Epitope sequences

Information

  • Patent Application
  • 20050221440
  • Publication Number
    20050221440
  • Date Filed
    February 25, 2005
    19 years ago
  • Date Published
    October 06, 2005
    19 years ago
Abstract
Disclosed herein are polypeptides, including epitopes, clusters, and antigens. Also disclosed are compositions that include said polypeptides and methods for their use.
Description
BACKGROUND OF THE INVENTION

1. Field of the Invention


The present invention generally relates to peptides, and nucleic acids encoding peptides, that are useful epitopes of target-associated antigens. More specifically, the invention relates to epitopes that have a high affinity for MHC class I and that are produced by target-specific proteasomes. The invention disclosed herein further relates to the identification of epitope cluster regions that are used to generate pharmaceutical compositions capable of inducing an immune response from a subject to whom the compositions have been administered.


2. Description of the Related Art


Neoplasia and the Immune System


The neoplastic disease state commonly known as cancer is thought to result generally from a single cell growing out of control. The uncontrolled growth state typically results from a multi-step process in which a series of cellular systems fail, resulting in the genesis of a neoplastic cell. The resulting neoplastic cell rapidly reproduces itself, forms one or more tumors, and eventually may cause the death of the host.


Because the progenitor of the neoplastic cell shares the host's genetic material, neoplastic cells are largely unassailed by the host's immune system. During immune surveillance, the process in which the host's immune system surveys and localizes foreign materials, a neoplastic cell will appear to the host's immune surveillance machinery as a “self” cell.


Viruses and the Immune System


In contrast to cancer cells, virus infection involves the expression of clearly non-self antigens. As a result, many virus infections are successfully dealt with by the immune system with minimal clinical sequela. Moreover, it has been possible to develop effective vaccines for many of those infections that do cause serious disease. A variety of vaccine approaches have been used successfully to combat various diseases. These approaches include subunit vaccines consisting of individual proteins produced through recombinant DNA technology. Notwithstanding these advances, the selection and effective administration of minimal epitopes for use as viral vaccines has remained problematic.


In addition to the difficulties involved in epitope selection stands the problem of viruses that have evolved the capability of evading a host's immune system. Many viruses, especially viruses that establish persistent infections, such as members of the herpes and pox virus families, produce immunomodulatory molecules that permit the virus to evade the host's immune system. The effects of these immunomodulatory molecules on antigen presentation may be overcome by the targeting of select epitopes for administration as immunogenic compositions. To better understand the interaction of neoplastic cells and virally infected cells with the host's immune system, a discussion of the system's components follows below.


The immune system functions to discriminate molecules endogenous to an organism (“self” molecules) from material exogenous or foreign to the organism (“non-self” molecules). The immune system has two types of adaptive responses to foreign bodies based on the components that mediate the response: a humoral response and a cell-mediated response. The humoral response is mediated by antibodies, while the cell-mediated response involves cells classified as lymphocytes. Recent anticancer and antiviral strategies have focused on mobilizing the host immune system as a means of anticancer or antiviral treatment or therapy.


The immune system functions in three phases to protect the host from foreign bodies: the cognitive phase, the activation phase, and the effector phase. In the cognitive phase, the immune system recognizes and signals the presence of a foreign antigen or invader in the body. The foreign antigen can be, for example, a cell surface marker from a neoplastic cell or a viral protein. Once the system is aware of an invading body, antigen specific cells of the immune system proliferate and differentiate in response to the invader-triggered signals. The last stage is the effector stage in which the effector cells of the immune system respond to and neutralize the detected invader.


An array of effector cells implements an immune response to an invader. One type of effector cell, the B cell, generates antibodies targeted against foreign antigens encountered by the host. In combination with the complement system, antibodies direct the destruction of cells or organisms bearing the targeted antigen. Another type of effector cell is the natural killer cell (NK cell), a type of lymphocyte having the capacity to spontaneously recognize and destroy a variety of virus infected cells as well as malignant cell types. The method used by NK cells to recognize target cells is poorly understood.


Another type of effector cell, the T cell, has members classified into three subcategories, each playing a different role in the immune response. Helper T cells secrete cytokines which stimulate the proliferation of other cells necessary for mounting an effective immune response, while suppressor T cells down-regulate the immune response. A third category of T cell, the cytotoxic T cell (CTL), is capable of directly lysing a targeted cell presenting a foreign antigen on its surface.


The Major Histocompatibility Complex and T Cell Target Recognition


T cells are antigen-specific immune cells that function in response to specific antigen signals. B lymphocytes and the antibodies they produce are also antigen-specific entities. However, unlike B lymphocytes, T cells do not respond to antigens in a free or soluble form. For a T cell to respond to an antigen, it requires the antigen to be processed to peptides which are then bound to a presenting structure encoded in the major histocompatibility complex (MHC). This requirement is called “MHC restriction” and it is the mechanism by which T cells differentiate “self” from “non-self” cells. If an antigen is not displayed by a recognizable MHC molecule, the T cell will not recognize and act on the antigen signal. T cells specific for a peptide bound to a recognizable MHC molecule bind to these MHC-peptide complexes and proceed to the next stages of the immune response.


There are two types of MHC, class I MHC and class II MHC. T Helper cells (CD4+) predominately interact with class II MHC proteins while cytolytic T cells (CD8+) predominately interact with class I MHC proteins. Both classes of MHC protein are transmembrane proteins with a majority of their structure on the external surface of the cell. Additionally, both classes of MHC proteins have a peptide binding cleft on their external portions. It is in this cleft that small fragments of proteins, endogenous or foreign, are bound and presented to the extracellular environment.


Cells called “professional antigen presenting cells” (pAPCs) display antigens to T cells using the MHC proteins but additionally express various co-stimulatory molecules depending on the particular state of differentiation/activation of the pAPC. When T cells, specific for the peptide bound to a recognizable MHC protein, bind to these MHC-peptide complexes on pAPCs, the specific co-stimulatory molecules that act upon the T cell direct the path of differentiation/activation taken by the T cell. That is, the co-stimulation molecules affect how the T cell will act on antigenic signals in future encounters as it proceeds to the next stages of the immune response.


As discussed above, neoplastic cells are largely ignored by the immune system. A great deal of effort is now being expended in an attempt to harness a host's immune system to aid in combating the presence of neoplastic cells in a host. One such area of research involves the formulation of anticancer vaccines.


Anticancer Vaccines


Among the various weapons available to an oncologist in the battle against cancer is the immune system of the patient. Work has been done in various attempts to cause the immune system to combat cancer or neoplastic diseases. Unfortunately, the results to date have been largely disappointing. One area of particular interest involves the generation and use of anticancer vaccines.


To generate a vaccine or other immunogenic composition, it is necessary to introduce to a subject an antigen or epitope against which an immune response may be mounted. Although neoplastic cells are derived from and therefore are substantially identical to normal cells on a genetic level, many neoplastic cells are known to present tumor-associated antigens (TuAAs). In theory, these antigens could be used by a subject's immune system to recognize these antigens and attack the neoplastic cells. In reality, however, neoplastic cells generally appear to be ignored by the host's immune system.


A number of different strategies have been developed in an attempt to generate vaccines with activity against neoplastic cells. These strategies include the use of tumor-associated antigens as immunogens. For example, U.S. Pat. No. 5,993,828, describes a method for producing an immune response against a particular subunit of the Urinary Tumor Associated Antigen by administering to a subject an effective dose of a composition comprising inactivated tumor cells having the Urinary Tumor Associated Antigen on the cell surface and at least one tumor associated antigen selected from the group consisting of GM-2, GD-2, Fetal Antigen and Melanoma Associated Antigen. Accordingly, this patent describes using whole, inactivated tumor cells as the immunogen in an anticancer vaccine.


Another strategy used with anticancer vaccines involves administering a composition containing isolated tumor antigens. In one approach, MAGE-A1 antigenic peptides were used as an immunogen. (See Chaux, P., et al., “Identification of Five MAGE-A1 Epitopes Recognized by Cytolytic T Lymphocytes Obtained by In Vitro Stimulation with Dendritic Cells Transduced with MAGE-A1,” J. Immunol., 163(5):2928-2936 (1999)). There have been several therapeutic trials using MAGE-A1 peptides for vaccination, although the effectiveness of the vaccination regimes was limited. The results of some of these trials are discussed in Vose, J. M., “Tumor Antigens Recognized by T Lymphocytes,” 10th European Cancer Conference, Day 2, Sep. 14, 1999.


In another example of tumor associated antigens used as vaccines, Scheinberg, et al. treated 12 chronic myelogenous leukemia (CML) patients already receiving interferon (IFN) or hydroxyurea with 5 injections of class I-associated bcr-abl peptides with a helper peptide plus the adjuvant QS-21. Scheinberg, D. A., et al., “BCR-ABL Breakpoint Derived Oncogene Fusion Peptide Vaccines Generate Specific Immune Responses in Patients with Chronic Myelogenous Leukemia (CML) [Abstract 1665], American Society of Clinical Oncology 35th Annual Meeting, Atlanta (1999). Proliferative and delayed type hypersensitivity (DTH) T cell responses indicative of T-helper activity were elicited, but no cytolytic killer T cell activity was observed within the fresh blood samples.


Additional examples of attempts to identify TuAAs for use as vaccines are seen in the recent work of Cebon, et al. and Scheibenbogen, et al. Cebon, et al. immunized patients with metastatic melanoma using intradermallly administered MART-126-35 peptide with IL-12 in increasing doses given either subcutaneously or intravenously. Of the first 15 patients, 1 complete remission, 1 partial remission, and 1 mixed response were noted. Immune assays for T cell generation included DTH, which was seen in patients with or without IL-12. Positive CTL assays were seen in patients with evidence of clinical benefit, but not in patients without tumor regression. Cebon, et al., “Phase I Studies of Immunization with Melan-A and IL-12 in HLA A2+Positive Patients with Stage III and IV Malignant Melanoma,” [Abstract 1671], American Society of Clinical Oncology 35th Annual Meeting, Atlanta (1999).


Scheibenbogen, et al. immunized 18 patients with 4 HLA class I restricted tyrosinase peptides, 16 with metastatic melanoma and 2 adjuvant patients. Scheibenbogen, et al., “Vaccination with Tyrosinase peptides and GM-CSF in Metastatic Melanoma: a Phase II Trial,” [Abstract 1680], American Society of Clinical Oncology 35th Annual Meeting, Atlanta (1999). Increased CTL activity was observed in 4/15 patients, 2 adjuvant patients, and 2 patients with evidence of tumor regression. As in the trial by Cebon, et al., patients with progressive disease did not show boosted immunity. In spite of the various efforts expended to date to generate efficacious anticancer vaccines, no such composition has yet been developed.


Antiviral Vaccines


Vaccine strategies to protect against viral diseases have had many successes. Perhaps the most notable of these is the progress that has been made against the disease small pox, which has been driven to extinction. The success of the polio vaccine is of a similar magnitude.


Viral vaccines can be grouped into three classifications: live attenuated virus vaccines, such as vaccinia for small pox, the Sabin poliovirus vaccine, and measles mumps and rubella; whole killed or inactivated virus vaccines, such as the Salk poliovirus vaccine, hepatitis A virus vaccine and the typical influenza virus vaccines; and subunit vaccines, such as hepatitis B. Due to their lack of a complete viral genome, subunit vaccines offer a greater degree of safety than those based on whole viruses.


The paradigm of a successful subunit vaccine is the recombinant hepatitis B vaccine based on the viruses envelope protein. Despite much academic interest in pushing the reductionist subunit concept beyond single proteins to individual epitopes, the efforts have yet to bear much fruit. Viral vaccine research has also concentrated on the induction of an antibody response although cellular responses also occur. However, many of the subunit formulations are particularly poor at generating a CTL response.


SUMMARY OF THE INVENTION

Previous methods of priming professional antigen presenting cells (pAPCs) to display target cell epitopes have relied simply on causing the pAPCs to express target-associated antigens (TAAs), or epitopes of those antigens which are thought to have a high affinity for MHC I molecules. However, the proteasomal processing of such antigens results in presentation of epitopes on the pAPC that do not correspond to the epitopes present on the target cells.


Using the knowledge that an effective cellular immune response requires that pAPCs present the same epitope that is presented by the target cells, the present invention provides epitopes that have a high affinity for MHC I, and that correspond to the processing specificity of the housekeeping proteasome, which is active in peripheral cells. These epitopes thus correspond to those presented on target cells. The use of such epitopes in vaccines can activate the cellular immune response to recognize the correctly processed TAA and can result in removal of target cells that present such epitopes. In some embodiments, the housekeeping epitopes provided herein can be used in combination with immune epitopes, generating a cellular immune response that is competent to attack target cells both before and after interferon induction. In other embodiments the epitopes are useful in the diagnosis and monitoring of the target-associated disease and in the generation of immunological reagents for such purposes.


In some aspects, the invention disclosed herein relates to the identification of epitope cluster regions that are used to generate pharmaceutical compositions capable of inducing an immune response from a subject to whom the compositions have been administered. One embodiment of the disclosed invention relates to an epitope cluster, the cluster being derived from an antigen associated with a target, the cluster including or encoding at least two sequences having a known or predicted affinity for an MHC receptor peptide binding cleft, wherein the cluster is an incomplete fragment of the antigen.


In one aspect of the invention, the target is a neoplastic cell.


In another aspect of the invention, the MHC receptor may be a class I HLA receptor.


In yet another aspect of the invention, the cluster includes or encodes a polypeptide having a length, wherein the length is at least 10 amino acids. Advantageously, the length of the polypeptide may be less than about 75 amino acids.


In still another aspect of the invention, there is provided an antigen having a length, wherein the cluster consists of or encodes a polypeptide having a length, wherein the length of the polypeptide is less than about 80% of the length of the antigen. Preferably, the length of the polypeptide is less than about 50% of the length of the antigen. Most preferably, the length of the polypeptide is less than about 20% of the length of the antigen.


Embodiments of the invention particularly relate to epitope clusters identified in the tumor-associated antigen PRAME (SEQ ID NO: 77). One embodiment of the invention relates to an isolated nucleic acid containing a reading frame with a first sequence encoding one or more segments of PRAME, wherein the whole antigen is not encoded, wherein each segment contains an epitope cluster, and wherein each cluster contains at least two amino acid sequences with a known or predicted affinity for a same MHC receptor peptide binding cleft. In various aspects of the invention the epitope cluster can be amino acids 18-59, 33-47, 71-81, 78-115, 99-108, 126-135, 222-238, 224-246, 290-303, 305-324, 343-363, 364-447, 394-409, 422-443, or 459-487 of PRAME.


In other aspects, the segments can consist of an epitope cluster; the first sequence can be a fragment of PRAME; the fragment can consist of a polypeptide having a length, wherein the length of the polypeptide is less than about 90%, 80%, 60%, 50%, 25%, or 10% of the length of PRAME. The fragment can consist essentially of an amino acid sequence beginning at amino acid 18, 33, 71, 78, 99, 126, 222, 224, 290, 305, 343, 364, 394, 422, or 459 of PRAME and ending at amino acid 47, 59, 81, 108, 115, 135, 238, 246, 303, 324, 363, 409, 443, 447, or 487 of PRAME. In some embodiments, the encoded fragment consists essentially of amino acids 18-47, 18-59, 18-81, 18-108, 18-115, 18-135, 18-238, 18-246, 18-303, 18-324, 18-363, 18-409, 18-443, 18-447, 18-487, 33-47, 33-59, 33-81, 33-108, 33-115, 33-135, 33-238, 33-246, 33-303, 33-324, 33-363, 33-409, 33-443, 33-447, 33-487, 71-81, 71-108, 71-115, 71-135, 71-238, 71-246, 71-303, 71-324, 71-363, 71-409, 71-443, 71-447, 71-487, 78-108, 78-115, 78-135, 78-238, 78-246, 78-303, 78-324, 78-363, 78-409, 78-443, 78-447, 78-487, 99-108, 99-115, 99-135, 99-238, 99-246, 99-303, 99-324, 99-363, 99-409, 99-443, 99-447, 99-487, 126-135, 126-238, 126-246, 126-303, 126-324, 126-363, 126-409, 126-443, 126-447, 126-487, 222-238, 222-246, 222-303, 222-324, 222-363, 222-409, 222-443, 222-447, 222-487, 224-238, 224-246, 224-303, 224-324, 224-363, 224-409, 224-443, 224-447, 224-487, 290-303, 290-324, 290-363, 290-409, 290-443, 290-447, 290-487, 305-324, 305-363, 305-409, 305-443, 305-447, 305-487, 343-363, 343-409, 343-443, 343-447, 343-487, 364-409, 364-443, 364-447, 364-487, 394-409, 394-443, 394-447, 394-487, 422-443, 422-447, 422-487, 459-487, 18-487, 224-487 of PRAME.


The first sequence can be a fragment of SSX-2. The fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 90%, 80%, 60%, 50%, 25%, or 10% of the length of SSX-2.


Further embodiments of the invention include a second sequence encoding essentially a housekeeping epitope. In one aspect of this embodiment the first and second sequences constitute a single reading frame. In aspects of the invention the reading frame is operably linked to a promoter. Other embodiments of the invention include the polypeptides encoded by the nucleic acid embodiments of the invention and immunogenic compositions containing the nucleic acids or polypeptides of the invention.


Other embodiments of the invention relate to isolated epitopes, and antigens or polypeptides that comprise the epitopes. Preferred embodiments include an epitope or antigen having the sequence as disclosed in Table 1. Other embodiments can include an epitope cluster comprising a polypeptide from Table 1. Further, embodiments include a polypeptide having substantial similarity to the already mentioned epitopes, polypeptides, antigens, or clusters. Other preferred embodiments include a polypeptide having functional similarity to any of the above. Still further embodiments relate to a nucleic acid encoding the polypeptide of any of the epitopes, clusters, antigens, and polypeptides from Table 1 and mentioned herein. For purposes of the following summary, discussions of other embodiments of the invention, when making reference to “the epitope,” or “the epitopes” may refer without limitation to all of the foregoing forms of the epitope.


The epitope can be immunologically active. The polypeptide comprising the epitope can be less than about 30 amino acids in length, more preferably, the polypeptide is 8 to 10 amino acids in length, for example. Substantial or functional similarity can include addition of at least one amino acid, for example, and the at least one additional amino acid can be at an N-terminus of the polypeptide. The substantial or functional similarity can include a substitution of at least one amino acid.


The epitope, cluster, or polypeptide comprising the same can have affinity to an HLA-A2 molecule. The affinity can be determined by an assay of binding, by an assay of restriction of epitope recognition, by a prediction algorithm, and the like. The epitope, cluster, or polypeptide comprising the same can have affinity to an HLA-B7, HLA-B51 molecule, and the like.


In preferred embodiments the polypeptide can be a housekeeping epitope. The epitope or polypeptide can correspond to an epitope displayed on a tumor cell, to an epitope displayed on a neovasculature cell, and the like. The epitope or polypeptide can be an immune epitope. The epitope, cluster and/or polypeptide can be a nucleic acid.


Other embodiments relate to pharmaceutical compositions comprising the polypeptides, including an epitope from Table 1, a cluster, or a polypeptide comprising the same, and a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like. The adjuvant can be a polynucleotide. The polynucleotide can include a dinucleotide, which can be CpG, for example. The adjuvant can be encoded by a polynucleotide. The adjuvant can be a cytokine and the cytokine can be, for example, GM-CSF.


The pharmaceutical compositions can further include a professional antigen-presenting cell (pAPC). The pAPC can be a dendritic cell, for example. The pharmaceutical composition can further include a second epitope. The second epitope can be a polypeptide, a nucleic acid, a housekeeping epitope, an immune epitope, and the like.


Still further embodiments relate to pharmaceutical compositions that include any of the nucleic acids discussed herein, including those that encode polypeptides that comprise epitopes or antigens from Table 1. Such compositions can include a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like.


Other embodiments relate to recombinant constructs that include such a nucleic acid as described herein, including those that encode polypeptides that comprise epitopes or antigens from Table 1. The constructs can further include a plasmid, a viral vector, an artificial chromosome, and the like. The construct can further include a sequence encoding at least one feature, such as for example, a second epitope, an IRES, an ISS, an NIS, a ubiquitin, and the like.


Further embodiments relate to purified antibodies that specifically bind to at least one of the epitopes in Table 1. Other embodiments relate to purified antibodies that specifically bind to a peptide-MHC protein complex comprising an epitope disclosed in Table 1 or any other suitable epitope. The antibody from any embodiment can be a monoclonal antibody or a polyclonal antibody.


Still other embodiments relate to multimeric MHC-peptide complexes that include an epitope, such as, for example, an epitope disclosed in Table 1. Also, contemplated are antibodies specific for the complexes.


Embodiments relate to isolated T cells expressing a T cell receptor specific for an MHC-peptide complex. The complex can include an epitope, such as, for example, an epitope disclosed in Table 1. The T cell can be produced by an in vitro immunization and can be isolated from an immunized animal. Embodiments relate to T cell clones, including cloned T cells, such as those discussed above. Embodiments also relate to polyclonal population of T cells. Such populations can include a T cell, as described above, for example.


Still further embodiments relate to pharmaceutical compositions that include a T cell, such as those described above, for example, and a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like.


Embodiments of the invention relate to isolated protein molecules comprising the binding domain of a T cell receptor specific for an MHC-peptide complex. The complex can include an epitope as disclosed in Table 1. The protein can be multivalent. Other embodiments relate to isolated nucleic acids encoding such proteins. Still further embodiments relate to recombinant constructs that include such nucleic acids.


Other embodiments of the invention relate to host cells expressing a recombinant construct as described herein, including constructs encoding an epitope, cluster or polypeptide comprising the same, disclosed in Table 1, for example. The host cell can be a dendritic cell, macrophage, tumor cell, tumor-derived cell, a bacterium, fungus, protozoan, and the like. Embodiments also relate to pharmaceutical compositions that include a host cell, such as those discussed herein, and a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like.


Still other embodiments relate to vaccines or immunotherapeutic compositions that include at least one component, such as, for example, an epitope disclosed in Table 1 or otherwise described herein; a cluster that includes such an epitope, an antigen or polypeptide that includes such an epitope; a composition as described above and herein; a construct as described above and herein, a T cell, or a host cell as described above and herein.


Further embodiments relate to methods of treating an animal. The methods can include administering to an animal a pharmaceutical composition, such as, a vaccine or immunotherapeutic composition, including those disclosed above and herein. The administering step can include a mode of delivery, such as, for example, transdermal, intranodal, perinodal, oral, intravenous, intradermal, intramuscular, intraperitoneal, mucosal, aerosol inhalation, instillation, and the like. The method can further include a step of assaying to determine a characteristic indicative of a state of a target cell or target cells. The method can include a first assaying step and a second assaying step, wherein the first assaying step precedes the administering step, and wherein the second assaying step follows the administering step. The method can further include a step of comparing the characteristic determined in the first assaying step with the characteristic determined in the second assaying step to obtain a result. The result can be for example, evidence of an immune response, a diminution in number of target cells, a loss of mass or size of a tumor comprising target cells, a decrease in number or concentration of an intracellular parasite infecting target cells, and the like.


Embodiments relate to methods of evaluating immunogenicity of a vaccine or immunotherapeutic composition. The methods can include administering to an animal a vaccine or immunotherapeutic, such as those described above and elsewhere herein, and evaluating immunogenicity based on a characteristic of the animal. The animal can be HLA-transgenic.


Other embodiments relate to methods of evaluating immunogenicity that include in vitro stimulation of a T cell with the vaccine or immunotherapeutic composition, such as those described above and elsewhere herein, and evaluating immunogenicity based on a characteristic of the T cell. The stimulation can be a primary stimulation.


Still further embodiments relate to methods of making a passive/adoptive immunotherapeutic. The methods can include combining a T cell or a host cell, such as those described above and elsewhere herein, with a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like.


Other embodiments relate to methods of determining specific T cell frequency, and can include the step of contacting T cells with a MHC-peptide complex comprising an epitope disclosed in Table 1, or a complex comprising a cluster or antigen comprising such an epitope. The contacting step can include at least one feature, such as, for example, immunization, restimulation, detection, enumeration, and the like. The method can further include ELISPOT analysis, limitirig dilution analysis, flow cytometry, in situ hybridization, the polymerase chain reaction, any combination thereof, and the like.


Embodiments relate to methods of evaluating immunologic response. The methods can include the above-described methods of determining specific T cell frequency carried out prior to and subsequent to an immunization step.


Other embodiments relate to methods of evaluating immunologic response. The methods can include determining frequency, cytokine production, or cytolytic activity of T cells, prior to and subsequent to a step of stimulation with MHC-peptide complexes comprising an epitope, such as, for example an epitope from Table 1, a cluster or a polypeptide comprising such an epitope.


Further embodiments relate to methods of diagnosing a disease. The methods can include contacting a subject tissue with at least one component, including, for example, a T cell, a host cell, an antibody, a protein, including those described above and elsewhere herein; and diagnosing the disease based on a characteristic of the tissue or of the component. The contacting step can take place in vivo or in vitro, for example.


Still other embodiments relate to methods of making a vaccine. The methods can include combining at least one component, an epitope, a composition, a construct, a T cell, a host cell; including any of those described above and elsewhere herein, with a pharmaceutically acceptable adjuvant, carrier, diluent, excipient, and the like.


Embodiments relate to computer readable media having recorded thereon the sequence of any one of SEQ ID NOS: 1-602, in a machine having a hardware or software that calculates the physical, biochemical, immunologic, molecular genetic properties of a molecule embodying said sequence, and the like.


Still other embodiments relate to methods of treating an animal. The methods can include combining the method of treating an animal that includes administering to the animal a vaccine or immunotherapeutic composition, such as described above and elsewhere herein, combined with at least one mode of treatment, including, for example, radiation therapy, chemotherapy, biochemotherapy, surgery, and the like.


Further embodiments relate to isolated polypeptides that include an epitope cluster. In preferred embodiments the cluster can be from a target-associated antigen having the sequence as disclosed in any one of Tables 25-44, wherein the amino acid sequence includes not more than about 80% of the amino acid sequence of the antigen.


Other embodiments relate to vaccines or immunotherapeutic products that include an isolated peptide as described above and elsewhere herein. Still other embodiments relate to isolated polynucleotides encoding a polypeptide as described above and elsewhere herein. Other embodiments relate vaccines or immunotherapeutic products that include these polynucleotides. The polynucleotide can be DNA, RNA, and the like.


Still further embodiments relate to kits comprising a delivery device and any of the embodiments mentioned above and elsewhere herein. The delivery device can be a catheter, a syringe, an internal or external pump, a reservoir, an inhaler, microinjector, a patch, and any other like device suitable for any route of delivery. As mentioned, the kit, in addition to the delivery device also includes any of the embodiments disclosed herein. For example, without limitations, the kit can include an isolated epitope, a polypeptide, a cluster, a nucleic acid, an antigen, a pharmaceutical composition that includes any of the foregoing, an antibody, a T cell, a T cell receptor, an epitope-MHC complex, a vaccine, an immunotherapeutic, and the like. The kit can also include items such as detailed instructions for use and any other like item.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a sequence alignment of NY-ESO-1 and several similar protein sequences.



FIG. 2 graphically represents a plasmid vaccine backbone useful for delivering nucleic acid-encoded epitopes.



FIGS. 3A and 3B are FACS profiles showing results of HLA-A2 binding assays for tyrosinase207-215 and tyrosinase208-216.



FIG. 3C shows cytolytic activity against a tyrosinase epitope by human CTL induced by in vitro immunization.



FIG. 4 is a T=120 min. time point mass spectrum of the fragments produced by proteasomal cleavage of SSX-231-68.



FIG. 5 shows a binding curve for HLA-A2:SSX-241-49 with controls.



FIG. 6 shows specific lysis of SSX-241-49-pulsed targets by CTL from SSX-241-49-immunized HLA-A2 transgenic mice.



FIG. 7A, B, and C show results of N-terminal pool sequencing of a T=60 min. time point aliquot of the PSMA163-192 proteasomal digest.



FIG. 8 shows binding curves for HLA-A2:PSMA168-177 and HLA-A2:PSMA288-297 with controls.



FIG. 9 shows results of N-terminal pool sequencing of a T=60 min. time point aliquot of the PSMA281-310 proteasomal digest.



FIG. 10 shows binding curves for HLA-A2:PSMA461-469, HLA-A2:PSMA460-469, and HLA-A2:PSMA663-671, with controls.



FIG. 11 shows the results of a γ-IFN-based ELISPOT assay detecting PSMA463-471-reactive HLA-A1+CD8+ T cells.



FIG. 12 shows blocking of reactivity of the T cells used in FIG. 10 by anti-HLA-A1 mAb, demonstrating HLA-A1-restricted recognition.



FIG. 13 shows a binding curve for HLA-A2:PSMA663-671, with controls.



FIG. 14 shows a binding curve for HLA-A2:PSMA662-671, with controls.



FIG. 15. Comparison of anti-peptide CTL responses following immunization with various doses of DNA by different routes of injection.



FIG. 16. Growth of transplanted gp33 expressing tumor in mice immunized by i.ln. injection of gp33 epitope-expressing, or control, plasmid.



FIG. 17. Amount of plasmid DNA detected by real-time PCR in injected or draining lymph nodes at various times after i.ln. of i.m. injection, respectively.



FIG. 18 depicts the sequence of Melan-A, showing clustering of class I HLA epitopes.



FIG. 19 depicts the sequence of SSX-2, showing clustering of class I HLA epitopes.



FIG. 20 depicts the sequence of NY-ESO, showing clustering of class I HLA epitopes.




DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT

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.


IMMUNE PROTEASOME—a proteasome normally active in pAPCs; the immune proteasome is also active in some peripheral cells in infected tissues.


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.


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.


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


TUMOR-ASSOCIATED ANTIGENS (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. Examples include inter alia, F(ab), single chain Fv, and Ig variable region-phage coat protein fusions.


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.


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 modest 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 do not meet 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. In testing for functional similarity of immunogenicity one would generally immunize with the “altered” antigen and test the ability of the elicited response (Ab, CTL, cytokine production, etc.) 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 are among the embodiments of the present invention.


Epitope Clusters


Embodiments of the invention disclosed herein provide epitope cluster regions (ECRs) for use in vaccines and in vaccine design and epitope discovery. Specifically, embodiments of the invention relate to identifying epitope clusters for use in generating immunologically active compositions directed against target cell populations, and for use in the discovery of discrete housekeeping epitopes and immune epitopes. In many cases, numerous putative class I MHC epitopes may exist in a single target-associated antigen (TAA). Such putative epitopes are often found in clusters (ECRs), MHC epitopes distributed at a relatively high density within certain regions in the amino acid sequence of the parent TAA. Since these ECRs include multiple putative epitopes with potential useful biological activity in inducing an immune response, they represent an excellent material for in vitro or in vivo analysis to identify particularly useful epitopes for vaccine design. And, since the epitope clusters can themselves be processed inside a cell to produce active MHC epitopes, the clusters can be used directly in vaccines, with one or more putative epitopes in the cluster actually being processed into an active MHC epitope.


The use of ECRs in vaccines offers important technological advances in the manufacture of recombinant vaccines, and further offers crucial advantages in safety over existing nucleic acid vaccines that encode whole protein sequences. Recombinant vaccines generally rely on expensive and technically challenging production of whole proteins in microbial fermentors. ECRs offer the option of using chemically synthesized polypeptides, greatly simplifying development and manufacture, and obviating a variety of safety concerns. Similarly, the ability to use nucleic acid sequences encoding ECRs, which are typically relatively short regions of an entire sequence, allows the use of synthetic oligonucleotide chemistry processes in the development and manipulation of nucleic acid based vaccines, rather than the more expensive, time consuming, and potentially difficult molecular biology procedures involved with using whole gene sequences.


Since an ECR is encoded by a nucleic acid sequence that is relatively short compared to that which encodes the whole protein from which the ECR is found, this can greatly improve the safety of nucleic acid vaccines. An important issue in the field of nucleic acid vaccines is the fact that the extent of sequence homology of the vaccine with sequences in the animal to which it is administered determines the probability of integration of the vaccine sequence into the genome of the animal. A fundamental safety concern of nucleic acid vaccines is their potential to integrate into genomic sequences, which can cause deregulation of gene expression and tumor transformation. The Food and Drug Administration has advised that nucleic acid and recombinant vaccines should contain as little sequence homology with human sequences as possible. In the case of vaccines delivering tumor-associated antigens, it is inevitable that the vaccines contain nucleic acid sequences that are homologous to those which encode proteins that are expressed in the tumor cells of patients. It is, however, highly desirable to limit the extent of those sequences to that which is minimally essential to facilitate the expression of epitopes for inducing therapeutic immune responses. The use of ECRs thus offers the dual benefit of providing a minimal region of homology, while incorporating multiple epitopes that have potential therapeutic value.


ECRs are Processed into MHC-Binding Epitopes in pAPCs


The immune system constantly surveys the body for the presence of foreign antigens, in part through the activity of pAPCs. The pAPCs endocytose matter found in the extracellular milieu, process that matter from a polypeptide form into shorter oligopeptides of about 3 to 23 amino acids in length, and display some of the resulting peptides to T cells via the MHC complex of the pAPCs. For example, a tumor cell upon lysis releases its cellular contents, including various proteins, into the extracellular milieu. Those released proteins can be endocytosed by pAPCs and processed into discrete peptides that are then displayed on the surface of the pAPCs via the MHC. By this mechanism, it is not the entire target protein that is presented on the surface of the pAPCs, but rather only one or more discrete fragments of that protein that are presented as MHC-binding epitopes. If a presented epitope is recognized by a T cell, that T cell is activated and an immune response results.


Similarly, the scavenger receptors on pAPC can take-up naked nucleic acid sequences or recombinant organisms containing target nucleic acid sequences. Uptake of the nucleic acid sequences into the pAPC subsequently results in the expression of the encoded products. As above, when an ECR can be processed into one or more useful epitopes, these products can be presented as MHC epitopes for recognition by T cells.


MHC-binding epitopes are often distributed unevenly throughout a protein sequence in clusters. Embodiments of the invention are directed to identifying epitope cluster regions (ECRs) in a particular region of a target protein. Candidate ECRs are likely to be natural substrates for various proteolytic enzymes and are likely to be processed into one or more epitopes for MHC display on the surface of an pAPC. In contrast to more traditional vaccines that deliver whole proteins or biological agents, ECRs can be administered as vaccines, resulting in a high probability that at least one epitope will be presented on MHC without requiring the use of a full length sequence.


The Use of ECRs in Identifying Discrete MHC-Binding Epitopes


Identifying putative MHC epitopes for use in vaccines often includes the use of available predictive algorithms that analyze the sequences of proteins or genes to predict binding affinity of peptide fragments for MHC. These algorithms rank putative epitopes according to predicted affinity or other characteristics associated with MHC binding. Exemplary algorithms for this kind of analysis include the Rammensee and NIH (Parker) algorithms. However, identifying epitopes that are naturally present on the surface of cells from among putative epitopes predicted using these algorithms has proven to be a difficult and laborious process. The use of ECRs in an epitope identification process can enormously simplify the task of identifying discrete MHC binding epitopes.


In a preferred embodiment, ECR polypeptides are synthesized on an automated peptide synthesizer and these ECRs are then subjected to in vitro digests using proteolytic enzymes involved in processing proteins for presentation of the epitopes. Mass spectrometry and/or analytical HPLC are then used to identify the digest products and in vitro MHC binding studies are used to assess the ability of these products to actually bind to MHC. Once epitopes contained in ECRs have been shown to bind MHC, they can be incorporated into vaccines or used as diagnostics, either as discrete epitopes or in the context of ECRs.


The use of an ECR (which because of its relatively short sequence can be produced through chemical synthesis) in this preferred embodiment is a significant improvement over what otherwise would require the use of whole protein. This is because whole proteins have to be produced using recombinant expression vector systems and/or complex purification procedures. The simplicity of using chemically synthesized ECRs enables the analysis and identification of large numbers of epitopes, while greatly reducing the time and expense of the process as compared to other currently used methods. The use of a defined ECR also greatly simplifies mass spectrum analysis of the digest, since the products of an ECR digest are a small fraction of the digest products of a whole protein.


In another embodiment, nucleic acid sequences encoding ECRs are used to express the polypeptides in cells or cell lines to assess which epitopes are presented on the surface. A variety of means can be used to detect the epitope on the surface. Preferred embodiments involve the lysis of the cells and affinity purification of the MHC, and subsequent elution and analysis of peptides from the MHC; or elution of epitopes from intact cells; (Falk, K. et al. Nature 351:290, 1991, and U.S. Pat. No. 5,989,565, respectively, both of which references are incorporated herein by reference in their entirety). A sensitive method for analyzing peptides eluted in this way from the MHC employs capillary or nanocapillary HPLC ESI mass spectrometry and on-line sequencing.


Target-Associated Antizens that Contain ECRs


TAAs from which ECRs may be defined include those from TuAAs, including oncofetal, cancer-testis, deregulated genes, fusion genes from errant translocations, differentiation antigens, embryonic antigens, cell cycle proteins, mutated tumor suppressor genes, and overexpressed gene products, including oncogenes. In addition, ECRs may be derived from virus gene products, particularly those associated with viruses that cause chronic diseases or are oncogenic, such as the herpes viruses, human papilloma viruses, human immunodeficiency virus, and human T cell leukemia virus. Also ECRs may be derived from gene products of parasitic organisms, such as Trypanosoma, Leishmania, and other intracellular or parasitic organisms.


Some of these TuAA include α-fetoprotein, carcinoembryonic antigen (CEA), esophageal cancer derived NY-ESO-1, and SSX genes, SCP-1, PRAME, MART-1/MelanA (MART-1), gp100 (Pmel 17), tyrosinase, TRP-1, TRP-2, MAGE-1, MAGE-2, MAGE-3, BAGE, GAGE-1, GAGE-2, p15; overexpressed oncogenes and mutated tumor-suppressor genes such as p53, Ras, HER-2/neu; unique tumor antigens resulting from chromosomal translocations such as BCR-ABL, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR1 and viral antigens, EBNA1, EBNA2, HPV-E6, -E7; prostate specific antigen (PSA), prostate stem cell antigen (PSCA), MAAT-1, GP-100, TSP-180, MAGE-4, MAGE-5, MAGE-6, RAGE, p185erbB-2, p185erbB-3, c-met, nm-23H1, TAG-72, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, β-Catenin, CDK4, Mum-1, p15, and p16.


Numerous other TAAs are also contemplated for both pathogens and tumors. In terms of TuAAs, a variety of methods are available and well known in the art to identify genes and gene products that are differentially expressed in neoplastic cells as compared to normal cells. Examples of these techniques include differential hybridization, including the use of microarrays; subtractive hybridization cloning; differential display, either at the level of mRNA or protein expression; EST sequencing; and SAGE (sequential analysis of gene expression). These nucleic acid techniques have been reviewed by Carulli, J. P. et al., J. Cellular Biochem Suppl. 30/31:286-296, 1998 (hereby incorporated by reference). Differential display of proteins involves, for example, comparison of two-dimensional poly-acrylamide gel electrophoresis of cell lysates from tumor and normal tissue, location of protein spots unique or overexpressed in the tumor, recovery of the protein from the gel, and identification of the protein using traditional biochemical- or mass spectrometry-based sequencing. An additional technique for identification of TAAs is the Serex technique, discussed in Türeci, Ö., Sahin, U., and Pfreundschuh, M., “Serological analysis of human tumor antigens: molecular definition and implications”, Molecular Medicine Today, 3:342, 1997, and hereby incorporated by reference.


Use of these and other methods provides one of skill in the art the techniques necessary to identify genes and gene products contained within a target cell that may be used as potential candidate proteins for generating the epitopes of the invention disclosed. However, it is not necessary, in practicing the invention, to identify a novel TuAA or TAA. Rather, embodiments of the invention make it possible to identify ECRs from any relevant protein sequence, whether the sequence is already known or is new. Protein Sequence Analysis to Identify Epitope Clusters


In preferred embodiments of the invention, identification of ECRs involves two main steps: (1) identifying good putative epitopes; and (2) defining the limits of any clusters in which these putative epitopes are located. There are various preferred embodiments of each of these two steps, and a selected embodiment for the first step can be freely combined with a selected embodiment for the second step. The methods and embodiments that are disclosed herein for each of these steps are merely exemplary, and are not intended to limit the scope of the invention in any way. Persons of skill in the art will appreciate the specific tools that can be applied to the analysis of a specific TAA, and such analysis can be conducted in numerous ways in accordance with the invention.


Preferred embodiments for identifying good putative epitopes include the use of any available predictive algorithm that analyzes the sequences of proteins or genes to predict binding affinity of peptide fragments for MHC, or to rank putative epitopes according to predicted affinity or other characteristics associated with MHC binding. As described above, available exemplary algorithms for this kind of analysis include the Rammensee and NIH (Parker) algorithms. Likewise, good putative epitopes can be identified by direct or indirect assays of MHC binding. To choose “good” putative epitopes, it is necessary to set a cutoff point in terms of the score reported by the prediction software or in terms of the assayed binding affinity. In some embodiments, such a cutoff is absolute. For example, the cutoff can be based on the measured or predicted half time of dissociation between an epitope and a selected MHC allele. In such cases, embodiments of the cutoff can be any half time of dissociation longer than, for example, 0.5 minutes; in a preferred embodiment longer than 2.5 minutes; in a more preferred embodiment longer than 5 minutes; and in a highly stringent embodiment can be longer than 10, or 20, or 25 minutes. In these embodiments, the good putative epitopes are those that are predicted or identified to have good MHC binding characteristics, defined as being on the desirable side of the designated cutoff point. Likewise, the cutoff can be based on the measured or predicted binding affinity between an epitope and a selected MHC allele. Additionally, the absolute cutoff can be simply a selected number of putative epitopes.


In other embodiments, the cutoff is relative. For example, a selected percentage of the total number of putative epitopes can be used to establish the cutoff for defining a candidate sequence as a good putative epitope. Again the properties for ranking the epitopes are derived from measured or predicted MHC binding; the property used for such a determination can be any that is relevant to or indicative of binding. In preferred embodiments, identification of good putative epitopes can combine multiple methods of ranking candidate sequences. In such embodiments, the good epitopes are typically those that either represent a consensus of the good epitopes based on different methods and parameters, or that are particularly highly ranked by at least one of the methods.


When several good putative epitopes have been identified, their positions relative to each other can be analyzed to determine the optimal clusters for use in vaccines or in vaccine design. This analysis is based on the density of a selected epitope characteristic within the sequence of the TAA. The regions with the highest density of the characteristic, or with a density above a certain selected cutoff, are designated as ECRs. Various embodiments of the invention employ different characteristics for the density analysis. For example, one preferred characteristic is simply the presence of any good putative epitope (as defined by any appropriate method). In this embodiment, all putative epitopes above the cutoff are treated equally in the density analysis, and the best clusters are those with the highest density of good putative epitopes per amino acid residue. In another embodiment, the preferred characteristic is based on the parameter(s) previously used to score or rank the putative epitopes. In this embodiment, a putative epitope with a score that is twice as high as another putative epitope is doubly weighted in the density analysis, relative to the other putative epitope. Still other embodiments take the score or rank into account, but on a diminished scale, such as, for example, by using the log or the square root of the score to give more weight to some putative epitopes than to others in the density analysis.


Depending on the length of the TAA to be analyzed, the number of possible candidate epitopes, the number of good putative epitopes, the variability of the scoring of the good putative epitopes, and other factors that become evident in any given analysis, the various embodiments of the invention can be used alone or in combination to identify those ECRs that are most useful for a given application. Iterative or parallel analyses employing multiple approaches can be beneficial in many cases. ECRs are tools for increased efficiency of identifying true MHC epitopes, and for efficient “packaging” of MHC epitopes into vaccines. Accordingly, any of the embodiments described herein, or other embodiments that are evident to those of skill in the art based on this disclosure, are useful in enhancing the efficiency of these efforts by using ECRs instead of using complete TAAs in vaccines and vaccine design.


Since many or most TAAs have regions with low density of predicted MHC epitopes, using ECRs provides a valuable methodology that avoids the inefficiencies of including regions of low epitope density in vaccines and in epitope identification protocols. Thus, useful ECRs can also be defined as any portion of a TAA that is not the whole TAA, wherein the portion has a higher density of putative epitopes than the whole TAA, or than any regions of the TAA that have a particularly low density of putative epitopes. In this aspect of the invention, therefore, an ECR can be any fragment of a TAA with elevated epitope density. In some embodiments, an ECR can include a region up to about 80% of the length of the TAA. In a preferred embodiment, an ECR can include a region up to about 50% of the length of the TAA. In a more preferred embodiment, an ECR can include a region up to about 30% of the length of the TAA. And in a most preferred embodiment, an ECR can include a region of between 5 and 15% of the length of the TAA.


In another aspect of the invention, the ECR can be defined in terms of its absolute length. Accordingly, by this definition, the minimal cluster for 9-mer epitopes includes 10 amino acid residues and has two overlapping 9-mers with 8 amino acids in common. In a preferred embodiment, the cluster is between about 15 and 75 amino acids in length. In a more preferred embodiment, the cluster is between about 20 and 60 amino acids in length. In a most preferred embodiment, the cluster is between about 30 and 40 amino acids in length.


In practice, as described above, ECR identification can employ a simple density function such as the number of epitopes divided by the number of amino acids spanned by the those epitopes. It is not necessarily required that the epitopes overlap, but the value for a single epitope is not significant. If only a single value for a percentage cutoff is used and an absolute cutoff in the epitope prediction is not used, it is possible to set a single threshold at this step to define a cluster. However, using both an absolute cutoff and carrying out the first step using different percentage cutoffs, can produce variations in the global density of candidate epitopes. Such variations can require further accounting or manipulation. For example, an overlap of 2 epitopes is more significant if only 3 candidate epitopes were considered, than if 30 candidates were considered for any particular length protein. To take this feature into consideration, the weight given to a particular cluster can further be divided by the fraction of possible peptides actually being considered, in order to increase the significance of the calculation. This scales the result to the average density of predicted epitopes in the parent protein.


Similarly, some embodiments base the scoring of good putative epitopes on the average number of peptides considered per amino acid in the protein. The resulting ratio represents the factor by which the density of predicted epitopes in the putative cluster differs from the average density in the protein. Accordingly, an ECR is defined in one embodiment as any region containing two or more predicted epitopes for which this ratio exceeds 2, that is, any region with twice the average density of epitopes. In other embodiments, the region is defined as an ECR if the ratio exceeds 1.5, 3, 4, or 5, or more.


Considering the average number of peptides per amino acid in a target protein to calculate the presence of an ECR highlights densely populated ECRs without regard to the score/affinity of the individual constituents. This is most appropriate for use of score-based cutoffs. However, an ECR with only a small number of highly ranked candidates can be of more biological significance than a cluster with several densely packed but lower ranking candidates, particularly if only a small percentage of the total number of candidate peptides were designated as good putative epitopes. Thus in some embodiments it is appropriate to take into consideration the scores of the individual peptides. This is most readily accomplished by substituting the sum of the scores of the peptides in the putative cluster for the number of peptides in the putative cluster in the calculation described above.


This sum of scores method is more sensitive to sparsely populated clusters containing high scoring epitopes. Because the wide range of scores (i.e. half times of dissociation) produced by the BIMAS-NIH/Parker algorithm can lead to a single high scoring peptide dwarfing the contribution of other potential epitopes, the log of the score rather than the score itself is preferably used in this procedure.


Various other calculations can be devised under one or another condition. Generally speaking, the epitope density function is constructed so that it is proportional to the number of predicted epitopes, their scores, their ranks, and the like, within the putative cluster, and inversely proportional to the number of amino acids or fraction of protein contained within that putative cluster. Alternatively, the function can be evaluated for a window of a selected number of contiguous amino acids. In either case the function is also evaluated for all predicted epitopes in the whole protein. If the ratio of values for the putative cluster (or window) and the whole protein is greater than, for example, 1.5, 2, 3, 4, 5, or more, an ECR is defined.


Analysis of Target Gene Products For MHC Binding


Once a TAA has been identified, the protein sequence can be used to identify putative epitopes with known or predicted affinity to the MHC peptide binding cleft. Tests of peptide fragments can be conducted in vitro, or using the sequence can be computer analyzed to determine MHC receptor binding of the peptide fragments. In one embodiment of the invention, peptide fragments based on the amino acid sequence of the target protein are analyzed for their predicted ability to bind to the MHC peptide binding cleft. Examples of suitable computer algorithms for this purpose include that found at the world wide web page of Hans-Georg Rammensee, Jutta Bachmann, Niels Emmerich, Stefan Stevanovic: SYFPEITHI: An Internet Database for MHC Ligands and Peptide Motifs (access via http://134.2.96.221/scripts/hlaserver.dll/EpPredict.htm). Results obtained from this method are discussed in Rammensee, et al., “MHC Ligands and Peptide Motifs,” Landes Bioscience Austin, Tex., 224-227, 1997, which is hereby incorporated by reference in its entirety. Another site of interest is http://bimas.dcrt.nih.gov/molbio/hla_bind, which also contains a suitable algorithm. The methods of this web site are discussed in Parker, et al., “Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains,” J. Immunol. 152:163-175, which is hereby incorporated by reference in its entirety.


As an alternative to predictive algorithms, a number of standard in vitro receptor binding affinity assays are available to identify peptides having an affinity for a particular allele of MHC. Accordingly, by the method of this aspect of the invention, the initial population of peptide fragments can be narrowed to include only putative epitopes having an actual or predicted affinity for the selected allele of MHC. Selected common alleles of MHC I, and their approximate frequencies, are reported in the tables below.

TABLE 1Estimated gene frequencies of HLA-A antigensCAUAFRASILATNATAntigenGfaSEbGfSEGfSEGfSEGfSEA115.18430.04895.72560.07714.48180.08467.40070.097812.03160.2533A228.65350.061918.88490.131724.63520.179428.11980.170029.34080.3585A313.38900.04638.44060.09252.64540.06558.07890.101911.02930.2437A284.46520.02809.92690.09971.76570.05378.94460.10675.38560.1750A360.02210.00201.88360.04480.01480.00490.15840.01480.15450.0303A231.82870.018110.20860.10100.32560.02312.92690.06281.99030.1080A249.32510.03952.96680.056022.03910.172213.26100.127112.66130.2590A9 unsplit0.08090.00380.03670.00630.08580.01190.05370.00860.03560.0145A9 total11.23470.042913.21210.112822.45050.173316.24160.138214.68720.2756A252.11570.01950.43290.02160.09900.01281.19370.04041.45200.0924A263.87950.02622.82840.05474.66280.08623.26120.06622.42920.1191A340.15080.00523.52280.06101.35290.04700.49280.02600.31500.0432A430.00180.00060.03340.00600.02310.00620.00550.00280.00590.0059A660.01730.00180.22330.01550.04780.00890.03990.00740.05340.0178A10 unsplit0.07900.00380.09390.01010.12550.01440.06470.00940.02980.0133A10 total6.24410.03287.13480.08506.31110.09935.05780.08164.28530.1565A293.57960.02523.20710.05821.12330.04294.51560.07743.43450.1410A302.50670.021213.09690.11292.20250.05984.48730.07722.53140.1215A312.73860.02211.65560.04203.60050.07614.83280.08006.08810.1855A323.69560.02561.53840.04051.03310.04112.70640.06042.55210.1220A331.20800.01486.56070.08229.27010.11912.65930.05991.07540.0796A740.02770.00221.99490.04610.05610.00960.20270.01670.10680.0252A19 unsplit0.05670.00320.20570.01490.09900.01280.12110.01290.04750.0168A19 total13.81290.046828.25930.150417.38460.155519.52520.148115.83580.2832AX0.82040.02974.95060.09632.99160.11771.63320.08781.84540.1925
aGene frequency.

bStandard error.









TABLE 2










Estimated gene frequencies for HLA-B antigens













CAU
AFR
ASI
LAT
NAT

















Antigen
Gfa
SEb
Gf
SE
Gf
SE
Gf
SE
Gf
SE




















B7
12.1782
0.0445
10.5960
0.1024
4.2691
0.0827
6.4477
0.0918
10.9845
0.2432


B8
9.4077
0.0397
3.8315
0.0634
1.3322
0.0467
3.8225
0.0715
8.5789
0.2176


B13
2.3061
0.0203
0.8103
0.0295
4.9222
0.0886
1.2699
0.0416
1.7495
0.1013


B14
4.3481
0.0277
3.0331
0.0566
0.5004
0.0287
5.4166
0.0846
2.9823
0.1316


B18
4.7980
0.0290
3.2057
0.0582
1.1246
0.0429
4.2349
0.0752
3.3422
0.1391


B27
4.3831
0.0278
1.2918
0.0372
2.2355
0.0603
2.3724
0.0567
5.1970
0.1721


B35
9.6614
0.0402
8.5172
0.0927
8.1203
0.1122
14.6516
0.1329
10.1198
0.2345


B37
1.4032
0.0159
0.5916
0.0252
1.2327
0.0449
0.7807
0.0327
0.9755
0.0759


B41
0.9211
0.0129
0.8183
0.0296
0.1303
0.0147
1.2818
0.0418
0.4766
0.0531


B42
0.0608
0.0033
5.6991
0.0768
0.0841
0.0118
0.5866
0.0284
0.2856
0.0411


B46
0.0099
0.0013
0.0151
0.0040
4.9292
0.0886
0.0234
0.0057
0.0238
0.0119


B47
0.2069
0.0061
0.1305
0.0119
0.0956
0.0126
0.1832
0.0159
0.2139
0.0356


B48
0.0865
0.0040
0.1316
0.0119
2.0276
0.0575
1.5915
0.0466
1.0267
0.0778


B53
0.4620
0.0092
10.9529
0.1039
0.4315
0.0266
1.6982
0.0481
1.0804
0.0798


B59
0.0020
0.0006
0.0032
0.0019
0.4277
0.0265
0.0055
0.0028
0c



B67
0.0040
0.0009
0.0086
0.0030
0.2276
0.0194
0.0055
0.0028
0.0059
0.0059


B70
0.3270
0.0077
7.3571
0.0866
0.8901
0.0382
1.9266
0.0512
0.6901
0.0639


B73
0.0108
0.0014
0.0032
0.0019
0.0132
0.0047
0.0261
0.0060
0c



B51
5.4215
0.0307
2.5980
0.0525
7.4751
0.1080
6.8147
0.0943
6.9077
0.1968


B52
0.9658
0.0132
1.3712
0.0383
3.5121
0.0752
2.2447
0.0552
0.6960
0.0641


B5 unsplit
0.1565
0.0053
0.1522
0.0128
0.1288
0.0146
0.1546
0.0146
0.1307
0.0278


B5 total
6.5438
0.0435
4.1214
0.0747
11.1160
0.1504
9.2141
0.1324
7.7344
0.2784


B44
13.4838
0.0465
7.0137
0.0847
5.6807
0.0948
9.9253
0.1121
11.8024
0.2511


B45
0.5771
0.0102
4.8069
0.0708
0.1816
0.0173
1.8812
0.0506
0.7603
0.0670


B12 unsplit
0.0788
0.0038
0.0280
0.0055
0.0049
0.0029
0.0193
0.0051
0.0654
0.0197


B12 total
14.1440
0.0474
11.8486
0.1072
5.8673
0.0963
11.8258
0.1210
12.6281
0.2584


B62
5.9117
0.0320
1.5267
0.0404
9.2249
0.1190
4.1825
0.0747
6.9421
0.1973


B63
0.4302
0.0088
1.8865
0.0448
0.4438
0.0270
0.8083
0.0333
0.3738
0.0471


B75
0.0104
0.0014
0.0226
0.0049
1.9673
0.0566
0.1101
0.0123
0.03560
0.0145


B76
0.0026
0.0007
0.0065
0.0026
0.0874
0.0120
0.0055
0.0028
0c



B77
0.0057
0.0010
0.0119
0.0036
0.0577
0.0098
0.0083
0.0034
0.0059
0.0059


B15 unsplit
0.1305
0.0049
0.0691
0.0086
0.4301
0.0266
0.1820
0.0158
0.0715
0.0206


B15 total
6.4910
0.0334
3.5232
0.0608
12.2112
0.1344
5.2967
0.0835
7.4290
0.2035


B38
2.4413
0.0209
0.3323
0.0189
3.2818
0.0728
1.9652
0.0517
1.1017
0.0806


B39
1.9614
0.0188
1.2893
0.0371
2.0352
0.0576
6.3040
0.0909
4.5527
0.1615


B16 unsplit
0.0638
0.0034
0.0237
0.0051
0.0644
0.0103
0.1226
0.0130
0.0593
0.0188


B16 total
4.4667
0.0280
1.6453
0.0419
5.3814
0.0921
8.3917
0.1036
5.7137
0.1797


B57
3.5955
0.0252
5.6746
0.0766
2.5782
0.0647
2.1800
0.0544
2.7265
0.1260


B58
0.7152
0.0114
5.9546
0.0784
4.0189
0.0803
1.2481
0.0413
0.9398
0.0745


B17 unsplit
0.2845
0.0072
0.3248
0.0187
0.3751
0.0248
0.1446
0.0141
0.2674
0.0398


B17 total
4.5952
0.0284
11.9540
0.1076
6.9722
0.1041
3.5727
0.0691
3.9338
0.1503


B49
1.6452
0.0172
2.6286
0.0528
0.2440
0.0200
2.3353
0.0562
1.5462
0.0953


B50
1.0580
0.0138
0.8636
0.0304
0.4421
0.0270
1.8883
0.0507
0.7862
0.0681


B21 unsplit
0.0702
0.0036
0.0270
0.0054
0.0132
0.0047
0.0771
0.0103
0.0356
0.0145


B21 total
2.7733
0.0222
3.5192
0.0608
0.6993
0.0339
4.3007
0.0755
2.3680
0.1174


B54
0.0124
0.0015
0.0183
0.0044
2.6873
0.0660
0.0289
0.0063
0.0534
0.0178


B55
1.9046
0.0185
0.4895
0.0229
2.2444
0.0604
0.9515
0.0361
1.4054
0.0909


B56
0.5527
0.0100
0.2686
0.0170
0.8260
0.0368
0.3596
0.0222
0.3387
0.0448


B22 unsplit
0.1682
0.0055
0.0496
0.0073
0.2730
0.0212
0.0372
0.0071
0.1246
0.0272


B22 total
2.0852
0.0217
0.8261
0.0297
6.0307
0.0971
1.3771
0.0433
1.9221
0.1060


B60
5.2222
0.0302
1.5299
0.0404
8.3254
0.1135
2.2538
0.0553
5.7218
0.1801


B61
1.1916
0.0147
0.4709
0.0225
6.2072
0.0989
4.6691
0.0788
2.6023
0.1231


B40 unsplit
0.2696
0.0070
0.0388
0.0065
0.3205
0.0230
0.2473
0.0184
0.2271
0.0367


B40 total
6.6834
0.0338
2.0396
0.0465
14.8531
0.1462
7.1702
0.0963
8.5512
0.2168


BX
1.0922
0.0252
3.5258
0.0802
3.8749
0.0988
2.5266
0.0807
1.9867
0.1634








aGene frequency.






bStandard error.






cThe observed gene count was zero.














TABLE 3










Estimated gene frequencies of HLA-DR antigens













CAU
AFR
ASI
LAT
NAT

















Antigen
Gfa
SEb
Gf
SE
Gf
SE
Gf
SE
Gf
SE




















DR1
10.2279
0.0413
6.8200
0.0832
3.4628
0.0747
7.9859
0.1013
8.2512
0.2139


DR2
15.2408
0.0491
16.2373
0.1222
18.6162
0.1608
11.2389
0.1182
15.3932
0.2818


DR3
10.8708
0.0424
13.3080
0.1124
4.7223
0.0867
7.8998
0.1008
10.2549
0.2361


DR4
16.7589
0.0511
5.7084
0.0765
15.4623
0.1490
20.5373
0.1520
19.8264
0.3123


DR6
14.3937
0.0479
18.6117
0.1291
13.4471
0.1404
17.0265
0.1411
14.8021
0.2772


DR7
13.2807
0.0463
10.1317
0.0997
6.9270
0.1040
10.6726
0.1155
10.4219
0.2378


DR8
2.8820
0.0227
6.2673
0.0800
6.5413
0.1013
9.7731
0.1110
6.0059
0.1844


DR9
1.0616
0.0139
2.9646
0.0559
9.7527
0.1218
1.0712
0.0383
2.8662
0.1291


DR10
1.4790
0.0163
2.0397
0.0465
2.2304
0.0602
1.8044
0.0495
1.0896
0.0801


DR11
9.3180
0.0396
10.6151
0.1018
4.7375
0.0869
7.0411
0.0955
5.3152
0.1740


DR12
1.9070
0.0185
4.1152
0.0655
10.1365
0.1239
1.7244
0.0484
2.0132
0.1086


DR5 unsplit
1.2199
0.0149
2.2957
0.0493
1.4118
0.0480
1.8225
0.0498
1.6769
0.0992


DR5 total
12.4449
0.0045
17.0260
0.1243
16.2858
0.1516
10.5880
0.1148
9.0052
0.2218


DRX
1.3598
0.0342
0.8853
0.0760
2.5521
0.1089
1.4023
0.0930
2.0834
0.2037








aGene frequency.






bStandard error.







It has been observed that predicted epitopes often cluster at one or more particular regions within the amino acid sequence of a TAA. The identification of such ECRs offers a simple and practicable solution to the problem of designing effective vaccines for stimulating cellular immunity. For vaccines in which immune epitopes are desired, an ECR is directly useful as a vaccine. This is because the immune proteasomes of the pAPCs can correctly process the cluster, liberating one or more of the contained MHC-binding peptides, in the same way a cell having immune proteasomes activity processes and presents peptides derived from the complete TAA. The cluster is also a useful a starting material for identification of housekeeping epitopes produced by the housekeeping proteasomes active in peripheral cells.


Identification of housekeeping epitopes using ECRs as a starting material is described in copending U.S. patent application Ser. No. 09/561,074 entitled “METHOD OF EPITOPE DISCOVERY,” filed Apr. 28, 2000, which is incorporated herein by reference in its entirety. Epitope synchronization technology and vaccines for use in connection with this invention are disclosed in copending U.S. patent application Ser. No. 09/560,465 entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” filed Apr. 28, 2000, which is incorporated herein by reference in its entirety. Nucleic acid constructs useful as vaccines in accordance with the present invention are disclosed in copending U.S. patent application Ser. No. 09/561,572 entitled “EXPRESSION VECTORS ENCODING EPITOPES ASSOClATED ANTIGENS,” filed Apr. 28, 2000, which is incorporated herein by reference in its entirety.

TABLE 1ASEQ ID NOS.* including epitopes in Examples1-7, 13.SEQIDNOIDENTITYSEQUENCE1Tyr 207-216FLPWHRLFLL2TyrosinaseAccession number**: P14679protein3SSX-2 proteinAccession number: NP_0031384PSMA proteinAccession number: NP_0044675TyrosinaseAccession number: NM_000372cDNA6SSX-2 cDNAAceession number: NM_0031477PSMA cDNAAceession number: NM_0044768Tyr 207-215FLPWHRLFL9Tyr 208-216LPWHRLFLL10SSX-2 31-68YFSKEEWEKMKASEKIFYVYMKRKYEAMTKLGFKATLP11SSX-2 32-40FSKEEWEKM12SSX-2 39-47KMKASEKIF13SSX-2 40-48MKASEKIFY14SSX-2 39-48KMKASEKIFY15SSX-2 41-49KASEKIFYV16SSX-2 40-49MKASEKIFYV17SSX-2 41-50KASEKIFYVY18SSX-2 42-49ASEKIFYVY19SSX-2 53-61RKYEAMTKL20SSX-2 52-61KRKYEAMTKL21SSX-2 54-63KYEAMTKLGF22SSX-2 55-63YEAMTKLGF23SSX-2 56-63EAMTKLGF24HBV18-27FLPSDYFPSV25HLA-B44AEMGKYSFYbinder26SSX-1 41-49KYSEKISYV27SSX-3 41-49KVSEKIVYV28SSX-4 41-49KSSEKIVYV29SSX-5 41-49KASEKIIYV30PSMA 163-192AFSPQGMPEGDLVYVNYARTEDFFKLERDM31PSMA 168-190GMPEGDLVYVNYARTEDFFKLER32PSMA 169-177MPEGDLVYV33PSMA 168-177GMPEGDLVYV34PSMA 168-176GMPEGDLVY35PSMA 167-176QGMPEGDLVY36PSMA 169-176MPEGDLVY37PSMA 171-179EGDLVYVNY38PSMA 170-179PEGDLVYVNY39PSMA 174-183LVYVNYARTE40PSMA 177-185VNYARTEDF41PSMA 176-185YVNYARTEDF42PSMA 178-186NYARTEDFF43PSMA 179-186YARTEDFF44PSMA 181-189RTEDFFKLE45PSMA 281-310RGIAEAVGLPSIPVHPIGYYDAQKLLEKMG46PSMA 283-307IAEAVGLPSIPVHPIGYYDAQKLLE47PSMA 289-297LPSIPVHPI48PSMA 288-297GLPSIPVHPI49PSMA 297-305IGYYDAQKL50PSMA 296-305PIGYYDAQKL51PSMA 291-299SIPVHPIGY52PSMA 290-299PSIPVHPIGY53PSMA 292-299IPVHPIGY54PSMA 299-307YYDAQKLLE55PSMA 454-481SSIEGNYTLRVDCTPLMYSLVHLTKEL56PSMA 456-464IEGNYTLRV57PSMA 455-464SIEGNYTLRV58PSMA 457-464EGNYTLRV59PSMA 461-469TLRVDCTPL60PSMA 460-469YTLRVDCTPL61PSMA 462-470LRVDCTPLM62PSMA 463-471RVDCTPLMY63PSMA 462-471LRVDCTPLMY64PSMA 653-687FDKSNPIVLRMMNDQLMFLERAFIDPLGLPDRPFY65PSMA 660-681VLRMMNDQLMFLERAFIDPLGL66PSMA 663-671MMNDQLMFL67PSMA 662-671RMMNDQLMFL68PSMA 662-670RMMNDQLMF69Tyr 1-17MLLAVLYCLLWSFQTSA










TABLE 1B










SEQ ID NOS.* including epitopes in Examples 14



and 15.










SEQ





ID




NO
IDENTITY
SEQUENCE













70
GP100 protein2
**Accession number: P40967






71
MAGE-1 protein
Accession number: P43355





72
MAGE-2 protein
Accession number: P43356





73
MAGE-3 protein
Accession number: P43357





74
NY-ESO-1 protein
Accession number: P78358





75
LAGE-1a protein
Accession number: CAA11116





76
LAGE-1b protein
Accession number: CAA11117





77
PRAME protein
Aceession number: NP 006106





78
PSA protein
Accession number: P07288





79
PSCA protein
Accession number: O43653





80
GP100 cds
Accession number: U20093





81
MAGE-1 cds
Accession number: M77481





82
MAGE-2 cds
Accession number: L18920





83
MAGE-3 cds
Accession number: U03735





84
NY-ESO-1 cDNA
Accession number: U87459





85
PRAME cDNA
Accession number: NM_006115





86
PSA cDNA
Accession number: NM_001648





87
PSCA cDNA
Accession number: AF043498





88
GP100 630-638
LPHSSSHWL





89
GP100 629-638
QLPHSSSHWL





90
GP100 614-622
LIYRRRLMK





91
GP100 613-622
SLIYRRRLMK





92
GP100 615-622
IYRRRLMK





93
GP100 630-638
LPHSSSHWL





94
GP100 629-638
QLPHSSSHWL





95
MAGE-1 95-102
ESLFRAVI





96
MAGE-1 93-102
ILESLFRAVI





97
MAGE-1 93-101
ILESLFRAV





98
MAGE-1 92-101
CILESLFRAV





99
MAGE-1 92-100
CILESLFRA





100
MAGE-1 263-271
EFLWGPRAL





101
MAGE-1 264-271
FLWGPRAL





102
MAGE-1 264-273
FLWGPRALAE





103
MAGE-1 265-274
LWGPRALAET





104
MAGE-1 268-276
PRALAETSY





105
MAGE-1 267-276
GPRALAETSY





106
MAGE-1 269-277
RALAETSYV





107
MAGE-1 271-279
LAETSYVKV





108
MAGE-1 270-279
ALAETSYVKV





109
MAGE-1 272-280
AETSYVKVL





110
MAGE-1 271-280
LAETSYVKVL





111
MAGE-1 274-282
TSYVKVLEY





112
MAGE-1 273-282
ETSYVKVLEY





113
MAGE-1 278-286
KVLEYVIKV





114
MAGE-1 168-177
SYVLVTCLGL





115
MAGE-1 169-177
YVLVTCLGL





116
MAGE-1 170-177
VLVTCLGL





117
MAGE-1 240-248
TQDLVQEKY





118
MAGE-1 239-248
LTQDLVQEKY





119
MAGE-1 232-240
YGEPRKLLT





120
MAGE-1 243-251
LVQEKYLEY





121
MAGE-1 242-251
DLVQEKYLEY





122
MAGE-1 230-238
SAYGEPRKL





123
MAGE-1 278-286
KVLEYVIKV





124
MAGE-1 277-286
VKVLEYVIKV





125
MAGE-1 276-284
YVKVLEYVI





126
MAGE-1 274-282
TSYVKVLEY





127
MAGE-1 273-282
ETSYVKVLEY





128
MAGE-1 283-291
VIKVSARVR





129
MAGE-1 282-291
YVIKVSARVR





130
MAGE-2 115-122
ELVHFLLL





131
MAGE-2 113-122
MVELVHFLLL





132
MAGE-2 109-116
ISRKMVEL





133
MAGE-2 108-116
AISRKMVEL





134
MAGE-2 107-116
AAISRKMVEL





135
MAGE-2 112-120
KMVELVHFL





136
MAGE-2 109-117
ISRKMVELV





137
MAGE-2 108-117
AISRKMVELV





138
MAGE-2 116-124
LVHFLLLKY





139
MAGE-2 115-124
ELVHFLLLKY





140
MAGE-2 111-119
RKMVELVHF





141
MAGE-2 158-166
LQLVFGIEV





142
MAGE-2 157-166
YLQLVFGIEV





143
MAGE-2 159-167
QLVFGIEVV





144
MAGE-2 158-167
LQLVFGIEVV





145
MAGE-2 164-172
IEVVEVVPI





146
MAGE-2 163-172
GIEVVEVVPI





147
MAGE-2 162-170
FGIEVVEVV





148
MAGE-2 154-162
ASEYLQLVF





149
MAGE-2 153-162
KASEYLQLVF





150
MAGE-2 218-225
EEKIWEEL





151
MAGE-2 216-225
APEEKIWEEL





152
MAGE-2 216-223
APEEKIWE





153
MAGE-2 220-228
KIWEELSML





154
MAGE-2 219-228
EKIWEELSML





155
MAGE-2 271-278
FLWGPRAL





156
MAGE-2 271-279
FLWGPRALI





157
MAGE-2 278-286
LIETSYVKV





158
MAGE-2 277-286
ALIETSYVKV





159
MAGE-2 276-284
RALIETSYV





160
MAGE-2 279-287
IETSYVKVL





161
MAGE-2 278-287
LIETSYVKVL





162
MAGE-3 271-278
FLWGPRAL





163
MAGE-3 270-278
EFLWGPRAL





164
MAGE-3 271-279
FLWGPRALV





165
MAGE-3 276-284
RALVETSYV





166
MAGE-3 272-280
LWGPRALVE





167
MAGE-3 271-280
FLWGPRALVE





168
MAGE-3 272-281
LWGPRALVET





169
NY-ESO-1 82-90
GPESRLLEF





170
NY-ESO-1 83-91
PESRLLEFY





171
NY-ESO-1 82-91
GPESRLLEFY





172
NY-ESO-1 84-92
ESRLLEFYL





173
NY-ESO-1 86-94
RLLEFYLAM





174
NY-ESO-1 88-96
LEFYLAMPF





175
NY-ESO-1 87-96
LLEFYLAMPF





176
NY-ESO-1 93-102
AMPFATPMEA





177
NY-ESO-1 94-102
MPFATPMEA





178
NY-ESO-1 115-123
PLPVPGVLL





179
NY-ESO-1 114-123
PPLPVPGVLL





180
NY-ESO-1 116-123
LPVPGVLL





181
NY-ESO-1 103-112
ELARRSLAQD





182
NY-ESO-1 118-126
VPGVLLKEF





183
NY-ESO-1 117-126
PVPGVLLKEF





184
NY-ESO-1 116-123
LPVPGVLL





185
NY-ESO-1 127-135
TVSGNILTI





186
NY-ESO-1 126-135
FTVSGNILTI





187
NY-ESO-1 120-128
GVLLKEFTV





188
NY-ESO-1 121-130
VLLKEFTVSG





189
NY-ESO-1 122-130
LLKEFTVSG





190
NY-ESO-1 118-126
VPGVLLKEF





191
NY-ESO-1 117-126
PVPGVLLKEF





192
NY-ESO-1 139-147
AADHRQLQL





193
NY-ESO-1 148-156
SISSCLQQL





194
NY-ESO-1 147-156
LSISSCLQQL





195
NY-ESO-1 138-147
TAADHRQLQL





196
NY-ESO-1 161-169
WITQCFLPV





197
NY-ESO-1 157-165
SLLMWITQC





198
NY-ESO-1 150-158
SSCLQQLSL





199
NY-ESO-1 154-162
QQLSLLMWI





200
NY-ESO-1 151-159
SCLQQLSLL





201
NY-ESO-1 150-159
SSCLQQLSLL





202
NY-ESO-1 163-171
TQCFLPVFL





203
NY-ESO-1 162-171
ITQCFLPVFL





204
PRAME 219-227
PMQDIKMIL





205
PRAME 218-227
MPMQDIKMIL





206
PRAME 428-436
QHLIGLSNL





207
PRAME 427-436
LQHLIGLSNL





208
PRAME 429-436
HLIGLSNL





209
PRAME 431-439
IGLSNLTHV





210
PRAME 430-439
LIGLSNLTHV





211
PSA 53-61
VLVHPQWVL





212
PSA 52-61
GVLVHPQWVL





213
PSA 52-60
GVLVHPQWV





214
PSA 59-67
WVLTAAHCI





215
PSA 54-63
LVHPQWVLTA





216
PSA 53-62
VLVHPQWVLT





217
PSA 54-62
LVHPQWVLT





218
PSA 66-73
CIRNKSVI





219
PSA 65-73
HCIRNKSVI





220
PSA 56-64
HPQWVLTAA





221
PSA 63-72
AAHCIRNKSV





222
PSCA 116-123
LLWGPGQL





223
PSCA 115-123
LLLWGPGQL





224
PSCA 114-123
GLLLWGPGQL





225
PSCA 99-107
ALQPAAAIL





226
PSCA 98-107
HALQPAAAIL





227
Tyr 128-137
APEKDKFFAY





228
Tyr 129-137
PEKDKFFAY





229
Tyr 130-138
EKDKFFAYL





230
Tyr 131-138
KDKFFAYL





231
Tyr 205-213
PAFLPWHRL





232
Tyr 204-213
APAFLPWHRL





233
Tyr 214-223
FLLRWEQEIQ





234
Tyr 212-220
RLFLLRWEQ





235
Tyr 191-200
GSEIWRDIDF





236
Tyr 192-200
SEIWRDIDF





237
Tyr 473-481
RIWSWLLGA





238
Tyr 476-484
SWLLGAAMV





239
Tyr 477-486
WLLGAAMVGA





240
Tyr 478-486
LLGAAMVGA





241
PSMA 4-12
LLHETDSAV





242
PSMA 13-21
ATARRPRWL





243
PSMA 53-61
TPKHNMKAF





244
PSMA 64-73
ELKAENIKKF





245
PSMA 69-77
NIKKFLH1NF





246
PSMA 68-77
ENIKKFLH1NF





247
PSMA 220-228
AGAKGVILY





248
PSMA 468-477
PLMYSLVHNL





249
PSMA 469-477
LMYSLVHNL





250
PSMA 463-471
RVDCTPLMY





251
PSMA 465-473
DCTPLMYSL





252
PSMA 507-515
SGMPRISKL





253
PSMA 506-515
FSGMPRISKL





254
NY-ESO-1 136-163
RLTAADHRQLQLSISSCLQQLSLLMWIT





255
NY-ESO-1 150-177
SSCLQQLSLLMWITQCFLPVFLAQPPSG








1This H was reported as Y in the SWISSPROT database.






2The amino acid at position 274 may be Pro or Leu depending upon the database. The particular analysis presented herein used the Pro.















TABLE 1C










SEQ ID NOS.* including epitopes in Example 14.











SEQ





ID




NO.
IDENTITY
SEQUENCE





256
Mage-1 125-
KAEMLESV




132





257
Mage-1 124-
TKAEMLESV



132





258
Mage-1 123-
VTKAEMLESV



132





259
Mage-1 128-
MLESVIKNY



136





260
Mage-1 127-
EMLESVIKNY



136





261
Mage-1 125-
KAEMLESVI



133





262
Mage-1 146-
KASESLQL



153





263
Mage-1 145-
GKASESLQL



153





264
Mage-1 147-
ASESLQLVF



155





265
Mage-1 153-
LVFGIDVKE



161





266
Mage-1 114-
LLKYRARE



121





267
Mage-1 106-
VADLVGFL



113





268
Mage-1 105-
KVADLVGFL



113





269
Mage-1 107-
ADLVGFLLL



115





270
Mage-1 106-
VADLVGFLLL



115





271
Mage-1 114-
LLKYRAREPV



123





272
Mage-3 278-
LVETSYVKV



286





273
Mage-3 277-
ALVETSYVKV



286





274
Mage-3 285-
KVLHHMVKI



293





275
Mage-3 283-
YVKVLHHMV



291





276
Mage-3 275-
PRALVETSY



283





277
Mage-3 274-
GPRALVETSY



283





278
Mage-3 278-
LVETSYVKVL



287





279
ED-B 4′-5
TIIPEVPQL





280
ED-B 5′-5
DTIIPEVPQL





281
ED-B 1-10
EVPQLTDLSF





282
ED-B 23-30
TPLNSSTI





283
ED-B 18-25
IGLRWTPL





284
ED-B 17-25
SIGLRWTPL





285
ED-B 25-33
LNSSTIIGY





286
ED-B 24-33
PLNSSTIIGY





287
ED-B 23-31
TPLNSSTII





288
ED-B 31-38
IGYRITVV





289
ED-B 30-38
IIGYRITVV





290
ED-B 29-38
TIIGYRITVV





291
ED-B 31-39
IGYRITVVA





292
ED-B 30-39
IIGYRITVVA





293
CEA 184-191
SLPVSPRL





294
CEA 183-191
QSLPVSPRL





295
CEA 186-193
PVSPRLQL





296
CEA 185-193
LPVSPRLQL





297
CEA 184-193
SLPVSPRLQL





298
CEA 185-192
LPVSPRLQ





299
CEA 192-200
QLSNGNRTL





300
CEA 191-200
LQLSNGNRTL





301
CEA 179-187
WVNNQSLPV





302
CEA 186-194
PVSPRLQLS





303
CEA 362-369
SLPVSPRL





304
CEA 361-369
QSLPVSPRL





305
CEA 364-371
PVSPRLQL





306
CEA 363-371
LPVSPRLQL





307
CEA 362-371
SLPVSPRLQL





308
CEA 363-370
LPVSPRLQ





309
CEA 370-378
QLSNDNRTL





310
CEA 369-378
LQLSNDNRTL





311
CEA 357-365
WVNNQSLPV





312
CEA 360-368
NQSLPVSPR





313
CEA 540-547
SLPVSPRL





314
CEA 539-547
QSLPVSPRL





315
CEA 542-549
PVSPRLQL





316
CEA 541-549
LPVSPRLQL





317
CEA 540-549
SLPVSPRLQL





318
CEA 541-548
LPVSPRLQ





319
CEA 548-556
QLSNGNRTL





320
CEA 547-556
LQLSNGNRTL





321
CEA 535-543
WVNGQSLPV





322
CEA 533-541
LWWVNGQSL





323
CEA 532-541
YLWWVNGQSL





324
CEA 538-546
GQSLPVSPR





325
Her-2 30-37
DMKLRLPA





326
Her-2 28-37
GTDMKLRLPA





327
Her-2 42-49
HLDMLRHL





328
Her-2 41-49
THLDMLRHL





329
Her-2 40-49
ETHLDMLRHL





330
Her-2 36-43
PASPETHL





331
Her-2 35-43
LPASPETHL





332
Her-2 34-43
RLPASPETHL





333
Her-2 38-46
SPETHLDML





334
Her-2 37-46
ASPETHLDML





335
Her-2 42-50
HLDMLRHLY





336
Her-2 41-50
THLDMLRHLY





337
Her-2 719-
ELRKVKVL



726





338
Her-2 718-
TELRKVKVL



726





339
Her-2 717-
ETELRKVKVL



726





340
Her-2 715-
LKETELRKV



723





341
Her-2 714-
ILKETELRKV



723





342
Her-2 712-
MRILKETEL



720





343
Her-2 711-
QMRILKETEL



720





344
Her-2 717-
ETELRKVKV



725





345
Her-2 716-
KETELRKVKV



725





346
Her-2 706-
MPNQAQMRI



714





347
Her-2 705-
AMPNQAQMRI



714





348
Her-2 706-
MPNQAQMRIL



715





349
HER-2 966-
RPRFRELV



973





350
HER-2 965-
CRPRFRELV



973





351
HER-2 968-
RFRELVSEF



976





352
HER-2 967-
PRFRELVSEF



976





353
HER-2 964-
ECRPRFREL



972





354
NY-ESO-1
GAASGLNGC



67-75





355
NY-ESO-1
RASGPGGGA



52-60





356
NY-ESO-1
PHGGAASGL



64-72





357
NY-ESO-1
GPHGGAASGL



63-72





358
NY-ESO-1
APRGPHGGAA



60-69





359
PRAME 112-
VRPRRWKL



119





360
PRAME 111-
EVRPRRWKL



119





361
PRAME 113-
RPRRWKLQV



121





362
PRAME 114-
PRRWKLQVL



122





363
PRAME 113-
RPRRWKLQVL



122





364
PRAME 116-
RWKLQVLDL



124





365
PRAME 115-
RRWKLQVLDL



124





366
PRAME 174-
PVEVLVDLF



182





367
PRAME 199-
VKRKKNVL



206





368
PRAME 198-
KVKRKKNVL



206





369
PRAME 197-
EKVKRKKNVL



206





370
PRAME 198-
KVKRKKNV



205





371
PRAME 201-
RKKNVLRL



208





372
PRAME 200-
KRKKNVLRL



208





373
PRAME 199-
VKRKKNVLRL



208





374
PRAME 189-
DELFSYLI



196





375
PRAME 205-
VLRLCCKKL



213





376
PRAME 204-
NVLRLCCKKL



213





377
PRAME 194-
YLIEKVKRK



202





378
PRAME 74-81
QAWPFTCL





379
PRAME 73-81
VQAWPFTCL





380
PRAME 72-81
MVQAWPFTCL





381
PRAME 81-88
LPLGVLMK





382
PRAME 80-88
CLPLGVLMK





383
PRAME 79-88
TCLPLGVLMK





384
PRAME 84-92
GVLMKGQHL





385
PRAME 81-89
LPLGVLMKG





386
PRAME 80-89
CLPLGVLMKG





387
PRAME 76-85
WPFTCLPLGV





388
PRAME 51-59
ELFPPLFMA





389
PRAME 49-57
PRELFPPLF





390
PRAME 48-57
LPRELFPPLF





391
PRAME 50-58
RELFPPLFM





392
PRAME 49-58
PRELFPPLFM





393
PSA 239-246
RPSLYTKV





394
PSA 238-246
ERPSLYTKV





395
PSA 236-243
LPERPSLY





396
PSA 235-243
ALPERPSLY





397
PSA 241-249
SLYTKVVHY





398
PSA 240-249
PSLYTKVVHY





399
PSA 239-247
RPSLYTKVV





400
PSMA 211-
GNKVKNAQ



218





401
PSMA 202-
IARYGKVF



209





402
PSMA 217-
AQLAGAKGV



225





403
PSMA 207-
KVFRGNKVK



215





404
PSMA 211-
GNKVKNAQL



219





405
PSMA 269-
TPGYPANEY



277





406
PSMA 268-
LTPGYPANEY



277





407
PSMA 271-
GYPANEYAY



279





408
PSMA 270-
PGYPANEYAY



279





409
PSMA 266-
DPLTPGYPA



274





410
PSMA 492-
SLYESWTKK



500





411
PSMA 491-
KSLYESWTKK



500





412
PSMA 486-
EGFEGKSLY



494





413
PSMA 485-
DEGFEGKSLY



494





414
PSMA 498-
TKKSPSPEF



506





415
PSMA 497-
WTKKSPSPEF



506





416
PSMA 492-
SLYESWTKKS



501





417
PSMA 725-
WGEVKRQI



732





418
PSMA 724-
AWGEVKRQI



732





419
PSMA 723-
KAWGEVKRQI



732





420
PSMA 723-
KAWGEVKR



730





421
PSMA 722-
SKAWGEVKR



730





422
PSMA 731-
QIYVAAFTV



739





423
PSMA 733-
YVAAFTVQA



741





424
PSMA 725-
WGEVKRQIY



733





425
PSMA 727-
EVKRQIYVA



735





426
PSMA 738-
TVQAAAETL



746





427
PSMA 737-
FTVQAAAETL



746





428
PSMA 729-
KRQIYVAAF



737





429
PSMA 721-
PSKAWGEVK



729





430
PSMA 723-
KAWGEVKRQ



731





431
PSMA 100-
WKEFGLDSV



108





432
PSMA 99-108
QWKEFGLDSV





433
PSMA 102-
EFGLDSVELA



111





434
SCP-1 126-
ELRQKESKL



134





435
SCP-1 125-
AELRQKESKL



134





436
SCP-1 133-
KLQENRKII



141





437
SCP-1 298-
QLEEKTKL



305





438
SCP-1 297-
NQLEEKTKL



305





439
SCP-1 288-
LLEESRDKV



296





440
SCP-1 287-
FLLEESRDKV



296





441
SCP-1 291-
ESRDKVNQL



299





442
SCP-1 290-
EESRDKVNQL



299





443
SCP-1 475-
EKEVHDLEY



483





444
SCP-1 474-
REKEVHDLEY



483





445
SCP-1 480-
DLEYSYCHY



488





446
SCP-1 477-
EVHDLEYSY



485





447
SCP-1 477-
EVHDLEYSYC



486





448
SCP-1 502-
KLSSKREL



509





449
SCP-1 508-
ELKNTEYF



515





450
SCP-1 507-
RELKNTEYF



515





451
SCP-1 496-
KRGQRPKL



503





452
SCP-1 494-
LPKRGQRPKL



503





453
SCP-1 509-
LKNTEYFTL



517





454
SCP-1 508-
ELKNTEYFTL



517





455
SCP-1 506-
KRELKNTEY



514





456
SCP-1 502-
KLSSKRELK



510





457
SCP-1 498-
GQRPKLSSK



506





458
SCP-1 497-
RGQRPKLSSK



506





459
SCP-1 500-
RPKLSSKRE



508





460
SCP-1 573-
LEYVREEL



580





461
SCP-1 572-
ELEYVREEL



580





462
SCP-1 571-
NELEYVREEL



580





463
SCP-1 579-
ELKQKREDEV



587





464
SCP-1 575-
YVREELKQK



583





465
SCP-1 632-
QLNVYEIKV



640





466
SCP-1 630-
SKQLNVYEI



638





467
SCP-1 628-
AESKQLNVY



636





468
SCP-1 627-
TAESKQLNVY



636





469
SCP-1 638-
IKVNKLEL



645





470
SCP-1 637-
EIKVNKLEL



645





471
SCP-1 636-
YEIKVNKLEL



645





472
SCP-1 642-
KLELELESA



650





473
SCP-1 635-
VYEIKVNKL



643





474
SCP-1 634-
NVYEIKVNKL



643





475
SCP-1 646-
ELESAKQKF



654





476
SCP-1 642-
KLELELESA



650





477
SCP-1 646-
ELESAKQKF



654





478
SCP-1 771-
KEKLKREA



778





479
SCP-1 777-
EAKENTATL



785





480
SCP-1 776-
REAKENTATL



785





481
SCP-1 773-
KLKREAKENT



782





482
SCP-1 112-
EAEKIKKW



119





483
SCP-1 101-
GLSRVYSKL



109





484
SCP-1 100-
EGLSRVYSKL



109





485
SCP-1 108-
KLYKEAEKI



116





486
SCP-1 98-
NSEGLSRVY



106





487
SCP-1 97-
ENSEGLSRVY



106





488
SCP-1 102-
LSRVYSKLY



110





489
SCP-1 101-
GLSRVYSKLY



110





490
SCP-1 96-
LENSEGLSRV



105





491
SCP-1 108-
KLYKEAEKIK



117





492
SCP-1 949-
REDRWAVI



956





493
SCP-1 948-
MREDRWAVI



956





494
SCP-1 947-
KMREDRWAVI



956





495
SCP-1 947-
KMREDRWAV



955





496
SCP-1 934-
TTPGSTLKF



942





497
SCP-1 933-
LTTPGSTLKF



942





498
SCP-1 937-
GSTLKGAI



945





499
SCP-1 945-
IRKMREDRW



953





500
SCP-1 236-
RLEMHFKL



243





501
SCP-1 235-
SRLEMHFKL



243





502
SCP-1 242-
KLKEDYEKI



250





503
SCP-1 249-
KIQHLEQEY



257





504
SCP-1 248-
EKIQHLEQEY



257





505
SCP-1 233-
ENSRLEMHF



242





506
SCP-1 236-
RLEMHFKLKE



245





507
SCP-1 324-
LEDIKVSL



331





508
SCP-1 323-
ELEDIKVSL



331





509
SCP-1 322-
KELEDIKVSL



331





510
SCP-1 320-
LTKELEDI



327





511
SCP-1 319-
HLTKELEDI



327





512
SCP-1 330-
SLQRSVSTQ



338





513
SCP-1 321-
TKELEDIKV



329





514
SCP-1 320-
LTKELEDIKV



329





515
SCP-1 326-
DIKVSLQRSV



335





516
SCP-1 281-
KMKDLTFL



288





517
SCP-1 280-
NKMKDLTFL



288





518
SCP-1 279-
ENKMKDLTFL



288





519
SCP-1 288-
LLEESRDKV



296





520
SCP-1 287-
FLLEESRDKV



296





521
SCP-1 291-
ESRDKVNQL



299





522
SCP-1 290-
EESRDKVNQL



299





523
SCP-1 277-
EKENKMKDL



285





524
SCP-1 276-
TEKENKMKDL



285





525
SCP-1 279-
ENKMKDLTF



287





526
SCP-1 218-
IEKMITAF



225





527
SCP-1 217-
NIEKMITAF



225





528
SCP-1 216-
SNIEKMITAF



225





529
SCP-1 223-
TAFEELRV



230





530
SCP-1 222-
ITAFEELRV



230





531
SCP-1 221-
MITAFEELRV



230





532
SCP-1 220-
KMITAFEEL



228





533
SCP-1 219-
EKMITAFEEL



228





534
SCP-1 227-
ELRVQAENS



235





535
SCP-1 213-
DLNSNIEKMI



222





536
SCP-1 837-
WTSAKNTL



844





537
SCP-1 846-
TPLPKAYTV



854





538
SCP-1 845-
STPLPKAYTV



854





539
SCP-1 844-
LSTPLPKAY



852





540
SCP-1 843-
TLSTPLPKAY



852





541
SCP-1 842-
NTLSTPLPK



850





542
SCP-1 841-
KNTLSTPLPK



850





543
SCP-1 828-
ISKDKRDY



835





544
SCP-1 826-
HGISKDKRDY



835





545
SCP-1 832-
KRDYLWTSA



840





546
SCP-1 829-
SKDKRDYLWT



838





547
SCP-1 279-
ENKMKDLT



286





548
SCP-1 260-
EINDKEKQV



268





549
SCP-1 274-
QITEKENKM



282





550
SCP-1 269-
SLLLIQITE



277





551
SCP-1 453-
FEKIAEEL



460





552
SCP-1 452-
QFEKIAEEL



460





553
SCP-1 451-
KQFEKIAEEL



460





554
SCP-1 449-
DNKQFEKI



456





555
SCP-1 448-
YDNKQFEKI



456





556
SCP-1 447-
LYDNKQFEKI



456





557
SCP-1 440-
LGEKETLL



447





558
SCP-1 439-
VLGEKETLL



447





559
SCP-1 438-
KVLGEKETLL



447





560
SCP-1 390-
LLRTEQQRL



398





561
SCP-1 389-
ELLRTEQQRL



398





562
SCP-1 393-
TEQQRLENY



401





563
SCP-1 392-
RTEQQRLENY



401





564
SCP-1 402-
EDQLIILTM



410





565
SCP-1 397-
RLENYEDQLI



406





566
SCP-1 368-
KARAAHSF



375





567
SCP-1 376-
VVTEFETTV



384





568
SCP-1 375-
FVVTEFETTV



384





569
SCP-1 377-
VTEFETTVC



385





570
SCP-1 376-
VVTEFETTVC



385





571
SCP-1 344-
DLQIATNTI



352





572
SCP-1 347-
IATNTICQL



355





573
SCP-1 346-
QIATNTICQL



355





574
SSX4 57-65
VMTKLGFKY





575
SSX4 53-61
LNYEVMTKL





576
SSX4 52-61
KLNYEVMTKL





577
SSX4 66-74
TLPPFMRSK





578
SSX4 110-
KIMPKKPAE





579
SSX4 103-
SLQRIFPKIM



112





580
Tyr 463-471
YIKSYLEQA





581
Tyr 459-467
SFQDYIKSY





582
Tyr 458-467
DSFQDYIKSY





583
Tyr 507-514
LPEEKQPL





584
Tyr 506-514
QLPEEKQPL





585
Tyr 505-514
KQLPEEKQPL





586
Tyr 507-515
LPEEKQPLL





587
Tyr 506-515
QLPEEKQPLL





588
Tyr 497-505
SLLCRHKRK





589
ED-B domain
EVPQLTDLSFVDITDSSIGLRWTPLNSSTIIGYRI



of
TVVAAGEGIPIFEDFVDSSVGYYTVTGLEPGID



Fibronectin
YDISVITLINGGESAPTTLTQQT





590
ED-B domain
CTFDNLSPGLEYNVSVYTVKDDKESVPISDTIIP



of
EVPQLTDLSFVDITDSSIGLRWTPLNSSTIIGYRI



Fibronectin
TVVAAGEGIPIFEDFVDSSVGYYTVTGLEPGID



with flank-
YDISVITLINGGESAPTTLTQQT



ing se-
AVPPPTDLRFTNIGPDTMRVTW



quence from



Fribronectin





591
ED-B domain
Accession number: X07717



of



Fibronectin



cds





592
CEA protein
Accession number: P06731





593
CEA cDNA
Accession number: NM_004363





594
Her2/Neu
Accession number: P04626



protein





595
Her2/Neu
Accession number: M11730



cDNA





596
SCP-1
Accession number: Q15431



protein





597
SCP-1 cDNA
Accession number: X95654





598
SSX-4
Accession number: O60224



protein





599
SSX-4 cDNA
Accession number: NM_005636







*Any of SEQ ID NOS. 1, 8, 9, 11-23, 26-29, 32-44, 47-54, 56-63, 66-68 88-253, and 256-588 can be useful as epitopes in any of the various embodiments of the invention. Any of SEQ ID NOS. 10, 30, 31, 45, 46, 55, 64, 65, 69, 254, and 255 can be useful as sequences containing epitopes or epitope clusters, as described in various embodiments of the invention.





**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.







Note that the following discussion sets forth the inventors' understanding of the operation of the invention. However, it is not intended that this discussion limit the patent to any particular theory of operation not set forth in the claims.


In pursuing the development of epitope vaccines others have generated lists of predicted epitopes based on MHC binding motifs. Such peptides can be immunogenic, but may not correspond to any naturally produced antigenic fragment. Therefore, whole antigen will not elicit a similar response or sensitize a target cell to cytolysis by CTL. Therefore such lists do not differentiate between those sequences that can be useful as vaccines and those that cannot. Efforts to determine which of these predicted epitopes are in fact naturally produced have often relied on screening their reactivity with tumor infiltrating lymphocytes (TIL). However, TIL are strongly biased to recognize immune epitopes whereas tumors (and chronically infected cells) will generally present housekeeping epitopes. Thus, unless the epitope is produced by both the housekeeping and immuno-proteasomes, the target cell will generally not be recognized by CTL induced with TIL-identified epitopes. The epitopes of the present invention, in contrast, are generated by the action of a specified proteasome, indicating that they can be naturally produced, and enabling their appropriate use. The importance of the distinction between housekeeping and immune epitopes to vaccine design is more fully set forth in PCT publication WO 01/82963A2, which is hereby incorporated by reference in its entirety.


The epitopes of the invention include or encode polypeptide fragments of TAAs that are precursors or products of proteasomal cleavage by a housekeeping or immune proteasome, and that contain or consist of a sequence having a known or predicted affinity for at least one allele of MHC I. In some embodiments, the epitopes include or encode a polypeptide of about 6 to 25 amino acids in length, preferably about 7 to 20 amino acids in length; more preferably about 8 to 15 amino acids in length, and still more preferably 9 or 10 amino acids in length. However, it is understood that the polypeptides can be larger as long as N-terminal trimming can produce the MHC epitope or that they do not contain sequences that cause the polypeptides to be directed away from the proteasome or to be destroyed by the proteasome. For immune epitopes, if the larger peptides do not contain such sequences, they can be processed in the pAPC by the immune proteasome. Housekeeping epitopes may also be embedded in longer sequences provided that the sequence is adapted to facilitate liberation of the epitope's C-terminus by action of the immunoproteasome. The foregoing discussion has assumed that processing of longer epitopes proceeds through action of the immunoproteasome of the pAPC. However, processing can also be accomplished through the contrivance of some other mechanism, such as providing an exogenous protease activity and a sequence adapted so that action of the protease liberates the MHC epitope. The sequences of these epitopes can be subjected to computer analysis in order to calculate physical, biochemical, immunologic, or molecular genetic properties such as mass, isoelectric point, predicted mobility in electrophoresis, predicted binding to other MHC molecules, melting temperature of nucleic acid probes, reverse translations, similarity or homology to other sequences, and the like.


In constructing the polynucleotides encoding the polypeptide epitopes of the invention, the gene sequence of the associated TAA can be used, or the polynucleotide can be assembled from any of the corresponding codons. For a 10 amino acid epitope this can constitute on the order of 106 different sequences, depending on the particular amino acid composition. While large, this is a distinct and readily definable set representing a miniscule fraction of the >1018 possible polynucleotides of this length, and thus in some embodiments, equivalents of a particular sequence disclosed herein encompass such distinct and readily definable variations on the listed sequence. In choosing a particular one of these sequences to use in a vaccine, considerations such as codon usage, self-complementarity, restriction sites, chemical stability, etc. can be used as will be apparent to one skilled in the art.


The invention contemplates producing peptide epitopes. Specifically these epitopes are derived from the sequence of a TAA, and have known or predicted affinity for at least one allele of MHC I. Such epitopes are typically identical to those produced on target cells or pAPCs.


Compositions Containing Active Epitopes


Embodiments of the present invention provide polypeptide compositions, including vaccines, therapeutics, diagnostics, pharmacological and pharmaceutical compositions. The various compositions include newly identified epitopes of TAAs, as well as variants of these epitopes. Other embodiments of the invention provide polynucleotides encoding the polypeptide epitopes of the invention. The invention further provides vectors for expression of the polypeptide epitopes for purification. In addition, the invention provides vectors for the expression of the polypeptide epitopes in an APC for use as an anti-tumor vaccine. Any of the epitopes or antigens, or nucleic acids encoding the same, from Table 1 can be used. Other embodiments relate to methods of making and using the various compositions.


A general architecture for a class I MHC-binding epitope can be described, and has been reviewed more extensively in Madden, D. R. Annu. Rev. Immunol. 13:587-622, 1995, which is hereby incorporated by reference in its entirety. Much of the binding energy arises from main chain contacts between conserved residues in the MHC molecule and the N- and C-termini of the peptide. Additional main chain contacts are made but vary among MHC alleles. Sequence specificity is conferred by side chain contacts of so-called anchor residues with pockets that, again, vary among MHC alleles. Anchor residues can be divided into primary and secondary. Primary anchor positions exhibit strong preferences for relatively well-defined sets of amino acid residues. Secondary positions show weaker and/or less well-defined preferences that can often be better described in terms of less favored, rather than more favored, residues. Additionally, residues in some secondary anchor positions are not always positioned to contact the pocket on the MHC molecule at all. Thus, a subset of peptides exists that bind to a particular MHC molecule and have a side chain-pocket contact at the position in question and another subset exists that show binding to the same MHC molecule that does not depend on the conformation the peptide assumes in the peptide-binding groove of the MHC molecule. The C-terminal residue (PΩ; omega) is preferably a primary anchor residue. For many of the better studied HLA molecules (e.g. A2, A68, B27, B7, B35, and B53) the second position (P2) is also an anchor residue. However, central anchor residues have also been observed including P3 and P5 in HLA-B8, as well as P5 and PΩ(omega)-3 in the murine MHC molecules H-2 Db and H-2 Kb, respectively. Since more stable binding will generally improve immunogenicity, anchor residues are preferably conserved or optimized in the design of variants, regardless of their position.


Because the anchor residues are generally located near the ends of the epitope, the peptide can buckle upward out of the peptide-binding groove allowing some variation in length. Epitopes ranging from 8-11 amino acids have been found for HLA-A68, and up to 13 amino acids for HLA-A2. In addition to length variation between the anchor positions, single residue truncations and extensions have been reported and the N- and C-termini, respectively. Of the non-anchor residues, some point up out of the groove, making no contact with the MHC molecule but being available to contact the TCR, very often P1, P4, and PΩ(omega)-1 for HLA-A2. Others of the non-anchor residues can become interposed between the upper edges of the peptide-binding groove and the TCR, contacting both. The exact positioning of these side chain residues, and thus their effects on binding, MHC fine conformation, and ultimately immunogenicity, are highly sequence dependent. For an epitope to be highly immunogenic it must not only promote stable enough TCR binding for activation to occur, but the TCR must also have a high enough off-rate that multiple TCR molecules can interact sequentially with the same peptide-MHC complex (Kalergis, A. M. et al., Nature Immunol. 2:229-234, 2001, which is hereby incorporated by reference in its entirety). Thus, without further information about the ternary complex, both conservative and non-conservative substitutions at these positions merit consideration when designing variants.


The polypeptide epitope variants can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations. Variants can be derived from substitution, deletion or insertion of one or more amino acids as compared with the native sequence. Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a threonine with a serine, for example. Such replacements are referred to as conservative amino acid replacements, and all appropriate conservative amino acid replacements are considered to be embodiments of one invention. Insertions or deletions can optionally be in the range of about 1 to 4, preferably 1 to 2, amino acids. It is generally preferable to maintain the “anchor positions” of the peptide which are responsible for binding to the MHC molecule in question. Indeed, immunogenicity of peptides can be improved in many cases by substituting more preferred residues at the anchor positions (Franco, et al., Nature Immunology, 1(2):145-150, 2000, which is hereby incorporated by reference in its entirety). Immunogenicity of a peptide can also often be improved by substituting bulkier amino acids for small amino acids found in non-anchor positions while maintaining sufficient cross-reactivity with the original epitope to constitute a useful vaccine. The variation allowed can be determined by routine insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the polypeptide epitope. Because the polypeptide epitope is often 9 amino acids, the substitutions preferably are made to the shortest active epitope, for example, an epitope of 9 amino acids.


Variants can also be made by adding any sequence onto the N-terminus of the polypeptide epitope variant. Such N-terminal additions can be from 1 amino acid up to at least 25 amino acids. Because peptide epitopes are often trimmed by N-terminal exopeptidases active in the pAPC, it is understood that variations in the added sequence can have no effect on the activity of the epitope. In preferred embodiments, the amino acid residues between the last upstream proteasomal cleavage site and the N-terminus of the MHC epitope do not include a proline residue. Serwold, T. at al., Nature Immunol. 2:644-651, 2001, which is hereby incorporated by reference in its entirety. Accordingly, effective epitopes can be generated from precursors larger than the preferred 9-mer class I motif.


Generally, peptides are useful to the extent that they correspond to epitopes actually displayed by MHC I on the surface of a target cell or a pACP. A single peptide can have varying affinities for different MHC molecules, binding some well, others adequately, and still others not appreciably (Table 2). MHC alleles have traditionally been grouped according to serologic reactivity which does not reflect the structure of the peptide-binding groove, which can differ among different alleles of the same type. Similarly, binding properties can be shared across types; groups based on shared binding properties have been termed supertypes. There are numerous alleles of MHC I in the human population; epitopes specific to certain alleles can be selected based on the genotype of the patient.

TABLE 2Predicted Binding of Tyrosinase207-216 (SEQID NO. 1) to Various MHC types*Half time ofMHC I typedissociation (min)A10.05A*02011311.A*020550.4A32.7A*1101 (part of the A3 supertype)0.012A246.0B74.0B88.0B14 (part of the B27 supertype)60.0B*27020.9B*270530.0B*3501 (part of the B7 supertype)2.0B*44030.1B*5101 (part of the B7 supertype)26.0B*510255.0B*58010.20B600.40B622.0
*HLA Peptide Binding Predictions (world wide web hypertext transfer protocol “access at bimas.dcrt.nih.gov/molbio/hla_bin”).


In further embodiments of the invention, the epitope, as peptide or encoding polynucleotide, can be administered as a pharmaceutical composition, such as, for example, a vaccine or an immunogenic composition, alone or in combination with various adjuvants, carriers, or excipients. It should be noted that although the term vaccine may be used throughout the discussion herein, the concepts can be applied and used with any other pharmaceutical composition, including those mentioned herein. Particularly advantageous adjuvants include various cytokines and oligonucleotides containing immunostimulatory sequences (as set forth in greater detail in the co-pending applications referenced herein). Additionally the polynucleotide encoded epitope can be contained in a virus (e.g. vaccinia or adenovirus) or in a microbial host cell (e.g. Salmonella or Listeria monocytogenes) which is then used as a vector for the polynucleotide (Dietrich, G. et al. Nat. Biotech. 16:181-185, 1998, which is hereby incorporated by reference in its entirety). Alternatively a pAPC can be transformed, ex vivo, to express the epitope, or pulsed with peptide epitope, to be itself administered as a vaccine. To increase efficiency of these processes, the encoded epitope can be carried by a viral or bacterial vector, or complexed with a ligand of a receptor found on pAPC. Similarly the peptide epitope can be complexed with or conjugated to a pAPC ligand. A vaccine can be composed of more than a single epitope.


Particularly advantageous strategies for incorporating epitopes and/or epitope clusters, into a vaccine or pharmaceutical composition are disclosed in U.S. patent application Ser. No. 09/560,465 entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” filed on Apr. 28, 2000, which is hereby incorporated by reference in its entirety. Epitope clusters for use in connection with this invention are disclosed in U.S. patent application Ser. No. 09/561,571 entitled “EPITOPE CLUSTERS,” filed on Apr. 28, 2000, which is hereby incorporated by reference in its entirety.


Preferred embodiments of the present invention are directed to vaccines and methods for causing a pAPC or population of pAPCs to present housekeeping epitopes that correspond to the epitopes displayed on a particular target cell. Any of the epitopes or antigens in Table 1, can be used for example. In one embodiment, the housekeeping epitope is a TuAA epitope processed by the housekeeping proteasome of a particular tumor type. In another embodiment, the housekeeping epitope is a virus-associated epitope processed by the housekeeping proteasome of a cell infected with a virus. This facilitates a specific T cell response to the target cells. Concurrent expression by the pAPCs of multiple epitopes, corresponding to different induction states (pre- and post-attack), can drive a CTL response effective against target cells as they display either housekeeping epitopes or immune epitopes.


By having both housekeeping and immune epitopes present on the pAPC, this embodiment can optimize the cytotoxic T cell response to a target cell. With dual epitope expression, the pAPCs can continue to sustain a CTL response to the immune-type epitope when the tumor cell switches from the housekeeping proteasome to the immune proteasome with induction by IFN, which, for example, may be produced by tumor-infiltrating CTLs.


In a preferred embodiment, immunization of a patient is with a vaccine that includes a housekeeping epitope. Many preferred TAAs are associated exclusively with a target cell, particularly in the case of infected cells. In another embodiment, many preferred TAAs are the result of deregulated gene expression in transformed cells, but are found also in tissues of the testis, ovaries and fetus. In another embodiment, useful TAAs are expressed at higher levels in the target cell than in other cells. In still other embodiments, TAAs are not differentially expressed in the target cell compare to other cells, but are still useful since they are involved in a particular function of the cell and differentiate the target cell from most other peripheral cells; in such embodiments, healthy cells also displaying the TAA may be collaterally attacked by the induced T cell response, but such collateral damage is considered to be far preferable to the condition caused by the target cell.


The vaccine contains a housekeeping epitope in a concentration effective to cause a pAPC or populations of pAPCs to display housekeeping epitopes. Advantageously, the vaccine can include a plurality of housekeeping epitopes or one or more housekeeping epitopes optionally in combination with one or more immune epitopes. Formulations of the vaccine contain peptides and/or nucleic acids in a concentration sufficient to cause pAPCs to present the epitopes. The formulations preferably contain epitopes in a total concentration of about 1 μg-1 mg/100 μl of vaccine preparation. Conventional dosages and dosing for peptide vaccines and/or nucleic acid vaccines can be used with the present invention, and such dosing regimens are well understood in the art. In one embodiment, a single dosage for an adult human may advantageously be from about 1 to about 5000 μl of such a composition, administered one time or multiple times, e.g., in 2, 3, 4 or more dosages separated by 1 week, 2 weeks, 1 month, or more. insulin pump delivers 1 ul per hour (lowest frequency) ref intranodal method patent.


The compositions and methods of the invention disclosed herein further contemplate incorporating adjuvants into the formulations in order to enhance the performance of the vaccines. Specifically, the addition of adjuvants to the formulations is designed to enhance the delivery or uptake of the epitopes by the pAPCs. The adjuvants contemplated by the present invention are known by those of skill in the art and include, for example, GMCSF, GCSF, IL-2, IL-12, BCG, tetanus toxoid, osteopontin, and ETA-1.


In some embodiments of the invention, the vaccines can include a recombinant organism, such as a virus, bacterium or parasite, genetically engineered to express an epitope in a host. For example, Listeria monocytogenes, a gram-positive, facultative intracellular bacterium, is a potent vector for targeting TuAAs to the immune system. In a preferred embodiment, this vector can be engineered to express a housekeeping epitope to induce therapeutic responses. The normal route of infection of this organism is through the gut and can be delivered orally. In another embodiment, an adenovirus (Ad) vector encoding a housekeeping epitope for a TuAA can be used to induce anti-virus or anti-tumor responses. Bone marrow-derived dendritic cells can be transduced with the virus construct and then injected, or the virus can be delivered directly via subcutaneous injection into an animal to induce potent T-cell responses. Another embodiment employs a recombinant vaccinia virus engineered to encode amino acid sequences corresponding to a housekeeping epitope for a TAA. Vaccinia viruses carrying constructs with the appropriate nucleotide substitutions in the form of a minigene construct can direct the expression of a housekeeping epitope, leading to a therapeutic T cell response against the epitope.


The immunization with DNA requires that APCs take up the DNA and express the encoded proteins or peptides. It is possible to encode a discrete class I peptide on the DNA. By immunizing with this construct, APCs can be caused to express a housekeeping epitope, which is then displayed on class I MHC on the surface of the cell for stimulating an appropriate CTL response. Constructs generally relying on termination of translation or non-proteasomal proteases for generation of proper termini of housekeeping epitopes have been described in U.S. patent application Ser. No. 09/561,572 entitled EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS, filed on Apr. 28, 2000.


As mentioned, it can be desirable to express housekeeping peptides in the context of a larger protein. Processing can be detected even when a small number of amino acids are present beyond the terminus of an epitope. Small peptide hormones are usually proteolytically processed from longer translation products, often in the size range of approximately 60-120 amino acids. This fact has led some to assume that this is the minimum size that can be efficiently translated. In some embodiments, the housekeeping peptide can be embedded in a translation product of at least about 60 amino acids. In other embodiments the housekeeping peptide can be embedded in a translation product of at least about 50, 30, or 15 amino acids.


Due to differential proteasomal processing, the immune proteasome of the pAPC produces peptides that are different from those produced by the housekeeping proteasome in peripheral body cells. Thus, in expressing a housekeeping peptide in the context of a larger protein, it is preferably expressed in the APC in a context other than its full length native sequence, because, as a housekeeping epitope, it is generally only efficiently processed from the native protein by the housekeeping proteasome, which is not active in the APC. In order to encode the housekeeping epitope in a DNA sequence encoding a larger protein, it is useful to find flanking areas on either side of the sequence encoding the epitope that permit appropriate cleavage by the immune proteasome in order to liberate that housekeeping epitope. Altering flanking amino acid residues at the N-terminus and C-terminus of the desired housekeeping epitope can facilitate appropriate cleavage and generation of the housekeeping epitope in the APC. Sequences embedding housekeeping epitopes can be designed de novo and screened to determine which can be successfully processed by immune proteasomes to liberate housekeeping epitopes.


Alternatively, another strategy is very effective for identifying sequences allowing production of housekeeping epitopes in APC. A contiguous sequence of amino acids can be generated from head to tail arrangement of one or more housekeeping epitopes. A construct expressing this sequence is used to immunize an animal, and the resulting T cell response is evaluated to determine its specificity to one or more of the epitopes in the array. By definition, these immune responses indicate housekeeping epitopes that are processed in the pAPC effectively. The necessary flanking areas around this epitope are thereby defined. The use of flanking regions of about 4-6 amino acids on either side of the desired peptide can provide the necessary information to facilitate proteasome processing of the housekeeping epitope by the immune proteasome. Therefore, a sequence ensuring epitope synchronization of approximately 16-22 amino acids can be inserted into, or fused to, any protein sequence effectively to result in that housekeeping epitope being produced in an APC. In alternate embodiments the whole head-to-tail array of epitopes, or just the epitopes immediately adjacent to the correctly processed housekeeping epitope can be similarly transferred from a test construct to a vaccine vector.


In a preferred embodiment, the housekeeping epitopes can be embedded between known immune epitopes, or segments of such, thereby providing an appropriate context for processing. The abutment of housekeeping and immune epitopes can generate the necessary context to enable the immune proteasome to liberate the housekeeping epitope, or a larger fragment, preferably including a correct C-terminus. It can be useful to screen constructs to verify that the desired epitope is produced. The abutment of housekeeping epitopes can generate a site cleavable by the immune proteasome. Some embodiments of the invention employ known epitopes to flank housekeeping epitopes in test substrates; in others, screening as described below are used whether the flanking regions are arbitrary sequences or mutants of the natural flanking sequence, and whether or not knowledge of proteasomal cleavage preferences are used in designing the substrates.


Cleavage at the mature N-terminus of the epitope, while advantageous, is not required, since a variety of N-terminal trimming activities exist in the cell that can generate the mature N-terminus of the epitope subsequent to proteasomal processing. It is preferred that such N-terminal extension be less than about 25 amino acids in length and it is further preferred that the extension have few or no proline residues. Preferably, in screening, consideration is given not only to cleavage at the ends of the epitope (or at least at its C-terminus), but consideration also can be given to ensure limited cleavage within the epitope.


Shotgun approaches can be used in designing test substrates and can increase the efficiency of screening. In one embodiment multiple epitopes can be assembled one after the other, with individual epitopes possibly appearing more than once. The substrate can be screened to determine which epitopes can be produced. In the case where a particular epitope is of concern a substrate can be designed in which it appears in multiple different contexts. When a single epitope appearing in more than one context is liberated from the substrate additional secondary test substrates, in which individual instances of the epitope are removed, disabled, or are unique, can be used to determine which are being liberated and truly constitute sequences ensuring epitope synchronization.


Several readily practicable screens exist. A preferred in vitro screen utilizes proteasomal digestion analysis, using purified immune proteasomes, to determine if the desired housekeeping epitope can be liberated from a synthetic peptide embodying the sequence in question. The position of the cleavages obtained can be determined by techniques such as mass spectrometry, HPLC, and N-terminal pool sequencing; as described in greater detail in U.S. Patent Applications entitled METHOD OF EPITOPE DISCOVERY, EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS, two Provisional U.S. Patent Applications entitled EPITOPE SEQUENCES, which are all cited and incorporated by reference above.


Alternatively, in vivo screens such as immunization or target sensitization can be employed. For immunization a nucleic acid construct capable of expressing the sequence in question is used. Harvested CTL can be tested for their ability to recognize target cells presenting the housekeeping epitope in question. Such targets cells are most readily obtained by pulsing cells expressing the appropriate MHC molecule with synthetic peptide embodying the mature housekeeping epitope. Alternatively, cells known to express housekeeping proteasome and the antigen from which the housekeeping epitope is derived, either endogenously or through genetic engineering, can be used. To use target sensitization as a screen, CTL, or preferably a CTL clone, that recognizes the housekeeping epitope can be used. In this case it is the target cell that expresses the embedded housekeeping epitope (instead of the pAPC during immunization) and it must express immune proteasome. Generally, the target cell can be transformed with an appropriate nucleic acid construct to confer expression of the embedded housekeeping epitope. Loading with a synthetic peptide embodying the embedded epitope using peptide loaded liposomes or a protein transfer reagent such as BIOPORTER™ (Gene Therapy Systems, San Diego, Calif.) represents an alternative.


Additional guidance on nucleic acid constructs useful as vaccines in accordance with the present invention are disclosed in U.S. patent application Ser. No. 09/561,572 entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS,” filed on Apr. 28, 2000. Further, expression vectors and methods for their design, which are useful in accordance with the present invention are disclosed in U.S. Patent Application No. 60/336,968 (attorney docket number CTLIMM.022PR) entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS AND METHODS FOR THEIR DESIGN,” filed on Nov. 7, 2001, which is incorporated by reference in its entirety.


A preferred embodiment of the present invention includes a method of administering a vaccine including an epitope (or epitopes) to induce a therapeutic immune response. The vaccine is administered to a patient in a manner consistent with the standard vaccine delivery protocols that are known in the art. Methods of administering epitopes of TAAs including, without limitation, transdermal, intranodal, perinodal, oral, intravenous, intradermal, intramuscular, intraperitoneal, and mucosal administration, including delivery by injection, instillation or inhalation. A particularly useful method of vaccine delivery to elicit a CTL response is disclosed in Australian Patent No. 739189 issued Jan. 17, 2002; U.S. patent application Ser. No. 09/380,534, filed on Sep. 1, 1999; and a Continuation-in-Part thereof U.S. patent application Ser. No. 09/776,232 both entitled “A METHOD OF INDUCING A CTL RESPONSE,” filed on Feb. 2, 2001.


Reagents Recognizing Epitopes


In another aspect of the invention, proteins with binding specificity for the epitope and/or the epitope-MHC molecule complex are contemplated, as well as the isolated cells by which they can be expressed. In one set of embodiments these reagents take the form of immunoglobulins: polyclonal sera or monoclonal antibodies (mAb), methods for the generation of which are well know in the art. Generation of mAb with specificity for peptide-MHC molecule complexes is known in the art. See, for example, Aharoni et al. Nature 351:147-150, 1991; Andersen et al. Proc. Natl. Acad. Sci. USA 93:1820-1824, 1996; Dadaglio et al. Immunity 6:727-738, 1997; Duc et al. Int. Immunol. 5:427-431,1993; Eastman et al. Eur. J. Immunol. 26:385-393, 1996; Engberg et al. Immunotechnology 4:273-278, 1999; Porgdor et al. Immunity 6:715-726, 1997; Puri et al. J. Immunol. 158:2471-2476, 1997; and Polakova, K., et al. J. Immunol. 165 342-348, 2000; all of which are hereby incorporated by reference in their entirety.


In other embodiments the compositions can be used to induce and generate, in vivo and in vitro, T-cells specific for the any of the epitopes and/or epitope-MHC complexes. In preferred embodiments the epitope can be any one or more of those listed in TABLE 1, for example. Thus, embodiments also relate to and include isolated T cells, T cell clones, T cell hybridomas, or a protein containing the T cell receptor (TCR) binding domain derived from the cloned gene, as well as a recombinant cell expressing such a protein. Such TCR derived proteins can be simply the extra-cellular domains of the TCR, or a fusion with portions of another protein to confer a desired property or function. One example of such a fusion is the attachment of TCR binding domains to the constant regions of an antibody molecule so as to create a divalent molecule. The construction and activity of molecules following this general pattern have been reported, for example, Plaksin, D. et al. J. Immunol. 158:2218-2227, 1997 and Lebowitz, M. S. et al. Cell Immunol. 192:175-184, 1999, which are hereby incorporated by reference in their entirety. The more general construction and use of such molecules is also treated in U.S. Pat. No. 5,830,755 entitled T CELL RECEPTORS AND THEIR USE IN THERAPEUTIC AND DIAGNOSTIC METHODS, which is hereby incorporated by reference in its entirety.


The generation of such T cells can be readily accomplished by standard immunization of laboratory animals, and reactivity to human target cells can be obtained by immunizing with human target cells or by immunizing HLA-transgenic animals with the antigen/epitope. For some therapeutic approaches T cells derived from the same species are desirable. While such a cell can be created by cloning, for example, a murine TCR into a human T cell as contemplated above, in vitro immunization of human cells offers a potentially faster option. Techniques for in vitro immunization, even using naive donors, are know in the field, for example, Stauss et al., Proc. Natl. Acad. Sci. USA 89:7871-7875, 1992; Salgaller et al. Cancer Res. 55:4972-4979, 1995; Tsai et al., J. Immunol. 158:1796-1802, 1997; and Chung et al., J Immunother. 22:279-287, 1999; which are hereby incorporated by reference in their entirety.


Any of these molecules can be conjugated to enzymes, radiochemicals, fluorescent tags, and toxins, so as to be used in the diagnosis (imaging or other detection), monitoring, and treatment of the pathogenic condition associated with the epitope. Thus a toxin conjugate can be administered to kill tumor cells, radiolabeling can facilitate imaging of epitope positive tumor, an enzyme conjugate can be used in an ELISA-like assay to diagnose cancer and confirm epitope expression in biopsied tissue. In a further embodiment, such T cells as set forth above, following expansion accomplished through stimulation with the epitope and/or cytokines, can be administered to a patient as an adoptive immunotherapy.


Reagents Comprising Epitopes


A further aspect of the invention provides isolated epitope-MHC complexes. In a particularly advantageous embodiment of this aspect of the invention, the complexes can be soluble, multimeric proteins such as those described in U.S. Pat. No. 5,635,363 (tetramers) or U.S. Pat. No. 6,015,884 (Ig-dimers), both of which are hereby incorporated by reference in their entirety. Such reagents are useful in detecting and monitoring specific T cell responses, and in purifying such T cells.


Isolated MHC molecules complexed with epitopic peptides can also be incorporated into planar lipid bilayers or liposomes. Such compositions can be used to stimulate T cells in vitro or, in the case of liposomes, in vivo. Co-stimulatory molecules (e.g. B7, CD40, LFA-3) can be incorporated into the same compositions or, especially for in vitro work, co-stimulation can be provided by anti-co-receptor antibodies (e.g. anti-CD28, anti-CD154, anti-CD2) or cytokines (e.g. IL-2, IL-12). Such stimulation of T cells can constitute vaccination, drive expansion of T cells in vitro for subsequent infusion in an immuotherapy, or constitute a step in an assay of T cell function.


The epitope, or more directly its complex with an MHC molecule, can be an important constituent of functional assays of antigen-specific T cells at either an activation or readout step or both. Of the many assays of T cell function current in the art (detailed procedures can be found in standard immunological references such as Current Protocols in Immunology 1999 John Wiley & Sons Inc., N.Y., which is hereby incorporated by reference in its entirety) two broad classes can be defined, those that measure the response of a pool of cells and those that measure the response of individual cells. Whereas the former conveys a global measure of the strength of a response, the latter allows determination of the relative frequency of responding cells. Examples of assays measuring global response are cytotoxicity assays, ELISA, and proliferation assays detecting cytokine secretion. Assays measuring the responses of individual cells (or small clones derived from them) include limiting dilution analysis (LDA), ELISPOT, flow cytometric detection of unsecreted cytokine (described in U.S. Pat. No. 5,445,939, entitled “METHOD FOR ASSESSMENT OF THE MONONUCLEAR LEUKOCYTE IMMUNE SYSTEM” and U.S. Pat. Nos. 5,656,446; and 5,843,689, both entitled “METHOD FOR THE ASSESSMENT OF THE MONONUCLEAR LEUKOCYTE IMMUNE SYSTEM,” reagents for which are sold by Becton, Dickinson & Company under the tradename ‘FASTIMMUNE’, which patents are hereby incorporated by reference in their entirety) and detection of specific TCR with tetramers or Ig-dimers as stated and referenced above. The comparative virtues of these techniques have been reviewed in Yee, C. et al. Current Opinion in Immunology, 13:141-146, 2001, which is hereby incorporated by reference in its entirety. Additionally detection of a specific TCR rearrangement or expression can be accomplished through a variety of established nucleic acid based techniques, particularly in situ and single-cell PCR techniques, as will be apparent to one of skill in the art.


These functional assays are used to assess endogenous levels of immunity, response to an immunologic stimulus (e.g. a vaccine), and to monitor immune status through the course of a disease and treatment. Except when measuring endogenous levels of immunity, any of these assays presume a preliminary step of immunization, whether in vivo or in vitro depending on the nature of the issue being addressed. Such immunization can be carried out with the various embodiments of the invention described above or with other forms of immunogen (e.g., pAPC-tumor cell fusions) that can provoke similar immunity. With the exception of PCR and tetramer/Ig-dimer type analyses which can detect expression of the cognate TCR, these assays generally benefit from a step of in vitro antigenic stimulation which can advantageously use various embodiments of the invention as described above in order to detect the particular functional activity (highly cytolytic responses can sometimes be detected directly). Finally, detection of cytolytic activity requires epitope-displaying target cells, which can be generated using various embodiments of the invention. The particular embodiment chosen for any particular step depends on the question to be addressed, ease of use, cost, and the like, but the advantages of one embodiment over another for any particular set of circumstances will be apparent to one of skill in the art.


The peptide MHC complexes described in this section have traditionally been understood to be non-covalent associations. However it is possible, and can be advantageous, to create a covalent linkages, for example by encoding the epitope and MHC heavy chain or the epitope, β2-microglobulin, and MHC heavy chain as a single protein (Yu, Y. L. Y., et al., J. Immunol. 168:3145-3149, 2002; Mottez, E., et at., J. Exp. Med. 181:493,1995; Dela Cruz, C. S., et al., Int. Immunol. 12:1293, 2000; Mage, M. G., et al., Proc. Natl. Acad. Sci. USA 89:10658,1992; Toshitani, K., et al., Proc. Natl. Acad. Sci. USA 93:236,1996; Lee, L., et al., Eur. J. Immunol. 24:2633,1994; Chung, D. H., et al., J. Immunol. 163:3699,1999; Uger, R. A. and B. H. Barber, J. Immunol. 160:1598, 1998; Uger, R. A., et al., J. Immunol. 162:6024,1999; and White, J., et al., J. Immunol. 162:2671, 1999; which are incorporated herein by reference in their entirety). Such constructs can have superior stability and overcome roadblocks in the processing-presentation pathway. They can be used in the already described vaccines, reagents, and assays in similar fashion.


Tumor Associated Antigens


Epitopes of the present invention are derived from the TuAAs tyrosinase (SEQ ID NO. 2), SSX-2, (SEQ ID NO. 3), PSMA (prostate-specific membrane antigen) (SEQ ID NO. 4), GP100, (SEQ ID NO. 70), MAGE-1, (SEQ ID NO. 71), MAGE-2, (SEQ ID NO. 72), MAGE-3, (SEQ ID NO. 73), NY-ESO-1, (SEQ ID NO. 74), PRAME, (SEQ ID NO. 77), PSA, (SEQ ID NO. 78), PSCA, (SEQ ID NO. 79), the ED-B domain of fibronectin (SEQ ID NOS 589 and 590), CEA (carcinoembryonic antigen) (SEQ ID NO. 592), Her2/Neu (SEQ ID NO. 594), SCP-1 (SEQ ID NO. 596) and SSX-4 (SEQ ID NO. 598). The natural coding sequences for these eleven proteins, or any segments within them, can be determined from their cDNA or complete coding (cds) sequences, SEQ ID NOS. 5-7, 80-87, 591, 593, 595, 597, and 599, respectively.


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 tyrosinase as a TuAA is taught in U.S. Pat. No. 5,747,271 entitled “METHOD FOR IDENTIFYING INDIVIDUALS SUFFERING FROM A CELLULAR ABNORMALITY 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.


GP100, also known as PMel17, 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.


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 have been found variously in 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 (see example 3 below).


MAGE-1, MAGE-2, and MAGE-3 are members of another family of cancer-testis antigens originally discovered in melanoma (MAGE is a contraction of melanoma-associated antigen) 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 so in many cases, an epitope from one can induce a cross-reactive response to other members of the family. A few of these 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.


NY-ESO-1, is a cancer-testis antigen found in a wide variety of tumors, also known as CTAG-1 (Cancer-Testis Antigen-1) and CAG-3 (Cancer Antigen-3). 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 (SEQ ID NO. 75) and LAGE-1b/L (SEQ ID NO. 76), have been disclosed in publicly available assemblies of the human genome, and have 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. The proteins are virtually identical through amino acid 70. From 71-134 the longest run of identities between NY-ESO-1 and LAGE is 6 residues, but potentially cross-reactive sequences are present. And from 135-180 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), plus LAGE2-A and LAGE2-B (both corresponding to CTAG-1 in the genome assemblies).


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, attorney docket number CTLIMM.015A, filed on Mar. 7, 2002, entitled “ANTI-NEOVASCULAR PREPARATIONS FOR CANCER,” both of which are hereby incorporated by reference in their 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 also 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 and has a different expression profile. Thus the most preferred epitopes are those with an N-terminus located from amino acid 58 to 308.


PRAME, also know as MAPE, DAGE, and OIP4, was originally observed as a melanoma antigen. Subsequently, it has been recognized as a CT antigen, but unlike many CT antigens (e.g., MAGE, GAGE, and BAGE) it 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.


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 kalikrein 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” and in U.S. patent application Ser. No. 10/094,699, attorney docket number CTLIMM.015A, entitled “ANTI-NEOVASCULATURE PREPARATIONS FOR CANCER, filed on date even with this application (Mar. 7, 2002). A tumor cell line is disclosed in Provisional U.S. Application No. 60/363,131, filed on Mar. 7, 2002, attorney docket number CTLIMM.028PR, 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.


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 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.


All references mentioned herein are hereby incorporated by reference in their entirety. Further, incorporated by reference in its entirety is U.S. patent application Ser. No. 10/005,905 (attorney docket number CTLIMM.021CP1) entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS,” filed on Nov. 7, 2001 and a continuation thereof, U.S. application Ser. No. 10/026,066, filed on Dec. 7, 2001, attorney docket number MANNK.021 CP1C, also entitled “EPITOPE SYNCHRONIZATION IN ANTIGEN PRESENTING CELLS.”


Useful epitopes were identified and tested as described in the following examples. However, these examples are intended for illustration purposes only, and should not be construed as limiting the scope of the invention in any way.


EXAMPLES

Sequences of Specific Preferred Epitopes


Example 1

Manufacture of Epitopes.


A. Synthetic Production of Epitopes


Peptides having an amino acid sequence of any of SEQ ID NO: 1, 8, 9, 11-23, 26-29, 32-44, 47-54, 56-63, 66-68 88-253, or 256-588 are synthesized using either FMOC or tBOC solid phase synthesis methodologies. After synthesis, the peptides are cleaved from their supports with either trifluoroacetic acid or hydrogen fluoride, respectively, in the presence of appropriate protective scavengers. After removing the acid by evaporation, the peptides are extracted with ether to remove the scavengers and the crude, precipitated peptide is then lyophilized. Purity of the crude peptides is determined by HPLC, sequence analysis, amino acid analysis, counterion content analysis and other suitable means. If the crude peptides are pure enough (greater than or equal to about 90% pure), they can be used as is. If purification is required to meet drug substance specifications, the peptides are purified using one or a combination of the following: re-precipitation; reverse-phase, ion exchange, size exclusion or hydrophobic interaction chromatography; or counter-current distribution.


Drug Product Formulation


GMP-grade peptides are formulated in a parenterally acceptable aqueous, organic, or aqueous-organic buffer or solvent system in which they remain both physically and chemically stable and biologically potent. Generally, buffers or combinations of buffers or combinations of buffers and organic solvents are appropriate. The pH range is typically between 6 and 9. Organic modifiers or other excipients can be added to help solubilize and stabilize the peptides. These include detergents, lipids, co-solvents, antioxidants, chelators and reducing agents. In the case of a lyophilized product, sucrose or mannitol or other lyophilization aids can be added. Peptide solutions are sterilized by membrane filtration into their final container-closure system and either lyophilized for dissolution in the clinic, or stored until use.


B. Construction of expression vectors for use as nucleic acid vaccines


The construction of three generic epitope expression vectors is presented below. The particular advantages of these designs are set forth in U.S. patent application Ser. No. 09/561,572 entitled “EXPRESSION VECTORS ENCODING EPITOPES OF TARGET-ASSOCIATED ANTIGENS,” which has been incorporated by reference in its entirety above.


A suitable E. coli strain was then transfected with the plasmid and plated out onto a selective medium. Several colonies were grown up in suspension culture and positive clones were identified by restriction mapping. The positive clone was then grown up and aliquotted into storage vials and stored at −70° C.


A mini-prep (QIAprep Spin Mini-prep: Qiagen, Valencia, Calif.) of the plasmid was then made from a sample of these cells and automated fluorescent dideoxy sequence analysis was used to confirm that the construct had the desired sequence.


B.1 Construction of pVAX-EP1-IRES-EP2


Overview:


The starting plasmid for this construct is pVAX1 purchased from Invitrogen (Carlsbad, Calif.). Epitopes EP1 and EP2 were synthesized by GIBCO BRL (Rockville, Md.). The IRES was excised from pIRES purchased from Clontech (Palo Alto, Calif.).


Procedure:




  • 1 pIRES was digested with EcoRI and NotI. The digested fragments were separated by agarose gel electrophoresis, and the IRES fragment was purified from the excised band.

  • 2 pVAX1 was digested with EcoRI and NotI, and the pVAX1 fragment was gel-purified.

  • 3 The purified pVAX1 and IRES fragments were then ligated together.


    4 Competent E. coli of strain DH5 cc were transformed with the ligation mixture.

  • 5 Minipreps were made from 4 of the resultant colonies.

  • 6 Restriction enzyme digestion analysis was performed on the miniprep DNA. One recombinant colony having the IRES insert was used for further insertion of EP 1 and EP2. This intermediate construct was called pVAX-IRES.

  • 7 Oligonucleotides encoding EP1 and EP2 were synthesized.

  • 8 EP1 was subcloned into pVAX-IRES between AflII and EcoRI sites, to make pVAX-EP 1-IRES;

  • 9 EP2 was subcloned into pVAX-EP1-IRES between SalI and NotI sites, to make the final construct pVAX-EP 1-IRES-EP2.

  • 10 The sequence of the EP 1-IRES-EP2 insert was confirmed by DNA sequencing.



B 2. Construction of pVAX-EP1-IRES-EP2-ISS-NIS


Overview:


The starting plasmid for this construct was pVAX-EP1-IRES-EP2 (Example 1). The ISS (immunostimulatory sequence) introduced into this construct is AACGTT, and the NIS (standing for nuclear import sequence) used is the SV40 72 bp repeat sequence. ISS-NIS was synthesized by GIBCO BRL. See FIG. 2.


Procedure:




  • 1 pVAX-EP1-IRES-EP2 was digested with NruI; the linearized plasmid was gel-purified.

  • 2 ISS-NIS oligonucleotide was synthesized.

  • 3 The purified linearized pVAX-EP1-IRES-EP2 and synthesized ISS-NIS were ligated together.

  • 4 Competent E. coli of strain DH5α were transformed with the ligation product.

  • 5 Minipreps were made from resultant colonies.

  • 6 Restriction enzyme digestions of the minipreps were carried out.

  • 7 The plasmid with the insert was sequenced.



B3. Construction of pVAX-EP2-UB-EP1


Overview:


The starting plasmid for this construct was pVAX1 (Invitrogen). EP2 and EP1 were synthesized by GIBCO BRL. Wild type Ubiquitin cDNA encoding the 76 amino acids in the construct was cloned from yeast.


Procedure:




  • 1 RT-PCR was performed using yeast mRNA. Primers were designed to amplify the complete coding sequence of yeast Ubiquitin.

  • 2 The RT-PCR products were analyzed using agarose gel electrophoresis. A band with the predicted size was gel-purified.

  • 3 The purified DNA band was subcloned into pZERO1 at EcoRV site. The resulting clone was named pZERO-UB.

  • 4 Several clones of pZERO-UB were sequenced to confirm the Ubiquitin sequence before further manipulations.

  • 5 EP1 and EP2 were synthesized.

  • 6 EP2, Ubiquitin and EP1 were ligated and the insert cloned into pVAX1 between BamHI and EcoRI, putting it under control of the CMV promoter.

  • 7 The sequence of the insert EP2-UB-EP1 was confirmed by DNA sequencing.



Example 2

Identification of Useful Epitope Variants.


The 10-mer FLPWHRLFLL (SEQ ID NO. 1) is identified as a useful epitope. Based on this sequence, numerous variants are made. Variants exhibiting activity in HLA binding assays (see Example 3, section 6) are identified as useful, and are subsequently incorporated into vaccines.


The HLA-A2 binding of length variants of FLPWHRLFLL have been evaluated. Proteasomal digestion analysis indicates that the C-terminus of the 9-mer FLPWHRLFL (SEQ ID NO. 8) is also produced. Additionally the 9-mer LPWHRLFLL (SEQ ID NO. 9) can result from N-terminal trimming of the 10-mer. Both are predicted to bind to the HLA-A*0201 molecule, however of these two 9-mers, FLPWHRLFL displayed more significant binding and is preferred (see FIGS. 3A and B).


In vitro proteasome digestion and N-terminal pool sequencing indicates that tyrosinase207-216 (SEQ ID NO. 1) is produced more commonly than tyrosinase207-215 (SEQ ID NO. 8), however the latter peptide displays superior immunogenicity, a potential concern in arriving at an optimal vaccine design. FLPWHRLFL, tyrosinase207-215 (SEQ ID NO. 8) was used in an in vitro immunization of HLA-A2+blood to generate CTL (see CTL Induction Cultures below). Using peptide pulsed T2 cells as targets in a standard chromium release assay it was found that the CTL induced by tyrosinase207-215 (SEQ ID NO. 8) recognize tyrosinase207-216 (SEQ ID NO. 1) targets equally well (see FIG. 3C). These CTL also recognize the HLA-A2+, tyrosinase+ tumor cell lines 624.38 and HTB64, but not 624.28 an HLA-A2-derivative of 624.38 (FIG. 3C). Thus the relative amounts of these two epitopes produced in vivo, does not become a concern in vaccine design.


CTL Induction Cultures


PBMCs from normal donors were purified by centrifugation in Ficoll-Hypaque from buffy coats. All cultures were carried out using the autologous plasma (AP) to avoid exposure to potential xenogeneic pathogens and recognition of FBS peptides. To favor the in vitro generation of peptide-specific CTL, we employed autologous dendritic cells (DC) as APCs. DC were generated and CTL were induced with DC and peptide from PBMCs as described (Keogh et al., 2001). Briefly, monocyte-enriched cell fractions were cultured for 5 days with GM-CSF and IL-4 and were cultured for 2 additional days in culture media with 2 μg/ml CD40 ligand to induce maturation. 2×106 CD8+-enriched T lymphocytes/well and 2×105 peptide-pulsed DC/well were co-cultured in 24-well plates in 2 ml RPMI supplemented with 10% AP, 10 ng/ml IL-7 and 20 IU/ml IL-2. Cultures were restimulated on days 7 and 14 with autologous irradiated peptide-pulsed DC.


Sequence variants of FLPWHRLFL are constructed as follow. Consistent with the binding coefficient table (see Table 3) from the NIH/BIMAS MHC binding prediction program (see reference in example 3 below), binding can be improved by changing the L at position 9, an anchor position, to V. Binding can also be altered, though generally to a lesser extent, by changes at non-anchor positions. Referring generally to Table 3, binding can be increased by employing residues with relatively larger coefficients. Changes in sequence can also alter immunogenicity independently of their effect on binding to MHC. Thus binding and/or immunogenicity can be improved as follows:


By substituting F,L,M,W, or Y for P at position 3; these are all bulkier residues that can also improve immunogenicity independent of the effect on binding. The amine and hydroxyl-bearing residues, Q and N; and S and T; respectively, can also provoke a stronger, cross-reactive response.


By substituting D or E for W at position 4 to improve binding; this addition of a negative charge can also make the epitope more immunogenic, while in some cases reducing cross-reactivity with the natural epitope. Alternatively the conservative substitutions of F or Y can provoke a cross-reactive response.


By substituting F for H at position 5 to improve binding. H can be viewed as partially charged, thus in some cases the loss of charge can hinder cross-reactivity. Substitution of the fully charged residues R or K at this position can enhance immunogenicity without disrupting charge-dependent cross-reactivity.


By substituting I, L, M, V, F, W, or Y for R at position 6. The same caveats and alternatives apply here as at position 5.


By substituting W or F for L at position 7 to improve binding. Substitution of V, I, S, T, Q, or N at this position are not generally predicted to reduce binding affinity by this model (the NIH algorithm), yet can be advantageous as discussed above.


Y and W, which are equally preferred as the Fs at positions 1 and 8, can provoke a useful cross-reactivity. Finally, while substitutions in the direction of bulkiness are generally favored to improve immunogenicity, the substitution of smaller residues such as A, S, and C, at positions 3-7 can be useful according to the theory that contrast in size, rather than bulkiness per se, is an important factor in immunogenicity. The reactivity of the thiol group in C can introduce other properties as discussed in Chen, J.-L., et al. J. Immunol. 165:948-955, 2000.

TABLE 39-mer Coefficient Table for HLA-A*0201*HLA Coefficient table for file “A_0201_standard”Amino AcidType1st2nd3rd4th5th6th7th8th9thA1.0001.0001.0001.0001.0001.0001.0001.0001.000C1.0000.4701.0001.0001.0001.0001.0001.0001.000D0.0750.1000.4004.1001.0001.0000.4901.0000.003E0.0751.4000.0644.1001.0001.0000.4901.0000.003F4.6000.0503.7001.0003.8001.9005.8005.5000.015G1.0000.4701.0001.0001.0001.0000.1301.0000.015H0.0340.0501.0001.0001.0001.0001.0001.0000.015I1.7009.9001.0001.0001.0002.3001.0000.4102.100K3.5000.1000.0351.0001.0001.0001.0001.0000.003L1.70072.0003.7001.0001.0002.3001.0001.0004.300M1.70052.0003.7001.0001.0002.3001.0001.0001.000N1.0000.4701.0001.0001.0001.0001.0001.0000.015P0.0220.4701.0001.0001.0001.0001.0001.0000.003Q1.0007.3001.0001.0001.0001.0001.0001.0000.003R1.0000.0100.0761.0001.0001.0000.2001.0000.003S1.0000.4701.0001.0001.0001.0001.0001.0000.015T1.0001.0001.0001.0001.0001.0001.0001.0001.500V1.7006.3001.0001.0001.0002.3001.0000.41014.000W4.6000.0108.3001.0001.0001.7007.5005.5000.015Y4.6000.0103.2001.0001.0001.5001.0005.5000.015
*This table and other comparable data that are publicly available are useful in designing epitope variants and in determining whether a particular variant is substantially similar, or is functionally similar.


Example 3

Cluster Analysis (SSX-231-68).


1. Epitope cluster region Prediction:


The computer algorithms: SYFPEITHI (internet http:// access at syfpeithi.bmi-heidelberg.com/Scripts/MHCServer.dll/EpPredict.htm), based on the book “MHC Ligands and Peptide Motifs” by H. G. Rammensee, J. Bachmann and S. Stevanovic; and HLA Peptide Binding Predictions (NIH) (internet http:// access at bimas.dcrt.nih.gov/molbio/hla_bin), described in Parker, K. C., et al., J. Immunol. 152:163, 1994; were used to analyze the protein sequence of SSX-2 (GI:10337583). Epitope clusters (regions with higher than average density of peptide fragments with high predicted MHC affinity) were defined as described fully in U.S. patent application Ser. No. 09/561,571 entitled “EPITOPE CLUSTERS,” filed on Apr. 28, 2000. Using a epitope density ratio cutoff of 2, five and two clusters were defined using the SYFPETHI and NIH algorithms, respectively, and peptides score cutoffs of 16 (SYFPETHI) and 5 (NIH). The highest scoring peptide with the NIH algorithm, SSX-241-49, with an estimated halftime of dissociation of >1000 min., does not overlap any other predicted epitope but does cluster with SSX-257-65 in the NIH analysis.


2. Peptide Synthesis and Characterization:


SSX-231-68, YFSKEEWEKMKASEKIFYVYMKRKYEAMTKLGFKATLP (SEQ ID NO. 10) was synthesized by MPS (Multiple Peptide Systems, San Diego, Calif. 92121) using standard solid phase chemistry. According to the provided ‘Certificate of Analysis’, the purity of this peptide was 95%.


3. Proteasome Digestion:


Proteasome was isolated from human red blood cells using the proteasome isolation protocol described in U.S. patent application Ser. No. 09/561,074 entitled “METHOD OF EPITOPE DISCOVERY,” filed on Apr. 28, 2000. SDS-PAGE, western-blotting, and ELISA were used as quality control assays. The final concentration of proteasome was 4 mg/ml, which was determined by non-interfering protein assay (Geno Technologies Inc.). Proteasomes were stored at −70° C. in 25 μl aliquots.


SSX-231-68 was dissolved in Milli-Q water, and a 2 mM stock solution prepared and 20 μL aliquots stored at −20° C.


1 tube of proteasome (25 μL) was removed from storage at −70° C. and thawed on ice. It was then mixed thoroughly with 12.5 μL of 2 mM peptide by repipetting (samples were kept on ice). A 5 μL sample was immediately removed after mixing and transferred to a tube containing 1.25 μL 10% TFA (final concentration of TFA was 2%); the T=0 min sample. The proteasome digestion reaction was then started and carried out at 37° C. in a programmable thermal controller. Additional 5 μL samples were taken out at 15, 30, 60, 120, 180 and 240 min respectively, the reaction was stopped by adding the sample to 1.25 μL 10% TFA as before. Samples were kept on ice or frozen until being analyzed by MALDI-MS. All samples were saved and stored at −20° C. for HPLC analysis and N-terminal sequencing. Peptide alone (without proteasome) was used as a blank control: 2 μL peptide+4 μL Tris buffer (20 mM, pH 7.6)+1.5 μL TFA.


4. MALDI-TOF MS Measurements:


For each time point 0.3 μL of matrix solution (10 mg/ml α-cyano-4-hydroxycinnamic acid in AcCN/H2O (70:30)) was first applied on a sample slide, and then an equal volume of digested sample was mixed gently with matrix solution on the slide. The slide was allowed to dry at ambient air for 3-5 min. before acquiring the mass spectra. MS was performed on a Lasermat 2000 MALDI-TOF mass spectrometer that was calibrated with peptide/protein standards. To improve the accuracy of measurement, the molecular ion weight (MH+) of the peptide substrate was used as an internal calibration standard. The mass spectrum of the T=120 min. digested sample is shown in FIG. 4.


5. MS Data Analysis and Epitope Identification:


To assign the measured mass peaks, the computer program MS-Product, a tool from the UCSF Mass Spectrometry Facility (http:// accessible at prospector.ucsf.edu/ucsfhtml3.4/msprod.htm), was used to generate all possible fragments (N- and C-terminal ions, and internal fragments) and their corresponding molecular weights. Due to the sensitivity of the mass spectrometer, average molecular weight was used. The mass peaks observed over the course of the digestion were identified as summarized in Table 4.


Fragments co-C-terminal with 8-10 amino acid long sequences predicted to bind HLA by the SYFPEITHI or NIH algorithms were chosen for further study. The digestion and prediction steps of the procedure can be usefully practiced in any order. Although the substrate peptide used in proteasomal digest described here was specifically designed to include predicted HLA-A2.1 binding sequences, the actual products of digestion can be checked after the fact for actual or predicted binding to other MHC molecules. Selected results are shown in Table 5.

TABLE 4SSX-231-68 Mass Peak Identification.MS PEAKCALCULATED(measured)PEPTIDESEQUENCEMASS (MH+)988.2331-37YFSKEEW989.081377.68 ± 2.3831-40YFSKEEWEKM1377.681662.45 ± 1.3031-43YFSKEEWEKMKAS1663.902181.72 ± 0.8531-47YFSKEEWEKMKASEKIF2181.522346.631-48YFSKEEWEKMKASEKIFY2344.711472.16 ± 1.5438-49       EKMKASEKIFYV1473.772445.78 ± 1.1831-49*YFSKEEWEKMKASEKIFYV2443.842607.31-50YFSKEEWEKMKASEKIFYVY2607.021563.350-61                   YMKRKYEAMTKL1562.933989.931-61YFSKEEWEKMKASEKIFYVYMKRKYEAMTKL3987.771603.74 ± 1.5351-63                    MKRKYEAMTKLGF1603.981766.45 ± 1.550-63                   YMKRKYEAMTKLGF1767.161866.32 ± 1.2249-63                  VYMKRKYEAMTKLGF1866.294192.631-63YFSKEEWEKMKASEKIFYVYMKRKYEAMTKLGF4192.004392.131-65**YFSKEEWEKMKASEKIFYVYMKRKYEAMTKLGFKA4391.25
Boldface sequence correspond to peptides predicted to bind to MHC.

*On the basis of mass alone this peak could also have been assigned to the peptide 32-50, however proteasomal removal of just the N-terminal amino acid is unlikely. N-terminal sequencing (below) verifies the assignment to 31-49.

**On the basis of mass this fragment might also represent 33-68. N-terminal sequencing below is consistent with the assignment to 31-65.










TABLE 5










Predicted HLA binding by proteasomally gen-



erated fragments











SEQ ID NO.
PEPTIDE
HLA
SYFPEITHI
NIH















11
FSKEEWEKM
B*3501
NP†
90






12
KMKASEKIF
B*08
17
<5





13 & (14)
(K)MKASEKIFY
A1
19(19)
<5





15 & (16)
(M)KASEKIFYV
A*0201
22(16)
1017







B*08
17
<5







B*5101
22(13)
60







B*5102
NP
133







B*5103
NP
121





17 & (18)
(K)ASEKIFYVY
A1
34(19)
14





19 & (20)
(K)RKYEAMTKL
A*0201
15
<5







A26
15
NP







B14
NP
45






(60)







B*2705
21
15







B*2709
16
NP







B*5101
15
<5





21
KYEAMTKLGF
A1
16
<5







A24
NP
300





22
 YEAMTKLGF
B*4403
NP
80





23
  EAMTKLGF
B*08
22
<5







†No prediction







As seen in Table 5, N-terminal addition of authentic sequence to epitopes can generate epitopes for the same or different MHC restriction elements. Note in particular the pairing of (K)RKYEAMTKL (SEQ ID NOS 19 and (20)) with HLA-B14, where the 10-mer has a longer predicted halftime of dissociation than the co-C-terminal 9-mer. Also note the case of the 10-mer KYEAMTKLGF (SEQ ID NO. 21) which can be used as a vaccine useful with several MHC types by relying on N-terminal trimming to create the epitopes for HLA-B*4403 and -B*08.


6. HLA-A0201 Binding Assay:


Binding of the candidate epitope KASEKIFYV, SSX-241-49, (SEQ ID NO. 15) to HLA-A2.1 was assayed using a modification of the method of Stauss et al., (Proc Natl Acad Sci USA 89(17):7871-5 (1992)). Specifically, T2 cells, which express empty or unstable MHC molecules on their surface, were washed twice with Iscove's modified Dulbecco's medium (IMDM) and cultured overnight in serum-free AIM-V medium (Life Technologies, Inc., Rockville, Md.) supplemented with human β2-microglobulin at 3 μg/ml (Sigma, St. Louis, Mo.) and added peptide, at 800, 400, 200, 100, 50, 25, 12.5, and 6.25 μg/ml.in a 96-well flat-bottom plate at 3×105 cells/200 μl/well. Peptide was mixed with the cells by repipeting before distributing to the plate (alternatively peptide can be added to individual wells), and the plate was rocked gently for 2 minutes. Incubation was in a 5% CO2 incubator at 37° C. The next day the unbound peptide was removed by washing twice with serum free RPMI medium and a saturating amount of anti-class I HLA monoclonal antibody, fluorescein isothiocyanate (FITC)-conjugated anti-HLA A2, A28 (One Lambda, Canoga Park, Calif.) was added. After incubation for 30 minutes at 4° C., cells were washed 3 times with PBS supplemented with 0.5% BSA, 0.05% (w/v) sodium azide, pH 7.4-7.6 (staining buffer). (Alternatively W6/32 (Sigma) can be used as the anti-class I HLA monoclonal antibody the cells washed with staining buffer and then incubated with fluorescein isothiocyanate (FITC)-conjugated goat F(ab′) antimouse-IgG (Sigma) for 30 min at 4° C. and washed 3 times as before.) The cells were resuspended in 0.5 ml staining buffer. The analysis of surface HLA-A2.1 molecules stabilized by peptide binding was performed by flow cytometry using a FACScan (Becton Dickinson, San Jose, Calif.). If flow cytometry is not to be performed immediately the cells can be fixed by adding a quarter volume of 2% paraformaldehyde and storing in the dark at 4° C.


The results of the experiment are shown in FIG. 5. SSX-241-49 (SEQ ID NO. 15) was found to bind HLA-A2.1 to a similar extent as the known A2.1 binder FLPSDYFPSV (HBV18-27; SEQ ID NO: 24) used as a positive control. An HLA-B44 binding peptide, AEMGKYSFY (SEQ ID NO: 25), was used as a negative control. The fluoresence obtained from the negative control was similar to the signal obtained when no peptide was used in the assay. Positive and negative control peptides were chosen from Table 18.3.1 in Current Protocols in Immunology p. 18.3.2, John Wiley and Sons, New York, 1998.


7. Immunogenicity:


A. In Vivo Immunization of Mice.


HHD1 transgenic A*0201 mice (Pascolo, S., et al. J. Exp. Med. 185:2043-2051, 1997) were anesthetized and injected subcutaneously at the base of the tail, avoiding lateral tail veins, using 100 μl containing 100 nmol of SSX-241-49 (SEQ ID NO. 15) and 20 μg of HTL epitope peptide in PBS emulsified with 50 μl of IFA (incomplete Freund's adjuvant).


B. Preparation of Stimulating Cells (LPS Blasts).


Using spleens from 2 naive mice for each group of immunized mice, un-immunized mice were sacrificed and the carcasses were placed in alcohol. Using sterile instruments, the top dermal layer of skin on the mouse's left side (lower mid-section) was cut through, exposing the peritoneum. The peritoneum was saturated with alcohol, and the spleen was aseptically extracted. The spleen was placed in a petri dish with serum-free media. Splenocytes were isolated by using sterile plungers from 3 ml syringes to mash the spleens. Cells were collected in a 50 ml conical tubes in serum-free media, rinsing dish well. Cells were centrifuged (12000 rpm, 7 min) and washed one time with RPMI. Fresh spleen cells were resuspended to a concentration of 1×106 cells per ml in RPMI-10% FCS (fetal calf serum). 25 g/ml lipopolysaccharide and 7 μg/ml Dextran Sulfate were added. Cell were incubated for 3 days in T-75 flasks at 37° C., with 5% CO2. Splenic blasts were collected in 50 ml tubes pelleted (12000 rpm, 7 min) and resuspended to 3×107/ml in RPMI. The blasts were pulsed with the priming peptide at 50 μg/ml, RT 4 hr. mitomycin C-treated at 25 μg/ml, 37° C., 20 min and washed three times with DMEM.


C. In Vitro Stimulation.


3 days after LPS stimulation of the blast cells and the same day as peptide loading, the primed mice were sacrificed (at 14 days post immunization) to remove spleens as above. 3×106 splenocytes were co-cultured with 1×106 LPS blasts/well in 24-well plates at 37° C., with 5% CO2 in DMEM media supplemented with 10% FCS, 5×10−5 M β-mercaptoethanol, 100 μg/ml streptomycin and 100 IU/ml penicillin. Cultures were fed 5% (vol/vol) ConA supernatant on day 3 and assayed for cytolytic activity on day 7 in a 51Cr-release assay.


D. Chromium-Release Assay Measuring CTL Activity.


To assess peptide specific lysis, 2×106 T2 cells were incubated with 100 μCi sodium chromate together with 50 μg/ml peptide at 37° C. for 1 hour. During incubation they were gently shaken every 15 minutes. After labeling and loading, cells were washed three times with 10 ml of DMEM-10% FCS, wiping each tube with a fresh Kimwipe after pouring off the supernatant. Target cells were resuspended in DMEM-10% FBS 1×105/ml. Effector cells were adjusted to 1×107/ml in DMEM-10% FCS and 100 μl serial 3-fold dilutions of effectors were prepared in U-bottom 96-well plates. 100 μl of target cells were added per well. In order to determine spontaneous release and maximum release, six additional wells containing 100 μl of target cells were prepared for each target. Spontaneous release was revealed by incubating the target cells with 100 μl medium; maximum release was revealed by incubating the target cells with 100 μl of 2% SDS. Plates were then centrifuged for 5 min at 600 rpm and incubated for 4 hours at 37° C. in 5% CO2 and 80% humidity. After the incubation, plates were then centrifuged for 5 min at 1200 rpm. Supernatants were harvested and counted using a gamma counter. Specific lysis was determined as follows: % specific release=[(experimental release−spontaneous release)/(maximum release−spontaneous release)]×100.


Results of the chromium release assay demonstrating specific lysis of peptide pulsed target cells are shown in FIG. 6.


8. Cross-Reactivity with Other SSX Proteins:


SSX-241-49 (SEQ ID NO. 15) shares a high degree of sequence identity with the same region of the other SSX proteins. The surrounding regions have also been generally well conserved. Thus the housekeeping proteasome can cleave following V49 in all five sequences. Moreover, SSX41-49 is predicted to bind HLA-A*0201 (see Table 6). CTL generated by immunization with SSX-241-49 cross-react with tumor cells expressing other SSX proteins.

TABLE 6SSX41-49 - A*0201 Predicted BindingFamilySYFPEITHINIHSEQ ID NO.MemberSequenceScoreScore15SSX-2KASEKIFYV22101726SSX-1KYSEKISYV181.727SSX-3KVSEKIVYV24110528SSX-4KSSEKIVYV208229SSX-5KASEKIIYV22175


Example 4

Cluster Analysis (PSMA163-192).


A peptide, AFSPQGMPEGDLVYVNYARTEDFFKLERDM, PSMA163-192, (SEQ ID NO. 30), containing an A1 epitope cluster from prostate specific membrane antigen, PSMA168-190 (SEQ ID NO. 31) was synthesized using standard solid-phase F-moc chemistry on a 433A ABI Peptide synthesizer. After side chain deprotection and cleavage from the resin, peptide first dissolved in formic acid and then diluted into 30% Acetic acid, was run on a reverse-phase preparative HPLC C4 column at following conditions: linear AB gradient (5% B/min) at a flow rate of 4 ml/min, where eluent A is 0.1% aqueous TFA and eluent B is 0.1% TFA in acetonitrile. A fraction at time 16.642 min containing the expected peptide, as judged by mass spectrometry, was pooled and lyophilized. The peptide was then subjected to proteasome digestion and mass spectrum analysis essentially as described above. Prominent peaks from the mass spectra are summarized in Table 7.

TABLE 7PSMA163-192 Mass Peak Identification.CALCULATEDPEPTIDESEQUENCEMASS (MH+)163-177AFSPQGMPEGDLVYV1610.0178-189               NYARTEDFFKLE1533.68170-189       PEGDLVYVNYARTEDFFKLE2406.66178-191NYARTEDFFKLERD1804.95170-191PEGDLVYVNYARTEDFFKLERD2677.93178-192NYARTEDFFKLERDM1936.17163-176AFSPQGMPEGDLVY1511.70177-192VNYARTEDFFKLERDM2035.30163-179AFSPQGMPEGDLVYVNY1888.12180-192ARTEDFFKLERDM1658.89163-183AFSPQGMPEGDLVYVNYARTE2345.61184-192DFFKLERDM1201.40176-192YVNYARTEDFFKLERDM2198.48167-185    QGMPEGDLVYVNYARTEDF2205.41178-186               NYARTEDFF1163.22
Boldface sequences correspond to peptides predicted to bind to MHC, see Table 8.


N-Terminal Pool Sequence Analysis


One aliquot at one hour of the proteasomal digestion (see Example 3 part 3 above) was subjected to N-terminal amino acid sequence analysis by an ABI 473A Protein Sequencer (Applied Biosystems, Foster City, Calif.). Determination of the sites and efficiencies of cleavage was based on consideration of the sequence cycle, the repetitive yield of the protein sequencer, and the relative yields of amino acids unique in the analyzed sequence. That is if the unique (in the analyzed sequence) residue X appears only in the nth cycle a cleavage site exists n−1 residues before it in the N-terminal direction. In addition to helping resolve any ambiguity in the assignment of mass to sequences, these data also provide a more reliable indication of the relative yield of the various fragments than does mass spectrometry.


For PSMA163-192 (SEQ ID NO. 30) this pool sequencing supports a single major cleavage site after V177 and several minor cleavage sites, particularly one after Y179. Reviewing the results presented in FIGS. 7A-C reveals the following:

    • S at the 3rd cycle indicating presence of the N-terminus of the substrate.
    • Q at the 5th cycle indicating presence of the N-terminus of the substrate.
    • N at the 1st cycle indicating cleavage after V177.
    • N at the 3rd cycle indicating cleavage after V175. Note the fragment 176-192 in Table 7.
    • T at the 5th cycle indicating cleavage after V177
    • T at the 1st-3rd cycles, indicating increasingly common cleavages after R181, A180 and Y179. Only the last of these correspond to peaks detected by mass spectrometry; 163-179 and 180-192, see Table 7. The absence of the others can indicate that they are on fragments smaller than were examined in the mass spectrum.
    • K at the 4th, 8th, and 10th cycles indicating cleavages after E183, Y179, and V177, respectively, all of which correspond to fragments observed by mass spectroscopy. See Table 7.
    • A at the 1st and 3rd cycles indicating presence of the N-terminus of the substrate and cleavage after V177, respectively.
    • P at the 4th and 8th cycles indicating presence of the N-terminus of the substrate.
    • G at the 6th and 10th cycles indicating presence of the N-terminus of the substrate.
    • M at the 7th cycle indicating presence of the N-terminus of the substrate and/or cleavage after F185.
    • M at the 15th cycle indicating cleavage after V177.
    • The 1st cycle can indicate cleavage after D191, see Table 7.
    • R at the 4th and 13th cycle indicating cleavage after V177.
    • R at the 2nd and 11th cycle indicating cleavage after Y179.
    • V at the 2nd, 6th, and 13th cycle indicating cleavage after V175, M169 and presence of the N-terminus of the substrate, respectively. Note fragments beginning at 176 and 170 in Table 7.
    • Y at the 1st, 2nd, and 14th cycles indicating cleavage after V175, V177, and presence of the N-terminus of the substrate, respectively.
    • L at the 11th and 12th cycles indicating cleavage after V177, and presence of the N-terminus of the substrate, respectively, is the interpretation most consistent with the other data. Comparing to the mass spectrometry results we see that L at the 2nd, 5th, and 9th cycles is consistent with cleavage after F186, E183 or M169, and Y179, respectively. See Table 7.


      Epitope Identification


Fragments co-C-terminal with 8-10 amino acid long sequences predicted to bind HLA by the SYFPEITHI or NIH algorithms were chosen for further analysis. The digestion and prediction steps of the procedure can be usefully practiced in any order. Although the substrate peptide used in proteasomal digest described here was specifically designed to include a predicted HLA-A1 binding sequence, the actual products of digestion can be checked after the fact for actual or predicted binding to other MHC molecules. Selected results are shown in Table 8.

TABLE 8Predicted HLA binding by proteasomally gen-erated fragmentsSEQ ID NOPEPTIDEHLASYFPEITHINIH32 & (33)(G)MPEGDLVA*020117(27)(2605)YVB*070220<5B*51012231434 & (35)(Q)GMPEGDLA124(26)<5VYA316(18)36B*2705172536 MPEGDLVYB*510115NP†37 & (38)(P)EGDLVYVA127(15)12NYA2623(17)NP39LVYVNYARTEA321<540 & (41)(Y)VNYARTEA26(20)NPDFB*0815<5B*2705125042NYARTEDFFA24NP†100Cw*0401NP12043 YARTEDFFB*0816<544RTEDFFKLEA121<5A2615NP
†No prediction


HLA-A*0201 Binding Assay:


HLA-A*0201 binding studies were preformed with PSMA168-177, GMPEGDLVYV, (SEQ ID NO. 33) essentially as described in Example 3 above. As seen in FIG. 8, this epitope exhibits significant binding at even lower concentrations than the positive control peptides. The Melan-A peptide used as a control in this assay (and throughout this disclosure), ELAGIGILTV, is actually a variant of the natural sequence (EAAGIGILTV) and exhibits a high affinity in this assay.


Example 5

Cluster Analysis (PSMA281-310).


Another peptide, RGIAEAVGLPSIPVHPIGYYDAQKLLEKMG, PSMA281-310, (SEQ ID NO. 45), containing an A1 epitope cluster from prostate specific membrane antigen, PSMA283-307 (SEQ ID NO. 46), was synthesized using standard solid-phase F-moc chemistry on a 433A ABI Peptide synthesizer. After side chain deprotection and cleavage from the resin, peptide in ddH2O was run on a reverse-phase preparative HPLC C18 column at following conditions: linear AB gradient (5% B/min) at a flow rate of 4 ml/min, where eluent A is 0.1% aqueous TFA and eluent B is 0.1% TFA in acetonitrile. A fraction at time 17.061 min containing the expected peptide as judged by mass spectrometry, was pooled and lyophilized. The peptide was then subjected to proteasome digestion and mass spectrum analysis essentially as described above. Prominent peaks from the mass spectra are summarized in Table 9.

TABLE 9PSMA281-310 Mass Peak Identification.CALCULATEDPEPTIDESEQUENCEMASS (MH+)281-297RGIAEAVGLPSIPVHPI*1727.07286-297     AVGLPSIPVHPI**1200.46287-297      VGLPSIPVHPI1129.38288-297       GLPSIPVHPI1030.25298-310GYYDAQKLLEKMG‡1516.5298-305                 GYYDAQKL§958.05281-305RGIAEAVGLPSIPVHPIGYYDAQKL2666.12281-307RGIAEAVGLPSIPVHPIGYYDAQKLLE2908.39286-307     AVGLPSIPVHPIGYYDAQKLLE¶2381.78287-307      VGLPSIPVHPIGYYDAQKLLE2310.70288-307       GLPSIPVHPIGYYDAQKLLE#2211.57281-299RGIAEAVGLPSIPVHPIGY1947286-299     AVGLPSIPVHPIGY1420.69287-299      VGLPSIPVHPIGY1349.61288-299       GLPSIPVHPIGY1250.48287-310VGLPSIPVHPIGYYDAQKLLEKMG2627.14288-310GLPSIPVHPIGYYDAQKLLEKMG2528.01
Boldface sequences correspond to peptides predicted to bind to MHC, see Table 10.

*By mass alone this peak could also have been 296-310 or 288-303.

**By mass alone this peak could also have been 298-307. Combination of HPLC and mass spectrometry show that at some later time points this peak is a mixture of both species.

†By mass alone this peak could also have been 289-298.

‡By mass alone this peak could also have been 281-295 or 294-306.

§By mass alone this peak could also have been 297-303.

¶By mass alone this peak could also have been 285-306.

#By mass alone this peak could also have been 288-303.

None of these alternate assignments are supported N-terminal pool sequence analysis.


N-Terminal Pool Sequence Analysis


One aliquot at one hour of the proteasomal digestion (see Example 3 part 3 above) was subjected to N-terminal amino acid sequence analysis by an ABI 473A Protein Sequencer (Applied Biosystems, Foster City, Calif.). Determination of the sites and efficiencies of cleavage was based on consideration of the sequence cycle, the repetitive yield of the protein sequencer, and the relative yields of amino acids unique in the analyzed sequence. That is if the unique (in the analyzed sequence) residue X appears only in the nth cycle a cleavage site exists n−1 residues before it in the N-terminal direction. In addition to helping resolve any ambiguity in the assignment of mass to sequences, these data also provide a more reliable indication of the relative yield of the various fragments than does mass spectrometry.


For PSMA281-310 (SEQ ID NO. 45) this pool sequencing supports two major cleavage sites after V287 and I297 among other minor cleavage sites. Reviewing the results presented in FIG. 9 reveals the following:

    • S at the 4th and 11th cycles indicating cleavage after V287 and presence of the N-terminus of the substrate, respectively.
    • H at the 8th cycle indicating cleavage after V287. The lack of decay in peak height at positions 9 and 10 versus the drop in height present going from 10 to 11 can suggest cleavage after A286 and E285 as well, rather than the peaks representing latency in the sequencing reaction.
    • D at the 2nd, 4th, and 7th cycles indicating cleavages after Y299, I297, and V294, respectively. This last cleavage is not observed in any of the fragments in Table 10 or in the alternate assignments in the notes below.
    • Q at the 6th cycle indicating cleavage after I297.
    • M at the 10th and 12th cycle indicating cleavages after Y299 and I297, respectively.


      Epitope Identification


Fragments co-C-terminal with 8-10 amino acid long sequences predicted to bind HLA by the SYFPEITHI or NIH algorithms were chosen for further study. The digestion and prediction steps of the procedure can be usefully practiced in any order. Although the substrate peptide used in proteasomal digest described here was specifically designed to include a predicted HLA-A1 binding sequence, the actual products of digestion can be checked after the fact for actual or predicted binding to other MHC molecules. Selected results are shown in Table 10.

TABLE 10Predicted HLA binding by proteasomally gener-ated fragments: PSMA281-310SEQ ID NO.PEPTIDEHLASYFPEITHINIH47 & (48)(G)LPSIPVHA*020116(24)PI(24)B*0702/B72312B*510124572Cw*0401NP†2049 & (50)(P)IGYYDAQA*0201(16)<5KLA26(20)NPB*27051625B*270915NPB*51012157Cw*0301NP2451 & (52)(P)SIPVHPIA121<5GY(27)A2622NPA316<553IPVHPIGYB*510116NP54YYDAQKLLEA122<5
†No prediction


As seen in Table 10, N-terminal addition of authentic sequence to epitopes can often generate still useful, even better epitopes, for the same or different MHC restriction elements. Note for example the pairing of (G)LPSIPVHPI with HLA-A*0201, where the 10-mer can be used as a vaccine useful with several MHC types by relying on N-terminal trimming to create the epitopes for HLA-B7, -B*5101, and Cw*0401.


HLA-A*0201 Binding Assay:


HLA-A*0201 binding studies were preformed with PSMA288-297, GLPSIPVHPI, (SEQ ID NO. 48) essentially as described in Examples 3 and 4 above. As seen in FIG. 8, this epitope exhibits significant binding at even lower concentrations than the positive control peptides.


Example 6

Cluster Analysis (PSMA454-481).


Another peptide, SSIEGNYTLRVDCTPLMYSLVHLTKEL, PSMA454-481, (SEQ ID NO. 55) containing an epitope cluster from prostate specific membrane antigen, was synthesized by MPS (purity >95%) and subjected to proteasome digestion and mass spectrum analysis as described above. Prominent peaks from the mass spectra are summarized in Table 11.

TABLE 11PSMA454-481 Mass Peak Identification.MS PEAKCALCULATED(measured)PEPTIDESEQUENCEMASS (MH+)1238.5454-464SSIEGNYTLRV1239.781768.38 ± 0.6454-469SSIEGNYTLRVDCTPL1768.99  01899.8454-470SSIEGNYTLRVDCTPLM1900.191097.63 ± 0.9463-471         RVDCTPLMY1098.32  12062.87 ± 0.6454-471*SSIEGNYTLRVDCTPLMY2063.36  81153472-481**                  SLVHNLTKEL1154.361449.93 ± 1.7470-481                MYSLVHNLTKEL1448.73  9
Boldface sequence correspond to peptides predicted to bind to MHC, see Table 12.

*On the basis of mass alone this peak could equally well be assigned to the peptide 455-472 however proteasomal removal of just the N-terminal amino acid is considered unlikely. If the issue were important it could be resolved by N-terminal sequencing.

**On the basis of mass this fragment might also represent 455-464.


Epitope Identification


Fragments co-C-terminal with 8-10 amino acid long sequences predicted to bind HLA by the SYFPEITHI or NIH algorithms were chosen for further study. The digestion and prediction steps of the procedure can be usefully practiced in any order. Although the substrate peptide used in proteasomal digest described here was specifically designated to include predicted HLA-A2.1 binding sequences, the actual products of digestion can be checked after the fact for actual or predicted binding to other MHC molecules. Selected results are shown in Table 12.

TABLE 12Predicted HLA binding by proteasomally gener-ated fragmentsSEQ ID NOPEPTIDEHLASYFPEITHINIH56 & (57)(S)IEGNYTLRA1(19)<5V58EGNYTLRVA*020116<5(22)B*510115NP†59 & (60)(Y)TLRVDCTPA*020120 (5)L(18)A2616NP(18)B71440B823<5B*27051230Cw*0301NP(30)61LRVDCTPLMB*270520600B*270920NP62 & (63)(L)RVDCTPLMA132125Y(22)(13.5)A325<5A2622NPB*2702NP(200) B*270513(1000) (NP)
†No prediction


As seen in Table 12, N-terminal addition of authentic sequence to epitopes can often generate still useful, even better epitopes, for the same or different MHC restriction elements. Note for example the pairing of (L)RVDCTPLMY (SEQ ID NOS 62 and (63)) with HLA-B*2702/5, where the 10-mer has substantial predicted halftimes of dissociation and the co-C-terminal 9-mer does not. Also note the case of SIEGNYTLRV (SEQ ID NO 57) a predicted HLA-A*0201 epitope which can be used as a vaccine useful with HLA-B*5101 by relying on N-terminal trimming to create the epitope.


HLA-A*0201 Binding Assay


HLA-A*0201 binding studies were preformed, essentially as described in Example 3 above, with PSMA460-469, TLRVDCTPL, (SEQ ID NO. 60). As seen in FIG. 10, this epitope was found to bind HLA-A2.1 to a similar extent as the known A2.1 binder FLPSDYFPSV (HBV18-27; SEQ ID NO: 24) used as a positive control. Additionally, PSMA461-469, (SEQ ID NO. 59) binds nearly as well.


ELISPOT Analysis: PSM 463-471 (SEQ ID NO. 62)


The wells of a nitrocellulose-backed microtiter plate were coated with capture antibody by incubating overnight at 4° C. using 50 μl/well of 4 μg/ml murine anti-human γ-IFN monoclonal antibody in coating buffer (35 mM sodium bicarbonate, 15 mM sodium carbonate, pH 9.5). Unbound antibody was removed by washing 4 times 5 min. with PBS. Unbound sites on the membrane then were blocked by adding 200 μl/well of RPMI medium with 10% serum and incubating 1 hr. at room temperature. Antigen stimulated CD8+ T cells, in 1:3 serial dilutions, were seeded into the wells of the microtiter plate using 10011/well, starting at 2×105 cells/well. (Prior antigen stimulation was essentially as described in Scheibenbogen, C. et al. Int. J. Cancer 71:932-936, 1997. PSMA462-471 (SEQ ID NO. 62) was added to a final concentration of 10 μg/ml and IL-2 to 100 U/ml and the cells cultured at 37° C. in a 5% CO2, water-saturated atmosphere for 40 hrs. Following this incubation the plates were washed with 6 times 200 μl/well of PBS containing 0.05% Tween-20 (PBS-Tween). Detection antibody, 50 μl/well of 2 g/ml biotinylated murine anti-human γ-IFN monoclonal antibody in PBS+10% fetal calf serum, was added and the plate incubated at room temperature for 2 hrs. Unbound detection antibody was removed by washing with 4 times 200 μl of PBS-Tween. 100 μl of avidin-conjugated horseradish peroxidase (Pharmingen, San Diego, Calif.) was added to each well and incubated at room temperature for 1 hr. Unbound enzyme was removed by washing with 6 times 200 μl of PBS-Tween. Substrate was prepared by dissolving a 20 mg tablet of 3-amino 9-ethylcoarbasole in 2.5 ml of N,N-dimethylformamide and adding that solution to 47,5 ml of 0.05 M phosphate-citrate buffer (pH 5.0). 25 μl of 30% H2O2 was added to the substrate solution immediately before distributing substrate at 100 μl/well and incubating the plate at room temperature. After color development (generally 15-30 min.), the reaction was stopped by washing the plate with water. The plate was air dried and the spots counted using a stereomicroscope.



FIG. 11 shows the detection of PSMA463-471 (SEQ ID NO. 62)-reactive HLA-A1+ CD8+ T cells previously generated in cultures of HLA-A1+ CD8+ T cells with autologous dendritic cells plus the peptide. No reactivity is detected from cultures without peptide (data not shown). In this case it can be seen that the peptide reactive T cells are present in the culture at a frequency between 1 in 2.2×104 and 1 in 6.7×104. That this is truly an HLA-A1-restricted response is demonstrated by the ability of anti-HLA-A1 monoclonal antibody to block γ-IFN production; see FIG. 12.


Example 7

Cluster Analysis (PSMA653-687).


Another peptide, FDKSNPIVLRMMNDQLMFLERAFIDPLGLPDRPFY PSMA653-687, (SEQ ID NO. 64) containing an A2 epitope cluster from prostate specific membrane antigen, PSMA660-681 (SEQ ID NO 65), was synthesized by MPS (purity >95%) and subjected to proteasome digestion and mass spectrum analysis as described above. Prominent peaks from the mass spectra are summarized in Table 13.

TABLE 13PSMA653-687 Mass Peak Identification.MS PEAKCALCULATED(measured)PEPTIDESEQUENCEMASS (MH+) 906.17 ± 0.65681-687**LPDRPFY 908.051287.73 ± 0.7677-687**DPLGLPDRPFY1290.47  6 1400.3 ± 1.79676-687IDPLGLPDRPFY1403.63 1548.0 ± 1.37675-687FIDPLGLPDRPFY1550.80 1619.5 ± 1.51674-687**AFIDPLGLPDRPFY1621.881775.48 ± 1.3673-687*RAFIDPLGLPDRPFY1778.07  2 2440.2 ± 1.3653-672FDKSNPIVLRMMNDQLMFLE2442.932313.821904.63 ± 1.5672-687*ERAFIDPLGLPDRPFY1907.19  6 2310.6 ± 2.5653-671FDKSNPIVLRMMNDQLMFL2313.82 2017.4 ± 1.94671-687LERAFIDPLGLPDRPFY2020.352197.43 ± 1.7653-670FDKSNPIVLRMMNDQLMF2200.66  8
Boldface sequence correspond to peptides predicted to bind to MHC, see Table 13.

*On the basis of mass alone this peak could equally well be assigned to a peptide beginning at 654, however proteasomal removal of just the N-terminal amino acid is considered unlikely. If the issue were important it could be resolved by N-terminal sequencing.

**On the basis of mass alone these peaks could have been assigned to internal fragments, but given the overall pattern of digestion it was considered unlikely.


Epitope Identification


Fragments co-C-terminal with 8-10 amino acid long sequences predicted to bind HLA by the SYFPEITHI or NIH algorithms were chosen for further study. The digestion and prediction steps of the procedure can be usefully practiced in any order. Although the substrate peptide used in proteasomal digest described here was specifically designed to include predicted HLA-A2.1 binding sequences, the actual products of digestion can be checked after the fact for actual or predicted binding to other MHC molecules. Selected results are shown in Table 14.

TABLE 14Predicted HLA binding by proteasomally gener-ated fragmentsSEQ ID NOPEPTIDEHLASYFPEITHINIH66 & (67)(R)MMNDQLMA*0201241360FL(23)(722)A*0205NP†71 (42)A2615NPB*2705125068RMMNDQLMFB*27051775
†No prediction


As seen in Table 14, N-terminal addition of authentic sequence to epitopes can generate still useful, even better epitopes, for the same or different MHC restriction elements. Note for example the pairing of (R)MMNDQLMFL (SEQ ID NOS. 66 and (67)) with HLA-A*02, where the 10-mer retains substantial predicted binding potential.


HLA-A*0201 Binding Assay


HLA-A*0201 binding studies were preformed, essentially as described in Example 3 above, with PSMA663-671, (SEQ ID NO. 66) and PSMA662-671, RMMNDQLMFL (SEQ NO. 67). As seen in FIGS. 10, 13 and 14, this epitope exhibits significant binding at even lower concentrations than the positive control peptide (FLPSDYFPSV (HBV18-27); SEQ ID NO: 24). Though not run in parallel, comparison to the controls suggests that PSMA662-671 (which approaches the Melan A peptide in affinity) has the superior binding activity of these two PSMA peptides.


Example 8

Vaccinating with Epitope Vaccines.


1. Vaccination with Peptide Vaccines:


A. Intranodal Delivery


A formulation containing peptide in aqueous buffer with an antimicrobial agent, an antioxidant, and an immunomodulating cytokine, was injected continuously over several days into the inguinal lymph node using a miniature pumping system developed for insulin delivery (MiniMed; Northridge, Calif.). This infusion cycle was selected in order to mimic the kinetics of antigen presentation during a natural infection.


B. Controlled Release


A peptide formulation is delivered using controlled PLGA microspheres as is known in the art, which alter the pharmacokinetics of the peptide and improve immunogenicity. This formulation is injected or taken orally.


C. Gene Gun Delivery


A peptide formulation is prepared wherein the peptide is adhered to gold microparticles as is known in the art. The particles are delivered in a gene gun, being accelerated at high speed so as to penetrate the skin, carrying the particles into dermal tissues that contain pAPCs.


D. Aerosol Delivery


A peptide formulation is inhaled as an aerosol as is known in the art, for uptake into appropriate vascular or lymphatic tissue in the lungs.


2. Vaccination with Nucleic Acid Vaccines:


A nucleic acid vaccine is injected into a lymph node using a miniature pumping system, such as the MiniMed insulin pump. A nucleic acid construct formulated in an aqueous buffered solution containing an antimicrobial agent, an antioxidant, and an immunomodulating cytokine, is delivered over a several day infusion cycle in order to mimic the kinetics of antigen presentation during a natural infection.


Optionally, the nucleic acid construct is delivered using controlled release substances, such as PLGA microspheres or other biodegradable substances. These substances are injected or taken orally. Nucleic acid vaccines are given using oral delivery, priming the immune response through uptake into GALT tissues. Alternatively, the nucleic acid vaccines are delivered using a gene gun, wherein the nucleic acid vaccine is adhered to minute gold particles. Nucleic acid constructs can also be inhaled as an aerosol, for uptake into appropriate vascular or lymphatic tissue in the lungs.


Example 9

Assays for the Effectiveness of Epitope Vaccines.


1. Tetramer Analysis:


Class I tetramer analysis is used to determine T cell frequency in an animal before and after administration of a housekeeping epitope. Clonal expansion of T cells in response to an epitope indicates that the epitope is presented to T cells by pAPCs. The specific T cell frequency is measured against the housekeeping epitope before and after administration of the epitope to an animal, to determine if the epitope is present on pAPCs. An increase in frequency of T cells specific to the epitope after administration indicates that the epitope was presented on pAPC.


2. Proliferation Assay:


Approximately 24 hours after vaccination of an animal with housekeeping epitope, pAPCs are harvested from PBMCs, splenocytes, or lymph node cells, using monoclonal antibodies against specific markers present on pAPCs, fixed to magnetic beads for affinity purification. Crude blood or splenoctye preparation is enriched for pAPCs using this technique. The enriched pAPCs are then used in a proliferation assay against a T cell clone that has been generated and is specific for the housekeeping epitope of interest. The pAPCs are coincubated with the T cell clone and the T cells are monitored for proliferation activity by measuring the incorporation of radiolabeled thymidine by T cells. Proliferation indicates that T cells specific for the housekeeping epitope are being stimulated by that epitope on the pAPCs.


3. Chromium Release Assay:


A human patient, or non-human animal genetically engineered to express human class I MHC, is immunized using a housekeeping epitope. T cells from the immunized subject are used in a standard chromium release assay using human tumor targets or targets engineered to express the same class I MHC. T cell killing of the targets indicates that stimulation of T cells in a patient would be effective at killing a tumor expressing a similar TuAA.


Example 10

Induction of CTL Response with Naked DNA is Efficient by Intra-Lymph Node Immunization.


In order to quantitatively compare the CD8+ CTL responses induced by different routes of immunization a plasmid DNA vaccine (pEGFPL33A) containing a well-characterized immunodominant CTL epitope from the LCMV-glycoprotein (G) (gp33; amino acids 33-41) (Oehen, S., et al. Immunology 99, 163-169 2000) was used, as this system allows a comprehensive assessment of antiviral CTL responses. Groups of 2 C57BL/6 mice were immunized once with titrated doses (200-0.02 μg) of pEGFPL33A DNA or of control plasmid pEGFP-N3, administered i.m. (intramuscular), i.d. (intradermal), i.spl. (intrasplenic), or i.ln. (intra-lymph node). Positive control mice received 500 pfu LCMV i.v. (intravenous). Ten days after immunization spleen cells were isolated and gp33-specific CTL activity was determined after secondary in vitro restimulation. As shown in FIG. 15, i.m. or i.d. immunization induced weakly detectable CTL responses when high doses of pEFGPL33A DNA (200 μg) were administered. In contrast, potent gp33-specific CTL responses were elicited by immunization with only 2 μg pEFGPL33A DNA i.spl. and with as little as 0.2 μg pEFGPL33A DNA given i.ln. (FIG. 15; symbols represent individual mice and one of three similar experiments is shown). Immunization with the control pEGFP-N3 DNA did not elicit any detectable gp33-specific CTL responses (data not shown).


Example 11

Intra-Lymph Node DNA Immunization Elicits Anti-Tumor Immunity.


To examine whether the potent CTL responses elicited following i.ln. immunization were able to confer protection against peripheral tumors, groups of 6 C57BL/6mice were immunized three times at 6-day intervals with 10 μg of pEFGPL33A DNA or control pEGFP-N3 DNA. Five days after the last immunization small pieces of solid tumors expressing the gp33 epitope (EL4-33) were transplanted s.c. into both flanks and tumor growth was measured every 3-4 d. Although the EL4-33 tumors grew well in mice that had been repetitively immunized with control pEGFP-N3 DNA (FIG. 16), mice which were immunized with pEFGPL33A DNA i.ln. rapidly eradicated the peripheral EL4-33 tumors (FIG. 16).


Example 12

Differences in Lymph Node DNA Content Mirrors Differences in CTL Response following Intra-Lymph Node and Intramuscular Injection.






    • pEFGPL33A DNA was injected i.ln. or i.m. and plasmid content of the injected or draining lymph node was assessed by real time PCR after 6, 12, 24, 48 hours, and 4 and 30 days. At 6, 12, and 24 hours the plasmid DNA content of the injected lymph nodes was approximately three orders of magnitude greater than that of the draining lymph nodes following i.m. injection. No plasmid DNA was detectable in the draining lymph node at subsequent time points (FIG. 17). This is consonant with the three orders of magnitude greater dose needed using i.m. as compared to i.ln. injections to achieve a similar levels of CTL activity. CD8−/− knockout mice, which do not develop a CTL response to this epitope, were also injected i.ln. showing clearance of DNA from the lymph node is not due to CD8+ CTL killing of cells in the lymph node. This observation also supports the conclusion that i.ln. administration will not provoke immunopathological damage to the lymph node.





Example 13

Administration of a DNA Plasmid Formulation of a Therapeutic Vaccine for Melanoma to Humans.


SYNCHROTOPE TA2M, a melanoma vaccine, encoding the HLA-A2-restricted tyrosinase epitope SEQ ID NO. 1 and epitope cluster SEQ ID NO. 69, was formulated in 1% Benzyl alcohol, 1% ethyl alcohol, 0.5 mM EDTA, citrate-phosphate, pH 7.6. Aliquots of 80, 160, and 320 μg DNA/ml were prepared for loading into MINIMED 407C infusion pumps. The catheter of a SILHOUETTE infusion set was placed into an inguinal lymph node visualized by ultrasound imaging. The assembly of pump and infusion set was originally designed for the delivery of insulin to diabetics and the usual 17 mm catheter was substituted with a 31 mm catheter for this application. The infusion set was kept patent for 4 days (approximately 96 hours) with an infusion rate of about 25 μl/hour resulting in a total infused volume of approximately 2.4 ml. Thus the total administered dose per infusion was approximately 200, and 400 μg; and can be 800 μg, respectively, for the three concentrations described above. Following an infusion subjects were given a 10 day rest period before starting a subsequent infusion. Given the continued residency of plasmid DNA in the lymph node after administration (as in example 12) and the usual kinetics of CTL response following disappearance of antigen, this schedule will be sufficient to maintain the immunologic CTL response.


Example 14

Additional Epitopes.


The methodologies described above, and in particular in examples 3-7, have been applied to additional synthetic peptide substrates, leading to the identification of further epitopes as set for the in tables 15-36 below. The substrates used here were designed to identify products of housekeeping proteasomal processing that give rise to HLA-A*0201 binding epitopes, but additional MHC-binding reactivities can be predicted, as discussed above. Many such reactivities are disclosed, however, these listings are meant to be exemplary, not exhaustive or limiting. As also discussed above, individual components of the analyses can be used in varying combinations and orders. The digests of the NY-ESO-1 substrates 136-163 and 150-177 (SEQ ID NOS. 254 and 255, respectively) yielded fragments that did not fly well in MALDI-TOF mass spectrometry. However, they were quite amenable to N-terminal peptide pool sequencing, thereby allowing identification of cleavage sites. Not all of the substrates necessarily meet the formal definition of an epitope cluster as referenced in example 3. Some clusters are so large, e.g. NY-ESO-186-171, that it was more convenient to use substrates spanning only a portion of this cluster. In other cases, substrates were extended beyond clusters meeting the formal definition to include neighboring predicted epitopes. In some instances, actual binding activity may have dictated what substrate was made, as with for example the MAGE epitopes reported here, where HLA binding activity was determined for a selection of peptides with predicted affinity, before synthetic substrates were designed.

TABLE 15GP100: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion†Scores are given from the two binding prediction programs referenced above(see example 3).HLA Binding PredictionsSEQ(SYFPEITHI/NIH)†SubstrateEpitopeSequenceID NOA*0201A1A3B7B8Comments609-644630-638*LPHSSSHWL8820/8016/<5*The digestion of629-638*QLPHSSSHW8921/117609-644 and 622-    L650 have614-622LIYRRRLMK9032/20generated the613-622SLIYRRRLMK9114/<529/60same epitopes.615-622IYRRRLMK9215/<5622-650630-638*LPHSSSHWL9320/8016/<5629-638*QLPHSSSHW9421/117    L










TABLE 16A










MAGE-1: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion


















SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†
















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















 86-109
 95-102
ESLFRAVI
95




16/<5








 93-102
ILESLFRAVI
96
21/<5

20/<5






 93-101
ILESLFRAV
97
23/<5






 92-101
CILESLFRAV
98
23/55






 92-100
CILESLFRA
99
20/138





263-292
263-271
EFLWGPRAL
100





A26 (R 21),














A24 (NIH 30)






264-271
FLWGPRAL
101




17/<5






264-273
FLWGPRALAE
102
16/<5

19/<5






265-274
LWGPRALAET
103
16/<5






268-276
PRALAETSY
104
15/<5






267-276
GPRALAETSY
105
15/<5


<15/<5

B4403 (NIH 7);














B3501 (NIH 120)






269-277
RALAETSYV
106
18/20






271-279
LAETSYVKV
107
19/<5






270-279
ALAETSYVKV
108
30/427

19/<5<5






272-280
AETSYVKVL
109
15/<5




B4403 (NIH 36)






271-280
LAETSYVKVL
110
18/<5


<15/<5






274-282
TSYVKVLEY
111

26/<5



B4403 (NIH 14)






273-282
ETSYVKVLEY
112

28/6



A26 (R 31),














B4403 (NIH 14)






278-286
KVLEYVIKV
113
26/743

16/<5

















TABLE 16B










MAGE-1: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















168-193
168-177
SYVLVTCLGL
114





A24 (NIH 300)







169-177
YVLVTCLGL
115
20/32

15/<5
<15/20






170-177
VLVTCLGL
116




17/<5





229-258
240-248
TQDLVQEKY
117

29/<5






239-248
LTQDLVQEKY
118

23/<5



A26 (R 22)






232-240
YGEPRKLLT
119

24/11






243-251
LVQEKYLEY
120

21/<5
21/<5


A26 (R 28)






242-251
DLVQEKYLEY
121

22/<5
19/<5


A26 (R 30)






230-238
SAYGEPRKL
122
21/<5




B5101 (25/121)





272-297
278-286
KVLEYVIKV
123
26/743

16/<5






277-286
VKVLEYVIKV
124
17/<5






276-284
YVKVLEYVI
125
15/<5

15/<5

17/<5






274-282
TSYVKVLEY
126

26/<5






273-282
ETSYVKVLEY
127

28/6






283-291
VIKVSARVR
128


20/<5






282-291
YVIKVSARVR
129


24/<5







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 17A










MAGE-2: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















107-126
115-122
ELVHFLLL
130




18/<5








113-122
MVELVHFLLL
131

21/<5



A26 (R 22)






109-116
ISRKMVEL
132




17/<5






108-116
AISRKMVEL
133
25/7

19/<5
16/12
26/<5






107-116
AAISRKMVEL
134
22/<5


14/36
n.p./16






112-120
KMVELVHFL
135
27/2800






109-117
ISRKMVELV
136
16/<5






108-117
AISRKMVELV
137
24/11






116-124
LVHFLLLKY
138

23/<5
19/<5


A26 (R 26)






115-124
ELVHFLLLKY
139

24/<5
19/5


A26 (R 29)






111-119
RKMVELVHF
140





145-175
158-166
LQLVFGIEV
141
17/168






157-166
YLQLVFGIEV
142
24/1215






159-167
QLVFGILEVV
143
25/32

18/<5






158-167
LQLVFGIEVV
144
18/20






164-172
IEVVEVVPI
145
16/<5






163-172
GIEVVEVVPI
146
22/<5






162-170
FGIEVVEVV
147
19/<5




B5101 (24/69.212)






154-162
ASEYLQLVF
148

22/68






153-162
KASEYLQLVF
149


15/<5







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 17B










MAGE-2: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion











HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence

A*0201
A1
A3
B7
B8
Other




















213-233
218-225
EEKIWEEL
150




22/<5








216-225
APEEKIWEEL
151
15/<5


22/72






216-223
APEEKIWE
152




18/<5






220-228
KIWEELSML
153
26/804

16/<5

16/<5
A26 (R 26)






219-228
EKIWEELSML
154





A26 (R 22)





271-291
271-278
FLWGPRAL
155




17/<5






271-279
FLWGPRALI
156
25/398

16/7






278-286
LIETSYVKV
157
23/<5






277-286
ALIETSYVKV
158
30/427

21/<5






276-284
RALIETSYV
159
18/19




B5101 (20/55)






279-287
IETSYVKVL
160
15/<5






278-287
LIETSYVKVL
161
22/<5




A26 (R 22)







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 18










MAGE-3: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other





267-286
271-278
FLWGPRAL
162




17/<5







270-278
EFLWGPRAL
163





A26 (R 21);














A24 (NIH 30)






271-279
FLWGPRALV
164
27/2655

16/<5






276-284
RALVETSYV
165
18/19




B5101 (20/55)






272-280
LWGPRALVE
166


15/<5






271-280
FLWGPRALVE
167
15/<5

22/<5






272-281
LWGPRALVET
168
16/<5







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 19A










NY-ESO-1: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/(NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















 81-113
 82-90
GPESRLLEF
169

16/11

18/<5
22/<5








 83-91
PESRLLEFY
170

15/<5



B4403 (NIH 18)






 82-91
GPESRLLEFY
171

25/11






 84-92
ESRLLEFYL
172




19/8






 86-94
RLLEFYLAM
173
21/430

21/<5






 88-96
LEFYLAMPF
174





B4403 (NIH 60)






 87-96
LLEFYLAMPF
175

<15/45
18/<5






 93-102
AMPFATPMEA
176
15/<5






 94-102
MPFATPMEA
177



17/<5





101-133
115-123
PLPVPGVLL
178
20/<5

17/<5
16/<5
18/<5






114-123
PPLPVPGVLL
179



23/12






116-123*
LPVPGVLL
180




16/<5
Comment






103-112
ELARRSLAQD
181
15/<5

20/<5


*Evidence of the






118-126*
VPGVLLKEF
182



17/<5
16/<5
same epitope






117-126*
PVPGVLLKEF
183


16/<5


obtained from





116-145
116-123*
LPVPGVLL
184




16/<5
two digests.






127-135
TVSGNILTI
185
21/<5

19/<5






126-135
FTVSGNILTI
186
20/<5






120-128
GVLLKEFTV
187
20/130

18/<5






121-130
VLLKEFTVSG
188
17/<5

18/<5






122-130
LLKEFTVSG
189
20/<5

18/<5






118-126*
VPGVLLKEF
190



17/<5
16/<5






117-126*
PVPGVLLKEF
191


16/<5







†Scores are given from the two binding prediction programs referenced above (see example 3).















TABLE 19B










NY-ESO-1: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















136-163
139-147
AADHRQLQL
192
17/<5
17/<5


22/<5







(SEQ ID NO 254)
148-156
SISSCLQQL
193
24/7




A26 (R 25)






147-156
LSISSCLQQL
194
18/<5






138-147
TAADHRQLQL
195
18/<5





150-177
161-169
WITQCFLPV
196
18/84





(SEQ ID NO 255)
157-165
SLLMWITQC
197
18/42

17/<5






150-158
SSCLQQLSL
198
15/<5






154-162
QQLSLLMWI
199
15/50






151-159
SCLQQLSLL
200
18/<5






150-159
SSCLQQLSLL
201
16/<5






163-171
TQCFLPVFL
202
<15/12






162-171
ITQCFLPVFL
203
18/<5




A26 (R 19)







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score















TABLE 20










PRAME: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















211-245
219-227
PMQDIKMIL
204
16/<5



16/n.d.
A26 (R 20)







218-227
MPMQDIKMIL
205



<15/240





411-446
428-436
QHLIGLSNL
206
18/<5






427-436
LQHLIGLSNL
207
16/8






429-436
HLIGLSNL
208




17/<5
B15 (R 21)






431-439
IGLSNLTHV
209
18/7




B*5101 (R 22)






430-439
LIGLSNLTHV
210
24/37







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 21










PSA: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















42-77
53-61
VLVHPQWVL
211
22/112


<15/6
17/<5








52-61
GVLVHPQWVL
212
17/21

16/<5
<15/30

A26 (R 18)






52-60
GVLVHPQWV
213
17/124






59-67
WVLTAAHCI
214
15/16






54-63
LVHPQWVLTA
215
19/<5

20/<5


A26 (R 16)






53-62
VLVHPQWVLT
216
17/22






54-62
LVHPQWVLT
217


17/n.d.





55-95
66-73
CIRNKSVI
218




26/20






65-73
HCIRNKSVI
219




<15/16






56-64
HPQWVLTAA
220



18/<5






63-72
AAHCIRNKSV
221
17/<5







†Scores are given from the two binding prediction programs referenced above (see example 3). R indicates a SYFPEITHI score.















TABLE 22










PSCA: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















93-123*
116-123
LLWGPGQL
222




16/<5








115-123
LLLWGPGQL
223
<15/18






114-123
GLLLWGPGQL
224
<15/10






 99-107
ALQPAAAIL
225
26/9

22/<5
<15/12
16/<5
A26 (R 19)






 98-107
HALQPAAAIL
226
18/<5


<15/12







*L123 is the C-terminus of the natural protein.





†Scores are given from the two binding prediction programs referenced above (see example 3).















TABLE 23










Tyrosinase: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















128-157
128-137
APEKDKFFAY
227

29/6

15/<5

B4403 (NIH 14)







129-137
PEKDKFFAY
228

18/<5


21/<5






130-138
EKDKFFAYL
229



15/<5






131-138
KDKFFAYL
230




20/<5





197-228
205-213
PAFLPWHRL
231




15/<5






204-213
APAFLPWHRL
232



23/360






207-216
FLPWHRLFLL
1
25/1310



<15/8






208-216
LPWHRLFLL
9
17/26


20/80
24/16






214-223
FLLRWEQEIQ
233


15/<5






212-220
RLFLLRWEQ
234


16/<5





191-211
191-200
GSEIWRDIDF
235

18/68






192-200
SEIWRDIDF
236




16/<5
B4403 (NIH 400)





207-230
207-215
FLWHRLFL
8
22/540


<15/6
17/<5





466-484
473-481
RIWSWLLGA
237
19/13

15/<5





476-497
476-484
SWLLGAAMV
238
18/<5






477-486
WLLGAAMVGA
239
21/194

18/<5






478-486
LLGAAMVGA
240
19/19

16/<5







†tScores are given from the two binding prediction programs referenced above (see example 3).















TABLE 24










PSMA: Preferred Epitopes Revealed by Housekeeping Proteasome Digestion












SEQ ID
HLA Binding Predictions (SYFPEITHI/NIH)†

















Substrate
Epitope
Sequence
NO
A*0201
A1
A3
B7
B8
Other




















 1-30
 4-12
LLHETDSAV
241
25/485

15/<5










 13-21
ATARRPRWL
242
18/<5



18/<5
A26 (R 19)





 53-80
 53-61
TPKHNMKAF
243




24/<5






 64-73
ELKAENIKKF
244


17/<5


A26 (R 30)






 69-77
NIKKFLH1NF
245





A26 (R 27)






 68-77
ENIKKFLH1NF
246





A26 (R 24)





215-244
220-228
AGAKGVILY
247

25/<5





457-489
468-477
PLMYSLVHNL
248
22/<5






469-477
LMYSLVHNL
249
27/193

<15/9






463-471
RVDCTPLMY
250

32/125
25/<5


A26 (R 22)






465-473
DCTPLMYSL
251





A26 (R 22)





503-533
507-515
SGMPRISKL
252
21/<5



21<5






506-5151
FSGMPRISKL
253
17/<5








1This H was reported as Y in the SWISSPROT database.





†Scores are given from the two binding prediction programs referenced above (see example 3).















TABLE 25A










MAGE-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





Mage-1
125-
KAEMLESV
256
B5101
19
n.a.



119-
132





146
124-
TKAEMLESV
257
A0201
20
<5



132






123-
VTKAEMLESV
258
A0201
20
<5



132






128-
MLESVIKNY
259
A1
28
45






136


A26
24
n.a.









A3
17
5






127-
EMLESVIKNY
260
A1
15
<1.0






136


A26
23
<1.0






125-
KAEMLESVI
261
B5101
23
100






133


A24
N.A.
4





Mage-1
146-
KASESLQL
262
B08
16
<1.0





143-
153


B5101
17
N.A.





170
145-
GKASESLQL
263
B2705
17
1






153


B2709
16
N.A.






147-
ASESLQLVF
264
A1
22
68



155






153-
LVFGIDVKE
265
A26
16
N.A.






161


A3
16
<1.0

















TABLE 25B










MAGE-1: Preferred Epitopes Revealed by House-



keeping in Proteasome Digestion











Seq.
Binding Prediction














Sub-
Epi-

ID
HLA




strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















Mage-1
114-
LLKYRARE
266
B8
25
<1.0



99-125
121






106-
VADLVGFL
267
B8
16
<1.0






113


B5101
21
N.A.






105-
KVADLVGFL
268
A0201
23
44






113


A26
25
N.A.









A3
16
<5









B0702
14
20









B2705
14
30






107-
ADLVGFLLL
269
A0201
17
<5






115


B0702
15
<5









B2705
16
1






106-
VADLVGFLLL
270
A0201
16
<5






115


A1
22
3






114-
LLKYRAREPV
271
A0201
20
2



123

















TABLE 26










MAGE-3: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















Mage-3
271-
FLWGPRAL
162
B08
17
<5



267-
278





295
270-
EFLWGPRAL
163
A26
21
N.A.






278


A24
N.A.
30









B1510
16
N.A.






271-
FLWGPRALV
164
A0201
27
2655






279


A3
16
2






278-
LVETSYVKV
272
A0201
19
<1.0






286


A26
17
N.A.






277-
ALVETSYVKV
273
A0201
28
428






286


A26
16
<5









A3
18
<5






285-
KVLHHMVKI
274
A0201
19
27






293


A3
19
<5






276-
RALVETSYV
165
A0201
18
20



284






283-
YVKVLHHMV
275
A0201
17
<1.0



291






275-
PRALVETSY
276
A1
17
<1.0



283






274-
GPRALVETSY
277
A1
15
<1.0



283






278-
LVETSYVKVL
278
A0201
18
<1.0



287






272-
LWGPRALVET
168
A0201
16
<1.0



281






271-
FLWGPRALVE
167
A3
22
<5



280

















TABLE 27A










Fibronectin ED-B: Preferred Epitopes Revealed



by Housekeeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















ED-B
4′-

TIIPEVPQL

279
A0201
27
7






14′-
5**


A26
28
N.A.





21*



A3
17
<5









B8
15
<5









B1510
15
N.A.









B2705
17
10









B2709
15
N.A.






5′-

DTIIPEVPQL

280
A0201
20
<5






5**


A26
32
N.A.






1-10
EVPQLTDLSF
281
A26
29
N.A.







*This substrate contains the 14 amino acids from fibronectin flanking ED-B to the N-terminal side.





**These peptides span the junction between the N-terminus of the ED-B domain and the rest of fibronectin.






The italicized lettering indicates sequence outside the ED-B domain.















TABLE 27B










Fibronectin ED-B: Preferred Epitopes Revealed



by Housekeeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH





ED-B
23-30
TPLNSSTI
282
B5101
22
N.A.






8-35
18-25
IGLRWTPL
283
B5101
18
N.A.






17-25
SIGLRWTPL
284
A0201
20
5









A26
18
N.A.









B08
25
<5






25-33
LNSSTIIGY
285
A1
19
<5









A26
16
<5






24-33
PLNSSTIIGY
286
A1
20
<5









A26
24
N.A.









A3
16
<5






23-31
TPLNSSTII
287
B0702
17
8









B5101
25
440

















TABLE 27C










Fibronectin ED-B: Preferred Epitopes Revealed



by Housekeeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





ED-B
31-38
IGYRITVV
288
B5101
25
N.A.






20-49
30-38
IIGYRITVV
289
A0201
23
15









A3
17
<1.0









B08
15
<1.0









B5101
15
3






29-38
TIIGYRITVV
290
A0201
26
9









A26
18
N.A.









A3
18
<5






23-30
TPLNSSTI
282
B5101
22
N.A.






25-33
LNSSTIIGY
285
A1
19
<5









A26
16
N.A.






24-33
PLNSSTIIGY
286
A26
24
N.A.









A3
16
<5






31-39
IGYRITVVA
291
A3
17
<5






30-39
IIGYRITVVA
292
A0201
15
<5









A3
18
<5






23-31
TPLNSSTII
287
B0702
17
8









B5101
25
440

















TABLE 28A










CEA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















CEA
184-
SLPVSPRL
293
B08
19
<5



176-
191





202
183-
QSLPVSPRL
294
A0201
15
<5






191


B1510
15









B2705
18
10









B2709
15






186-
PVSPRLQL
295
B08
18
<5



193






185-
LPVSPRLQL
296
B0702
26
180






193


B08
16
<5









B5101
19
130






184-
SLPVSPRLQL
297
A0201
23
21






193


A26
18
N.A.









A3
18
<5






185-
LPVSPRLQ
298
B5101
17
N.A.



192






192-
QLSNGNRTL
299
A0201
21
4






200


A26
16
N.A.









A3
19
<5









B08
17
<5









B1510
15






191-
LQLSNGNRTL
300
A0201
16
3



200






179-
WVNNQSLPV
301
A0201
16
28


187






186-
PVSPRLQLS
302
A26
17
N.A.






194


A3
15
<5

















TABLE 28B










CEA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















CEA
362-
SLPVSPRL
303
B08
19
<1.0



354-
369





380
361-
QSLPVSPRL
304
A0201
15
<1.0






369


B2705
18
10









B2709
15






364-
PVSPRLQL
305
B08
18
<1.0



371






363-
LPVSPRLQL
306
B0702
26
180






371


B08
16
<1.0









B5101
19
130






362-
SLPVSPRLQL
307
A0201
23
21






371


A26
18
N.A.









A24
N.A.
6









A3
18
<5






363-
LPVSPRLQ
308
B5101
17
N.A.



370






370-
QLSNDNRTL
309
A0201
22
4






378


A26
16
N.A.









A3
17
<1.0









B08
17
<1.0






369-
LQLSNDNRTL
310
A0201
16
3



378






357-
WVNNQSLPV
311
A0201
16
28



365






360-
NQSLPVSPR
312
B2705
14
100



368

















TABLE 28C










CEA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















CEA
540-
SLPVSPRL
313
B08
19
<5



532-
547





558
539-
QSLPVSPRL
314
A0201
15
<5






547


B1510
15
<5









B2705
18
10









B2709
15






542-
PVSPRLQL
315
B08
18
<5



549






541-
LPVSPRLQL
316
B0702
26
180






549


B08
16
<1.0









B5101
19
130






540-
SLPVSPRLQL
317
A0201
23
21






549


A26
18
N.A.









A3
18
<5



541-
LPVSPRLQ
318
B5101
17
N.A.



548






548-
QLSNGNRTL
319
A0201
24
4






556


A26
16
N.A.









A3
19
<1.0









B08
17
<1.0









B1510
15






547-
LQLSNGNRTL
320
A0201
16
3



556






535-
WVNGQSLPV
321
A0201
18
28



543


A3
15
<1.0






533-
LWWVNGQSL
322
A0201
15
<5



541

















TABLE 28D










CEA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















CEA
532-
YLWWVNGQSL
323
A0201
25
816






532-
541


A26
18
N.A.





558
538-
GQSLPVSPR
324
B2705
17
100


(con-
546


tin-


ued)

















TABLE 29A










HER2/NEU: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















Her-2
30-37
DMKLRLPA
325
B08
19
8






25-52
28-37
GTDMKLRLPA
326
A1
23
6






42-49
HLDMLRHL
327
B08
17
<5






41-49
THLDMLRHL
328
A0201
17
<5









B1510
24
N.A.






40-49
ETHLDMLRHL
329
A26
29
N.A.






36-43
PASPETHL
330
B5101
17
N.A.






35-43
LPASPETHL
331
A0201
15
<5









B5101
20
130









B5102
N.A.
100






34-43
RLPASPETHL
332
A0201
20
21






38-46
SPETHLDML
333
A0201
15
<5









B0702
20
24









B08
18
<5









B5101
18
110






37-46
ASPETHLDML
334
A0201
18
<5






42-50
HLDMLRHLY
335
A1
29
25









A26
20
N.A.









A3
17
4






41-50
THLDMLRHLY
336
A1
18
<1.0

















TABLE 29B










HER2/NEU: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















Her-2
719-
ELRKVKVL
337
B08
24
16



705-
726





732
718-
TELRKVKVL
338
A0201
16
1






726


B08
22
<5









B5101
16
<5






717-
ETELRKVKVL
339
A1
18
2






726


A26
28
6






715-
LKETELRKV
340
A0201
17
<5






723


B5101
15
<5






714-
ILKETELRKV
341
A0201
29
8



723






712-
MRILKETEL
342
A0201
15
<5






720


B08
22
<5









B2705
27
2000









B2709
21
N.A.






711-
QMRILKETEL
343
A0201
20
2






720


B0702
13
40






717-
ETELRKVKV
344
A1
18
5






725


A26
18
N.A.






716-
KETELRKVKV
345
A0201
16
19



725






706-
MPNQAQMRI
346
B0702
16
8






714


B5101
22
629






705-
AMPNQAQMRI
347
A0201
18
8



714






706-
MPNQAQMRIL
348
B0702
20
80



715
















TABLE 29C










HER2/NEU: Preferred Epitopes Revealed by House-


keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















Her-2
966-
RPRFRELV
349
B08
20
24



954-
973


B5101
18
N.A.





982
965-
CRPRFRELV
350
B2709
18



973






968-
RFRELVSEF
351
A26
25
N.A.






976


A24
N.A.
32









A3
15
<5









B08
16
<5









B2705
19






967-
PRFRELVSEF
352
A26
18
N.A.



976






964-
ECRPRFREL
353
A26
21
N.A.






972


A24
N.A.
6









B0702
15
40









B8
27
640









B1510
16
<5

















TABLE 30










NY-ESO-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















NY-
67-75
GAASGLNGC
354
A0201
15
<5






ESO-1
52-60
RASGPGGGA
355
B0702
15
<5





51-77
64-72
PHGGAASGL
356
B1510
21
N.A.






63-72
GPHGGAASGL
357
B0702
22
80






60-69
APRGPHGGAA
358
B0702
23
60

















TABLE 31A










PRAME: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





PRAME
112-
VRPRRWKL
359
B08
19




103-
119





135
111-
EVRPRRWKL
360
A26
27
N.A.






119


A24
N.A.
5









A3
19
N.A.









B0702
15
(B7)











300.00









B08
26
160






113-
RPRRWKLQV
361
B0702
21
(B7)






121




40.00









B5101
19
110






114-
PRRWKLQVL
362
B08
26
<5






122


B2705
23
200






113-
RPRRWKLQVL
363
B0702
24
(B7)






122




800.00









B8
N.A.
160









B5101
N.A.
61









B5102
N.A.
61









A24
N.A.
10






116-
RWKLQVLDL
364
B08
22
<5






124


B2705
17
3






115-
RRWKLQVLDL
365
A0201
16
<5



124





PRAME
174-
PVEVLVDLF
366
A26
25
N.A.


161-
182


187

















TABLE 31B










PRAME: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















PRAME
199-
VKRKKNVL
367
B08
27
8



185-
206





215
198-
KVKRKKNVL
368
A0201
16
<1.0






206


A26
20
N.A.









A3
22
<1.0









B08
30
40









B2705
16






197-
EKVKRKKNVL
369
A26
15
N.A.



206






198-
KVKRKKNV
370
B08
20
6



205






201-
RKKNVLRL
371
B08
20
<5



208






200-
KRKKNVLRL
372
A0201
15
<1.0



208


A26
15
N.A.









B0702
15
<1.0









B08
21
<1.0









B2705
28









B2709
25






199-
VKRKKNVLRL
373
A0201
16
<1.0






208


B0702
16
4






189-
DELFSYLI
374
B5101
15
N.A.



196






205-
VLRLCCKKL
375
A0201
22
3






213


A26
17
N.A.









B08
25
8






204-
NVLRLCCKKL
376
A0201
17
7






213


A26
19
N.A.

















TABLE 31C










PRAME: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















PRAME
194-
YLIEKVKRK
377
A0201
20
<1.0






185-
202


A26
18
N.A.





215



A3
25
68





(con-



B08
20
<1.0





tin-



B2705
17





ued)





PRAME
 74-
QAWPFTCL
378
B5101
17
n.a.


 71-
 81





 98
 73-
VQAWPFTCL
379
A0201
14
7






 81


A24
n.a.
5









B0702
16
6






 72-
MVQAWPFTCL
380
A26
22
n.a.






 81


A24
n.a.
7









B0702
13
30






 81-
LPLGVLMK
381
B5101
18
n.a.



 88






 80-
CLPLGVLMK
382
A0201
17
<1.0






 88


A3
27
120






 79-
TCLPLGVLMK
383
A1
12
10






 88


A3
19
3






 84-
GVLMIKGQHL
384
A0201
18
7






 92


A26
21
n.a.









B08
21
4






 81-
LPLGVLMKG
385
B5101
20
2



 89






 80-
CLPLGVLMKG
386
A0201
16
<1.0



 89






 76-
WPFTCLPLGV
387
B0702
18
4



 85

















TABLE 31D










PRAME: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















PRAME
51-59
ELFPPLFMA
388
A0201
19
18






39-65



A26
23
N.A.






49-57
PRELFPPLF
389
B2705
22









B2709
19






48-57
LPRELFPPLF
390
B0702
19
4



50-58
RELFPPLFM
391
B2705
16









B2705
15






49-58
PRELFPPLFM
392
A1
16
<1.0

















TABLE 32










PSA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





PSA
239-
RPSLYTKV
393
B5101
21
N.A.



232-
246





258
238-
ERPSLYTKV
394
B2705
15
60



246






236-
LPERPSLY
395
B5101
18
N.A.



243






235-
ALPERPSLY
396
A1
19
<1.0






243


A26
22
N.A.









A3
26
6









B08
16
<1.0









B2705
11
15









B2709
19
N.A.






241-
SLYTKVVHY
397
A0201
20
<1.0






249


A1
19
<1.0









A26
25
N.A.









A3
26
60









B08
20
<1.0









B2705
13
75






240-
PSLYTKVVHY
398
A1
20
<1.0






249


A26
16
N.A.






239-
RPSLYTKVV
399
B0702
21
4






247


B5101
23
110

















TABLE 33A










PSMA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















PSMA
211-
GNKVKNAQ
400
B08
22
<5



202-
218





228
202-
IARYGKVF
401
B08
18
<5



209






217-
AQLAGAKGV
402
A0201
16
26



225






207-
KVFRGNKVK
403
A3
32
15



215






211-
GNKVKNAQL
404
B8
33
80






219


B2705
17
20





PSMA
269-
TPGYPANEY
405
A1
16
<5


255-
277





282
268-
LTPGYPANEY
406
A1
21
1






277


A26
24
N.A.






271-
GYPANEYAY
407
A1
15
<5



279






270-
PGYPANEYAY
408
A1
19
<5



279






266-
DPLTPGYPA
409
B0702
21
3






274


B5101
17
20





PSMA
492-
SLYESWTKK
410
A0201
17
<5





483-
500


A3
27
150





509



B2705
18
150






491-
KSLYESWTKK
411
A3
16
<5



500






486-
EGFEGKSLY
412
A1
19
<5






494


A26
21
N.A.









B2705
16
<5






485-
DEGFEGKSLY
413
A1
17
<5






494


A26
17
N.A.






498-
TKKSPSPEF
414
B08
17
<5



506

















TABLE 33B










PSMA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





PSMA
497-
WTKKSPSPEF
415
A26
24
N.A.



483-
506





509
492-
SLYESWTKKS
416
A0201
16
<5





(con-
501


A3
16
<5





tin-





ued)





PSMA
725-
WGEVKRQI
417
B08
17
<5





721-
732


B5101
17
N.A.





749
724-
AWGEVKRQI
418
B5101
15
6



732






723-
KAWGEVKRQI
419
A0201
16
<1.0



732






723-
KAWGEVKR
420
B5101
15
N.A.



730






722-
SKAWGEVKR
421
B2705
15
<5



730






731-
QIYVAAFTV
422
A0201
21
177






739


A3
21
<1.0









B5101
15
5






733-
YVAAFTVQA
423
A0201
17
6






741


A3
20
<1.0






725-
WGEVKRQIY
424
A1
26
11



733






727-
EVKRQIYVA
425
A26
22
N.A.






735


A3
18
<1.0






738-
TVQAAAETL
426
A26
18
N.A.






746


A3
19
<1.0






737-
FTVQAAAETL
427
A0201
17
<1.0






746


A26
19
N.A.

















TABLE 33C










PSMA: Preferred Epitopes Revealed by Housekeep-



ing Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





PSMA
729-
KRQIYVAAF
428
A26
16
N.A.






721-
737


B2705
24
3000





749



B2709
21
N.A.





(con-
721-
PSKAWGEVK
429
A3
20
<1.0


tin-
729





ued)
723-
KAWGEVKRQ
430
B5101
16
<1.0



731





PSMA
100-
WKEFGLDSV
431
A0201
16
<5


 95-
108





122
 99-
QWKEFGLDSV
432
A0201
17
<5



108






102-
EFGLDSVELA
433
A26
16
N.A.



111

















TABLE 34A










SCP-1: Preferred Epitopes Revealed by Housekeeping Proteasome



Digestion










Binding Prediction














Substrate
Epitope
Sequence
Seq. ID No.
HLA type
SYFPEITHI
NIH

















SCP-1 117-143
126-134
ELRQKESKL
434
A0201
20
<5










A26
26
N.A.









A3
17
<5









B0702
13
(B7)








40.00









B8
34
320






125-134
AELRQKESKL
435
A0201
16
<5






133-141
KLQENRKII
436
A0201
20
61





SCP-1 281-308
298-305
QLEEKTKL
437
B08
28
2






297-305
NQLEEKTKL
438
A0201
16
33









B2705
19
200






288-296
LLEESRDKV
439
A0201
25
15









B5101
15
3






287-296
FLLEESRDKV
440
A0201
27
2378






291-299
ESRDKVNQL
441
A26
21
N.A.









B08
29
240






290-299
EESRDKVNQL
442
A26
19
N.A.





SCP-1 471-498
475-483
EKEVHDLEY
443
A1
31
11









A26
17
N.A.






474-483
REKEVHDLEY
444
A1
21
<1.0






480-488
DLEYSYCHY
445
A1
26
45









A26
30
N.A.









A3
16
<5






477-485
EVHDLEYSY
446
A1
15
1

















TABLE 34B










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





SCP-1
477-
EVHDLEYSY

A26
29
N.A.






471-
485


A3
19
<1.0





498
477-
EVHDLEYSYC
447
A26
22
N.A.


(con-
486





tin-





ued)





SCP-1
502-
KLSSKREL
448
B08
26
4


493-
509





520
508-
ELKNTEYF
449
B08
24
<1.0



515






507-
RELKNTEYF
450
B2705
18
45






515


B4403
N.A.
120






496-
KRGQRPKL
451
B08
18
<1.0



503






494-
LPKRGQRPKL
452
B0702
22
120






503


B8
N.A.
16









B5101
N.A.
130









B3501
N.A.
60






509-
LKNTEYFTL
453
A0201
15
<5



517






508-
ELKNTEYFTL
454
A0201
18
<1.0






517


A26
27
N.A.









A3
16
<1.0






506-
KRELKNTEY
455
A1
26
2






514


B2705
26
3000






502-
KLSSKRELK
456
A3
25
60



510






498-
GQRPKLSSK
457
A3
22
4






506


B2705
18
200






497-
RGQRPKLSSK
458
A3
22
<1.0



506






500-
RPKLSSKRE
459
B08
18
<1.0



508

















TABLE 34C










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















SCP-1
573-
LEYVREEL
460
B08
19
<5



570-
580





596
572-
ELEYVREEL
461
A0201
17
<1.0






580


A26
23
N.A.









A24
N.A.
9









B08
20
N.A.






571-
N ELEYVREEL
462
A0201
16
4



580






579-
ELKQKRDEV
463
A0201
19
<1.0






587


A26
18
N.A.









B08
29
48






575-
YVREELKQK
464
A26
17
N.A.






583


A3
27
2





SCP-1
632-
QLNVYEIKV
465
A0201
24
70


618-
640





645
630-
SKQLNVYEI
466
A0201
17
<5



638






628-
AESKQLNVY
467
A1
19
<5






636


A26
16
N.A.






627-
TAESKQLNVY
468
A1
26
45






636


A26
15
N.A.

















TABLE 34D










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















SCP-1
638-
IKVNKLEL
469
B08
21
<1.0



633-
645





660
637-
EIKVNKLEL
470
A0201
17
<1.0






645


A26
26
N.A.









B08
28
8









B1510
15
N.A.






636-
YEIKVNKLEL
471
A0201
17
2



645






642-
KLELELESA
472
A0201
20
1






650


A3
16
<1.0






635-
VYEIKVNKL
473
A0201
18
<1.0






643


A24
N.A.
396









B08
22
<1.0






634-
NVYEIKVNKL
474
A0201
24
56






643


A26
25
N.A.









A24
N.A.
6









A3
15
<5









B0702
11
(B7)











20









B08
N.A.
6






646-
ELESAKQKF
475
A26
27
N.A.



654





SCP-1
642-
KLELELESA
476
A0201
20
1





640-
650


A3
16
<1.0





668
646-
ELESAKQKF
477
A26
27
N.A.






654

















TABLE 34E










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















SCP-1
771-
KEKLKREA
478
B08
21
<5



768-
778





796
777-
EAKENTATL
479
A0201
18
<5






785


A26
18
N.A.









A24
N.A.
5









B0702
13
12









B08
28
48









B5101
20
121






776-
REAKENTATL
480
A0201
16
<5



785






773-
KLKREAKENT
481
A3
17
<5



782





SCP-1
112-
EAEKIKKW
482
B5101
17
N.A.


 92-
119





125
101-
GLSRVYSKL
483
A0201
23
32






109


A26
22
N.A









A24
N.A.
6









A3
17
3









B08
17
<1.0






100-
EGLSRVYSKL
484
A26
21
N.A.






109


A24
N.A.
9






108-
KLYKEAEKI
485
A0201
22
57






116


A3
20
9









B5101
18
5






 98-
NSEGLSRVY
486
A1
31
68



106






 97-
ENSEGLSRVY
487
A26
18
N.A.



106






102-
LSRVYSKLY
488
A1
22
<1.0



110

















TABLE 34F










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















SCP-1
101-
GLSRVYSKLY
489
A1
18
<1.0






 92-
110


A26
18
N.A.





125



A3
19
18





(con-
96-
LENSEGLSRV
490
A0201
17
5





tin-
105





ued)
108-
KLYKEAEKIK
491
A3
27
150



117





SCP-1
949-
REDRWAVI
492
B5101
15
N.A.


931-
956





958
948-
MREDRWAVI
493
B2705
18
600






956


B2709
18
N.A.









B5101
15
1






947-
KMREDRWAVI
494
A0201
21
6






956


B08
N.A.
15






947-
KMREDRWAV
495
A0201
22
411



955






934-
TTPGSTLKF
496
A26
25
N.A.



942






933-
LTTPGSTLKF
497
A26
23
N.A.



942






937-
GSTLKFGAI
498
B08
19
1



945






945-
IRKMREDRW
499
B08
19
<5



953





SCP-1
236-
RLEMHFKL
500
B08
16
<5


232-
243





259
235-
SRLEMHFKL
501
A0201
18
<5






243


B2705
25
2000









B2709
22






242-
KILKEDYEKI
502
A0201
22
4



250

















TABLE 34G










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





SCP-1



A26
16
N.A.






232-259



A3
15
3





(con-



B08
24
<5





tinued)



B5101
14
2






249-
KIQHLEQEY
503
A1
15
<5






257


A26
23
N.A.









A3
17
<5






248-
EKIQHLEQEY
504
A1
15
<5






257


A26
21
N.A.






233-
ENSRLEMHF
505
A26
19
N.A.



242






236-
RLEMHFKLKE
506
A1
19
<5






245


A3
17
<5






324-LEDIKVSL
507
B08
20
<1.0


SCP-1
331





310-340
323-
ELEDIKVSL
508
A0201
21
<1.0






331


A26
25
N.A.









A24
N.A.
10









A3
17
<1.0









B08
19
<1.0









B1510
16
N.A.






322-
KELEDIKVSL
509
A0201
19
22



331






320-
LTKELEDI
500
B08
18
<5



327






319-
HLTKELEDI
511
A0201
21
<1.0



327






330-
SLQRSVSTQ
512
A0201
18
<1.0



338

















TABLE 34H










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















SCP-1
321-
TKELEDIKV
513
A1
16
<1.0



310-
329





340
320-
LTKELEDIKV
514
A0201
19
<1.0


(con-
329





tin-
326-
DIKVSLQRSV
515
A26
18
N.A.


ued)
335





SCP-1
281-
KMKDLTFL
516
B08
20
3


272-
288





305
280-
NKMKDLTFL
517
A0201
15
1



288






279-
ENKMKDLTFL
518
A26
19
N.A.



288






288-
LLEESRDKV
519
A0201
25
15






296


B5101
15
3






287-
FLLEESRDKV
520
A0201
27
2378



296






291-
ESRDKVNQL
521
A26
21
N.A.






299


B08
29
240






290-
EESRDKVNQL
522
A26
19
N.A.



299






277-
EKENKMKDL
523
A26
19
N.A.






285


B08
23
<1.0






276-
TEKENKMKDL
524
A26
15
N.A.



285






279-
ENKMKDLTF
525
A26
18
N.A.






287


B08
28
4





SCP-1
218-
IEKMITAF
526
B08
17
<5


211-
225





239
217-
NIEKMITAF
527
A26
26
N.A.



225






216-
SNIEKMITAF
528
A26
19
N.A.



225






223-
TAFEELRV
529
B5101
23
N.A.



230






222-
ITAFEELRV
530
A0201
18
2



230






221-
MITAFEELRV
531
A0201
18
16



230

















TABLE 34I










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















SCP-1
220-
KMITAFEEL
532
A0201
23
50






211-
228


A26
15
N.A.





239



A24
N.A.
16





(con-
219-
EKMITAFEEL
533
A26
19
N.A.


tin-
228





ued)
227-
ELRVQAENS
534
A3
16
<1.0






235


B08
15
<1.0






213-
DLNSNIEKMI
535
A0201
17
<1.0






222


A26
16
N.A.





SCP-1
837-
WTSAKNTL
536
B08
20
4


836-
844





863
846-
TPLPKAYTV
537
A0201
18
2






854


B0702
17
4









B08
16
2









B5101
25
220






845-
STPLPKAYTV
538
A0201
19
<5



854






844-
LSTPLPKAY
539
A1
23
8



852






843-
TLSTPLPKAY
540
A1
16
<1.0






852


A26
19
N.A.









A3
18
2






842-
NTLSTPLPK
541
A3
16
3



850






841-
KNTLSTPLPK
542
A3
18
<1.0



850

















TABLE 34J










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















SCP-1
828-
ISKDKRDY
543
B08
21
3






819-
835


A26
21
N.A.





845
826-
HGISKDKRDY
544
A1
15
<5



835






832-
KRDYLWTSA
545
B2705
16
600



840






829-
SKDKRDYLWT
546
A1
18
<5



838





SCP-1
279-
ENKMKDLT
547
B08
22
8


260-
286





288
260-
EINDKEKQV
548
A0201
17
3






268


A26
19
N.A.









B08
17
<5






274-
QITEKENKM
549
A0201
17
3






282


A26
22
N.A.









B08
16
<5






269-
SLLLIQITE
550
A0201
16
<1.0






277


A3
18
<1.0





SCP-1
453-
FEKIAEEL
551
B08
21
<1.0


437-
460





464
452-
QFEKIAEEL
552
B2705
15



460






451-
KQFEKIAEEL
553
A0201
16
56



460






449-
DNKQFEKI
554
B08
16
2






456


B5101
16
N.A.






448-
YDNKQFEKI
555
B5101
16
1



456






447-
LYDNKQFEKI
556
A1
15
<1.0



456

















TABLE 34K










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH





SCP-1
440-
LGEKETLL
557
B5101
16
N.A.



437-
447





464
439-
VLGEKETLL
558
A0201
24
149





(con-
447


A26
19
N.A.





tin-



B08
29
12





ued)
438-
KVLGEKETLL
559
A0201
19
24






447


A26
20
N.A.









A24
N.A.
12









A3
18
<1.0









B0702
14
20





SCP-1
390-
LLRTEQQRL
560
A0201
22
3





383-
398


A26
18
N.A.





412



B08
22
1.6









B2705
15
30






389-
ELLRTEQQRL
561
A0201
19
6






398


A26
24
N.A.









A3
15
<1.0






393-
TEQQRLENY
562
A1
15
<5






401


A26
16
N.A.






392-
RTEQQRLENY
563
A1
31
113






401


A26
26
N.A.






402-
EDQLIILTM
564
A26
18
N.A.



410






397-
RLENYEDQLI
565
A0201
17
<1.0






406


A3
15
<1.0

















TABLE 34L










SCP-1: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion











Seq.
Binding Prediction















Sub-
Epi-

ID
HLA





strate
tope
Sequence
No.
type
SYFPEITHI
NIH

















SCP-1
368-
KARAAHSF
566
B08
16
<1.0



366-
375





394
376-
VVTEFETTV
567
A0201
19
161






384


A3
16
<1.0






375-
FVVTEFETTV
568
A0201
17
106



384






377-
VTEFETTVC
569
A1
18
2



385






376-
VVTEFETTVC
570
A3
16
<5



385





SCP-1
344-
DLQLATNTI
571
A0201
22
<5





331-
352


A3
15
<1.0





357



B5101
17
11






347-
IATNTICQL
572
A0201
19
1






355


B08
16
<1.0









B5101
20
79






346-
QIATNTICQL
573
A0201
24
7






355


A26
24
N.A.

















TABLE 35










SSX-4: Preferred Epitopes Revealed by House-



keeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















SSX4
 57-
VMTKIGFKV
574
A0201
21
495



45-76
 65






 53-
LNYEVMTKL
575
A0201
17
7



 61






 52-
KNYEVMTK
576
A0201
23
172






 61
L

A26
21
N.A.









A24
N.A.
18









A3
14
4









B7
N.A.
4






 66-
TLPPFMRSK
577
A26
16
N.A.






 74


A3
25
14





SSX4
110-
KIMPKKPAE
578
A0201
15
<5





98-124
118


A26
15
N.A.









A3
16
<5






103-
SLQRIFPKIM
579
A0201
15
8






112


A26
16
N.A.









A3
15
<5

















TABLE 36










Tyrosinase: Preferred Epitopes Revealed by



Housekeeping Proteasome Digestion










Binding Prediction















Sub-
Epi-

Seq.
HLA





strate
tope
Sequence
ID No.
type
SYFPEITHI
NIH

















Tyr
463-
YIKSYLEQA
580
A0201
18
<5






445-
471


A26
17
N.A.





474
459-
SFQDYIKSY
581
A1
18
<5






467


A26
22
N.A.






458-
DSFQDYIKSY
582
A1
19
<5






467


A26
24
N.A.





Tyr
507-
LPEEKQPL
583
B08
28
5





490-
514


B5101
18
N.A.





518
506-
QLPEEKQPL
584
A0201
22
88






514


A26
20
N.A.









A24
N.A.
9









B08
18
<5






505-
KQLPEEKQPL
585
A0201
15
28






514


A24
N.A.
17






507-
LPEEKQPLL
586
A0201
15
<5






515


B0702
21
24









B08
28
5









B5101
21
157






506-
QLPEEKQPLL
587
A0201
23
88






515


A26
20
N.A.









A24
N.A.
7






497-
SLLCRHKRK
588
A3
25
15



505









Example 15

Evaluating Likelihood of Epitope Cross-reactivity on Non-target Tissues.


As noted above PSA is a member of the kallikrein family of proteases, which is itself a subset of the serine protease family. While the members of this family sharing the greatest degree of sequence identity with PSA also share similar expression profiles, it remains possible that individual epitope sequences might be shared with proteins having distinctly different expression profiles. A first step in evaluating the likelihood of undesirable cross-reactivity is the identification of shared sequences. One way to accomplish this is to conduct a BLAST search of an epitope sequence against the SWISSPROT or Entrez non-redundant peptide sequence databases using the “Search for short nearly exact matches” option; hypertext transfer protocol accessible on the world wide web (http://www) at “ncbi.nlm.nih.gov/blast/index.html”. Thus searching SEQ ID NO. 214, WVLTAAHCI, against SWISSPROT (limited to entries for homo sapiens) one finds four exact matches, including PSA. The other three are from kallikrein 1 (tissue kallikrein), and elastase 2A and 2B. While these nine amino acid segments are identical, the flanking sequences are quite distinct, particularly on the C-terminal side, suggesting that processing may proceed differently and that thus the same epitope may not be liberated from these other proteins. (Please note that kallikrein naming is confused. Thus the kallikrein 1 [accession number P06870] is a different protein than the one [accession number AAD13817] mentioned in the paragraph on PSA above in the section on tumor-associated antigens).


It is possible to test this possibility in several ways. Synthetic peptides containing the epitope sequence embedded in the context of each of these proteins can be subjected to in vitro proteasomal digestion and analysis as described above. Alternatively, cells expressing these other proteins, whether by natural or recombinant expression, can be used as targets in a cytotoxicity (or similar) assay using CD8+ T cells that recognize the epitope, in order to determine if the epitope is processed and presented.


Epitope Clusters.


Known and predicted epitopes are generally not evenly distributed across the sequences of protein antigens. As referred to above, we have defined segments of sequence containing a higher than average density of (known or predicted) epitopes as epitope clusters. Among the uses of epitope clusters is the incorporation of their sequence into substrate peptides used in proteasomal digestion analysis as described herein. Epitope clusters can also be useful as vaccine components. A fuller discussion of the definition and uses of epitope clusters is found in U.S. patent application Ser. No. 09/561,571 entitled EPITOPE CLUSTERS, previously incorporated by reference in its entirety.


Example 16

Melan-A/MART-1


This melanoma tumor-associated antigen (TAA) is 118 amino acids in length. Of the 110 possible 9-mers, 16 are given a score >16 by the SYFPEITHI/Rammensee algorithm. (See Table 37). These represent 14.5% of the possible peptides and an average epitope density on the protein of 0.136 per amino acid. Twelve of these overlap, covering amino acids 22-49 resulting in epitope density for the cluster of 0.428, giving a ratio, as described above, of 3.15. Another two predicted epitopes overlap amino acids 56-69, giving an epitope denisty for the cluster of 0.143, which is not appreciably different than the average, with a ratio of just 1.05. See FIG. 18.

TABLE 37SYFPEITHI (Rammensee algorithm)Results for Melan-A/MART-1RankStartScore131272562633526432255272562924734238612093319102219119918123618132818148717154117164016


Restricting the analysis to the 9-mers predicted to have a half time of dissociation of ≧5 minutes by the BIMAS-NIH/Parker algorithm leaves only 5. (See Table 38). The average density of epitopes in the protein is now only 0.042 per amino acid. Three overlapping peptides cover amino acids 31-48 and the other two cover 56-69, as before, giving ratios of 3.93 and 3.40, respectively. (See Table 39).

TABLE 38BIMAS-NIH/Parker algorithm Results for Melan-A/MART-1RankStartScoreLog(Score)1401289.013.112561055.1043.0233181.3851.9143520.7531.325614.9680.70









TABLE 39










Predicted Epitope Clusters for Melan-A/MART-1


Calculations(Epitopes/AAs)


















Whole




Cluster
AA
Peptides
Cluster
protein
Ratio







1
31-48
3, 4, 1
0.17
0.042
3.93



2
56-69
2, 5
0.14
0.042
3.40










Example 17

SSX-2/HOM-MEL-40


This melanoma tumor-associated antigen (TAA) is 188 amino acids in length. Of the 180 possible 9-mers, 11 are given a score ≧16 by the SYFPEITHI/Rammensee algorithm. These represent 6.1% of the possible peptides and an average epitope density on the protein of 0.059 per amino acid. Three of these overlap, covering amino acids 99-114 resulting in an epitope density for the cluster of 0.188, giving a ratio, as described above, of 3.18. There are also overlapping pairs of predicted epitopes at amino acids 16-28, 57-67, and 167-183, giving ratios of 2.63, 3.11, and 2.01, respectively. There is an additional predicted epitope covering amino acids 5-28. Evaluating the region 5-28 containing three epitopes gives an epitope density of 0.125 and a ratio 2.14.


Restricting the analysis to the 9-mers predicted to have a half time of dissociation of >5 minutes by the BIMAS-NIH/Parker algorithm leaves only 6. The average density of epitopes in the protein is now only 0.032 per amino acid. Only a single pair overlap, at 167-180, with a ratio of 4.48. However the top ranked peptide is close to another single predicted epitope if that region, amino acids 41-65, is evaluated the ratio is 2.51, representing a substantial difference from the average. See FIG. 19.

TABLE 40SYFPEITHI/Rammensee algorithm for SSX-2/HOM-MEL-40RankStartScore110323216722341224162159920659197201785179175161010616115716









TABLE 41










Calculations (Epitopes/AAs)


Calculations(Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio















1
5 to 28
8, 4, 7
0.125
0.059
2.14


2
16-28
4, 7
0.15
0.059
2.63


3
57-67
11, 6
0.18
0.059
3.11


4
 99-114
5, 1, 10
0.19
0.059
3.20


5
167-183
2, 9
0.12
0.059
2.01
















TABLE 42










BIMAS-NIH/Parker algorithm












Rank
Start
Score
Log(Score)
















1
41
1017.062
3.01



2
167
21.672
1.34



3
57
20.81
1.32



4
103
10.433
1.02



5
172
10.068
1.00



6
16
6.442
0.81

















TABLE 43










Calculations(Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio





1
41-65
1, 3
0.08
0.032
2.51


2
167-180
2, 5
0.14
0.032
4.48









Example 18

NY-ESO


This tumor-associated antigen (TAA) is 180 amino acids in length. Of the 172 possible 9-mers, 25 are given a score ≧16 by the SYFPEITHI/Rammensee algorithm. Like Melan-A above, these represent 14.5% of the possible peptides and an average epitope density on the protein of 0.136 per amino acid. However the distribution is quite different. Nearly half the protein is empty with just one predicted epitope in the first 78 amino acids. Unlike Melan-A where there was a very tight cluster of highly overlapping peptides, in NY-ESO the overlaps are smaller and extend over most of the rest of the protein. One set of 19 overlapping peptides covers amino acids 108-174, resulting in a ratio of 2.04. Another 5 predicted epitopes cover 79-104, for a ratio of just 1.38.


If instead one takes the approach of considering only the top 5% of predicted epitopes, in this case 9 peptides, one can examine whether good clusters are being obscured by peptides predicted to be less likely to bind to MHC. When just these predicted epitopes are considered we see that the region 108-140 contains 6 overlapping peptides with a ratio of 3.64. There are also 2 nearby peptides in the region 148-167 with a ratio of 2.00. Thus the large cluster 108-174 can be broken into two smaller clusters covering much of the same sequence.


Restricting the analysis to the 9-mers predicted to have a half time of dissociation of ≧5 minutes by the BIMAS-NIH/Parker algorithm brings 14 peptides into consideration. The average density of epitopes in the protein is now 0.078 per amino acid. A single set of 10 overlapping peptides is observed, covering amino acids 144-171, with a ratio of 4.59. All 14 peptides fall in the region 86-171 which is still 2.09 times the average density of epitopes in the protein. While such a large cluster is larger than we consider ideal it still offers a significant advantage over working with the whole protein. See FIG. 20.

TABLE 44SYFPEITHI (Rammensee algorithm) Results for NY-ESORankStartScore1108252148243159214127215862161322071222081202091152010962011113191291191316618141611815157181615118171371818791819139172013117218717221521623144162412916251516









TABLE 45










Calculations(Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio





1
108-140
1, 9, 8, 7, 4, 6
0.18
0.05
3.64


2
148-167
2, 3
0.10
0.05
2.00


3
 79-104
5 12, 10, 18, 21
0.19
0.14
1.38


4
108-174
1, 11, 9, 8, 7,
0.28
0.14
2.04




4, 6, 17, 2, 16,




15, 3, 14, 13,




24, 20, 19, 23,




22
















TABLE 46










BIMAS-NIH/Parker algorithm Results for NY-ESO












Rank
Start
Score
Log(Score)
















1
159
1197.321
3.08



2
86
429.578
2.63



3
120
130.601
2.12



4
161
83.584
1.92



5
155
52.704
1.72



6
154
49.509
1.69



7
157
42.278
1.63



8
108
21.362
1.33



9
132
19.425
1.29



10
145
13.624
1.13



11
163
11.913
1.08



12
144
11.426
1.06



13
148
6.756
0.83



14
152
4.968
0.70

















TABLE 47










Calculations(Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio















1
 86-171
2, 8, 3, 9, 10, 12,
0.163
0.078
2.09




13, 14, 6, 5, 7, 1,




4, 11


2
144-171
10, 12, 13, 14, 6,
0.36
0.078
4.59




5, 7, 1, 4, 11









Example 19

Tyrosinase


This melanoma tumor-associated antigen (TAA) is 529 amino acids in length. Of the 521 possible 9-mers, 52 are given a score >16 by the SYFPEITHI/Rammensee algorithm. These represent 10% of the possible peptides and an average epitope density on the protein of 0.098 per amino acid. There are 5 groups of overlapping peptides containing 2 to 13 predicted epitopes each, with ratios ranging from 2.03 to 4.41, respectively. There are an additional 7 groups of overlapping peptides, containing 2 to 4 predicted epitopes each, with ratios ranging from 1.20 to 1.85, respectively. The 17 peptides in the region 444-506, including the 13 overlapping peptides above, constitutes a cluster with a ratio of 2.20.


Restricting the analysis to the 9-mers predicted to have a half time of dissociation of ≧5 minutes by the BIMAS-NIH/Parker algorithm brings 28 peptides into consideration. The average density of epitopes in the protein under this condition is 0.053 per amino acid. At this density any overlap represents more than twice the average density of epitopes. There are 5 groups of overlapping peptides containing 2 to 7 predicted epitopes each, with ratios ranging from 2.22 to 4.9, respectively. Only three of these clusters are common to the two algorithms. Several, but not all, of these clusters could be enlarged by evaluating a region containing them and nearby predicted epitopes.

TABLE 48SYFPEITHI/Rammensee algorithm Results for TyrosinaseRankStartScore1490342491313487284127522564822373802383692392142310506221134322122072213137221457221516920161182017920184881919483192048019214791922478192347319243651925287192620019275192848418294761830463183144418324251833316183418718354021736388173734617383361739225174022417412081742186174317117445141645494164640616473851648349164918416501671651145165213916









TABLE 49










Calculations(Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio















1
1 to 17
4, 5, 27, 17
0.24
0.098
2.39


2
137-153
13, 52, 51
0.18
0.098
1.80


3
167-179
15, 43, 50
0.23
0.098
2.35


4
184-195
34, 42, 49
0.25
0.098
2.54


5
200-222
26, 41, 9, 12
0.17
0.098
1.77


6
224-233
39, 40
0.20
0.098
2.03


7
336-357
38, 11, 37, 48
0.18
0.098
1.85


8
365-377
24, 8
0.15
0.098
1.57


9
380-396
7, 47, 36
0.18
0.098
1.80


10
402-414
35, 46
0.15
0.098
1.57


11
473-502
29, 28, 23, 22,
0.43
0.098
4.41




21, 20, 6, 19, 3,




18, 1, 2, 45


12
506-522
10, 44
0.12
0.098
1.20



444-522
31, 30, 23, 29,
0.22
0.098
2.20




22, 21, 20, 6, 19,




28, 3, 18, 1, 2,




45, 10, 44
















TABLE 50










BIMAS-NIH/Parker algorithm Results












Rank
Start
Score
Log(Score)
















1
207
540.469
2.73



2
369
531.455
2.73



3
1
309.05
2.49



4
9
266.374
2.43



5
490
181.794
2.26



6
214
177.566
2.25



7
224
143.451
2.16



8
171
93.656
1.97



9
506
87.586
1.94



10
487
83.527
1.92



11
491
83.527
1.92



12
2
54.474
1.74



13
137
47.991
1.68



14
200
30.777
1.49



15
208
26.248
1.42



16
460
21.919
1.34



17
478
19.425
1.29



18
365
17.14
1.23



19
380
16.228
1.21



20
444
13.218
1.12



21
473
13.04
1.12



22
57
10.868
1.04



23
482
8.252
0.92



24
483
7.309
0.86



25
5
6.993
0.84



26
225
5.858
0.77



27
343
5.195
0.72



28
514
5.179
0.71

















TABLE 51










Calculations (Epitopes/AAs)
















Whole



Cluster
AA
Peptides
Cluster
protein
Ratio















1
1 to 17
3, 12, 25, 4
0.24
0.053
4.45


2
200-222
14, 1, 15, 6
0.17
0.053
3.29


3
224-233
7, 26
0.20
0.053
3.78


4
365-377
18, 2
0.15
0.053
2.91


5
473-499
21, 17, 23, 24,
0.26
0.053
4.90




10, 5, 11


6
506-522
9, 28
0.12
0.053
2.22


7
365-388
18, 2, 19
0.13
0.053
2.36


8
444-499
20, 16, 21, 17,
0.16
0.053
3.03




23, 24, 10, 5, 11


9
444-522
20, 16, 21, 17,
0.14
0.053
2.63




23, 24, 10, 5, 11,




9, 28


10
200-233
14, 1, 15, 6, 7, 26
0.18
0.053
3.33









Example 20

The following tables (52-75) present 9-mer epitopes predicted for HLA-A2 binding using both the SYFPEITHI and NIH algorithms and the epitope density of regions of overlapping epitopes, and of epitopes in the whole protein, and the ratio of these two densities. (The ratio must exceed one for there to be a cluster by the above definition; requiring higher values of this ratio reflect preferred embodiments). Individual 9-mers are ranked by score and identified by the position of their first amino in the complete protein sequence. Each potential cluster from a protein is numbered. The range of amino acid positions within the complete sequence that the cluster covers is indicated as are the rankings of the individual predicted epitopes it is made up of.

TABLE 52BIMAS-NIH/Parker algorithm Results for gp100RankStartScore1619149326024133162226418118517811862731177601818243639606601037350115443612291291359229142682915472716585261757621184652119570202091921416192225182356617246031525384142613142729012286371029639930485931453832102833399834456735113736622737697386046393506405835









TABLE 53










SYFPEITHI (Rammensee algorithm) Results for gp100









Rank
Start
Score












1
606
30


2
162
29


3
456
28


4
18
28


5
602
27


6
598
27


7
601
26


8
597
26


9
13
26


10
585
25


11
449
25


12
4
25


13
603
24


14
576
24


15
453
24


16
178
24


17
171
24


18
11
24


19
619
23


20
280
23


21
268
23


22
592
22


23
544
22


24
465
22


25
399
22


26
373
22


27
273
22


28
243
22


29
566
21


30
563
21


31
485
21


32
384
21


33
350
21


34
9
21


35
463
20


36
397
20


37
291
20


38
269
20


39
2
20


40
610
19


41
594
19


42
591
19


43
583
19


44
570
19


45
488
19


46
446
19


47
322
19


48
267
19


49
250
19


50
205
19


51
180
19


52
169
19


53
88
19


54
47
19


55
10
19


56
648
18


57
605
18


58
604
18


59
595
18


60
571
18


61
569
18


62
450
18


63
409
18


64
400
18


65
371
18


66
343
18


67
298
18


68
209
18


69
102
18


70
97
18


71
76
18


72
69
18


73
60
18


74
17
18


75
613
17


76
599
17


77
572
17


78
557
17


79
556
17


80
512
17


81
406
17


82
324
17


83
290
17


84
101
17


85
95
17


86
635
16


87
588
16


88
584
16


89
577
16


90
559
16


91
539
16


92
494
16


93
482
16


94
468
16


95
442
16


96
413
16


97
408
16


98
402
16


99
286
16


100
234
16


101
217
16


102
211
16


103
176
16


104
107
16


105
96
16


106
80
16


107
16
16


108
14
16


109
7
16
















TABLE 54








Prediction of clusters for gp100















Total AAs: 661


Total 9-mers: 653


SYFPEITHI ≧ 16: 109 9-mers


NIH ≧ 5: 40 9-mers












Epitopes/AA
















Epitopes

Whole




Cluster #
AAs
(by Rank)
Cluster
Pr
Ratio





SYFPEITHI
1
2 to 26
39, 12, 109, 34, 55, 11,
0.440
0.165
2.668





9, 108, 107, 74, 4



2
 69-115
72, 71, 106, 53, 85, 105,
0.213
0.165
1.290





70, 84, 69, 104



3
 95-115
85, 105, 70, 84, 69
0.238
0.165
1.444



4
162-188
2, 52, 17, 103, 16, 51
0.222
0.165
1.348



5
205-225
50, 68, 102, 101
0.190
0.165
1.155



6
243-258
28, 49
0.125
0.165
0.758



7
267-306
48, 21, 38, 27, 20, 99,
0.225
0.165
1.364





83, 37, 67



8
322-332
47, 82
0.182
0.165
1.103



9
343-358
66, 33
0.125
0.165
0.758



10 
371-381
65, 26
0.182
0.165
1.103



11 
397-421
36, 25, 64, 98, 81, 97,
0.320
0.165
1.941





63, 96



12 
442-476
95, 46, 11, 62, 15, 3,
0.257
0.165
1.559





35, 24, 94



13 
482-502
93, 31, 45, 93
0.190
0.165
1.155



14 
539-552
91,23
0.143
0.165
0.866



15 
556-627
79, 78, 90, 30, 29, 61,
0.431
0.165
2.611





44, 60, 77, 14, 89, 43,





88, 10, 87, 42, 22, 41,





59, 8, 6, 76, 7, 5, 13, 58,





57, 1, 40, 75, 19


NIH
1
9 to 33
20, 26, 4, 22
0.160
0.061
2.644



2
268-281
14, 6
0.143
0.061
2.361



3
290-299
27, 12
0.200
0.061
3.305



 4*
102-121
32, 35
0.100
0.061
1.653



 5*
373-392
10, 25
0.100
0.061
1.653



6
453-473
31, 34, 18
0.143
0.061
2.361



7
566-600
23, 19, 17, 40, 16, 13
0.171
0.061
2.833



8
601-614
7, 2, 24, 38, 9
0.357
0.061
5.902



9
619-630
1, 36
0.17
0.061
2.754



10 
637-647
28, 29
0.18
0.061
3.005







*Nearby but not overlapping epitopes














TABLE 55










BIMAS-NIH/Parker algorithm Results for PSMA









Rank
Start
Score












1
663
1360


2
711
1055


3
4
485


4
27
400


5
26
375


6
668
261


7
707
251


8
469
193


9
731
177


10
35
67


11
33
64


12
554
59


13
427
50


14
115
47


15
20
40


16
217
26


17
583
24


18
415
19


19
193
14


20
240
12


21
627
11


22
260
10


23
130
10


24
741
9


25
3
9


26
733
8


27
726
7


28
286
6


29
174
5


30
700
5
















TABLE 56










SYFPEITHI (Rammensee algorithm) Results for PSMA









Rank
Start
Score












1
469
27


2
27
27


3
741
26


4
711
26


5
354
25


6
4
25


7
663
24


8
130
24


9
57
24


10
707
23


11
260
23


12
20
23


13
603
22


14
218
22


15
109
22


16
731
21


17
668
21


18
660
21


19
507
21


20
454
21


21
427
21


22
358
21


23
284
21


24
115
21


25
33
21


26
606
20


27
568
20


28
473
20


29
461
20


30
200
20


31
26
20


32
3
20


33
583
19


34
579
19


35
554
19


36
550
19


37
547
19


38
390
19


39
219
19


40
193
19


41
700
18


42
472
18


43
364
18


44
317
18


45
253
18


46
91
18


47
61
18


48
13
18


49
733
17


50
673
17


51
671
17


52
642
17


53
571
17


54
492
17


55
442
17


56
441
17


57
397
17


58
391
17


59
357
17


60
344
17


61
305
17


62
304
17


63
286
17


64
282
17


65
169
17


66
142
17


67
122
17


68
738
16


69
634
16


70
631
16


71
515
16


72
456
16


73
440
16


74
385
16


75
373
16


76
365
16


77
361
16


78
289
16


79
278
16


80
258
16


81
247
16


82
217
16


83
107
16


84
100
16


85
75
16


86
37
16


87
30
16


88
21
16
















TABLE 57








Prediction of clusters for prostate-specific membrane antigen (PSMA)















Total AAs: 750


Total 9-mers: 742


SYFPEITHI ≧ 16: 88 9-mers


NIH ≧ 5: 30 9-mers












Epitopes/AA
















Epitopes

Whole




Cluster #
Aas
(by rank)
Cluster
Pr
Ratio





SYFPEITHI
1
3 to 12
32, 6
0.200
0.117
1.705



2
13-45
13, 12, 88, 31, 2, 87, 25, 86
0.242
0.117
2.066



3
57-69
9, 47
0.154
0.117
1.311



4
100-138
84, 83, 15, 24, 67, 8
0.154
0.117
1.311



5
193-208
40, 30
0.125
0.117
1.065



6
217-227
82, 14, 39
0.273
0.117
2.324



7
247-268
81, 45, 80, 11
0.182
0.117
1.550



8
278-297
79, 64, 23, 63, 78
0.250
0.117
2.131



9
354-381
5, 59, 22, 77, 43, 76, 75
0.250
0.117
2.131



10 
385-405
74, 38, 58, 57
0.190
0.117
1.623



11 
440-450
73, 56, 55
0.273
0.117
2.324



12 
454-481
20, 72, 29, 1, 42, 28
0.214
0.117
1.826



13 
507-523
17, 71
0.118
0.117
1.003



14 
547-562
37, 36, 35
0.188
0.117
1.598



15 
568-591
27, 53, 34, 33
0.167
0.117
1.420



16 
603-614
13, 26
0.167
0.117
1.420



17 
631-650
70, 69, 52
0.150
0.117
1.278



18 
660-681
18, 7, 17, 51, 50
0.227
0.117
1.937



19 
700-719
41, 10, 4
0.150
0.117
1.278



20 
731-749
16, 49, 68, 3
0.211
0.117
1.794


NIH
1
3 to 12
25, 3
0.200
0.040
5.000



2
20-43
15, 5, 4, 11, 10
0.208
0.040
5.208



 3*
415-435
18, 13
0.095
0.040
2.381



4
663-676
1, 6
0.143
0.040
3.571



5
700-715
30, 7, 3
0.188
0.040
4.688



6
726-749
27, 9, 26, 24
0.167
0.040
4.167







*Nearby but not overlapping epitopes














TABLE 58










BIMAS-NIH/Parker algorithm Results for PSA









Rank
Start
Score












1
7
607


2
170
243


3
52
124


4
53
112


5
195
101


6
165
23


7
72
18


8
245
18


9
2
16


10
59
16


11
122
15


12
125
15


13
191
13


14
9
8


15
14
6


16
175
5


17
130
5
















TABLE 59










SYFPEITHI (Rammensee algorithm) Results for PSA









Rank
Start
Score












1
72
26


2
170
22


3
53
22


4
7
22


5
234
21


6
166
21


7
140
21


8
66
21


9
241
20


10
175
20


11
12
20


12
41
19


13
20
19


14
14
19


15
130
18


16
124
18


17
121
18


18
47
18


19
17
18


20
218
17


21
133
17


22
125
17


23
122
17


24
118
17


25
110
17


26
67
17


27
52
17


28
21
17


29
16
17


30
2
17


31
184
16


32
179
16


33
158
16


34
79
16


35
73
16


36
4
16
















TABLE 60








Prediction of clusters for prostate specific antigen (PSA)















Total AAs: 261


Total 9-mers: 253


SYFPEITHI ≧ 16: 36 9-mers


NIH ≧ 5: 17 9-mers












Epitopes/AA
















Epitopes

Whole




Cluster #
AAs
(by rank)
Cluster
Pr
Ratio





SYFPEITHI
1
2 to 29
30, 36, 4, 11, 14, 29, 19, 13, 28
0.321
0.138
2.330



2
41-61
12, 18, 27, 3
0.190
0.138
1.381



3
66-87
8, 26, 1, 35, 34
0.227
0.138
1.648



4
110-148
25, 24, 17, 23, 16, 22, 15, 21, 7
0.184
0.138
1.332



5
158-192
33, 6, 2, 10, 32, 31
0.171
0.138
1.243



6
234-249
5, 9
0.125
0.138
0.906



 7*
118-133
24, 17, 23, 16, 22
0.313
0.138
2.266



 8*
118-138
24, 17, 23, 16, 22, 15
0.286
0.138
2.071


NIH
1
 2-22
9, 1, 14, 15
0.190
0.065
2.924



2
52-67
3, 4, 10
0.188
0.065
2.879



3
122-138
11, 12, 17
0.176
0.065
2.709



4
165-183
6, 2, 16
0.158
0.065
2.424



5
191-203
13, 5
0.154
0.065
2.362



 6**
52-80
3, 4, 10, 7
0.138
0.065
2.118







*These clusters are internal to the less preferred cluster #4.





**Includes a nearby but not overlapping epitope.














TABLE 61










BIMAS-NIH/Parker algorithm Results for PSCA









Rank
Start
Score












1
43
153


2
5
84


3
7
79


4
109
36


5
105
25


6
108
24


7
14
21


8
20
18


9
115
17


10
42
15


11
36
15


12
99
9


13
58
8
















TABLE 62










SYFPEITHI (Rammensee algorithm) Results for PSCA









Rank
Start
Score












1
108
30


2
14
30


3
105
29


4
5
28


5
115
26


6
99
26


7
7
26


8
109
24


9
53
23


10
107
21


11
20
21


12
8
21


13
13
20


14
102
19


15
60
19


16
57
19


17
54
19


18
12
19


19
4
19


20
1
19


21
112
18


22
101
18


23
98
18


24
51
18


25
43
18


26
106
17


27
104
17


28
83
17


29
63
17


30
50
17


31
3
17


32
9
16


33
92
16
















TABLE 63








Prediction of clusters for prostate stem cell antigen (PSCA)















Total AAs: 123


Total 9-mers: 115


SYFPEITHI ≧ 16: 33;


SYFPEITHI ≧ 20: 13


NIH ≧ 5: 13













Epitopes
Epitopes/AA














Cluster #
AAs
(by rank)
Cluster
Whole Pr.
Ratio





SYFPEITHI > 16
1
1 to 28
20, 31, 19, 4, 7, 12, 33, 18, 13,
0.393
0.268
1.464





2, 11



2
43-71
25, 30, 24, 9, 17, 16, 15, 29
0.276
0.268
1.028



3
 92-123
32, 23, 6, 27, 14, 22, 3, 26,
0.406
0.268
1.514





10, 1, 8, 21, 5


SYFPEITHI > 20
1
5 to 28
4, 7, 12, 13, 2, 11
0.250
0.106
2.365



2
 99-123
6, 3, 10, 1, 8, 5
0.240
0.106
2.271


NIH
1
5 to 28
2, 3, 7, 8
0.167
0.106
1.577



2
36-51
11, 10, 1
0.188
0.106
1.774



3
 99-123
12, 5, 6, 4, 9
0.200
0.106
1.892



 4*
105-116
5, 6, 4
0.250
0.106
2.365







*This cluster is internal to the less preferred cluster #3.







In tables 49-60 epitope prediction and cluster analysis data for each algorithm are presented together in a single table.

TABLE 64Prediction of clusters for MAGE-1 (NIH algorithm)Total AAs: 309Total 9-mers: 301NIH ≧ 5: 19 9-mersEpitopes/AAClusterEpitopeStartNIHWhole#AAsRankPositionScoreClusterPr.Ratio118-32161890.1330.0632.112192472101-11314101110.1540.0632.4427105443146-1599146320.1430.0632.26331511694169-20210169320.1760.0632.796131741618181817187861887451941105264-27722641900.1430.0632.26312269206278-29012787430.1540.0632.4371128228









TABLE 65








Prediction of clusters for MAGE-1 (SYFPEITHI algorithm)















Total AAs: 309


Total 9-mers: 301


SYFPEITHI ≧ 16: 46 9-mers















Epitope
Start
SYFPEITHI
Epitopes/AA














Cluster #
Aas
Rank
Position
Score
Cluster
Whole
Ratio





1
 7-49
22
7
19
0.233
0.153
1.522




9
15
22




27
18
18




16
20
20




28
22
18




29
24
18




33
31
17




30
35
18




2
38
26




17
41
20


2
 89-132
10
89
22
0.273
0.153
1.783




18
92
20




7
93
23




23
96
19




43
98
16




4
101
25




8
105
23




34
107
17




35
108
17




36
113
17




37
118
17




19
124
20


3
167-203
44
167
16
0.270
0.153
1.766




20
169
20




12
174
21




24
181
19




6
187
24




31
188
18




25
191
19




38
192
17




1
194
27




13
195
21


4
230-246
14
230
21
0.118
0.153
0.769




39
238
17


5
264-297
15
264
21
0.235
0.153
1.538




32
269
18




40
270
17




26
271
19




46
275
16




3
278
26




21
282
20




41
289
17
















TABLE 66








Prediction of clusters for MAGE-2 (NIH algorithm)















Total AAs: 314


Total 9-mers: 308


NIH >= 5: 20 9-mers















Epitope
Start
NIH
Epitope/AA














Cluster #
AAs
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
101-120
18
101
5.373
0.150
0.065
2.310




16
108
6.756




1
112
2800.697


2
153-167
8
153
31.883
0.200
0.065
3.080




4
158
168.552




7
159
32.138


3
169-211
14
169
8.535
0.209
0.065
3.223




19
174
5.346




6
176
49.993




11
181
15.701




15
188
7.536




12
195
12.809




5
200
88.783




10
201
16.725




17
203
5.609


4
271-284
3
271
398.324
0.143
0.065
2.200




9
276
19.658
















TABLE 67








Prediction of clusters for MAGE-2 (SYFPEITHI algorithm)















Total AAs: 314


Total 9-mers: 308


SYFPEITHI ≧ 16: 52 9-mers















Epitope
Start
SYFPEITHI
Epitopes/AA














Cluster #
AAs
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
15-32
13
15
21
0.278
0.169
1.645




29
18
18




43
20
16




30
22
18




21
24
19


2
37-56
31
37
18
0.250
0.169
1.481




16
40
20




44
44
16




14
45
21




22
48
19


3
 96-133
36
96
17
0.211
0.169
1.247




46
101
16




6
108
25




47
109
16




2
112
27




37
120
17




38
125
17




17
131
20


4
153-216
12
153
22
0.344
0.169
2.036




39
158
17




7
159
25




23
161
19




24
162
19




48
164
16




49
167
16




32
170
18




50
171
16




4
174
26




9
176
24




51
177
16




15
181
21




25
188
19




18
194
20




33
195
18




19
198
20




3
200
27




1
201
28




40
202
17




10
203
23




52
208
16


5
237-254
26
237
19
0.167
0.169
0.987




27
245
19




34
246
18


6
271-299
8
271
25
0.241
0.169
1.430




35
276
18




41
277
17




11
278
23




28
283
19




20
285
20




42
291
17
















TABLE 68








Prediction of clusters for MAGE-3 (NIH algorithm)















Total AAs: 314


Total 9-mers: 308


NIH ≧ 5: 22 9-mers















Epitope
Start
NIH
Epitopes/AA














Cluster #
AAs
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
101-120
15
101
11.002
0.200
0.071
2.800




21
105
6.488




8
108
49.134




2
112
339.313


2
153-167
18
153
7.776
0.200
0.071
2.800




6
158
51.77




22
159
5.599


3
174-209
17
174
8.832
0.194
0.071
2.722




7
176
49.993




13
181
15.701




19
188
7.536




14
195
12.809




5
200
88.783




12
201
16.725


4
237-251
16
237
10.868
0.200
0.071
2.800




4
238
148.896




20
243
6.88


5
271-284
1
271
2655.495
0.143
0.071
2.000




11
276
19.658
















TABLE 69








Prediction of clusters for MAGE-3 (SYFPEITHI algorithm)















Total AAs: 314


Total 9-mers: 308


SYFPEITHI ≧ 16: 47 9-mers















Epitope
Start
SYFPEITHI
Epitopes/AA














Cluster #
AAs
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
15-32
12
15
21
0.278
0.153
1.820




26
18
18




37
20
16




27
22
18




18
24
19


2
38-56
38
38
16
0.263
0.153
1.725




15
40
20




39
44
16




13
45
21




19
48
19


3
101-142
28
101
18
0.190
0.153
1.248




40
105
16




1
108
31




6
112
25




31
120
17




32
125
17




16
131
20




41
134
16


4
153-216
20
153
19
0.313
0.153
2.048




29
156
18




33
158
17




21
159
19




34
161
17




42
164
16




43
167
16




10
174
22




8
176
23




14
181
21




22
188
19




44
193
16




11
194
22




23
195
19




45
197
16




17
198
20




3
200
27




2
201
28




35
202
17




46
208
16


5
220-230
5
220
26
0.182
0.153
1.191




47
222
16


6
237-246
7
237
25
0.200
0.153
1.311




9
238
23


7
271-293
4
271
27
0.217
0.153
1.425




30
276
18




24
278
19




36
283
17




25
285
19
















TABLE 70








Prediction of clusters for PRAME (NIH algorithm)















Total AAs: 509


Total 9-mers: 501


NIH ≧ 5: 40 9-mers












Epitopes/AA














Cluster

Epitope
Start
NIH

Whole



#
AAs
Rank
Position
Score
Cluster
Pr.
Ratio





1
33-47
20
33
18
0.133
0.080
1.670




17
39
21


2
71-81
9
71
50
0.2
0.07984
2.505




32
73
7


3
 99-108
23
100
15
0.2
0.07984
2.505




24
99
13


4
126-135
38
126
5
0.2
0.07984
2.505




35
127
6


5
224-246
5
224
124
0.130
0.080
1.634




8
230
63




39
238
5


6
290-303
18
290
18
0.214
0.080
2.684




14
292
23




7
295
66


7
305-324
28
305
10
0.200
0.080
2.505




30
308
8




25
312
13




36
316
6


8
394-409
2
394
182
0.188
0.080
2.348




12
397
42




31
401
7


9
422-443
10
422
49
0.227
0.080
2.847




3
425
182




34
431
7




29
432
9




4
435
160


10
459-487
15
459
21
0.172
0.080
2.159




11
462
45




22
466
15




40
472
5




37
479
6
















TABLE 71








Prediction of clusters for PRAME (SYFPEITHI algorithm)















Total AAs: 509


Total 9-mers: 501


SYFPEITHI ≧ 17: 80 9-mers















Epitope
Start
SYFPEITHI
Epitopes/AA














Cluster #
AAs
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
18-59
65
18
17
0.238
0.160
1.491




50
21
18




66
26
17




35
33
20




22
34
22




51
37
18




5
39
27




23
40
22




13
44
24




46
51
19


2
 78-115
36
78
20
0.263
0.160
1.648




67
80
17




52
84
18




24
86
22




53
91
18




25
93
22




9
99
25




8
100
26




54
103
18




55
107
18


3
191-202
56
191
18
0.167
0.160
1.044




38
194
20


4
205-215
26
205
22
0.182
0.160
1.139




27
207
22


5
222-238
47
222
19
0.235
0.160
1.474




14
224
24




69
227
17




57
230
18


6
241-273
70
241
17
0.212
0.160
1.328




15
248
24




71
255
17




30
258
21




39
259
20




58
261
18




40
265
20


7
290-342
72
290
17
0.208
0.160
1.300




48
293
19




31
298
21




73
301
17




18
305
23




6
308
27




10
312
25




19
316
23




28
319
22




41
326
20




74
334
17


8
343-363
59
343
18
0.238
0.160
1.491




60
348
18




75
351
17




20
353
23




76
355
17


9
364-447
49
364
19
0.250
0.160
1.566




32
371
21




11
372
25




61
375
18




77
382
17




21
390
23




78
391
17




1
394
30




42
397
20




62
403
18




33
410
21




43
418
20




34
419
21




7
422
27




2
425
29




79
426
17




63
428
18




64
431
18




12
432
25




16
435
24




80
439
17


10
455-474
29
455
22
0.200
0.160
1.253




17
459
24




4
462
28




3
466
29
















TABLE 72








Prediction of clusters for CEA (NIH algorithm)















Total AAs: 702


Total 9-mers: 694


NIH ≧ 5: 30 9-mers















Peptides
Start

Peptides/AAs














Cluster #
AA
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
17-32
5
17
79.041
0.188
0.043
4.388




7
18
46.873




20
24
12.668


2
113-129
2
113
167.991
0.118
0.043
2.753




15
121
21.362


3
172-187
25
172
9.165
0.125
0.043
2.925




14
179
27.995


4
278-291
30
278
5.818
0.143
0.043
3.343




17
283
19.301


5
350-365
9
350
43.075
0.125
0.043
2.925




12
357
27.995


6
528-543
8
528
43.075
0.125
0.043
2.925




13
535
27.995


7
631-645
23
631
9.563
0.200
0.043
4.680




19
634
13.381




24
637
9.245


8
691-702
1
691
196.407
0.167
0.043
3.900




27
694
7.769
















TABLE 73








Prediction of clusters for CEA (SYFPEITHI algorithm)















Total AAs: 702


Total 9-mers: 694


SYFPEITHI ≧ 16: 81 9-mers












Peptides/AAs














Cluster

Peptides
Start


Whole



#
AA
Rank
Position
Score
Cluster
Pr.
Ratio





1
 5-36
67
5
16
0.250
0.117
2.140




23
12
19




24
16
19




9
17
22




25
18
19




32
19
18




68
23
16




33
28
18


2
37-62
41
37
17
0.269
0.117
2.305




20
44
20




26
45
19




42
46
17




27
50
19




43
53
17




44
54
17


3
 99-115
14
99
21
0.235
0.117
2.014




5
100
23




45
104
17




34
107
18


4
116-129
69
116
16
0.143
0.117
1.223




21
121
20


5
172-187
46
172
17
0.125
0.117
1.070




70
179
16


6
192-202
3
192
24
0.182
0.117
1.557




47
194
17


7
226-241
48
226
17
0.188
0.117
1.605




49
229
17




15
233
21


8
307-318
11
307
22
0.250
0.117
2.140




71
308
16




51
310
17


9
319-349
52
319
17
0.129
0.117
1.105




53
327
17




72
335
16




35
341
18


10
370-388
12
370
22
0.211
0.117
1.802




54
372
17




74
375
16




6
380
23


11
403-419
56
403
17
0.235
0.117
2.014




57
404
17




58
407
17




28
411
19


12
427-442
59
427
17
0.188
0.117
1.605




75
432
16




76
434
16


13
450-462
77
450
16
0.154
0.117
1.317




13
454
22


14
488-505
36
488
18
0.167
0.117
1.427




18
492
21




60
497
17


15
548-558
4
548
24
0.182
0.117
1.557




61
550
17


16
565-577
62
565
17
0.154
0.117
1.317




19
569
21


17
579-597
78
579
16
0.143
0.117
1.223




79
582
16




7
589
23


18
605-618
2
605
25
0.143
0.117
1.223




38
610
18


19
631-669
29
631
19
0.154
0.117
1.317




63
637
17




80
644
16




64
652
17




39
660
18




81
661
16


20
675-702
22
675
20
0.286
0.117
2.446




30
683
19




31
687
19




40
688
18




65
690
17




1
691
31




66
692
17




8
694
23
















TABLE 74








Prediction of clusters for SCP-1 (NIH algorithm)















Total AAs: 976


Total 9-mers: 968


NIH ≧ 5: 37 9-mers















Peptides
Start

Peptides/AAs














Cluster #
AA
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
101-116
15
101
40.589
0.125
0.038
3.270




13
108
57.255


 2*
281-305
14
281
44.944
0.12
0.038
3.139




24
288
15.203




17
297
32.857


3
431-447
8
431
80.217
0.073
0.038
1.914




26
438
11.861




4
439
148.896


4
557-579
11
557
64.335
0.174
0.038
4.550




19
560
24.937




6
564
87.586




18
571
32.765


5
635-650
10
635
69.552
0.125
0.038
3.270




34
642
6.542


6
755-767
36
755
5.599
0.154
0.038
4.025




35
759
5.928


7
838-854
2
838
284.517
0.118
0.038
3.078




28
846
11.426
















TABLE 75








Prediction of clusters for SCP-1















Total AAs: 976


Total 9-mers: 968


Rammensee ≧ 16: 118 9-mers















Peptides
Start

Peptides/AAs














Cluster #
AA
Rank
Position
Score
Cluster
Whole Pr.
Ratio





1
 8-28
99
8
16
0.143
0.121
1.182




77
15
17




100
20
16


2
63-80
78
63
17
0.222
0.121
1.838




50
66
19




102
69
16




60
72
18


3
 94-123
79
94
17
0.133
0.121
1.103




12
101
23




17
108
22




103
115
16


4
126-158
35
126
20
0.182
0.121
1.504




36
133
20




51
139
19




80
140
17




61
143
18




37
150
20


5
161-189
38
161
20
0.207
0.121
1.711




52
165
19




81
171
17




82
177
17




62
178
18




39
181
20


6
213-230
40
213
20
0.167
0.121
1.379




13
220
23




28
222
21


7
235-250
63
235
18
0.125
0.121
1.034




18
242
22


8
260-296
83
260
17
0.243
0.121
2.012




105
262
16




84
267
17




106
269
16




41
270
20




64
271
18




85
274
17




19
281
22




3
288
25


9
312-338
108
312
16
0.148
0.121
1.225




29
319
21




30
323
21




65
330
18


10
339-355
66
339
18
0.235
0.121
1.946




31
340
21




42
344
20




53
347
19


11
376-447
54
376
19
0.194
0.121
1.608




43
382
20




44
386
20




20
390
22




55
397
19




6
404
24




86
407
17




45
411
20




67
417
18




21
425
22




46
431
20




68
432
18




32
438
21




7
439
24


12
455-488
33
455
21
0.235
0.121
1.946




47
459
20




56
462
19




87
463
17




88
466
17




14
470
23




109
473
16




34
480
21


13
515-530
57
515
19
0.125
0.121
1.034




22
522
22


14
557-590
8
557
24
0.147
0.121
1.216




23
564
22




9
571
24




90
575
17




58
582
19


15
610-625
69
610
18
0.125
0.121
1.034




91
617
17


16
633-668
92
633
17
0.222




10
635
24




70
638
18




93
640
17




48
642
20




49
645
20




111
652
16




112
660
16


17
674-685
71
674
18
0.167
0.121
1.379




11
677
24


18
687-702
1
687
26
0.125
0.121
1.034




94
694
17


19
744-767
113
744
16
0.250
0.121
2.068




95
745
17




4
745
25




24
752
22




2
755
26




72
759
18


20
812-827
97
812
17
0.125
0.121
1.034




115
819
16


21
838-857
116
838
16
0.150
0.121
1.241




25
846
22




74
849
18


22
896-913
117
896
16
0.222
0.121
1.838




98
899
17




26
902
22




76
905
18









The embodiments of the invention are applicable to and contemplate variations in the sequences of the target antigens provided herein, including those disclosed in the various databases that are accessible by the world wide web. Specifically for the specific sequences disclosed herein, variation in sequences can be found by using the provided accession numbers to access information for each antigen.


All patents and publications mentioned in the specification are indicative of the levels of those skilled in the art to which the invention pertains. The entire contents of all patents and publications discussed herein are incorporated by reference in their entirety to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety.


The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. The terms and expressions which have been employed are used as the terms of description and not of limitation, and there is no intention that in the use of such terms and expressions indicates the exclusion of equivalents of the features shown and described or portions thereof. It is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.

Claims
  • 1. An isolated nucleic acid comprising a reading frame comprising a first sequence, wherein said first sequence encodes one or more segments of tumor-associated antigen PRAME (SEQ ID NO: 77), wherein the first sequence does not encode a complete PRAME antigen, and each segment comprises an epitope cluster, said cluster comprising or encoding at least two amino acid sequences having a known or predicted affinity for a same MHC receptor peptide binding cleft.
  • 2. The nucleic acid of claim 1, wherein said epitope cluster is selected from the group consisting of amino acids 18-59, 33-47, 71-81, 78-115, 99-108, 126-135, 222-238, 224-246, 290-303, 305-324, 343-363, 364-447, 394-409, 422-443, and 459-487 of PRAME.
  • 3. The nucleic acid of claim 1, wherein said one or more segments consist of said epitope cluster.
  • 4. The nucleic acid of claim 1, wherein said first sequence encodes a fragment of PRAME.
  • 5. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 90% of the length of PRAME.
  • 6. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 80% of the length of PRAME.
  • 7. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 60% of the length of PRAME.
  • 8. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 50% of the length of PRAME.
  • 9. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 25% of the length of PRAME.
  • 10. The nucleic acid of claim 4, wherein said encoded fragment consists of a polypeptide having a length, wherein the length of the polypeptide is less than about 10% of the length of PRAME.
  • 11. The nucleic acid of claim 4, wherein said encoded fragment consists essentially of an amino acid sequence beginning at one of amino acids selected from the group consisting of 18, 33, 71, 78, 99, 126, 222, 224, 290, 305, 343, 364, 394, 422, and 459 of PRAME, and ending at one of the amino acids selected from the group consisting of amino acid 47, 59, 81, 108, 115, 135, 238, 246, 303, 324, 363, 409, 443, 447, and 487 of PRAME.
  • 12. The nucleic acid of claim 11, wherein said encoded fragment consists essentially ofamino acids 18-47, 18-59, 18-81, 18-108, 18-115, 18-135, 18-238, 18-246, 18-303, 18-324, 18-363, 18-409, 18-443, 18-447, 18-487, 33-47, 33-59, 33-81, 33-108, 33-115, 33-135, 33-238, 33-246, 33-303, 33-324, 33-363, 33-409, 33-443, 33-447, 33-487, 71-81, 71-108, 71-115, 71-135, 71-238, 71-246, 71-303, 71-324, 71-363, 71-409, 71-443, 71-447, 71-487, 78-108, 78-115, 78-135, 78-238, 78-246, 78-303, 78-324, 78-363, 78-409, 78-443, 78-447, 78-487, 99-108, 99-115, 99-135, 99-238, 99-246, 99-303, 99-324, 99-363, 99-409, 99-443, 99-447, 99-487, 126-135, 126-238, 126-246, 126-303, 126-324, 126-363, 126-409, 126-443, 126-447, 126-487, 222-238, 222-246, 222-303, 222-324, 222-363, 222-409, 222-443, 222-447, 222-487, 224-238, 224-246, 224-303, 224-324, 224-363, 224-409, 224-443, 224-447, 224-487, 290-303, 290-324, 290-363, 290-409, 290-443, 290-447, 290-487, 305-324, 305-363, 305-409, 305-443, 305-447, 305-487, 343-363, 343-409, 343-443, 343-447, 343-487, 364-409, 364-443, 364-447, 364-487, 394-409, 394-443, 394-447, 394-487, 422-443, 422-447, 422-487, 459-487, 18-487, or 224-487 of PRAME.
  • 13. The nucleic acid of claim 1, wherein said reading frame is operably linked to a promoter.
  • 14. The nucleic acid of claim 1, further comprising a second sequence, wherein the second sequence encodes essentially a housekeeping epitope.
  • 15. The nucleic acid of claim 14, wherein said first and second sequences constitute a single reading frame.
  • 16. The nucleic acid of claim 15, wherein said reading frame is operably linked to a promoter.
  • 17. An isolated polypeptide comprising the amino acid sequence encoded in said reading frame of claim 16.
  • 18. An immunogenic composition comprising the nucleic acid of claim 17.
  • 19. An immunogenic composition comprising the polypeptide of claim 18.
  • 20. The nucleic acid of claim 1, wherein said reading frame further comprises a second sequence encoding a polypeptide sequence consisting essentially of an epitope or epitope array.
  • 21. An expression vector comprising a promoter operably linked to means for encoding an amino acid sequence comprising at least one PRAME epitope cluster, wherein said means do not encode the complete PRAME antigen.
CROSS REFERENCE

This application is a continuation of U.S. patent application Ser. No. 10/117,937, filed Apr. 4, 2002, which claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application Ser. No. 60/282,211, filed on Apr. 6, 2001; U.S. Provisional Patent Application Ser. No. 60/337,017, filed on Nov. 7, 2001; and U.S. Provisional Patent Application Ser. No. 60/363,210, filed on Mar. 7, 2002; all entitled “EPITOPE SEQUENCES,” and all of which are hereby incorporated by reference in their entirety.

Provisional Applications (3)
Number Date Country
60282211 Apr 2001 US
60337017 Nov 2001 US
60363210 Mar 2002 US
Continuations (1)
Number Date Country
Parent 10117937 Apr 2002 US
Child 11067159 Feb 2005 US