embodiments, the density of epitopes within the cluster is greater than the density of all known or predicted epitopes with binding affinity for the shared MHC restriction element within the complete protein sequence. Epitope clusters are disclosed and more fully defined in U.S. patent application Ser. No. 09/561,571 entitled EPITOPE CLUSTERS, which is incorporated herein by reference in its entirety.


LIBERATION SEQUENCE—a designed or engineered sequence comprising or encoding a housekeeping epitope embedded in a larger sequence that provides a context allowing the housekeeping epitope to be liberated by processing activities including, for example, immunoproteasome activity, N terminal trimming, and/or other processes or activities, alone or in any combination.


CTLp —CTL precursors are T cells that can be induced to exhibit cytolytic activity. Secondary in vitro lytic activity, by which CTLp are generally observed, can arise from any combination of naïve, effector, and memory CTL in vivo.


MEMORY T CELL—A T cell, regardless of its location in the body, that has been previously activated by antigen, but is in a quiescent physiologic state requiring re-exposure to antigen in order to gain effector function. Phenotypically they are generally CD62L CD44hi CD107α IGN-γ LTβTNF-α and is in G0 of the cell cycle.


EFFECTOR T CELL—A T cell that, upon encountering antigen antigen, readily exhibits effector function. Effector T cells are generally capable of exiting the lymphatic system and entering the immunological periphery. Phenotypically they are generally CD62L CD44hi CD107α+ IGN-γ+LTβ+ TNF-α+ and actively cycling.


EFFECTOR FUNCTION—Generally, T cell activation generally, including acquisition of cytolytic activity and/or cytokine secretion.


INDUCING a T cell response—Includes in many embodiments the process of generating a T cell response from naïve, or in some contexts, quiescent cells; activating T cells.


AMPLIFYING a T cell response—Includes in many embodiments the process or increasing the number of cells, the number of activated cells, the level of activity, rate of proliferation, or similar parameter of T cells involved in a specific response.


ENTRAINMENT—Includes in many embodiments an induction that confers particular stability on the immune profile of the induced lineage of T cells.


TOLL-LIKE RECEPTOR (TLR)—Toll-like receptors (TLRs) are a family of pattern recognition receptors that are activated by specific components of microbes and certain host molecules. As part of the innate immune system, they contribute to the first line of defense against many pathogens, but also play a role in adaptive immunity.


TOLL-LIKE RECEPTOR (TLR) LIGAND—Any molecule capable of binding and activating a toll-like recepetor. Examples include, without limitation: poly IC A synthetic, double-stranded RNA know for inducing interferon. The polymer is made of one strand each of polyinosinic acid and polycytidylic acid, double-stranded RNA, unmethylated CpG oligodeoxyribonucleotide or other immunostimulatory sequences (ISSs), lipopolysacharide (LPS), β-glucans, and imidazoquinolines, as well as derivatives and analogues thereof.


IMMUNOPOTENTIATING ADJUVANTS—Adjuvants that activate pAPC or T cells including, for example: TLR ligands, endocytic-Pattern Recognition Receptor (PRR) ligands, quillaja saponins, tucaresol, cytokines, and the like. Some preferred adjuvants are disclosed in Marciani, D. J. Drug Discovery Today 8:934-943, 2003, which is incorporated herein by reference in its entirety.


IMMUNOSTIMULATORY SEQUENCE (ISS)—Generally an oligodeoxyribonucleotide containing an unmethlylated CpG sequence. The CpG may also be embedded in bacterially produced DNA, particularly plasmids. Further embodiments include various analogues; among preferred embodiments are molecules with one or more phosphorothioate bonds or non-physiologic bases.


VACCINE—In preferred embodiments a vaccine can be an immunogenic composition providing or aiding in prevention of disease. In other embodiments, a vaccine is a composition that can provide or aid in a cure of a disease. In others, a vaccine composition can provide or aid in amelioration of a disease. Further embodiments of a vaccine immunogenic composition can be used as therapeutic and/or prophylactic agents.


IMMUNIZATION—a process to induce partial or complete protection against a disease. Alternatively, a process to induce or amplify an immune system response to an antigen. In the second definition it can connote a protective immune response, particularly proinflammatory or active immunity, but can also include a regulatory response. Thus in some embodiments immunization is distinguished from tolerization (a process by which the immune system avoids producing proinflammatory or active immunity) while in other embodiments this term includes tolerization.


ENCODE—an open-ended term such that a nucleic acid encoding a particular amino acid sequence can consist of codons specifying that (poly)peptide, but can also comprise additional sequences either translatable, or for the control of transcription, translation, or replication, or to facilitate manipulation of some host nucleic acid construct.


COVERAGE—the fraction or proportion of tumor cells expressing a particular TuAA or at least one TuAA from a set of selected TuAAs.


REDUNDANCY—the degree to which a population of tumor cells, or some subset of them, express more than one of a selected set of TuAAs.


Tumor Associated Antigens


Examples of TuAAs useful in embodiments disclosed herein include tyrosinase (SEQ. ID NO. 1), melan-A, (SEQ. ID NO. 2), SSX-2, (SEQ. ID NO. 3), PSMA (prostate-specific membrane antigen) (SEQ. ID NO. 4), MAGE-1, (SEQ. ID NO. 5), MAGE-3 (SEQ. ID NO. 6), NY-ESO-1, (SEQ. ID NO. 7), PRAME, (SEQ. ID NO. 8), and Her2/Neu (SEQ. ID NO. 9). The natural coding sequences for these nine proteins, or any segments within them, can be determined from their cDNA or complete coding (cds) sequences, SEQ. ID NO. 10-18, respectively.

TABLE 1SEQ. ID NOS.ACCESSIONSEQ. ID NO.IDENTITYNUMBER**1Tyrosinase proteinP146792Melan-A proteinQ166553SSX-2 proteinNP_0031384PSMA proteinNP_0044675MAGE-1 proteinP433556MAGE-3 proteinP433577NY-ESO-1 proteinP783588PRAME proteinNP 0061069Her2/Neu proteinP0462610Tyrosinase cDNANM_00037211Melan-A cDNAU0645212SSX-2 cDNANM_00314713PSMA cDNANM_00447614MAGE-1 cdsM7748115MAGE-3 cdsU0373516NY-ESO-1 cDNAU8745917PRAME cDNANM_00611518Her2/Neu cDNAM11730
**All accession numbers used here and throughout can be accessed through the NCBI databases, for example, through the Entrez seek and retrieval system on the world wide web.


Tyrosinase is a melanin biosynthetic enzyme that is considered one of the most specific markers of melanocytic differentiation. Tyrosinase is expressed in few cell types, primarily in melanocytes, and high levels are often found in melanomas. The usefulness of tyrosine as a TuAA is taught in U.S. Pat. No. 5,747,271 entitled “METHOD FOR IDENTIFYING INDIVIDUALS SUFFERING FROM A CELLULAR ABNORMALLY SOME OF WHOSE ABNORMAL CELLS PRESENT COMPLEXES OF HLA-A2/TYROSINASE DERIVED PEPTIDES, AND METHODS FOR TREATING SAID INDIVIDUALS” which is hereby incorporated by reference in its entirety.


GP10, also known as PMell7, also is a melanin biosynthetic protein expressed at high levels in melanomas. GP100 as a TuAA is disclosed in U.S. Pat. No. 5,844,075 entitled “MELANOMA ANTIGENS AND THEIR USE IN DIAGNOSTIC AND THERAPEUTIC METHODS,” which is hereby incorporated by reference in its entirety.


Melan-A, also called MART-1 (Melanoma Antigen Recognized by T cells), is another melanin biosynthetic protein expressed at high levels in melanomas. The usefulness of Melan-A/MART-1 as a TuAA is taught in U.S. Pat. Nos. 5,874,560 and 5,994,523 both entitled “MELANOMA ANTIGENS AND THEIR USE IN DIAGNOSTIC AND THERAPEUTIC METHODS,” as well as U.S. Pat. No. 5,620,886, entitled “ISOLATED NUCLEIC ACID SEQUENCE CODING FOR A TUMOR REJECTION ANTIGEN PRECURSOR PROCESSED TO AT LEAST ONE TUMOR REJECTION ANTIGEN PRESENTED BY HLA-A2”, each of which is hereby incorporated by reference in its entirety.


SSX-2, also know as Hom-Mel-40, is a member of a family of highly conserved cancer-testis antigens (Gure, A. O. et al. Int. J. Cancer 72:965-971, 1997, which is hereby incorporated by reference in its entirety). Its identification as a TuAA is taught in U.S. Pat. No. 6,025,191 entitled “ISOLATED NUCLEIC ACID MOLECULES WHICH ENCODE A MELANOMA SPECIFIC ANTIGEN AND USES THEREOF,” which is hereby incorporated by reference in its entirety. Cancer-testis antigens are found in a variety of tumors, but are generally absent from normal adult tissues except testis. Expression of different members of the SSX family has been found in various tumor cell lines. Due to the high degree of sequence identity among SSX family members, similar epitopes from more than one member of the family will be generated and able to bind to an MHC molecule, so that some vaccines directed against one member of this family can cross-react and be effective against other members of this family.


MAGE-1 (melanoma-associated antigen-1), MAGE-2 (melanoma-associated antigen-2), and MAGE-3 (melanoma-associated antigen-3) are members of another family of cancer-testis antigens originally discovered in melanoma but found in a variety of tumors. The identification of MAGE proteins as TuAAs is taught in U.S. Pat. No. 5,342,774 entitled NUCLEOTIDE SEQUENCE ENCODING THE TUMOR REJECTION ANTIGEN PRECURSOR, MAGE-1, which is hereby incorporated by reference in its entirety, and in numerous subsequent patents. Currently there are 17 entries for (human) MAGE in the SWISS Protein database. There is extensive similarity among these proteins, such that in many cases, an epitope from one can induce a cross-reactive response to other members of the family. A few members of the MAGE family have not been observed in tumors, most notably MAGE-H1 and MAGE-D1, which are expressed in testes and brain, and bone marrow stromal cells, respectively. The possibility of cross-reactivity on normal tissue is ameliorated by the fact that they are among the least similar to the other MAGE proteins.


GAGE-1 is a member of the GAGE family of cancer testis antigens (Van den Eynde, B., et al., J. Exp. Med. 182: 689-698, 1995; U.S. Pat. Nos. 5,610,013; 5,648,226; 5,858,689; 6,013,481; and 6,069,001). The PubGene database currently lists 12 distinct accessible members, some of which are synonymously known as PAGE or XAGE. GAGE-1 through GAGE-8 have a very high degree of sequence identity, so most epitopes can be shared among multiple members of the family.


BAGE is a cancer-testis antigen commonly expressed in melanoma, particularly metastatic melanoma, as well as in carcinomas of the lung, breast, bladder, and squamous cells of the head and neck. Its usefulness as a TuAA is taught in U.S. Pat. Nos. 5,683,88 entitled “TUMOR REJECTION ANTIGENS WHICH CORRESPOND TO AMINO ACID SEQUENCES IN TUMOR REJECTION ANTIGEN PRECURSOR BAGE, AND USES THEREOF” and U.S. Pat. No. 5,571,711 entitled “ISOLATED NUCLEIC ACID MOLECULES CODING FOR BAGE TUMOR REJECTION ANTIGEN PRECURSORS,” each of which is hereby incorporated by reference in its entirety.


NY-ESO-1, also known as CTAG-1 (Cancer-Testis Antigen-1) and CAG-3 (Cancer Antigen-3), is a cancer-testis antigen found in a wide variety of tumors. NY-ESO-1 as a TuAA is disclosed in U.S. Pat. No. 5,804,381 entitled ISOLATED NUCLEIC ACID MOLECULE ENCODING AN ESOPHAGEAL CANCER ASSOCIATED ANTIGEN, THE ANTIGEN ITSELF, AND USES THEREOF which is hereby incorporated by reference in its entirety. A paralogous locus encoding antigens with extensive sequence identity, LAGE-1a/s and LAGE-1b/L, has been disclosed in publicly available assemblies of the human genome, and has been concluded to arise through alternate splicing. Additionally, CT-2 (or CTAG-2, Cancer-Testis Antigen-2) appears to be either an allele, a mutant, or a sequencing discrepancy of LAGE-1b/L. Due to the extensive sequence identity, many epitopes from NY-ESO-1 can also induce immunity to tumors expressing these other antigens. See FIG. 1. NY-ESO-1 and LAGE are virtually identical through amino acid 70. From amino acid 71 through 134 the longest run of identity between the two proteins is 6 residues, but potentially cross-reactive sequences are present. From amino acid 135 through 180, NY-ESO and LAGE-1a/s are identical except for a single residue, but LAGE-1b/L is unrelated due to the alternate splice. The CAMEL and LAGE-2 antigens appear to derive from the LAGE-1 mRNA, but from alternate reading frames, thus giving rise to unrelated protein sequences. More recently, GenBank Accession AF277315.5, Homo sapiens chromosome X clone RP5-865E18, RP5-1087L19, complete sequence, reports three independent loci in this region which are labeled as LAGE1 (corresponding to CTAG-2 in the genome assemblies), LAGE2-A and LAGE2-B (both corresponding to CTAG-1 in the genome assemblies).


PRAME, also know as MAPE, DAGE, and OIP4, was originally observed as a melanoma antigen. Subsequently, it has been recognized as a cancer-testis (CT) antigen, but unlike many CT antigens, such as, MAGE, GAGE and BAGE, PRAME is expressed in acute myeloid leukemias. PRAME is a member of the MAPE family, which consists largely of hypothetical proteins with which it shares limited sequence similarity. The usefulness of PRAME as a TuAA is taught in U.S. Pat. No. 5,830,753 entitled “ISOLATED NUCLEIC ACID MOLECULES CODING FOR TUMOR REJECTION ANTIGEN PRECURSOR DAGE AND USES THEREOF,” which is hereby incorporated by reference in its entirety.


PSMA (prostate-specific membranes antigen), a TuAA described in U.S. Pat. No. 5,538,866 entitled “PROSTATE-SPECIFIC MEMBRANES ANTIGEN” which is hereby incorporated by reference in its entirety, is expressed by normal prostate epithelium and, at a higher level, in prostatic cancer. It has also been found in the neovasculature of non-prostatic tumors. PSMA can thus form the basis for vaccines directed to both prostate cancer and to the neovasculature of other tumors. This later concept is more fully described in a provisional U.S. Patent Application No. 60/274,063 entitled “ANTI-NEOVASCULAR VACCINES FOR CANCER,” filed Mar. 7, 2001, and U.S. application Ser. No. 10/094,699 (Pub. No. 20030046714 A1), filed on Mar. 7, 2002, entitled “ANTI-NEOVASCULAR PREPARATIONS FOR CANCER,” each of which is hereby incorporated by reference in its entirety. Briefly, as tumors grow they recruit ingrowth of new blood vessels. This is understood to be necessary to sustain growth as the centers of unvascularized tumors are generally necrotic and angiogenesis inhibitors have been reported to cause tumor regression. Such new blood vessels, or neovasculature, express antigens not found in established vessels, and thus can be specifically targeted. By inducing CTL against neovascular antigens the vessels can be disrupted, interrupting the flow of nutrients to, and removal of wastes from, tumors, leading to regression.


Alternate splicing of the PSMA mRNA leads to a protein with an apparent start at Met58, thereby deleting the putative membrane anchor region of PSMA as described in U.S. Pat. No. 5,935,818 entitled “ISOLATED NUCLEIC ACID MOLECULE ENCODING ALTERNATIVELY SPLICED PROSTATE-SPECIFIC MEMBRANES ANTIGEN AND USES THEREOF,” which is hereby incorporated by reference in its entirety. A protein termed PSMA-like protein, Genbank accession number AF261715, is nearly identical to amino acids 309-750 of PSMA, but has a different expression profile. Thus the most preferred epitopes are those with an N-terminus located from amino acid 58 to 308.


PSA, prostate specific antigen, is a peptidase of the kallikrein family and a differentiation antigen of the prostate. Expression in breast tissue has also been reported. Alternate names include gamma-seminoprotein, kallikrein 3, seminogelase, seminin, and P-30 antigen. PSA has a high degree of sequence identity with the various alternate splicing products prostatic/glandular kallikrein-1 and -2, as well as kallikrein 4, which is also expressed in prostate and breast tissue. Other kallikreins generally share less sequence identity and have different expression profiles. Nonetheless, cross-reactivity that might be provoked by any particular epitope, along with the likelihood that that epitope would be liberated by processing in non-target tissues (most generally by the housekeeping proteasome), should be considered in designing a vaccine.


PSCA, prostate stem cell antigen, and also known as SCAH-2, is a differentiation antigen preferentially expressed in prostate epithelial cells, and overexpresssed in prostate cancers. Lower level expression is seen in some normal tissues including neuroendocrine cells of the digestive tract and collecting ducts of the kidney. PSCA is described in U.S. Pat. No. 5,856,136 entitled “HUMAN STEM CELL ANTIGENS,” which is hereby incorporated by reference in its entirety.


Synaptonemal complex protein 1 (SCP-1), also known as HOM-TES-14, is a meiosis-associated protein and also a cancer-testis antigen (Tureci, O., et al. Proc. Natl. Acad. Sci. USA 95:5211-5216, 1998). As a cancer antigen its expression is not cell-cycle regulated and it is found frequently in gliomas, breast, renal cell, and ovarian carcinomas. It has some similarity to myosins, but with few enough identities that cross-reactive epitopes are not an immediate prospect.


The ED-B domain of fibronectin is also a potential target. Fibronectin is subject to developmentally regulated alternative splicing, with the ED-B domain being encoded by a single exon that is used primarily in oncofetal tissues (Matsuura, H. and S. Hakomori Proc. Natl. Acad. Sci. USA 82:6517-6521, 1985; Carnemolla, B. et al. J. Cell Biol. 108:1139-1148, 1989; Loridon-Rosa, B. et al. Cancer Res. 50:1608-1612, 1990; Nicolo, G. et al. Cell Differ. Dev. 32:401-408, 1990; Borsi, L. et al. Exp. Cell Res. 199:98-105, 1992; Oyama, F. et al. Cancer Res. 53:2005-2011, 1993; Mandel, U. et al. APMIS 102:695-702, 1994; Farnoud, M. R. et al. Int. J. Cancer 61:27-34, 1995; Pujuguet, P. et al. Am. J. Pathol. 148:579-592, 1996; Gabler, U. et al. Heart 75:358-362, 1996; Chevalier, X. Br. J. Rheumatol. 35:407-415, 1996; Midulla, M. Cancer Res. 60:164-169, 2000).


The ED-B domain is also expressed in fibronectin of the neovasculature (Kaczmarek, J. et al. Int. J. Cancer 59:11-16, 1994; Castellani, P. et al. Int. J. Cancer 59:612-618, 1994; Neri, D. et al. Nat. Biotech. 15:1271-1275, 1997; Karelina, T. V. and A. Z. Eisen Cancer Detect. Prev. 22:438-444, 1998; Tarli, L. et al. Blood 94:192-198, 1999; Castellani, P. et al. Acta Neurochir. (Wien) 142:277-282, 2000). As an oncofetal domain, the ED-B domain is commonly found in the fibronectin expressed by neoplastic cells in addition to being expressed by the neovasculature. Thus, CTL-inducing vaccines targeting the ED-B domain can exhibit two mechanisms of action: direct lysis of tumor cells, and disruption of the tumor's blood supply through destruction of the tumor-associated neovasculature. As CTL activity can decay rapidly after withdrawal of vaccine, interference with normal angiogenesis can be minimal. The design and testing of vaccines targeted to neovasculature is described in Provisional U.S. Patent Application No. 60/274,063 entitled “ANTI-NEOVASCULATURE VACCINES FOR CANCER,” filed on Mar. 7, 2001, and in U.S. patent application Ser. No. 10/094,699, (Pub. No. 20030046714 A1), entitled “ANTI-NEOVASCULATURE PREPARATIONS FOR CANCER,” filed on Mar. 7, 2002, each of which is hereby incorporated by reference in its entirety. A tumor cell line is disclosed in Provisional U.S. Application No. 60/363,131, filed on Mar. 7, 2002, entitled “HLA-TRANSGENIC MURINE TUMOR CELL LINE,” which is hereby incorporated by reference in its entirety.


Carcinoembryonic antigen (CEA) is a paradigmatic oncofetal protein first described in 1965 (Gold and Freedman, J. Exp. Med. 121: 439-462, 1965. Fuller references can be found in the Online Medelian Inheritance in Man; record *114890. It has officially been renamed carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5). Its expression is most strongly associated with adenocarcinomas of the epithelial lining of the digestive tract and in fetal colon. CEA is a member of the immunoglobulin supergene family and the defining member of the CEA subfamily.


Survivin, also known as Baculoviral IAP Repeat-Containing Protein 5 (BIRC5), is another protein with an oncofetal pattern of expression. It is a member of the inhibitor of apoptosis protein (IAP) gene family. It is widely over-expressed in cancers (Ambrosini, G. et al., Nat. Med. 3:917-921, 1997; Velculiscu V. E. et al., Nat. Genet. 23:387-388, 1999) and its function as an inhibitor of apoptosis is believed to contribute to the malignant phenotype.


HER2/NEU is an oncogene related to the epidermal growth factor receptor (van de Vijver, et al., New Eng. J. Med. 319:1239-1245, 1988), and apparently identical to the c-ERBB2 oncogene (Di Fiore, et al., Science 237: 178-182, 1987). The over-expression of ERBB2 has been implicated in the neoplastic transformation of prostate cancer. As with HER2, it is amplified and over-expressed in 25-30% of breast cancers among other tumors where expression level is correlated with the aggressiveness of the tumor (Slamon, et al., New Eng. J. Med. 344:783-792, 2001). A more detailed description is available in the Online Medelian Inheritance in Man; record *164870.


Further examples of tumor-associated antigens include MelanA (MART-I), gp100 (Pmel 17), tyrosinase, TRP-1, TRP-2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, p15(58), CEA, RAGE, NY-ESO (LAGE), SCP-1, Hom/Mel-40, PRAME, p53, H-Ras, HER-2/neu, BCR-ABL, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens E6 and E7, TSP-180, MAGE-4, MAGE-5, MAGE-6, p185erbB2, p180erbB-3, c-met, nm-23H1, PSA, TAG-72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, β-Catenin, CDK4, Mum-1, p16, TAGE, PSMA, PSCA, CT7, telomerase, 43-9F, 5T4, 791Tgp72, alpha-fetoprotein, β-HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29\BCAA), CA 195, CA 242, CA-50, CAM43, CD68\KP1, CO-029, FGF-5, G250, Ga733 (EpCAM), HTgp-175, M344, MA-50, MG7-Ag, MOV18, NB/70K, NY—CO-1, RCAS1, SDCCAG16, TA-90 (Mac-2 binding protein\cyclophilin C-associated protein), TAAL6, TAG72, TLP, TPS, and the like.


Additional tumor-associated antigens are described in Chen, YT, “Identification of human tumor antigens by serological expression cloning: an online review on SEREX” Cancer Immun. 2004 [updated 2004 Mar 10; cited 2004 Apr 1] at world wide web cancerimmunotherapy.org/SEREX/; and Renkvist, N. et al., “A listing of tumor antigens recognized by T cells,” Cancer Immunology Immunotherapy, 50:3-15 (2001), each of which is hereby incorporated by reference in its entirety.


Table 2, adapted from Scanlan et al., “The cancer/testis genes: Review, standardization, and commentary,” Cancer Immunity 4:1 (Jan. 23, 2004), which is hereby incorporated by reference in its entirety, provides a listing of CT Antigens. Table 3 provides the frequency of mRNA expression in various tumor types for the CT antigens in Table 2. Scanlan et al., “The cancer/testis genes: Review, standardization, and commentary,” Cancer Immunity 4:1 (Jan. 23, 2004), which is hereby incorporated by reference in its entirety.

TABLE 2Listing of CT genesCTTranscript/TranscriptIdentifierfamilyFamily Members/CT Identifier (Synonyms)CT1MAGEAMAGEA1/CT1.1, MAGEA2/CT1.2,MAGEA3/CT1.3, MAGEA4/CT1.4,MAGEA5/CT1.5, MAGEA6/CT1.6,MAGEA7/CT1.7, MAGEA8/CT1.8,MAGEA9/CT.9, MAGEA10/CT1.10,MAGEA11/CT1.11, MAGEA12/CT1.12CT2BAGEBAGE/CT2.1, BAGE2/CT2.2, BAGE3/CT2.3,BAGE4/CT2.4, BAGE5/CT2.5CT3MAGEBMAGEB1/CT3.1, MAGEB2/CT3.2,MAGEB5/CT3.3, MAGEB6/CT3.4CT4GAGE1GAGE1/CT4.1, GAGE2/CT4.2, GAGE3/CT4.3,GAGE4/CT4.4, GAGE5/CT4.5, GAGE6/CT4.6,GAGE7/CT4.7, GAGE8/CT4.8CT5SSXSSX1/CT5.1, SSX2/CT5.2a, SSX2/CT5.2b,SSX3/CT5.3, SSX4/CT5.4CT6NY-ESO-1NY-ESO-1/CT6.1, LAGE-1a/CT6.2a, LAGE-1b/CT6.2bCT7MAGEC1MAGEC1/CT7.1, MAGEC3/CT7.2CT8SYCP1SYCP1/CT8CT9BRDTBRDT/CT9CT10MAGEE1MAGEE1/CT10CT11CTp11/SPANXSPANXA1/CT11.1, SPANXB1/CT11.2,SPANXC/CT11.3, SPANXD/CT11.4CT12XAGE-1/GAGEDXAGE-1a/CT12.1a, XAGE-1b/CT12.1b, XAGE-1c/CT12.1c, XAGE-1d/CT12.1d, XAGE-2/CT12.2,XAGE-3a/CT12.3a, XAGE-3b/CT12.3b, XAGE-4/CT12.4CT13HAGEHAGE/CT13CT14SAGESAGE/CT14CT15ADAM2ADAM2/CT15CT16PAGE-5PAGE-5/CT16.1, CT16.2CT17LIP1LIP1/CT17CT18NA88NA88/CT12CT19IL13RA1IL13RA1/CT19CT20TSP50TSP50/CT20CT21CTAGE-1CTAGE-1/CT21.1, CTAGE-2/CT21.2CT22SPA17SPA17/CT22CT23OY-TES-1OY-TES-1/CT23CT24CSAGECSAGE/CT24.1, TRAG3/CT24.2CT25MMA1/DSCR8MMA-1a/CT25.1a, MMA-1b/CT25.1bCT26CAGECAGE/CT26CT27BORISBORIS/CT27CT28HOM-TES-85HOM-TES-85/CT28CT29AF15q14/D40D40/CT29CT30E2F-like/HCA661HCA661/CT30CT31PLU-1PLU-1/CT31CT32LDHCLDHC/CT32CT33MORCMORC/CT33CT34SGY-1SGY-1/CT34CT35SPO11SPO11/CT35CT36TPX1TPX-1/CT36CT37NY-SAR-35NY-SAR-35/CT37CT38FTHL17FTHL17/CT38CT39NXF2NXF2/CT39CT40TAF7LTAF7L/CT40CT41TDRD1TDRD1/CT41.1, NY-CO-45/CT41.2CT42TEX15TEX15/CT42CT43FATEFATE/CT43CT44TPTETPTE/CT44PRAME(MAPE, DAGE)











TABLE 3











Frequency (%) of Expression in Tumor Type

























CT Family








Leuk/
Lung









(Member)
Blad
Brn
Brst
Col
Eso
Gas
H/N
Liver
Lymph
(NSCLC)
Mel
Ov
Pancr
Pros
Renal
Sarc
Ref





MAGEA1/CT1.1
22

18
 2
53
29
28
80
 0
49
48
28

15
 0
14
44


BAGE1/CT2.1
15

10
 0


 8

 0
 4
26
15

 0
 0
 6
44


MAGEB1/CT3.1
 0
 0
17
 0

 0
 0

 0
14
22


 0
 0
 9
45


GAGE/CT4.1
12

 9
 0


19
38b
 1
19
28
31

10
 0
25
44


SSX2/CT5.2
44
 6
 7
12


35
 9b
36
16
35


40
 5
50
46


NY-ESO-1/CT6.1
80
 0
30
 0

 0

29
 0
17
34
25
 0
25
 9
 0
 8


MAGEC1/CT7.1
44

30
10


36


33
70




60
20


SYCP1/CT8

47
20
 0

 7

28b
 0
 7
14
 0

 0
 8
 0
 9


BRDT/CT9
 0

 0
 0
 8

 8


25
 0



 0

16


MAGEE1/CT10
44

38
 0


36


24
50




 0
12


SPANXC/CT11.3
 9

25
22
 0




33
70

 0



14


XAGE-1a/CT12.1a










 8




22
47


HAGE/CT13
24
37
 5
31
27


20
 9
32
17


22
 6
20
13


SAGE/CT14
12
 0
 5
 0
20

17

 4
22
 4


 0
 5
 5
13


ADAM2/CT15


 0
 0





 0
 0
 0


12

17


PAGE-5/CT16


 5
11





39
22
 0


44

17


LIP1/CT17


 5
 0





 0
 0
 0


25

17


NA88/CT18










11





48


TSP50/CT20


28













49


CTAGE-1/CT21.1








35







50


SPA17/CT22








26







51


OYTES1/CT23
28

40
15

 0

40

20




 0

52


MMA1a/CT25.1a


 0
 0
 0




40
26

 0


18
15


CAGE/CT26





89



100 






53


HOMTES85/CT28

35
 0
10



19

28
36
32

 0


54


D40/CT29

20

13

 0



41

36
27



55


HCA661/CT30
 0




 0
 0
29


20





56


PLU-1/CT31


86













27


LDHC/CT32


35
15





47
44
42

37
57

18


MORC/CT33


 0
 0





18
18
14

 0
 0

18


SGY-1/CT34


20
 0





12
25
57

12
 0

18


SPO11/CT35


 0
 0





 0
 6
 0

 0
 0

18


TPX1/CT36


15
 0






 6
14

37
14

18


NYSAR35/CT37
42

23
 0
 8




17
 6
 8


 0
 8
57


FTHL17/CT38
22

14
 0
 0

10

 0
25
 0


 0
 0
 0
58


NXF2/CT39
19

 0
11
12

 5

 0
15
55


14
 0
27
58


TAF7L/CT40
10

 0
 0
 0

10

 0
 9
21


 0
 0
12
58


TDRD1/CT41.1
28

37
 0
10

22

 5
 5
 0


38
 0
 0
58


TEX15/CT42
21

 0
 0
20

11

 0
21
27


12
33
28
58


FATE/CT43



21

 7

66

 0






19


TPTE/CT44



 0

 0

39

36






19








aAbbreviations: Blad, bladder; Brn, brain; Brst, breast; Col, colon; Gas, gastric; H/N, head and neck; Leuk, leukemia; Lymph, lymphoma, NSCLC, non-small cell lung carcinoma; Mel, melanoma; Ov, ovarian; Pancr, pancreatic; Pros, prostate; Sarc, sarcoma; Ref, reference.






bReference 59.







Additional antigens associated with tumor neovasculature are VEGFR2 (vascular endothelial growth factor receptor 2) described in U.S. Pat. No. 6,342,221, which is hereby incorporated by reference in its entirety; and Tie-2, an endothelium specific receptor tyrosine kinase which is described in WO9943801, which is hereby incorporated by reference in its entirety.


One of skill in the art will appreciate that any other antigen or protein associated with vascular cells can be a target for the immunogenic compositions, including those that are presently known and those yet to be identified.


Compositions


Immunogenic compositions, including, for example, vaccines, can be prepared using whole antigen or an epitopic peptide. Peptide immunogens can be readily prepared using standard peptide synthesis means known in the art, for example. Immunogens can be prepared commercially by one of numerous companies that do chemical synthesis. An example such a company is American Peptides, Inc., where the distributor is CLINALFA AG (Laufelfingen, Switzerland). The antigens or immunogens can be prepared in accordance with GMP standards and purity can be assessed by analytical HPLC. The product can be characterized by amino-acid analysis and tested for sterility and the absence of pyrogens.


An antigen may be delivered to an animal's system either directly or indirectly. For example, a polypeptide may be delivered directly as the polypeptide, or it may be delivered indirectly, for example, using a DNA construct or vector, or a recombinant virus that codes for the desired antigen. Any vector driving expression in a professional antigen presenting cell can be suitable for this purpose. In indirect delivery, the antigen is expressed in the cell, then presented by the MHC Class I on the surface of the cell to stimulate a CTL response. Expression of a secreted form of the antigen can be useful to induce an antibody response recognizing antigens that are membrane proteins.


In a preferred embodiment, an encoded antigen can be delivered in the form of a naked plasmid expression vector. Particularly useful constructs are disclosed in U.S. patent application Ser. No. 09/561,572, entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS;” U.S. Patent Application No. 10/292,413 (Pub. No. 20030228634 A1), entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS AND METHODS FOR THEIR DESIGN;” U.S. patent application Ser. No. 10/225,568 (Pub No. 2003-0138808); PCT Application No. PCT/U.S. 2003/026231 (Pub. No. WO 2004/018666); U.S. Pat. No. 6,709,844, entitled AVOIDANCE OF UNDESIRABLE REPLICATION INTERMEDIATES IN PLASMIND PROPAGATION, and in U.S. patent application Ser. No. 10/026,066 (Pub. No. 20030215425 A1), entitled EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS, each of which is hereby incorporated by reference in its entirety. Additional methodology, compositions, peptides, and peptide analogues are disclosed in U.S. Provisional Application No. 60/______ (Attorney Docket MANNK.038PR), filed on the same date as the instant application, entitled “SSX-2 PEPTIDE ANALOGS;” which is incorporated herein by reference in its entirety. Further methodology, compositions, peptides, and peptide analogues are disclosed in U.S. Provisional Application No. 60/______ (Attorney Docket MANNK.039PR), filed on the same date as the instant application, entitled “NY-ESO PEPTIDE ANALOGS;” which is incorporated herein by reference in its entirety. The feasibility of and general procedures related to the use of naked DNA for immunization are described in U.S. Pat. No. 5,589,466, entitled “INDUCTION OF A PROTECTIVE IMMUNE RESPONSE IN A MAMMAL BY INJECTING A DNA SEQUENCE” and in U.S. Pat. No. 5,679,647, entitled “METHODS AND DEVICES FOR IMMUNIZING A HOST AGAINST TUMOR-ASSOCIATED ANTIGENS THROUGH ADMINISTRATIONS OF NAKED POLYNUCLEOTIDES WHICH ENCODE TUMOR-ASSOCIATED ANTIGENIC PEPTIDES,” each of which is hereby incorporated by reference in its entirety. The former teaches only intramuscular or intradermal injection while the latter teaches only administration to skin or mucosa.


In a preferred embodiment, the antigen can be administered directly to the lymphatic system. Intranodal administration for the generation of CTL is taught in U.S. patent application Ser. Nos. 09/380,534 and 09/776,232 (Pub. No. 20020007173 A1), and in PCT Application No. PCTUS98/14289 (Pub. No. WO9902183A2) each entitled “A METHOD OF INDUCING A CTL RESPONSE,” each of which is hereby incorporated by reference in its entirety. Single bolus injection intra lymph node (i.ln.) required only 0.1% of the dose required in order to obtain a similar level of CTL response by intramuscular (i.m.) injection. Therefore a protective response can be established against systemic viral infection with a single bolus delivered i.ln., but not with a dose nearing the practical limit delivered i.m. Repeated bolus injections i.m. failed to establish a protective response against a peripheral virus infection or transplanted tumor, whereas lower doses administered i.ln. were completely effective. Particularly useful intranodal immunization protocols are taught in Provisional U.S. Patent Application No. 60/479,393, entitled “METHODS TO CONTROL MAGNITUDE AND QUALITY THE MHC CLASS I-RESTRICTED IMMUNE RESPONSE,” and in U.S. Patent Application No. 60/______ entitled “METHODS TO ELICIT, ENHANCE AND SUSTAIN IMMUNE RESPONSES AGAINST MHC CLASS I-RESTRICTED EPITOPES, FOR PROPHYLACTIC OR THERAPEUTIC PURPOSE” (Attorney Docket No. MANNK.034A) (Pub. No. ______), filed on date even with the instant Application, each of which is hereby incorporated by reference in its entirety.


A class of epitopes that can be advantageous in anti-cancer immunogenic compositions are housekeeping epitopes. These are produced through the action of the housekeeping (or standard) proteasome. Housekeeping epitopes can be liberated from the translation product of expression vectors through proteolytic processing by the immunoproteasome of professional antigen presenting cells (pAPC). In one embodiment of the invention, sequences flanking the housekeeping epitope(s) can be altered to promote cleavage by the immunoproteasome at the desired location(s). Housekeeping epitopes, their uses, and identification are described in U.S. patent application Ser. No. 09/560,465 filed on Apr. 28, 2000, and U.S. patent application Ser. No. 10/026,066 (Pub. No. 20030215425 A1), filed on Dec. 7, 2001, entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” and U.S. patent application Ser. No. 09/561,074, filed on Apr. 28, 2000, entitled “METHOD OF EPITOPE DISCOVERY,” each of which is hereby incorporated by reference in its entirety.


Examples of housekeeping epitopes are disclosed in Provisional U.S. patent Applications No. 60/282,211, filed on Apr. 6, 2001; 60/337,017, filed on Nov. 7, 2001; 60/363,210 filed Mar. 7, 2002; and 60/409,123, filed on Sep. 5, 2002; U.S. patent application Ser. No. 10/117,937 (Publication No. 20030220239A1), filed on Apr. 4, 2002; and U.S. patent application Ser. No. 10/657,022, and PCT Application No. PCT/U.S. 2003/027706 (Pub. No. WO04022709A2) both entitled EPITOPE SEQUENCES; each of which is hereby incorporated by reference in its entirety.


In other embodiments of the invention, the housekeeping epitope(s) can be flanked by arbitrary sequences or by sequences incorporating residues known to be favored in immunoproteasome cleavage sites. As used herein the term “arbitrary sequences” refers to sequences chosen without reference to the native sequence context of the epitope, their ability to promote processing, or immunological function. In further embodiments of the invention multiple epitopes can be arrayed head-to-tail. These arrays can be made up entirely of housekeeping epitopes. Likewise, the arrays can include alternating housekeeping and immune epitopes. Alternatively, the arrays can include housekeeping epitopes flanked by immune epitopes, whether complete or distally truncated. Further, the arrays can be of any other similar arrangement. There is no restriction on placing a housekeeping epitope at the terminal positions of the array. The vectors can additionally contain authentic protein coding sequences or segments thereof containing epitope clusters as a source of immune epitopes. The term “authentic” refers to natural protein sequences.


Epitope clusters and their uses are described in U.S. patent application Ser. Nos. 09/561,571, entitled “EPITOPE CLUSTERS,” filed on Apr. 28, 2000; Ser. No. 10/005,905, entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” filed on Nov. 7, 2001; and Ser. No. 10/026,066, filed on Dec. 7, 2001, also entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” each of which is hereby incorporated by reference in its entirety.


In another embodiment of the invention an encoded antigen can be delivered in the form of a viral vector. A wide array of viruses with modified genomes adapted to express interposed reading frames but often no, or at least a reduced number of, viral proteins are known in the art, including without limitation, retroviruses including lentiviruses, adenoviruses, parvoviruses including adeno-associated virus, herpesviruses, and poxviruses including vaccinia virus. Such viral vectors facilitate delivery of the nucleic acid component into the cell allowing for expression. A subset of these vectors, such as retroviruses and parvoviruses, promote integration of their nucleic acid component into the host genome, whereas others do not.


Bacteria can also serve as vectors, that is they can be used to deliver a nucleic acid molecule capable of causing expression of an antigen. For example, a strain of Listeria monocytogenes has been devised that effects its own lysis upon entering the cytosol of macrophages (its normal target), thereby releasing plasmid from which antigen is subsequently expressed (Dietrich, G. et al., Biotechnology 16:181-185, 1998 which is hereby incorporated by reference in its entirety). Shigela flexneri and Escherichia coli have been similarly used (Sizemore, D. R. et al., Science 270:299-302, 1995, and Courvalin, P. et al., Life Sci. 318:1207-1212, 1995, respectively, each of which is hereby incorporated by reference in its entirety).


The use of microbial vectors for nucleic acid delivery can be complicated by the immune reactions the vectors themselves provoke. When prolonged or repeated administration is required, antibody elicited by the earlier treatment can prevent useful quantities of the vector from ever reaching its intended host. However, by direct administration into a lymph node, for example, the combination of proximity to host cells and the much reduced effective dose makes it possible to administer a dose capable of evading or overwhelming an existing antibody titer.


The word vector has been used, here and elsewhere, in reference to several modalities and variously modified (e.g., expression vector, viral vector, delivery vector, etc.). The underlying principle is that a nucleic acid capable of causing expression of an antigen, rather than the antigen itself, ultimately arrives in an APC. Unless modified, explicitly or by local context, the term vector as used herein is intended to encompass all such possibilities.


The techniques discussed above are distinct from the approach of modifying the microbial genome, including extra-chromosomal DNA, such that the antigen is produced as a component of the microbe, which is then itself administered as the immunogen. Examples of microbes used in the genomic modification approach include viruses, bacteria, fungi, and protazoa. In embodiments of the invention described herein, the compositions, including the vaccines, can include the already synthesized antigen or a nucleic acid capable of causing an APC to express the antigen in vivo. In alternative embodiments, combinations of these two techniques are used. For example, one embodiment contemplates the use of a virus vector as discussed above that also incorporates a target epitope into a capsid or envelope protein.


Antigens may be used alone or may be delivered in combination with other antigens or with other compounds such as cytokines. Cytokines that are known to enhance immune stimulation of CTL responses, include, for example, GM-CSF, IL-12, IL-2, TNF, IFN, IL-18, IL-3, IL-4, IL-8, IL-9, IL-13, IL-10, IL-14, IL-15, G-SCF, IFN alpha, IFN beta, IFN gamma, TGF alpha, TGF beta, and the like. Cytokines are known in the art and are readily available in the literature or commercially. Many animal and human tumors have been shown to produce cytokines, such as IL-4, IL-10, TGF-B, that are potent modulators of the immune response and that protect tumors from immune-mediated destruction. The production of IL-4, IL-10 or TGF-B by tumors may achieve this protective effect by suppressing the induction of cellular immunity, including the elaboration of CTL responses. Alternatively, cytokines that support CTL responses can be exogenously added to help in the balance between induction of anti-tumor cell mediated and non-tumor-destructive humoral responses. Several such exogenous cytokines show utility in experimental mouse vaccination models which are known to enhance CTL responses, including GM-CSF, IFN and IL-2. An example of an effective exogenous cytokine that may be used is GM-CSF. GM-CSF is reported to enhance the expression of the so called “co-stimulatory” molecules, such as B7-1 or B7-2 on antigen presenting cells (APC). These co-stimulatory molecules are important players in the variety of interactions that occur during stimulation of CTL by APC. Moreover, GM-CSF is known to induce activation of APCs and to facilitate growth and differentiation of APCs, thereby making these APCs important CTL stimulating cells available both in greater numbers and potency.


Delivery of the Antigen


While not wanting to be bound by any particular theory, it is thought that T cells do not have a functional memory that is long-lived. Antibody-mediated B-cell memory, on the other hand, appears to have a long-lived effector memory. Thus, delivering an antigen that induces a CTL response is most preferably done over time to keep the patient's immune system appropriately stimulated to attack the target cells. In one approach the presence of antigen is maintained virtually continuously within the lymphatic system to maintain effector CTL function as disclosed in U.S. patent application Ser. No. 09/776,232 (Pub. No. 20020007173 A1), entitled “A METHOD OF INDUCING A CTL RESPONSE,” which is hereby expressly incorporated by reference. In another approach T cell memory is repeatedly induced, and re-amplified and reactivated as described in Provisional U.S. patent Application No. 60/479,393, entitled “METHODS TO CONTROL MAGNITUDE AND QUALITY THE MHC CLASS I-RESTRICTED IMMUNE RESPONSE,” and in U.S. patent application No. ______ entitled “METHODS TO ELICIT, ENHANCE AND SUSTAIN IMMUNE RESPONSES AGAINST MHC CLASS I-RESTRICTED EPITOPES, FOR PROPHYLACTIC OR THERAPEUTIC PURPOSE” (Attorney Docket No. MANNK.034A) (Pub. No. ______), filed on date even with the instant Application, each of which is hereby incorporated by reference in its entirety. While it has been suggested that antigens and adjuvants can be prepared as biodegradable microspheres or liposomes, none of these preparations have thus far provided a CTL response that is useful for attacking cancer cells or pathogens on a long term basis. Preferably, delivery of the antigen is sustained over the desired period of time at a level sufficient to maintain the antigen level to obtain the desired response. In one embodiment, a reservoir having fluid antigen composition can be used to deliver the antigen such that it reaches the animal's lymphatic system. While much of the following discussion focuses on the use of infusion to deliver the antigen it is also possible to use bolus injections directly into the lymphatic system, the number and frequency of which will depend on the persistence of antigen conferred by the particular form and formulation of antigen used.


Ultimately antigen finds its way into the lymphatic system in order to most efficiently stimulate CTL. Delivery of antigen can involve infusion into various compartments of the body, including but not limited to subcutaneous, intravenous, intraperitoneal and intralymphatic, the latter being preferred. While each of these points of infusion results in antigen uptake into the lymphatic system, the relative amounts of antigen needed to induce a beneficial CTL response varies according to the site of infusion. In general, direct infusion of antigen into the lymph system is deemed to be the most efficient means of inducing a CTL response, however, any delivery route may be used. Pump systems are capable of delivering material quantities of antigen in a range that is suitable for inducing a CTL response through delivery to all compartments of the body. CTL stimulation following delivery of antigen via the various routes will vary depending on the properties of different antigens, including factors that influence antigen behavior in the body and its rate of equilibration to (or longevity in) the lymph, such as antigen stability in the body fluid, solubility of antigen in body fluid, binding affinity for HLA and potency as a stimulator of CTL.


In a preferred embodiment, introduction of the antigen is done as directly as possible to the lymphatic system to avoid the destruction of the antigen by metabolism in the body. When introduction of a fluid antigen composition occurs subcutaneously, larger quantities of antigen are needed to assure enough antigen reaches the lymphatic system. Such subcutaneous injection is contemplated by the invention disclosed herein, depending on factors such as cost, stability of the antigen, how quickly the antigen gets to the lymph system, how well it equilibrates with the lymph, and other factors that the attending doctor or specialist will recognize. Subcutaneous delivery generally can require 100 to 1000 times more antigen than direct delivery to the lymph system. It is preferable, therefore, that the antigen composition is introduced through a device for local administration to the lymphatic system, e.g., the spleen, a lymph node, or a lymph vessel. The device for local administration can be positioned outside the patient or implanted into the patient. In either case, the device can have a reservoir to hold the fluid antigen-containing composition, a pump to transfer the composition, and a transmission channel leading from the reservoir to be directed to the preferred region of administration in the patient's body. In either case it is preferably portable.


For the device positioned outside the patient's body (the external device), there are numerous devices used for delivering insulin to diabetic patients that are useful in delivering antigen according to the embodiments described herein. Generally these devices can be comprised of a reservoir for holding the antigen composition (instead of insulin), a programmable pump to pump the composition out of the reservoir, a transmission channel or line for transmitting the composition, and a means to introduce the composition into the animal's body to ultimately reach the lymphatic system.


Preferably, the reservoir for the antigen composition should be large enough for delivery of the desired amount of antigen over time and easily refillable or replaceable without requiring the user to reinsert the means for introducing the antigen composition to the lymph system.


In preparing the antigen compositions of embodiments of the invention disclosed herein, a composition (preferably aqueous) can be prepared to be compatible with the lymph system and physiologically acceptable to the animal being treated. Relevant considerations include, for example, the physicochemical properties of the antigen, such as the isoelectric point, molecular weight, glycosylation or other post-translational modification, and overall amino acid composition. These properties along with any known behavior of the drug in different solutions (e.g., different buffers, cofactors, etc.) as well as its in vivo behavior can help guide the choice of formulation components. One parameter that impacts all the major degradation pathways is the solution pH. Thus, the initial formulations also assess the pH dependence of the degradation reactions and the mechanism for degradation, which can often be determined from the pH dependence to determine the stability of the protein in each solution. Rapid screening methods usually involve the use of accelerated stability at elevated temperatures (e.g., 40° C.) using techniques known in the art.


In general the antigen compositions useful in embodiments described herein can be suitable for parenteral injection, in very small quantities. As such a composition should be free of contamination and have a pH compatible with the lymphatic system. However, because very small quantities of the antigenic composition will be delivered it need not be the same pH as blood or lymph, and it need not be aqueous-based. The preferable pH range that is compatible is from about 6.7-7.3 and can be prepared using water for injection to meet USP specifications (see Remington: The Science and Practice of Pharmacy, Nineteenth Edition; Chapters 86-88). For antigens that are less soluble, a suitable cosolvent or surfactant may be used, such as dimethyl sulfoxide (DMSO) or PLURONIC brand surfactants. Generally, a standard saline solution that is buffered with a physiologically acceptable weak acid and its base conjugate, e.g., a phosphate or citrate buffering system, will be the basis of the antigen composition. In some cases, a small amount of an antioxidant may be useful to stabilize the composition and prevent oxidation. Factors to consider in preparing the antigen compositions may be found in the 1994 American Chemical Society book entitled “Formulation and Delivery of Proteins and Peptides” (Acs Symposium Series, No. 567) by Jeffery L. Cleland and Robert Langer (Editor)).


For nucleic acid encoded antigens similar considerations can apply, although the variety of physico-chemical properties encountered with polypeptides is absent, so that acceptable formulations will have nearly universal applicability. As seen in Examples 6-10, plasmid DNA in standard phosphate buffered saline (PBS) is an acceptable and effective formulation. In some embodiments of the invention, DNA is administered continuously or intermittently at short intervals, from a reservoir worn on, or implanted in, the patient's body. It is preferable that the DNA be maintained in a soluble, stable form at or near body temperature over a period of time measured minimally in days. In such applications where the formulated nucleic acid will be delivered from a reservoir over a period of several days or longer, the stability of the nucleic acid at room or body temperature for that period of time, as well as its continued sterility, take on increased importance. The addition of bacteriostatic agents (e.g., benzyl or ethyl alcohol) and chelating agents (e.g. EDTA) is useful toward these ends. Formulations containing about 0.5-2% ethyl alcohol, 0.25-0.5 mM EDTA generally perform well. Such formulations are also appropriate for bolus injections.


Generally the amount of the antigen in the antigen composition will vary from patient to patient and from antigen to antigen, depending on such factors as the activity of the antigen in inducing a response and the flow rate of the lymph through the patient's system. In general the antigen composition may be delivered at a rate of from about 1 to about 500 microliters/hour or about 24 to about 12000 microliters/day. The concentration of the antigen is such that about 0.1 micrograms to about 10,000 micrograms of the antigen will be delivered during 24 hours. The flow rate is based on the knowledge that each minute approximately about 100 to about 1000 microliters of lymph fluid flows through an adult inguinal lymph node. The objective is to maximize local concentration of vaccine formulation in the lymph system. A certain amount of empirical investigation on patients will be necessary to determine the most efficacious level of infusion for a given vaccine preparation in humans.


To introduce the antigen composition into the lymphatic system of the patient the composition is preferably directed to a lymph vessel, lymph node, the spleen, or other appropriate portion of the lymph system. Preferably, the composition is directed to a lymph node such as an inguinal or axillary node by inserting a catheter or needle to the node and maintaining the catheter or needle throughout the delivery. Suitable needles or catheters are available made of metal or plastic (e.g., polyurethane, polyvinyl chloride [PVC], TEFLON, polyethylene, and the like). In inserting the catheter or needle into the inguinal node for example, the inguinal node is punctured under ultrasonographic control using a Vialon™ Insyte-™ cannula and catheter of 24G3/4 (Becton Dickinson, USA) which is fixed using Tegaderm™ transparent dressing (Tegaderm™ 1624, 3M, St. Paul, Minn. 55144, USA). This procedure is generally done by an experienced radiologist. The location of the catheter tip inside the inguinal lymph node is confirmed by injection of a minimal volume of saline, which immediately and visibly increases the size of the lymph node. The latter procedure allows confirmation that the tip is inside the node. This procedure can be performed to ensure that the tip does not slip out of the lymph node and can be repeated on various days after implantation of the catheter. In the event that the tip does slip out of location inside the lymph node, a new catheter can be implanted.


Formulation and Treatment protocol


There are several approaches to utilizing the combination of TuAAs with DNA vaccines. A first approach is to include all the antigens or epitopes from all the antigens in a given combination into a single DNA expression vector. This approach has the advantages of simplicity for manufacturing and administration to patients. However, in some instances, epitope competition can limit the usefulness of this approach. That is, it is possible that only the most immunogenic epitope will elicit an immune response when a vaccine with several epitopes representing all TuAAs in the combination is given to patients. It is also more difficult to design and construct a DNA vaccine in which all epitopes are expressed at high efficiencies. Nevertheless, because the procedure for treating patients is simple and uniform within each type of cancer, the cost is likely to be lower than for the other approaches described below.


An alternate approach is to include only one antigen or epitopes of one antigen in a DNA expression vector. This approach has the advantages of simplicity in designing and constructing the DNA vector, flexibility, and customized administration to patients. If a large number of individual TuAA vaccines are available, then one can customize treatment for each individual patient based on the TuAA expression profile of his or her tumor. For example, if the standard combination for treating a given type of cancer is TuAA A, B, and C (where A, B, and C designate different tumor associated antigens), but a patient's tumor expresses TuAA A, C, and Z (but not B), then the patient can be treated with separate vaccines for each of A, C, and Z. It is expected that this flexibility and customizability will improve the success rate of immunotherapy because antigen redundancy can be achieved for each patient. However, the procedure of treating the patient may be more complex. For example, delivery using this approach may include a sequential administration scheme (one antigen at a time), or injection into multiple, anatomically separate sites of the patient at about the same time.


Still another approach is to combine epitopes from multiple TuAAs that have similar immunogenicity into a DNA expression vector (more than one vector may be used for some combinations). This approach can have some of the advantages of the above two approaches but also can suffer from the disadvantages of the previous two.


A profile of the antigen expression of a particular tumor can be used to determine which antigen or combination of antigens to use. Exemplary methodology is found in U.S. Provisional Application No. 60/______ (Attorney Docket: MANNK.035PR2), filed on even date herewith, entitled “COMBINATIONS OF TUMOR-ASSOCIATED ANTIGENS IN DIAGNOTISTICS FOR VARIOUS TYPES OF CANCERS;” and which is hereby incorporated by reference in its entirety.


Patients that can benefit from such methods of immunization can be recruited using methods to define their MHC protein expression profile and general level of immune responsiveness. In addition, their level of immunity can be monitored using standard techniques in conjunction with access to peripheral blood. Finally, treatment protocols can be adjusted based on the responsiveness to induction or amplification phases and variation in antigen expression. For example, repeated entrainment doses preferably can be administered until a detectable response is obtained, and then administering the amplifying peptide dose(s), rather than amplifying after some set number of entrainment doses. Similarly, scheduled amplifying or maintenance doses of peptide can be discontinued if their effectiveness wanes, antigen-specific regulatory T cell numbers rise, or some other evidence of tolerization is observed, and further entrainment can be administered before resuming amplification with the peptide. The integration of diagnostic techniques to assess and monitor immune responsiveness with methods of immunization is discussed more fully in Provisional U.S. Patent Application No. 60/______ (Atty. Docket No. MANNK.040PR), entitled IMPROVED EFFICACY OF ACTIVE IMMUNOTHERAPY BY INTEGRATING DIAGNOSTIC WITH THERAPEUTIC METHODS, which was filed on date even with the present application and is hereby incorporated by reference in its entirety.


Many variations and alternative elements of the invention have been disclosed. Still further variations and alternate elements will be apparent to one of skill in the art. Various embodiments of the invention can specifically include or exclude any of these variation or elements.


Each of the references cited herein is hereby incorporated herein by reference in its entirety.


The following examples are for illustrative purposes only and are not intended to limit the scope of the embodiments in any way.


EXAMPLES
TuAA Analysis and Selection of Combinations

The presence of TuAAs was measured by Real-Time PCR (RT-PCR). Briefly, total RNA was isolated from tumor specimens by standard methods and cDNA was made with standard reverse transcription procedures. Complementary DNA (cDNA) was amplified with specially designed, gene specific, primers that anneal only to cDNA but not genomic DNA. TuAA expression patterns of 12 ovarian and 7 colorectal tumor specimens were analyzed by RT-PCR. The results are summarized in the Table 4 below.

TABLE 4Total #PRAMENY-ESO-1SSX-2PSMAMAGE1MAGE3Ovarian121256643Colorectal 7 512501


Example 1
Ovarian Cancer

In the case of ovarian cancer, all samples analyzed were positive for PRAME. Thus the inclusion of PRAME in the combination improves coverage of the cases with ovarian cancer.


In order to achieve antigen redundancy and improve coverage in a large population, combinations of other antigens were considered in addition to PRAME. SSX-2 as well as PSMA were present in 6 of the 12 cases individually, but the combination of SSX-2 and PSMA provided coverage in 9 of 12 cases. Although NY-ESO-1 and SSX-2 were only present in 5 and 6 of the 12 cases, respectively, either NYESO-1 or SSX-2 was detected in 7 of the 12 cases.


Thus, the combination of PRAME, SSX-2, and PSMA or PRAME, NY-ESO-1, and SSX-2 provided preferable coverage and redundancy compared to the combination of PRAME and PSMA or the combination of PRAME and SSX-2. The combination of PRAME, SSX-2, and PSMA provided excellent coverage of cases and good antigen redundancy because the majority of ovarian tumor samples analyzed had at least two of the four TuAA in the combination present. The combination of PRAME, SSX-2, PSMA, and NY-ESO-1 provided more preferred antigen redundancy, and thus, lower possibility of tumor escape.


Example 2
Colorectal Cancer

In the case of colorectal cancer, PRAME and PSMA were each detected in 5 of the 7 samples analyzed. In 6 of the 7 cases, either PRAME or PSMA was detected. Although SSX-2 was only detected in 2 of 7 cases, both SSX-2-PRAME and SSX2-PSMA combinations increased coverage to 6 of 7. Similarly, although NYESO-1 was detected in only 1 of 7 cases, the combination of NY-ESO-1-PRAME as well as the NYESO-1-PSMA combination increased coverage to 6 of 7. The addition of SSX-2 or NYESO-1 to the PRAME and PSMA combination improved coverage to 7 of 7. Thus, the combination of PRAME, PSMA, and NYESO-1, or the combination of PRAME, PSMA, and SSX-2 provided good coverage of cases and redundancy of antigens for a majority of patients. The combination of PRAME, PSMA, NY-ESO-1, and SSX-2 provided further redundancy.


Example 3
Pancreatic Cancer

In pancreatic cancer specimens, the presence of NYESO-1 and SSX-2 was detected in 40% and 20% of the specimens, respectively. PSMA and over-expression of HER2-/neu were reported to be present in 100% and 21% of pancreatic tumors, respectively (Chang S S et al, Cancer Res 1999, 59:3192; Safran H et al, Am J Clin Oncol. 2001, 24:496). Although over-expression of HER2/neu may render the cancer tissue a preferred target, thus providing some specificity for immunotherapy, low level expression of HER2/neu in normal tissues remains a concern. Thus, the combination of NYESO-1, SSX-2, and PSMA provides excellent coverage and some redundancy for treating pancreatic cancer.


Example 4
Non-Small Cell Lung Cancer

For non-small cell lung cancer, the reported presence of NYESO-1, SSX-2, MAGE-3, BAGE, over-expression of Her2/neu, and PSMA was 21, 15, 60, 6, 16, and 100%, respectively (Scanlan M J et al, Cancer lett 2000, 150:155; Chang S S et al, Cancer Res 1999, 59:3192; Selvaggi G et al, Cancer 2002, 94:2669, which are hereby incorporated by reference in their entireties). Thus, the combination of NYESO-1, SSX-2, MAGE-3, and PSMA provides coverage and antigen redundancy for the immunotherapy of non-small cell lung cancer.


Example 5
Renal Cell Carcinoma

For renal cell carcinoma, SSX-2, PSMA and PRAME were detected with frequencies of 5, 100 and 40%, respectively (Sahin, U et al, Clin Cancer Res. 2000, 6:3916; Chang S S et al, Urology 2001, 57:801; Neumann E et al, Cancer Res. 1998, 58:4090, which are hereby incorporated by reference in their entireties). Thus, the combination of PSMA and PRAME provides excellent coverage and redundancy for renal cell carcinoma. Adding SSX-2 to the combination of PSMA and PRAME improves redundancy.


Example 6
Melanoma

For melanoma, Melan A, Tyrosinase, NYESO-1, and SSX-2 were reported to be present in 92, 92, 41, and 35% of tumor specimens, respectively (Fetsch P A, et al, Cancer 1999, 87:37; Fetsch P A, et al, Cancer 2000, 90:252; Schultz-Thater E et al, Br J Cancer 2000, 83:204; Sahin, U et al, Clin Cancer Res. 2000, 6:3916). Therefore, the combination of Melan A, Tyrosinase, NYESO-1, and SSX-2 provides excellent coverage and antigen redundancy for the immunotherapy of melanoma. Significant redundancy is achieved using tyrosinase and melan-A together, or by combining NY-ESO-1 and SSX-2 with either of tyrosinase or melan-A.


Example 7
Schedule of Immunization with Plasmids Expressing Epitopes from Two Antigens

Two groups of HHD mice (n=4) were immunized via intra lymph node injection with either pSEM expressing Melan-A26-35A27L (ELA) and pCBP expressing SSX-241-49 as a mixture; or with pSEM in the left inguinal lymph node and pCBP in the right inguinal lymph node, twice, at day 0 and 4 as shown in FIG. 1. The amount of the plasmid was 25 μg/plasmid/dose. Two weeks later, the animals were sacrificed, and cytotoxicity was measured against T2 cells pulsed or not with peptide.


Example 8
Co-Administration of Different Vectors Carrying Distinct Antigens

The animals immunized as described in Example 7, were sacrificed and splenocytes from each group pooled and stimulated with the two peptides (ELA or SSX-241-49) in parallel. The cytotoxicity was measured by incubation with Cr51-tagged, peptide loaded T2 target cells. Data in FIG. 2 show mean of specific cytotoxicity (n=4/group) against various target cells.


The results show that use of plasmid mixture interferes with the response elicited by pCBP plasmid; however, segregating the two plasmids relative to site of administration rescues the activity of pCBP. Thus, the co-administration of different vectors carrying distinct antigens can result in establishment of a hierarchy with regard to immunogenicity. Vector segregation can rescue the immunogenicity of the less dominant component, resulting in a multivalent response.


Example 9
Rescue of Multivalent Response by Addition of Peptide Boost Steps

Four groups of HHD mice (n=6) were immunized via intra lymph node injection with either pSEM and pCBP as a mixture; or with pSEM in the left inguinal lymph node and pCBP in the right inguinal lymph node, twice, at day 0 and 4 as shown in FIG. 3. As a control, mice were immunized with either pSEM or pCBP plasmid. The amount of the plasmid was 25 μg/plasmid/dose. Two weeks later, the animals were boosted with melan A and/or SSX-2 peptides, mirroring the plasmid immunization dose and combination. Two weeks later, the animals were challenged with splenocytes stained with CFSE and loaded or not with Melan A or SSX-2 peptide, for evaluation of in vivo cytotoxicity.


Example 10
Peptide Amplification Rescues the Immunogenicity of the Less Dominant Epitope

Mice were immunized as described in Example 9 and challenged with HHD littermate splenocytes coated with ELA or SSX-2 peptide, employing a triple peak CFSE in vivo cytotoxicity assay that allows the assessment of the specific lysis of two antigen targets simultaneously. Equal numbers of control-CFSElo, SSX-241-49-CFSEmed, and ELA-CFSEhi cells were intravenously infused into immunized mice and 18 hours later the mice were sacrificed and target cell elimination was measured in the spleen (FIG. 4) by CFSE fluorescence using a flow cytometry. FIG. 4 shows the percent specific lysis of the SSX2 and Melan-A antigen targets from individual mice, as well as the mean and SEM for each group.


The results show that immunizing the animals with a mixture of the two vaccines comprising plasmids followed by peptides generated immunity to both antigens and resulted in the highest immune response, representing an average SSX-2 percent specific lysis in the spleen of 30+/−11, and an average Melan-A percent specific lysis of 97+/−1.


Example 11
Clinical Practice for Entrain-and-Amplify Immunization

The data in FIGS. 2 and 4 suggest two scenarios for achieving a strong multivalent response in the clinic, shown in FIG. 5. In the first scenario (A), use of peptides for boosting restores multivalent immune responses even if plasmids and peptides are used as mixtures. In the second scenario (B), segregation of plasmid and peptide components respectively, allows induction of multivalent immune responses.


Example 12
Antigen Sequences

Below are sequences of the antigens listed in Table 1.

SEQ. ID NO. 1SIZE: 529 aaDEFINITION Tyrosinase precursorACCESSION NO.: P14679mllavlycll   wsfqtsaghf   pracvssknl   mekeccppws   gdrspcgqlsgrgscqnill   snaplgpqfp   ftgvddresw   psvfynrtcq   csgnfmgfncgnckfgfwgp   ncterrllvr   rnifdlsape   kdkffayltl   akhtissdyvipigtygqmk   ngstpmfndi   niydlfvwmh   yyvsmdallg   gseiwrdidfaheapaflpw    hrlfllrweq    eiqkltgden   ftipywdwrd   aekcdictdeymggqhptnp   nllspasffs   swqivcsrle   eynshqslcn   gtpegplrrnpgnhdksrtp   rlpssadvef   clsltqyesg   smdkaanfsf   rntlegfaspltgiadasqs   smhnalhiym   ngtmsqvqgs   andpifllhh    afvdsifeqwlrrhrplqev   ypeanapigh   nresymvpfi    plyrngdffi    sskdlgydysylqdsdpdsf   qdyiksyleq   asriwswllg   aamvgavlta   llaglvsllcrhkrkqlpee kqpllmeked yhslyqshlSEQ. ID NO. 2SIZE: 118 aaDEFINITION Melanoma antigen recognized by T-cells 1 (MART-1)(Melan-A protein)ACCESSION NO.: Q16655mpredahfiy    gypkkghghs   yttaeeaagi   giltvilgvl   lligcwycrrrngyralmdk   slhvgtqcal   trrcpqegfd   hrdskvslqe   kncepvvpnappayeklsae qspppyspSEQ. ID NO. 3SIZE: 223 aaDEFINITION: synovial sarcoma, X breakpoint 2 isoform a;sarcoma, synovial, X-chromosome-related 2; SSX2 protein [Homosapiens].ACCESSION NO.: NP_003138mngddafarr   ptvgaqipek   iqkafddiak   yfskeewekm   kasekifyvymkrkyeamtk   lgfkatlppf   mcnkraedfq   gndldndpnr   gnqverpqmtfgrlqgispk   impkkpaeeg   ndseevpeas   gpqndgkelc   ppgkpttsekihersgnrea   qekeerrgta   hrwssqnthn   igrfslstsm   gavhgtpktithnrdpkggn mpgptdcvre nswSEQ. ID NO. 4SIZE: 750 aaDEFINITION folate hydrolase (prostate-specific membraneantigen) [Homo sapiens].ACCESSION NO.: NP_004467mwnllhetds   avatarrprw   lcagalvlag   gffllgflfg     wfikssneatnitpkhnmka   fldelkaeni    kkflynftqi    phlagteqnf   qlakqiqsqwkefgldsvel   ahydvllsyp   nkthpnyisi   inedgneifn   tslfeppppgyenvsdivpp   fsafspqgmp   egdlvyvnya   rtedffkler   dmkincsgkiviarygkvfr   gnkvknaqla   gakgvilysd   padyfapgvk   sypdgwnlpgggvqrgniln   lngagdpltp   gypaneyayr   rgiaeavglp   sipvhpigyydaqkllekmg   gsappdsswr   gslkvpynvg   pgftgnfstq   kvkmhihstnevtriynvig   tlrgavepdr   yvilgghrds   wvfggidpqs   gaavvheivrsfgtlkkegw   rprrtilfas   wdaeefgllg   stewaeensr   llqergvayinadssiegny   tlrvdctplm   yslvhnltke   lkspdegfeg   kslyeswtkkspspefsgmp   risklgsgnd   fevffqrlgi   asgrarytkn   wetnkfsgyplyhsvyetye   lvekfydpmf   kyhltvaqvr   ggmvfelans   ivlpfdcrdyavvlrkyadk   iysismkhpq   emktysvsfd   slfsavknft   eiaskfserlqdfdksnpiv   lrmmndqlmf   lerafidplg    lpdrpfyrhv   iyapsshnkyagesfpgiyd alfdieskvd pskawgevkr qiyvaaftvq aaaetlsevaSEQ. ID NO. 5SIZE: 309 aaDEFINITION Melanoma-associated antigen 1 (MAGE-1 antigen)(Antigen MZ2-E).ACCESSION NO.: P43355msleqrslhc   kpeealeaqq   ealglvcvqa   atssssplvl   gtleevptagstdppqspqg   asafpttinf   trqrqpsegs   ssreeegpst   scileslfravitkkvadlv   gflllkyrar    epvtkaemle   sviknykhcf   peifgkaseslqlvfgidvk   eadptghsyv   lvtclglsyd   gllgdnqimp   ktgfliivlvmiamegghap   eeeiweelsv   mevydgrehs   aygeprkllt   qdlvqekyleyrqvpdsdpa ryeflwgpra  laetsyvkvl eyvikvsarv rfffpslreaalreeeegvSEQ. ID NO. 6SIZE: 314 aaDEFINITION Melanoma-associated antigen 3 (MAGE-3 antigen)(Antigen MZ2-D).ACCESSION NO.: P43357mpleqrsqhc   kpeeglearg   ealglvgaqa   pateeqeaas   ssstlvevtlgevpaaespd   ppqspqgass   lpttmnyplw   sqsyedssnq   eeegpstfpdlesefqaals   rkvaelvhfl    llkyrarepv   tkaemlgsvv   gnwqyffpvifskassslql   vfgielmevd   pighlyifat   clglsydgll   gdnqimpkaglliivlaiia   regdcapeek   iweelsvlev   fegredsilg   dpkklltqhfvqenyleyrq   vpgsdpacye   flwgpralve    tsyvkvlhhm   vkisggphisypplhewvlr egeeSEQ. ID NO. 7SIZE: 180 aaDEFINITION Cancer/testis antigen 1 (Autoimmunogeniccancer/testis antigen NY-ESO-1).ACCESSION NO.: P78358mqaegrgtgg   stgdadgpgg   pgipdgpggn   aggpgeagat   ggrgprgagaarasgpggga   prgphggaas   glngccrcga   rgpesrllef   ylampfatpmeaelarrsla   qdapplpvpg   vllkeftvsg   niltirltaa   dhrqlqlsissclqqlsllm witqcflpvf laqppsgqrrSEQ. ID NO. 8SIZE: 509 aaDEFINITION preferentially expressed antigen in melanoma;melanoma antigen preferentially expressed in tumors; Opa-interacting protein OIP4; preferentially expressed antigen ofmelanoma [Homo sapiens].ACCESSION NO.: NP_006106merrrlwgsi   qsryismsvw   tsprrlvela   gqsllkdeal   aiaalellprelfpplfmaa   fdgrhsqtlk   amvqawpftc   lplgvlmkgq   hlhletfkavldgldvllaq   evrprrwklq   vldlrknshq   dfwtvwsgnr   aslysfpepeaaqpmtkkrk   vdglsteaeq   pfipvevlvd    lflkegacde    lfsyliekvkrkknvlrlcc   kklkifampm   qdikmilkmv   qldsiedlev   tctwklptlakfspylgqmi   nlrrlllshi   hassyispek   eeqyiaqfts   qflslqclqalyvdslfflr    grldqllrhv   mnpletlsit   ncrlsegdvm   hlsqspsvsqlsvlslsgvm   ltdvspeplq   allerasatl   qdlvfdecgi   tddqllallpslshcsqltt   lsfygnsisi   salqsllqhl   iglsnlthvl   ypvplesyedihgtlhlerl aylharlrel lcelgrpsmv wlsanpcphc gdrtfydpepilcpcfmpnSEQ. ID NO. 9SIZE: 1255 aa3DEFINITION Receptor protein-tyrosine kinase erbB-2 precursor(p185erbB2) (NEU proto-oncogene) (C-erbB-2) (Tyrosine kinase-type cell surface receptor HER2) (MLN 19).ACCESSION NO.: P04626melaalcrwg   lllallppga   astqvctgtd   mklrlpaspe   thldmlrhlyqgcqvvqgnl   eltylptnas   lsflqdiqev    qgyvliahnq   vrqvplqrlrivrgtqlfed   nyalavldng   dplnnttpvt   gaspgglrel   qlrslteilkggvliqrnpq   lcyqdtilwk   difhknnqla   ltlidtnrsr   achpcspmckgsrcwgesse   dcqsltrtvc   aggcarckgp   lptdccheqc   aagctgpkhsdclaclhfnh   sgicelhcpa   lvtyntdtfe   smpnpegryt   fgascvtacpynylstdvgs   ctlvcplhnq   evtaedgtqr   cekcskpcar   vcyglgmehlrevravtsan   iqefagckki   fgslaflpes    fdgdpasnta   plqpeqlqvfetleeitgyl   yisawpdslp   dlsvfqnlqv   irgrilhnga   ysltlqglgiswlglrslre   lgsglalihh   nthlcfvhtv   pwdqlfrnph   qallhtanrpedecvgegla   chqlcarghc   wgpgptqcvn   csqflrgqec    veecrvlqglpreyvnarhc   lpchpecqpq   ngsvtcfgpe   adqcvacahy   kdppfcvarcpsgvkpdlsy   mpiwkfpdee   gacqpcpinc   thscvdlddk   gcpaeqraspltsiisavvg   illvvvlgvv   fgilikrrqq   kirkytmrrl   lqetelvepltpsgampnqa   qmrilketel   rkvkvlgsga   fgtvykgiwi   pdgenvkipvaikvlrents   pkankeilde   ayvmagvgsp   yvsrllgicl   tstvqlvtqlmpygclldhv   renrgrlgsq   dllnwcmqia   kgmsyledvr   lvhrdlaarnvlvkspnhvk   itdfglarll   dideteyhad   ggkvpikwma   lesilrrrfthqsdvwsygv   tvwelmtfga   kpydgipare   ipdllekger   lpqppictidvymimvkcwm   idsecrprfr   elvsefsrma   rdpqrfvviq   nedlgpaspldstfyrslle   dddmgdlvda   eeylvpqqgf   fcpdpapgag   gmvhhrhrssstrsgggdlt   lglepseeea   prsplapseg   agsdvfdgdl   gmgaakglqslpthdpsplq   rysedptvpl   psetdgyvap   ltcspqpeyv   nqpdvrpqppspregplpaa   rpagatlerp   ktlspgkngv   vkdvfafgga   venpeyltpqggaapqphpp pafspafdnl yywdqdpper gappstfkgt ptaenpeylgldvpvSEQ. ID NO. 10SIZE: 2384 bpDEFINITION Homo sapiens tyrosinase (oculocutaneous albinismIA) (TYR), mRNA.ACCESSION NO.: NM_000372tattgagttc   ttcaaacatt   gtagcctctt   tatggtctct   gagaaataactaccttaaac   ccataatctt   taatacttcc   taaactttct   taataagagaagctctattc   ctgacactac   ctctcatttg   caaggtcaaa   tcatcattagttttgtagtc   tattaactgg   gtttgcttag   gtcaggcatt   attattactaaccttattgt   taatattcta   accataagaa   ttaaactatt   aatggtgaatagagtttttc   actttaacat   aggcctatcc   cactggtggg   atacgagccaattcgaaaga   aaagtcagtc   atgtgctttt   cagaggatga   aagcttaagataaagactaa   aagtgtttga   tgctggaggt   gggagtggta   ttatataggtctcagccaag   acatgtgata   atcactgtag   tagtagctgg   aaagagaaatctgtgactcc   aattagccag   ttcctgcaga   ccttgtgagg   actagaggaagaatgctcct   ggctgttttg   tactgcctgc   tgtggagttt   ccagacctccgctggccatt   tccctagagc   ctgtgtctcc   tctaagaacc   tgatggagaaggaatgctgt   ccaccgtgga   gcggggacag   gagtccctgt   ggccagctttcaggcagagg   ttcctgtcag   aatatccttc   tgtccaatgc   accacttgggcctcaatttc   ccttcacagg   ggtggatgac   cgggagtcgt   ggccttccgtcttttataat   aggacctgcc   agtgctctgg   caacttcatg   ggattcaactgtggaaactg   caagtttggc   ttttggggac   caaactgcac   agagagacgactcttggtga   gaagaaacat   cttcgatttg   agtgccccag   agaaggacaaattttttgcc   tacctcactt   tagcaaagca   taccatcagc   tcagactatgtcatccccat   agggacctat   ggccaaatga   aaaatggatc   aacacccatgtttaacgaca   tcaatattta   tgacctcttt   gtctggatgc   attattatgtgtcaatggat   gcactgcttg   ggggatctga   aatctggaga   gacattgattttgcccatga   agcaccagct   tttctgcctt   ggcatagact   cttcttgttgcggtgggaac   aagaaatcca   gaagctgaca   ggagatgaaa   acttcactattccatattgg   gactggcggg   atgcagaaaa   gtgtgacatt   tgcacagatgagtacatggg   aggtcagcac   cccacaaatc   ctaacttact   cagcccagcatcattcttct   cctcttggca   gattgtctgt   agccgattgg   aggagtacaacagccatcag   tctttatgca   atggaacgcc   cgagggacct   ttacggcgtaatcctggaaa   ccatgacaaa   tccagaaccc   caaggctccc   ctcttcagctgatgtagaat   tttgcctgag   tttgacccaa   tatgaatctg   gttccatggataaagctgcc   aatttcagct   ttagaaatac   actggaagga   tttgctagtccacttactgg   gatagcggat   gcctctcaaa   gcagcatgca   caatgccttgcacatctata   tgaatggaac   aatgtcccag   gtacagggat   ctgccaacgatcctatcttc   cttcttcacc   atgcatttgt   tgacagtatt   tttgagcagtggctccgaag   gcaccgtcct   cttcaagaag   tttatccaga   agccaatgcacccattggac   ataaccggga   atcctacatg   gttcctttta   taccactgtacagaaatggt   gatttcttta   tttcatccaa   agatctgggc   tatgactatagctatctaca   agattcagac   ccagactctt   ttcaagacta   cattaagtcctatttggaac   aagcgagtcg   gatctggtca   tggctccttg   gggcggcgatggtaggggcc   gtcctcactg   ccctgctggc   agggcttgtg   agcttgctgtgtcgtcacaa   gagaaagcag   cttcctgaag   aaaagcagcc   actcctcatggagaaagagg   attaccacag   cttgtatcag   agccatttat   aaaaggcttaggcaatagag   tagggccaaa   aagcctgacc   tcactctaac   tcaaagtaatgtccaggttc   ccagagaata   tctgctggta   tttttctgta   aagaccatttgcaaaattgt   aacctaatac   aaagtgtagc   cttcttccaa   ctcaggtagaacacacctgt   ctttgtcttg   ctgttttcac   tcagcccttt   taacattttcccctaagccc   atatgtctaa   ggaaaggatg   ctatttggta   atgaggaactgttatttgta tgtgaattaa agtgctctta ttttSEQ. ID NO. 11SIZE: 1524 bpDEFINITION Human melanoma antigen recognized by T-cells(MART-1) mRNA.ACCESSION NO.: U06452agcagacaga   ggactctcat   taaggaaggt   gtcctgtgcc   ctgaccctacaagatgccaa   gagaagatgc   tcacttcatc   tatggttacc   ccaagaaggggcacggccac   tcttacacca   cggctgaaga   ggccgctggg   atcggcatcctgacagtgat   cctgggagtc   ttactgctca   tcggctgttg   gtattgtagaagacgaaatg   gatacagagc   cttgatggat   aaaagtcttc   atgttggcactcaatgtgcc   ttaacaagaa   gatgcccaca   agaagggttt   gatcatcgggacagcaaagt   gtctcttcaa   gagaaaaact   gtgaacctgt   ggttcccaatgctccacctg   cttatgagaa   actctctgca   gaacagtcac   caccaccttattcaccttaa   gagccagcga   gacacctgag   acatgctgaa   attatttctctcacactttt   gcttgaattt   aatacagaca   tctaatgttc   tcctttggaatggtgtagga   aaaatgcaag   ccatctctaa   taataagtca   gtgttaaaattttagtaggt   ccgctagcag   tactaatcat   gtgaggaaat   gatgagaaatattaaattgg   gaaaactcca   tcaataaatg   ttgcaatgca   tgatactatctgtgccagag   gtaatgttag   taaatccatg   gtgttatttt   ctgagagacagaattcaagt   gggtattctg   gggccatcca   atttctcttt   acttgaaatttggctaataa   caaactagtc   aggttttcga   accttgaccg   acatgaactgtacacagaat   tgttccagta   ctatggagtg   ctcacaaagg   atacttttacaggttaagac   aaagggttga   ctggcctatt   tatctgatca   agaacatgtcagcaatgtct   ctttgtgctc   taaaattcta   ttatactaca   ataatatattgtaaagatcc   tatagctctt   tttttttgag   atggagtttc   gcttttgttgcccaggctgg   agtgcaatgg   cgcgatcttg   gctcaccata   acctccgcctcccaggttca   agcaattctc   ctgccttagc   ctcctgagta   gctgggattacaggcgtgcg   ccactatgcc   tgactaattt   tgtagtttta   gtagagacggggtttctcca   tgttggtcag   gctggtctca   aactcctgac   ctcaggtgatctgcccgcct   cagcctccca   aagtgctgga   attacaggcg   tgagccaccacgcctggctg   gatcctatat   cttaggtaag   acatataacg   cagtctaattacatttcact   tcaaggctca   atgctattct   aactaatgac   aagtattttctactaaacca   gaaattggta   gaaggattta   aataagtaaa   agctactatgtactgcctta   gtgctgatgc   ctgtgtactg   ccttaaatgt   acctatggcaatttagctct   cttgggttcc   caaatccctc   tcacaagaat   gtgcagaagaaatcataaag gatcagagat tctgSEQ. ID NO. 12SIZE: 1466 bpDEFINITION Homo sapiens synovial sarcoma, X breakpoint 2(SSX2), transcript variant 1, mRNA.ACCESSION NO.: NM_003147gcatgctctg   actttctctc   tctttcgatt   cttccatact   cagagtacgcacggtctgat   tttctctttg   gattcttcca   aaatcagagt   cagactgctcccggtgccat   gaacggagac   gacgcctttg   caaggagacc   cacggttggtgctcaaatac   cagagaagat   ccaaaaggcc   ttcgatgata   ttgccaaatacttctctaag   gaagagtggg   aaaagatgaa   agcctcggag   aaaatcttctatgtgtatat   gaagagaaag   tatgaggcta   tgactaaact   aggtttcaaggccaccctcc   cacctttcat   gtgtaataaa   cgggccgaag   acttccaggggaatgatttg   gataatgacc   ctaaccgtgg   gaatcaggtt   gaacgtcctcagatgacttt   cggcaggctc   cagggaatct   ccccgaagat   catgcccaagaagccagcag   aggaaggaaa   tgattcggag   gaagtgccag   aagcatctggcccacaaaat   gatgggaaag   agctgtgccc   cccgggaaaa   ccaactacctctgagaagat   tcacgagaga   tctggaaata   gggaggccca   agaaaaggaagagagacgcg   gaacagctca   tcggtggagc   agtcagaaca   cacacaacattggtcgattc   agtttgtcaa   cttctatggg   tgcagttcat   ggtacccccaaaacaattac   acacaacagg   gacccaaaag   gggggaacat   gcctggacccacagactgcg   tgagagaaaa   cagctggtga   tttatgaaga   gatcagcgaccctgaggaag   atgacgagta   actcccctca   gggatacgac   acatgcccatgatgagaagc   agaacgtggt   gacctttcac   gaacatgggc   atggctgcggacccctcgtc   atcaggtgca   tagcaagtga   aagcaagtgt   tcacaacagtgaaaagttga   gcgtcatttt   tcttagtgtg   ccaagagttc   gatgttagcgtttacgttgt   attttcttac   actgtgtcat   tctgttagat   actaacattttcattgatga   gcaagacata   cttaatgcat   attttggttt   gtgtatccatgcacctacct   tagaaaacaa   gtattgtcgg   ttacctctgc   atggaacagcattaccctcc   tctctcccca   gatgtgacta   ctgagggcag   ttctgagtgtttaatttcag   attttttcct   ctgcatttac   acacacacgc   acacaaaccacaccacacac   acacacacac   acacacacac   acacacacac   acacaccaagtaccagtata   agcatctgcc   atctgctttt   cccattgcca   tgcgtcctggtcaagctccc   ctcactctgt   ttcctggtca   gcatgtactc   ccctcatccgattcccctgt   agcagtcact   gacagttaat   aaacctttgc   aaacgttcaaaaaaaaaaaa aaaaaaSEQ. ID NO. 13SIZE: 2653 bpDEFINITION Homo sapiens folate hydrolase (prostate-specificmembrane antigen) 1 (FOLH1), mRNA.ACCESSION NO.: NM_004476ctcaaaaggg   gccggatttc   cttctcctgg   aggcagatgt   tgcctctctctctcgctcgg   attggttcag   tgcactctag   aaacactgct   gtggtggagaaactggaccc   caggtctgga   gcgaattcca   gcctgcaggg   ctgataagcgaggcattagt   gagattgaga   gagactttac   cccgccgtgg   tggttggagggcgcgcagta   gagcagcagc   acaggcgcgg   gtcccgggag   gccggctctgctcgcgccga   gatgtggaat   ctccttcacg   aaaccgactc   ggctgtggccaccgcgcgcc   gcccgcgctg   gctgtgcgct   ggggcgctgg   tgctggcgggtggcttcttt   ctcctcggct   tcctcttcgg   gtggtttata   aaatcctccaatgaagctac   taacattact   ccaaagcata   atatgaaagc   atttttggatgaattgaaag   ctgagaacat   caagaagttc   ttatataatt   ttacacagataccacattta   gcaggaacag   aacaaaactt   tcagcttgca   aagcaaattcaatcccagtg   gaaagaattt   ggcctggatt   ctgttgagct   agcacattatgatgtcctgt   tgtcctaccc   aaataagact   catcccaact   acatctcaataattaatgaa   gatggaaatg   agattttcaa   cacatcatta   tttgaaccacctcctccagg   atatgaaaat   gtttcggata   ttgtaccacc   tttcagtgctttctctcctc   aaggaatgcc   agagggcgat   ctagtgtatg   ttaactatgcacgaactgaa   gacttcttta   aattggaacg   ggacatgaaa   atcaattgctctgggaaaat   tgtaattgcc   agatatggga   aagttttcag   aggaaataaggttaaaaatg   cccagctggc   aggggccaaa   ggagtcattc   tctactccgaccctgctgac   tactttgctc   ctggggtgaa   gtcctatcca   gatggttggaatcttcctgg   aggtggtgtc   cagcgtggaa   atatcctaaa   tctgaatggtgcaggagacc   ctctcacacc   aggttaccca   gcaaatgaat   atgcttataggcgtggaatt   gcagaggctg   ttggtcttcc   aagtattcct   gttcatccaattggatacta   tgatgcacag   aagctcctag   aaaaaatggg   tggctcagcaccaccagata   gcagctggag   aggaagtctc   aaagtgccct   acaatgttggacctggcttt   actggaaact   tttctacaca   aaaagtcaag   atgcacatccactctaccaa   tgaagtgaca   agaatttaca   atgtgatagg   tactctcagaggagcagtgg   aaccagacag   atatgtcatt   ctgggaggtc   accgggactcatgggtgttt   ggtggtattg   accctcagag   tggagcagct   gttgttcatgaaattgtgag   gagctttgga   acactgaaaa   aggaagggtg   gagacctagaagaacaattt   tgtttgcaag   ctgggatgca   gaagaatttg   gtcttcttggttctactgag   tgggcagagg   agaattcaag   actccttcaa   gagcgtggcgtggcttatat   taatgctgac   tcatctatag   aaggaaacta   cactctgagagttgattgta   caccgctgat   gtacagcttg   gtacacaacc   taacaaaagagctgaaaagc   cctgatgaag   gctttgaagg   caaatctctt   tatgaaagttggactaaaaa   aagtccttcc   ccagagttca   gtggcatgcc   caggataagcaaattgggat   ctggaaatga   ttttgaggtg   ttcttccaac   gacttggaattgcttcaggc   agagcacggt   atactaaaaa   ttgggaaaca   aacaaattcagcggctatcc   actgtatcac   agtgtctatg   aaacatatga   gttggtggaaaagttttatg   atccaatgtt   taaatatcac   ctcactgtgg   cccaggttcgaggagggatg   gtgtttgagc   tagccaattc   catagtgctc   ccttttgattgtcgagatta   tgctgtagtt   ttaagaaagt   atgctgacaa   aatctacagtatttctatga   aacatccaca   ggaaatgaag   acatacagtg   tatcatttgattcacttttt   tctgcagtaa   agaattttac   agaaattgct   tccaagttcagtgagagact   ccaggacttt   gacaaaagca   acccaatagt   attaagaatgatgaatgatc   aactcatgtt   tctggaaaga   gcatttattg   atccattagggttaccagac   aggccttttt   ataggcatgt   catctatgct   ccaagcagccacaacaagta   tgcaggggag   tcattcccag   gaatttatga   tgctctgtttgatattgaaa   gcaaagtgga   cccttccaag   gcctggggag   aagtgaagagacagatttat   gttgcagcct   tcacagtgca   ggcagctgca   gagactttgagtgaagtagc   ctaagaggat   tctttagaga   atccgtattg   aatttgtgtggtatgtcact   cagaaagaat   cgtaatgggt   atattgataa   attttaaaattggtatattt gaaataaagt tgaatattat atataaaaaa aaaaaaaaaa aaaSEQ. ID NO. 14SIZE: 2420 bpDEFINITION Human antigen (MAGE-1) gene, complete cds.ACCESSION NO.: M77481ggatccaggc   cctgccagga   aaaatataag   ggccctgcgt   gagaacagagggggtcatcc   actgcatgag   agtggggatg   tcacagagtc   cagcccaccctcctggtagc   actgagaagc   cagggctgtg   cttgcggtct   gcaccctgagggcccgtgga   ttcctcttcc   tggagctcca   ggaaccaggc   agtgaggccttggtctgaga   cagtatcctc   aggtcacaga   gcagaggatg   cacagggtgtgccagcagtg   aatgtttgcc   ctgaatgcac   accaagggcc   ccacctgccacaggacacat   aggactccac   agagtctggc   ctcacctccc   tactgtcagtcctgtagaat   cgacctctgc   tggccggctg   taccctgagt   accctctcacttcctccttc   aggttttcag   gggacaggcc   aacccagagg   acaggattccctggaggcca   cagaggagca   ccaaggagaa   gatctgtaag   taggcctttgttagagtctc   caaggttcag   ttctcagctg   aggcctctca   cacactccctctctccccag   gcctgtgggt   cttcattgcc   cagctcctgc   ccacactcctgcctgctgcc   ctgacgagag   tcatcatgtc   tcttgagcag   aggagtctgcactgcaagcc   tgaggaagcc   cttgaggccc   aacaagaggc   cctgggcctggtgtgtgtgc   aggctgccac   ctcctcctcc   tctcctctgg   tcctgggcaccctggaggag   gtgcccactg   ctgggtcaac   agatcctccc   cagagtcctcagggagcctc   cgcctttccc   actaccatca   acttcactcg   acagaggcaacccagtgagg   gttccagcag   ccgtgaagag   gaggggccaa   gcacctcttgtatcctggag   tccttgttcc   gagcagtaat   cactaagaag   gtggctgatttggttggttt   tctgctcctc   aaatatcgag   ccagggagcc   agtcacaaaggcagaaatgc   tggagagtgt   catcaaaaat   tacaagcact   gttttcctgagatcttcggc   aaagcctctg   agtccttgca   gctggtcttt   ggcattgacgtgaaggaagc   agaccccacc   ggccactcct   atgtccttgt   cacctgcctaggtctctcct   atgatggcct   gctgggtgat   aatcagatca   tgcccaagacaggcttcctg   ataattgtcc   tggtcatgat   tgcaatggag   ggcggccatgctcctgagga   ggaaatctgg   gaggagctga   gtgtgatgga   ggtgtatgatgggagggagc   acagtgccta   tggggagccc   aggaagctgc   tcacccaagatttggtgcag   gaaaagtacc   tggagtaccg   gcaggtgccg   gacagtgatcccgcacgcta   tgagttcctg   tggggtccaa   gggccctcgc   tgaaaccagctatgtgaaag   tccttgagta   tgtgatcaag   gtcagtgcaa   gagttcgctttttcttccca   tccctgcgtg   aagcagcttt   gagagaggag   gaagagggagtctgagcatg   agttgcagcc   aaggccagtg   ggagggggac   tgggccagtgcaccttccag   ggccgcgtcc   agcagcttcc   cctgcctcgt   gtgacatgaggcccattctt   cactctgaag   agagcggtca   gtgttctcag   tagtaggtttctgttctatt   gggtgacttg   gagatttatc   tttgttctct   tttggaattgttcaaatgtt   tttttttaag   ggatggttga   atgaacttca   gcatccaagtttatgaatga   cagcagtcac   acagttctgt   gtatatagtt   taagggtaagagtcttgtgt   tttattcaga   ttgggaaatc   cattctattt   tgtgaattgggataataaca   gcagtggaat   aagtacttag   aaatgtgaaa   aatgagcagtaaaatagatg   agataaagaa   ctaaagaaat   taagagatag   tcaattcttgccttatacct   cagtctattc   tgtaaaattt   ttaaagatat   atgcatacctggatttcctt   ggcttctttg   agaatgtaag   agaaattaaa   tctgaataaagaattcttcc   tgttcactgg   ctcttttctt   ctccatgcac   tgagcatctgctttttggaa   ggccctgggt   tagtagtgga   gatgctaagg   taagccagactcatacccac   ccatagggtc   gtagagtcta   ggagctgcag   tcacgtaatcgaggtggcaa   gatgtcctct   aaagatgtag   ggaaaagtga   gagaggggtgagggtgtggg   gctccgggtg   agagtggtgg   agtgtcaatg   ccctgagctggggcattttg   ggctttggga   aactgcagtt   ccttctgggg   gagctgattgtaatgatctt gggtggatccSEQ. ID NO. 15SIZE: 4204 bp DNA linear PRI APR. 07, 1994DEFINITION Human MAGE-3 antigen (MAGE-3) gene, complete cds.ACCESSION NO.: U03735acgcaggcag   tgatgtcacc   cagaccacac   cccttccccc   aatgccacttcagggggtac   tcagagtcag   agacttggtc   tgaggggagc   agaagcaatctgcagaggat   ggcggtccag   gctcagccag   gcatcaactt   caggaccctgagggatgacc   gaaggccccg   cccacccacc   cccaactccc   ccgaccccaccaggatctac   agcctcagga   cccccgtccc   aatccttacc   ccttgccccatcaccatctt   catgcttacc   tccaccccca   tccgatcccc   atccaggcagaatccagttc   cacccctgcc   cggaacccag   ggtagtaccg   ttgccaggatgtgacgccac   tgacttgcgc   attggaggtc   agaagaccgc   gagattctcgccctgagcaa   cgagcgacgg   cctgacgtcg   gcggagggaa   gccggcccaggctcggtgag   gaggcaaggt   aagacgctga   gggaggactg   aggcgggcctcacctcagac   agagggcctc   aaataatcca   gtgctgcctc   tgctgccgggcctgggccac   cccgcagggg   aagacttcca   ggctgggtcg   ccactacctcaccccgccga   cccccgccgc   tttagccacg   gggaactctg   gggacagagcttaatgtggc   cagggcaggg   ctggttagaa   gaggtcaggg   cccacgctgtggcaggaatc   aaggtcagga   ccccgagagg   gaactgaggg   cagcctaaccaccaccctca   ccaccattcc   cgtcccccaa   cacccaaccc   cacccccatcccccattccc   atccccaccc   ccacccctat   cctggcagaa   tccgggctttgcccctggta   tcaagtcacg   gaagctccgg   gaatggcggc   caggcacgtgagtcctgagg   ttcacatcta   cggctaaggg   agggaagggg   ttcggtatcgcgagtatggc   cgttgggagg   cagcgaaagg   gcccaggcct   cctggaagacagtggagtcc   tgaggggacc   cagcatgcca   ggacaggggg   cccactgtacccctgtctca   aaccgaggca   ccttttcatt   cggctacggg   aatcctagggatgcagaccc   acttcagcag   ggggttgggg   cccagccctg   cgaggagtcatggggaggaa   gaagagggag   gactgagggg   accttggagt   ccagatcagtggcaaccttg   ggctggggga   tgctgggcac   agtggccaaa   tgtgctctgtgctcattgcg   ccttcagggt   gaccagagag   ttgagggctg   tggtctgaagagtgggactt   caggtcagca   gagggaggaa   tcccaggatc   tgcagggcccaaggtgtacc   cccaaggggc   ccctatgtgg   tggacagatg   cagtggtcctaggatctgcc   aagcatccag   gtgaagagac   tgagggagga   ttgagggtacccctgggaca   gaatgcggac   tgggggcccc   ataaaaatct   gccctgctcctgctgttacc   tcagagagcc   tgggcagggc   tgtcagctga   ggtccctccattatcctagg   atcactgatg   tcagggaagg   ggaagccttg   gtctgagggggctgcactca   gggcagtaga   gggaggctct   cagaccctac   taggagtggaggtgaggacc   aagcagtctc   ctcacccagg   gtacatggac   ttcaataaatttggacatct   ctcgttgtcc   tttccgggag   gacctgggaa   tgtatggccagatgtgggtc   ccctcatgtt   tttctgtacc   atatcaggta   tgtgagttcttgacatgaga   gattctcagg   ccagcagaag   ggagggatta   ggccctataaggagaaaggt   gagggccctg   agtgagcaca   gaggggatcc   tccaccccagtagagtgggg   acctcacaga   gtctggccaa   ccctcctgac   agttctgggaatccgtggct   gcgtttgctg   tctgcacatt   gggggcccgt   ggattcctctcccaggaatc   aggagctcca   ggaacaaggc   agtgaggact   tggtctgaggcagtgtcctc   aggtcacaga   gtagaggggg   ctcagatagt   gccaacggtgaaggtttgcc   ttggattcaa   accaagggcc   ccacctgccc   cagaacacatggactccaga   gcgcctggcc   tcaccctcaa   tactttcagt   cctgcagcctcagcatgcgc   tggccggatg   taccctgagg   tgccctctca   cttcctccttcaggttctga   ggggacaggc   tgacctggag   gaccagaggc   ccccggaggagcactgaagg   agaagatctg   taagtaagcc   tttgttagag   cctccaaggttccattcagt   actcagctga   ggtctctcac   atgctccctc   tctccccaggccagtgggtc   tccattgccc   agctcctgcc   cacactcccg   cctgttgccctgaccagagt   catcatgcct   cttgagcaga   ggagtcagca   ctgcaagcctgaagaaggcc   ttgaggcccg   aggagaggcc   ctgggcctgg   tgggtgcgcaggctcctgct   actgaggagc   aggaggctgc   ctcctcctct   tctactctagttgaagtcac   cctgggggag   gtgcctgctg   ccgagtcacc   agatcctccccagagtcctc   agggagcctc   cagcctcccc   actaccatga   actaccctctctggagccaa   tcctatgagg   actccagcaa   ccaagaagag   gaggggccaagcaccttccc   tgacctggag   tccgagttcc   aagcagcact   cagtaggaaggtggccgagt   tggttcattt   tctgctcctc   aagtatcgag   ccagggagccggtcacaaag   gcagaaatgc   tggggagtgt   cgtcggaaat   tggcagtatttctttcctgt   gatcttcagc   aaagcttcca   gttccttgca   gctggtctttggcatcgagc   tgatggaagt   ggaccccatc   ggccacttgt   acatctttgccacctgcctg   ggcctctcct   acgatggcct   gctgggtgac   aatcagatcatgcccaaggc   aggcctcctg   ataatcgtcc   tggccataat   cgcaagagagggcgactgtg   cccctgagga   gaaaatctgg   gaggagctga   gtgtgttagaggtgtttgag   gggagggaag   acagtatctt   gggggatccc   aagaagctgctcacccaaca   tttcgtgcag   gaaaactacc   tggagtaccg   gcaggtccccggcagtgatc   ctgcatgtta   tgaattcctg   tggggtccaa   gggccctcgttgaaaccagc   tatgtgaaag   tcctgcacca   tatggtaaag   atcagtggaggacctcacat   ttcctaccca   cccctgcatg   agtgggtttt   gagagagggggaagagtgag   tctgagcacg   agttgcagcc   agggccagtg   ggagggggtctgggccagtg   caccttccgg   ggccgcatcc   cttagtttcc   actgcctcctgtgacgtgag   gcccattctt   cactctttga   agcgagcagt   cagcattcttagtagtgggt   ttctgttctg   ttggatgact   ttgagattat   tctttgtttcctgttggagt   tgttcaaatg   ttccttttaa   cggatggttg   aatgagcgtcagcatccagg   tttatgaatg   acagtagtca   cacatagtgc   tgtttatatagtttaggagt   aagagtcttg   ttttttactc   aaattgggaa   atccattccattttgtgaat   tgtgacataa   taatagcagt   ggtaaaagta   tttgcttaaaattgtgagcg   aattagcaat   aacatacatg   agataactca   agaaatcaaaagatagttga   ttcttgcctt   gtacctcaat   ctattctgta   aaattaaacaaatatgcaaa   ccaggatttc   cttgacttct   ttgagaatgc   aagcgaaattaaatctgaat   aaataattct   tcctcttcac   tggctcgttt   cttttccgttcactcagcat   ctgctctgtg   ggaggccctg   ggttagtagt   ggggatgctaaggtaagcca   gactcacgcc   tacccatagg   gctgtagagc   ctaggacctgcagtcatata   attaaggtgg   tgagaagtcc   tgtaagatgt   agaggaaatgtaagagaggg gtgagggtgt ggcgctccgg gtgagagtag tggagtgtca gtgcSEQ. ID NO. 16SIZE: 752 bpDEFINITION Human autoimmunogenic cancer/testis antigen NY-ESO-1 mRNA, complete cds.ACCESSION NO.: U87459atcctcgtgg   gccctgacct   tctctctgag   agccgggcag   aggctccggagccatgcagg   ccgaaggccg   gggcacaggg   ggttcgacgg   gcgatgctgatggcccagga   ggccctggca   ttcctgatgg   cccagggggc   aatgctggcggcccaggaga   ggcgggtgcc   acgggcggca   gaggtccccg   gggcgcaggggcagcaaggg   cctcggggcc   gggaggaggc   gccccgcggg   gtccgcatggcggcgcggct   tcagggctga   atggatgctg   cagatgcggg   gccagggggccggagagccg   cctgcttgag   ttctacctcg   ccatgccttt   cgcgacacccatggaagcag   agctggcccg   caggagcctg   gcccaggatg   ccccaccgcttcccgtgcca   ggggtgcttc   tgaaggagtt   cactgtgtcc   ggcaacatactgactatccg   actgactgct   gcagaccacc   gccaactgca   gctctccatcagctcctgtc   tccagcagct   ttccctgttg   atgtggatca   cgcagtgctttctgcccgtg   tttttggctc   agcctccctc   agggcagagg   cgctaagcccagcctggcgc   cccttcctag   gtcatgcctc   ctcccctagg   gaatggtcccagcacgagtg   gccagttcat   tgtgggggcc   tgattgtttg   tcgctggaggaggacggctt acatgtttgt ttctgtagaa aataaaactg agctacgaaa aaSEQ. ID NO. 17SIZE: 2148 bpDEFINITION Homo sapiens preferentially expressed antigen inmelanoma (PRAME), mRNA.ACCESSION NO.: NM_006115gcttcagggt   acagctcccc   cgcagccaga   agccgggcct   gcagcgcctcagcaccgctc   cgggacaccc   cacccgcttc   ccaggcgtga   cctgtcaacagcaacttcgc   ggtgtggtga   actctctgag   gaaaaaccat   tttgattattactctcagac   gtgcgtggca   acaagtgact   gagacctaga   aatccaagcgttggaggtcc   tgaggccagc   ctaagtcgct   tcaaaatgga   acgaaggcgtttgtggggtt   ccattcagag   ccgatacatc   agcatgagtg   tgtggacaagcccacggaga   cttgtggagc   tggcagggca   gagcctgctg   aaggatgaggccctggccat   tgccgccctg   gagttgctgc   ccagggagct   cttcccgccactcttcatgg   cagcctttga   cgggagacac   agccagaccc   tgaaggcaatggtgcaggcc   tggcccttca   cctgcctccc   tctgggagtg   ctgatgaagggacaacatct   tcacctggag   accttcaaag   ctgtgcttga   tggacttgatgtgctccttg   cccaggaggt   tcgccccagg   aggtggaaac   ttcaagtgctggatttacgg   aagaactctc   atcaggactt   ctggactgta   tggtctggaaacagggccag   tctgtactca   tttccagagc   cagaagcagc   tcagcccatgacaaagaagc   gaaaagtaga   tggtttgagc   acagaggcag   agcagcccttcattccagta   gaggtgctcg   tagacctgtt   cctcaaggaa   ggtgcctgtgatgaattgtt   ctcctacctc   attgagaaag   tgaagcgaaa   gaaaaatgtactacgcctgt   gctgtaagaa   gctgaagatt   tttgcaatgc   ccatgcaggatatcaagatg   atcctgaaaa   tggtgcagct   ggactctatt   gaagatttggaagtgacttg   tacctggaag   ctacccacct   tggcgaaatt   ttctccttacctgggccaga   tgattaatct   gcgtagactc   ctcctctccc   acatccatgcatcttcctac   atttccccgg   agaaggaaga   gcagtatatc   gcccagttcacctctcagtt   cctcagtctg   cagtgcctgc   aggctctcta   tgtggactctttatttttcc   ttagaggccg   cctggatcag   ttgctcaggc   acgtgatgaaccccttggaa   accctctcaa   taactaactg   ccggctttcg   gaaggggatgtgatgcatct   gtcccagagt   cccagcgtca   gtcagctaag   tgtcctgagtctaagtgggg   tcatgctgac   cgatgtaagt   cccgagcccc   tccaagctctgctggagaga   gcctctgcca   ccctccagga   cctggtcttt   gatgagtgtgggatcacgga   tgatcagctc   cttgccctcc   tgccttccct   gagccactgctcccagctta   caaccttaag   cttctacggg   aattccatct   ccatatctgccttgcagagt   ctcctgcagc   acctcatcgg   gctgagcaat   ctgacccacgtgctgtatcc   tgtccccctg   gagagttatg   aggacatcca   tggtaccctccacctggaga   ggcttgccta   tctgcatgcc   aggctcaggg   agttgctgtgtgagttgggg   cggcccagca   tggtctggct   tagtgccaac   ccctgtcctcactgtgggga   cagaaccttc   tatgacccgg   agcccatcct   gtgcccctgtttcatgccta   actagctggg   tgcacatatc   aaatgcttca   ttctgcatacttggacacta   aagccaggat   gtgcatgcat   cttgaagcaa   caaagcagccacagtttcag   acaaatgttc   agtgtgagtg   aggaaaacat   gttcagtgaggaaaaaacat   tcagacaaat   gttcagtgag   gaaaaaaagg   ggaagttggggataggcaga   tgttgacttg   aggagttaat   gtgatctttg   gggagatacatcttatagag   ttagaaatag   aatctgaatt   tctaaaggga   gattctggcttgggaagtac   atgtaggagt   taatccctgt   gtagactgtt   gtaaagaaactgttgaaaat aaagagaagc aatgtgaagc aaaaaaaaaa aaaaaaaaSEQ. ID NO. 18SIZE: 4530 bpDEFINITION Human tyrosine kinase-type receptor (HER2) mRNA,complete cds.ACCESSION NO.: M11730aattctcgag   ctcgtcgacc   ggtcgacgag   ctcgagggtc   gacgagctcgagggcgcgcg   cccggccccc   acccctcgca   gcaccccgcg   ccccgcgccctcccagccgg   gtccagccgg   agccatgggg   ccggagccgc   agtgagcaccatggagctgg   cggccttgtg   ccgctggggg   ctcctcctcg   ccctcttgccccccggagcc   gcgagcaccc   aagtgtgcac   cggcacagac   atgaagctgcggctccctgc   cagtcccgag   acccacctgg   acatgctccg   ccacctctaccagggctgcc   aggtggtgca   gggaaacctg   gaactcacct   acctgcccaccaatgccagc   ctgtccttcc   tgcaggatat   ccaggaggtg   cagggctacgtgctcatcgc   tcacaaccaa   gtgaggcagg   tcccactgca   gaggctgcggattgtgcgag   gcacccagct   ctttgaggac   aactatgccc   tggccgtgctagacaatgga   gacccgctga   acaataccac   ccctgtcaca   ggggcctccccaggaggcct   gcgggagctg   cagcttcgaa   gcctcacaga   gatcttgaaaggaggggtct   tgatccagcg   gaacccccag   ctctgctacc   aggacacgattttgtggaag   gacatcttcc   acaagaacaa   ccagctggct   ctcacactgatagacaccaa   ccgctctcgg   gcctgccacc   cctgttctcc   gatgtgtaagggctcccgct   gctggggaga   gagttctgag   gattgtcaga   gcctgacgcgcactgtctgt   gccggtggct   gtgcccgctg   caaggggcca   ctgcccactgactgctgcca   tgagcagtgt   gctgccggct   gcacgggccc   caagcactctgactgcctgg   cctgcctcca   cttcaaccac   agtggcatct   gtgagctgcactgcccagcc   ctggtcacct   acaacacaga   cacgtttgag   tccatgcccaatcccgaggg   ccggtataca   ttcggcgcca   gctgtgtgac   tgcctgtccctacaactacc   tttctacgga   cgtgggatcc   tgcaccctcg   tctgccccctgcacaaccaa   gaggtgacag   cagaggatgg   aacacagcgg   tgtgagaagtgcagcaagcc   ctgtgcccga   gtgtgctatg   gtctgggcat   ggagcacttgcgagaggtga   gggcagttac   cagtgccaat   atccaggagt   ttgctggctgcaagaagatc   tttgggagcc   tggcatttct   gccggagagc   tttgatggggacccagcctc   caacactgcc   ccgctccagc   cagagcagct   ccaagtgtttgagactctgg   aagagatcac   aggttaccta   tacatctcag   catggccggacagcctgcct   gacctcagcg   tcttccagaa   cctgcaagta   atccggggacgaattctgca   caatggcgcc   tactcgctga   ccctgcaagg   gctgggcatcagctggctgg   ggctgcgctc   actgagggaa   ctgggcagtg   gactggccctcatccaccat   aacacccacc   tctgcttcgt   gcacacggtg   ccctgggaccagctctttcg   gaacccgcac   caagctctgc   tccacactgc   caaccggccagaggacgagt   gtgtgggcga   gggcctggcc   tgccaccagc   tgtgcgcccgagggcactgc   tggggtccag   ggcccaccca   gtgtgtcaac   tgcagccagttccttcgggg   ccaggagtgc   gtggaggaat   gccgagtact   gcaggggctccccagggagt   atgtgaatgc   caggcactgt   ttgccgtgcc   accctgagtgtcagccccag   aatggctcag   tgacctgttt   tggaccggag   gctgaccagtgtgtggcctg   tgcccactat   aaggaccctc   ccttctgcgt   ggcccgctgccccagcggtg   tgaaacctga   cctctcctac   atgcccatct   ggaagtttccagatgaggag   ggcgcatgcc   agccttgccc   catcaactgc   acccactcctgtgtggacct   ggatgacaag   ggctgccccg   ccgagcagag   agccagccctctgacgtcca   tcgtctctgc   ggtggttggc   attctgctgg   tcgtggtcttgggggtggtc   tttgggatcc   tcatcaagcg   acggcagcag   aagatccggaagtacacgat   gcggagactg   ctgcaggaaa   cggagctggt   ggagccgctgacacctagcg   gagcgatgcc   caaccaggcg   cagatgcgga   tcctgaaagagacggagctg   aggaaggtga   aggtgcttgg   atctggcgct   tttggcacagtctacaaggg   catctggatc   cctgatgggg   agaatgtgaa   aattccagtggccatcaaag   tgttgaggga   aaacacatcc   cccaaagcca   acaaagaaatcttagacgaa   gcatacgtga   tggctggtgt   gggctcccca   tatgtctcccgccttctggg   catctgcctg   acatccacgg   tgcagctggt   gacacagcttatgccctatg   gctgcctctt   agaccatgtc   cgggaaaacc   gcggacgcctgggctcccag   gacctgctga   actggtgtat   gcagattgcc   aaggggatgagctacctgga   ggatgtgcgg   ctcgtacaca   gggacttggc   cgctcggaacgtgctggtca   agagtcccaa   ccatgtcaaa   attacagact   tcgggctggctcggctgctg   gacattgacg   agacagagta   ccatgcagat   gggggcaaggtgcccatcaa   gtggatggcg   ctggagtcca   ttctccgccg   gcggttcacccaccagagtg   atgtgtggag   ttatggtgtg   actgtgtggg   agctgatgacttttggggcc   aaaccttacg   atgggatccc   agcccgggag   atccctgacctgctggaaaa   gggggagcgg   ctgccccagc   cccccatctg   caccattgatgtctacatga   tcatggtcaa   atgttggatg   attgactctg   aatgtcggccaagattccgg   gagttggtgt   ctgaattctc   ccgcatggcc   agggacccccagcgctttgt   ggtcatccag   aatgaggact   tgggcccagc   cagtcccttggacagcacct   tctaccgctc   actgctggag   gacgatgaca   tgggggacctggtggatgct   gaggagtatc   tggtacccca   gcagggcttc   ttctgtccagaccctgcccc   gggcgctggg   ggcatggtcc   accacaggca   ccgcagctcatctaccagga   gtggcggtgg   ggacctgaca   ctagggctgg   agccctctgaagaggaggcc   cccaggtctc   cactggcacc   ctccgaaggg   gctggctccgatgtatttga   tggtgacctg   ggaatggggg   cagccaaggg   gctgcaaagcctccccacac   atgaccccag   ccctctacag   cggtacagtg   aggaccccacagtacccctg   ccctctgaga   ctgatggcta   cgttgccccc   ctgacctgcagcccccagcc   tgaatatgtg   aaccagccag   atgttcggcc   ccagcccccttcgccccgag   agggccctct   gcctgctgcc   cgacctgctg   gtgccactctggaaagggcc   aagactctct   ccccagggaa   gaatggggtc   gtcaaagacgtttttgcctt   tgggggtgcc   gtggagaacc   ccgagtactt   gacaccccagggaggagctg   cccctcagcc   ccaccctcct   cctgccttca   gcccagccttcgacaacctc   tattactggg   accaggaccc   accagagcgg   ggggctccacccagcacctt   caaagggaca   cctacggcag   agaacccaga   gtacctgggtctggacgtgc   cagtgtgaac   cagaaggcca   agtccgcaga   agccctgatgtgtcctcagg   gagcagggaa   ggcctgactt   ctgctggcat   caagaggtgggagggccctc   cgaccacttc   caggggaacc   tgccatgcca   ggaacctgtcctaaggaacc   ttccttcctg   cttgagttcc   cagatggctg   gaaggggtccagcctcgttg   gaagaggaac   agcactgggg   agtctttgtg   gattctgaggccctgcccaa   tgagactcta   gggtccagtg   gatgccacag   cccagcttggccctttcctt   ccagatcctg   ggtactgaaa   gccttaggga   agctggcctgagaggggaag   cggccctaag   ggagtgtcta   agaacaaaag   cgacccattcagagactgtc   cctgaaacct   agtactgccc   cccatgagga   aggaacagcaatggtgtcag   tatccaggct   ttgtacagag   tgcttttctg   tttagtttttactttttttg   ttttgttttt   ttaaagacga   aataaagacc   caggggagaatgggtgttgt   atggggaggc   aagtgtgggg   ggtccttctc   cacacccactttgtccattt gcaaatatat tttggaaaac


Claims
  • 1. A method of treating ovarian or colorectal cancer comprising immunizing against PRAME and at least one other tumor-associated antigen.
  • 2. The method of claim 1, wherein the at least one other tumor-associated antigen is selected from the group consisting of SSX-2, NY-ESO-1, and PSMA.
  • 3. The method of claim 1, wherein the at least one other tumor-associated antigen comprises NY-ESO-1 and SSX-2.
  • 4. The method of claim 1, wherein the at least one other tumor-associated antigen comprises NY-ESO-1, SSX-2, and PSMA.
  • 5. The method of claim 1, wherein a cytolytic T cell response is induced.
  • 6. The method of claim 1, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 7. The method of claim 6, wherein the antigen associated with tumor neovasculature is selected from the group consisting of PSMA, VEGFR2, and Tie-2.
  • 8. A method of treating pancreatic cancer comprising immunizing against PSMA and at least one other tumor-associated antigen.
  • 9. The method of claim 8 wherein the at least one other tumor-associated antigen is selected from the group consisting of SSX-2 and NY-ESO-1.
  • 10. The method of claim 8, wherein the at least one other tumor-associated antigen comprises NY-ESO-1 and SSX-2.
  • 11. The method of claim 8, wherein a cytolytic T cell response is induced.
  • 12. The method of claim 8, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 13. The method of claim 12, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 14. A method of treating non-small cell lung cancer comprising immunizing against PSMA and at least one other tumor-associated antigen.
  • 15. The method of claim 14, wherein the at least one other tumor-associated antigen is selected from the group consisting of MAGE-3, SSX-2 and NY-ESO-1.
  • 16. The method of claim 14, wherein the at least one other tumor-associated antigen comprises NY-ESO-1 and SSX-2.
  • 17. The method of claim 14, wherein the at least one other tumor-associated antigen comprises MAGE-3, SSX-2 and NY-ESO-1.
  • 18. The method of claim 14, wherein a cytolytic T cell response is induced.
  • 19. The method of claim 14, further comprising immunizing against at least one other antigen associated with tumor neovasculature.
  • 20. The method of claim 19, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 21. A method of treating renal cell carcinoma comprising immunizing against PSMA and at least one other tumor-associated antigen.
  • 22. The method of claim 21, wherein the at least one other tumor-associated antigen is selected from the group consisting of PRAME and SSX-2.
  • 23. The method of claim 21, wherein the at least one other tumor-associated antigen comprises PRAME and SSX-2.
  • 24. The method of claim 21, wherein a cytolytic T cell response is induced.
  • 25. The method of claim 21, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 26. The method of claim 25, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 27. A method of treating melanoma comprising immunizing against at least one first tumor-associated antigen and at least one second tumor-associated antigen, wherein the at least one first tumor-associated antigen is selected from the group consisting of tyrosinase and melan-A.
  • 28. The method of claim 27, and wherein the at least one second tumor-associated antigen is selected from the group consisting of SSX-2, PRAME, a MAGE protein, and NY-ESO-1.
  • 29. The method of claim 27, wherein the at least one first tumor-associated antigen is Melan-A, and the at least one second tumor-associated antigen comprises SSX-2 and NY-ESO.
  • 30. The method of claim 27, wherein the at least one first tumor-associated antigen is Melan-A and tyrosinase, and the at least one second tumor-associated antigen comprises SSX-2.
  • 31. The method of claim 27, wherein a cytolytic T cell response is induced.
  • 32. The method of claim 27, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 33. The method of claim 32, wherein the antigen associated with tumor neovasculature is selected from the group consisting of PSMA, VEGFR2, and Tie-2.
  • 34. An immunogenic composition for the treatment of ovarian or colorectal cancer comprising, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens are PRAME and at least one other tumor-associated antigen.
  • 35. The composition of claim 34, wherein the at least one other tumor-associated antigen is selected from the group consisting of SSX-2, NY-ESO-1, and PSMA.
  • 36. The composition of claim 34, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 37. The composition of claim 36, wherein the antigen associated with tumor neovasculature is selected from the group consisting of PSMA, VEGFR2, and Tie-2.
  • 38. An immunogenic composition for the treatment of pancreatic cancer comprising, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens are PSMA and at least one other tumor-associated antigen.
  • 39. The composition of claim 38, wherein the at least one other tumor-associated antigen is selected from the group consisting of SSX-2 and NY-ESO-1.
  • 40. The composition of claim 38, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 41. The composition of claim 40, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 42. An immunogenic composition for the treatment of non-small cell lung cancer comprising, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens are PSMA and at least one other tumor-associated antigen.
  • 43. The composition of claim 42, wherein the at least one other tumor-associated antigen is selected from the group consisting of MAGE-3, SSX-2 and NY-ESO-1.
  • 44. The composition of claim 42, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 45. The composition of claim 44, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 46. An immunogenic composition for the treatment of renal cell carcinoma comprising, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens are PSMA and at least one other tumor-associated antigen.
  • 47. The composition of claim 46, wherein the at least one other tumor-associated antigen is selected from the group consisting of PRAME and SSX-2.
  • 48. The composition of claim 46, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 49. The composition of claim 48, wherein the antigen associated with tumor neovasculature is selected from the group consisting of VEGFR2 and Tie-2.
  • 50. An immunogenic composition for the treatment of melanoma comprising, individually or in combination, 1) whole antigens, 2) fragments of antigens, 3) epitope clusters derived from antigens, 4) epitopes derived from antigens, or 5) nucleic acids encoding any of 1 to 4; wherein the antigens are selected from each of a first group of antigens and a second group of antigens; wherein the first group of antigens consists of tyrosinase and melan-A.
  • 51. The composition of claim 50, wherein the second group of antigens consists of SSX-2, PRAME, a MAGE protein, and NY-ESO-1.
  • 52. The composition of claim 50, further comprising immunizing against at least one antigen associated with tumor neovasculature.
  • 53. The composition of claim 52, wherein the antigen associated with tumor neovasculature is selected from the group consisting of PSMA, VEGFR2, and Tie-2.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 60/479,554, filed on Jun. 17, 2003, entitled COMBINATIONS OF TUMOR-ASSOCIATED ANTIGENS IN COMPOSITIONS FOR VARIOUS TYPES OF CANCERS; the disclosure of which is incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
60479554 Jun 2003 US