WO2005037854A2 - Immunogenic peptides - Google Patents

Immunogenic peptides Download PDF

Info

Publication number
WO2005037854A2
WO2005037854A2 PCT/US2004/033241 US2004033241W WO2005037854A2 WO 2005037854 A2 WO2005037854 A2 WO 2005037854A2 US 2004033241 W US2004033241 W US 2004033241W WO 2005037854 A2 WO2005037854 A2 WO 2005037854A2
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
antibody
peptide
antigen
Prior art date
Application number
PCT/US2004/033241
Other languages
French (fr)
Other versions
WO2005037854A3 (en
Inventor
Charles A. Nicolette
Original Assignee
Genzyme Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genzyme Corporation filed Critical Genzyme Corporation
Publication of WO2005037854A2 publication Critical patent/WO2005037854A2/en
Publication of WO2005037854A3 publication Critical patent/WO2005037854A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids

Definitions

  • the invention relates to compounds useful in therapeutic, diagnostic and screening methods for human cancers and related malignancies.
  • Therapeutic cancer vaccines seek to activate a patient's own immune system to kill cancer cells.
  • Antigen-specific T cell activity has provided a venue for developing novel strategies for anti-cancer vaccines based on the observation that antigenic epitopes presented by molecules of the Major Histocompatibility Complex (MHC) play a central role in the establishment, maintenance and execution ofthe mammalian immune response.
  • MHC Major Histocompatibility Complex
  • Antigen-specific cytotoxic T lymphocytes (CTLs) that recognize certain cancer antigens have been used to attack cells expressing these antigens to reject established tumors.
  • Anti-tumor T cells are localized within various cancer patient's tissues, including in the blood (where they can be found in the peripheral blood mononuclear cell fraction), in primary and secondary lymphoid tissue, e.g., the spleen, in ascites fluid in ovarian cancer patients (tumor associated lymphocytes or TALs) or within the tumor itself (tumor infiltrating lymphocytes or TILs).
  • TILs have been the most useful in the identification of tumor antigens and tumor antigen-derived peptides recognized by T cells.
  • TILs manipulated to specifically recognize, bind and lyse cancer cells have been used as a means of adoptive T-cell therapy. (Yee and Greenbert (June 2002) supra).
  • Conventional methods to generate TILs involve mincing tumor biopsy tissue and culturing the cell suspension in vitro in the presence of the T cell growth factor interleukin-2 (IL-2). Over a period of several days, the combination of the tumor cells and IL-2 can stimulate the proliferation of tumor specific T cells at the expense of tumor cells. In this way, the T cell population is expanded.
  • IL-2 T cell growth factor interleukin-2
  • the T cells derived from the first expansion are subsequently mixed with either mitomycin C-treated or irradiated tumor cells and cultured in vitro with IL-2 to promote further proliferation and enrichment of tumor reactive T cells.
  • IL-2 IL-2-induced tumor reactive T cells.
  • a potent anti-tumor T cell population can be recovered and used to identify tumor antigens via conventional but tedious expression cloning methodology.
  • Dissis and Cheever report that many newly defined tumor antigens are self proteins (e.g., MAGE, MART, gplOO, and tyrosinase) and therefore will not induce an effective immune response. (Dissi and Cheever (1998) Critical Rev. Immunol. 18:37-45). These therapeutic failures are believed to be the result of immune tolerance which reduces the ability of cancer vaccines to trigger a robust and sustained immune response against cancer cells. In other words, cancer cells and therefore therapeutics based on cancer-specific peptides, are not typically seen by a patient's immune system as being non-self or foreign (i.e., different from normal cells) and thus, do not activate the immune system in the same way a "foreign" protein can.
  • tolerance as a state of immunologic unresponsiveness to antigens, whether self or foreign.
  • a recent publication explains tolerance as resulting in the journal "from one of three inhibitory influences on T and B cells: (1) clonal deletion, in which antigenic recognition leads to the activation-induced death of specific lymphocytes; (2) clonal anergy, in which lymphocytes are not killed but are rendered unresponsive to the recognized antigen; and (3) T cell-mediated suppression, in which regulatory T cells actively inhibit an immune response to an antigen.
  • Immature lymphocytes are more susceptible to induction of tolerance than are mature lymphocytes. Tolerance can be induced in immature lymphocytes either centrally or in the periphery.
  • T cells thymus
  • B cells bone marrow
  • T cells recognize antigens that have been processed into peptides and presented in a complex with MHC molecules (self-MHC-peptide complexes). Consequently, immature T cells must be screened for their ability to recognize self-MHC. This screening takes place in the thymus gland.
  • T cells bearing receptors that recognize self-MHC are subjected to the processes of positive and negative selection. Positive selection occurs when T cells bearing receptors with a moderate affinity for self-MHC-peptide complexes receive survival and maturation signals after receptor ligation.
  • T cells bearing receptors with a high affinity for self-MHC-peptide complexes undergo activation-induced death.
  • the thymus gland is capable of presenting many self-antigens that are normally expressed outside of the thymus or during restricted developmental stages. This allows the elimination of most T cells bearing high-affinity receptors for self-MHC-peptide complexes and plays a major role in preventing autoimmunity in peripheral organs and thus explains the failure of recent therapeutic approaches. (See Ochoa-Garay, et al. (1997) Mol. Immunol. 34(3):273-281).
  • T cells bearing receptors capable of low-affinity interactions with self-MHC-peptide complexes potentially self-reactive T cells are normally found in peripheral lymphoid organs. Peripheral tolerance prevents these cells from inducing autoimmune disease. Peripheral tolerance is most often caused by the failure of T cells bearing low-affinity receptors to recognize self-antigen in the periphery. In this situation, the potentially self-reactive T cell is not activated and remains functionally naive. These cells are functional, however, as is shown by the fact that they can be activated by immunization with self-antigen delivered in the presence of immune adjuvants.
  • a second mechanism of peripheral tolerance involves the elimination of self-reactive T cells by apoptosis. This process is analogous to clonal deletion in the thymus. Whether peripheral deletion plays an important role in tolerance to self-antigens is not known.
  • a third mechanism of peripheral tolerance involves the acquisition of anergy after ligation of the T cell receptor complex. This antigen-nonresponsive state can be induced in several distinct ways. The most extensively characterized mechanism of anergy induction occurs when the T cell receptor is ligated in the absence of costimulation.
  • T cells After a T cell has bound with an antigen, the cell requires a so-called second signal delivered by one or more costimulatory molecules to be primed for an immune response.
  • T cells express several surface molecules that can transmit this second signal. These costimulatory receptors are engaged by ligands expressed on the surface of APCs.
  • T cells that are activated in the absence of costimulation acquire defects in the transcriptional control pathways that allow the production of IL-2, an important T cell autocrine growth factor. In vitro anergy can often be overcome by supplying exogenous IL-2 to anergic T cells.
  • Costimulatory signals can be delivered to T cells by soluble factors or cell-surface molecules expressed on APCs.
  • the most potent costimulatory signals are delivered when CD28, CD 154, or both are ligated on the surface of T cells.
  • the ligand for CD 154 is CD40, a protein expressed on the surface of activated B cells, DCs, and maerophages.
  • the ligands for CD28 (B7-1 (CD80), B7-2 (CD86), and related proteins) are expressed on the surface of APCs, such as DCs, monocytes, and B cells. Their expression is induced when APCs are activated in the course of microbial infection. This property heightens the immune response in the setting of perceived danger.
  • B7-1 and B7-2 have overlapping immunostimulatory roles: mice lacking either protein are only partially deficient in generating an immune response to foreign antigen.
  • CTLA-4 a structurally related protein that turns off activated T cells.
  • the activated T cell initiates a program that will ensure its elimination at the conclusion of the immune response.
  • CTLA-4 has a higher affinity for B7-1 and B7-2.
  • CD28-B7 interactions may play a role in promoting tolerance to self-antigens.
  • CD28-B7 interactions are required for the maturation of a distinct class of regulatory T cells that help maintain peripheral tolerance.
  • These regulatory T cells (Tr) are included in a subpopulation (5%-15%) of peripheral blood CD4 + T cells that express CD25, a subunit of the IL-2 receptor.
  • CD4 + and CD25 + T cells that mature in the thymus gland bear receptors that have an intermediate affinity for self-MHC-peptide complexes.
  • antigen exposure confers the ability to suppress the activation of CD4 + and CD25 + T cells in an antigen-independent, cell contact-dependent manner.
  • these cells secrete IL-10, a potent anti-inflammatory cytokine, their suppressive activity is cytokine independent.
  • CD4 + and CD25 + T cells can suppress graft versus host disease in allotransplants and they can prevent autoimmune disease in several different animal models. Consequently, these cells probably play an essential role in maintaining peripheral tolerance to self-antigens.”
  • WebMD Scientific American Medicine (samed.com/sam/forms/index/htm; ISSN: 0194-9063; Section 6: Immunology/ Alergy).
  • CTLS cancer killing cytotoxic T cells
  • this invention is a peptide wherein the sequence of the peptide is represented by the group comprising SEQ ID NOs: 2 through 44 (shown in Table 1, infra).
  • the peptides can be combined with a carrier such as a pharmaceutically acceptable carrier.
  • polynucleotides encoding the peptides of the invention for example the nucleic acid sequences provided in SEQ ID NOs: 1 through 43, complements and variants thereof.
  • the polynucleotides can be combined with a carrier such as a pharmaceutically available carrier.
  • gene delivery vehicles and/or host cells comprising these polynucleotides.
  • compositions comprising at least one of the biological materials represented by SEQ ID NOS. 1 through 44 and a carrier, such as a pharmaceutically acceptable carrier.
  • a host cell comprising at least one peptide and/or polynucleotide encoding said peptide, wherein said peptide comprises a sequence selected from SEQ ID NOs: 2 through 44.
  • the host cell is an antigen presenting cell, e.g., a dendritic cell.
  • Compositions comprising such host cells and a carrier such as a pharmaceutically acceptable carrier, are also provided.
  • the invention provides methods for inducing an immune response in a subject by delivering to the subject the peptides of the invention, and delivering these in the context of an MHC molecule.
  • the peptides of the invention are also useful to generate antibodies that specifically recognize and bind to these polypeptides.
  • the invention also provides immune effector cells raised in vivo or in vitro in the presence and at the expense of an antigen presenting cell that presents one or more poly ⁇ eptide(s) of the invention in the context of an MHC molecule and a method of adoptive immunotherapy comprising administering an effective amount of these immune effector cells to a subject.
  • a method for inducing an immune response in a subject by delivering to the subject a composition comprising an effective amount of at least one polypeptide shown in Table 1.
  • the present invention additionally provides methods and compositions for detecting, diagnosing, prognosing and monitoring the progress of cancers and malignancies and kits for use in said methods. Further provided are methods for screening to identify agonists and antagonists of cancer antigens associated with cancers and malignancies.
  • MOLECULAR CLONING A LABORATORY MANUAL, 2 nd edition (1989); CURRENT PROTOCOLS IN MOLECULAR BIOLOGY (F. M. Ausubel, et al. eds. (1987)); the series METHODS IN ENZYMOLOGY (Academic Press, Inc.); 005/037854
  • PCR A PRACTICAL APPROACH (M. MacPherson, et al. IRL Press at Oxford University Press (1991)); PCR 2: A PRACTICAL APPROACH (M.J. MacPherson, B.D. Hames and G.R. Taylor eds. (1995)); ANTIBODIES, A LABORATORY MANUAL (Hariow and Lane eds. (1988)); USING ANTIBODIES, A LABORATORY MANUAL (Hariow and Lane eds. ( 1999)); and ANIMAL CELL CULTURE (R.I. Freshney ed. (1987)).
  • compositions and methods include the recited elements, but do not exclude others.
  • Consisting essentially of when used to define compositions and methods shall mean excluding other elements of any essential significance to the combination.
  • compositions consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like.
  • Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention. Embodiments defined by each of these transition terms are within the scope of this invention.
  • a "native” or “natural” or “wild-type” antigen is a polypeptide, protein or a fragment which contains an epitope, which has been isolated from a natural biological source, and which can specifically bind to an antigen receptor, in particular a T cell antigen receptor (TCR), in a subject.
  • TCR T cell antigen receptor
  • an “antigen” is a substance that is antigenic.
  • Antigenic refers to the ability of a peptide to bind to its ligand. As an example, the antigenicity of a peptide or epitope is determined by its ability to bind to a T cell.
  • a "self-antigen” also referred to herein as a native or wild-type antigen is an antigenic peptide that induces little or no immune response in the subject due to self-tolerance to the antigen.
  • An example of a self-antigen is the melanoma specific antigen gplOO. Any substance that can elicit an immune response is said to be
  • immunogenic and is referred to as an "immunogen”. All immunogens are antigens, however, not all antigens are immunogenic.
  • An immune response of this invention can be humoral (via antibody activity) or cell-mediated (via T cell activation).
  • ligand refers to any molecule that binds to a specific site on another molecule. In other words, the ligand confers the specificity of the protein in a reaction with an immune effector cell. It is the ligand site within the protein that combines directly with the complementary binding site on the immune effector cell.
  • tumor associated antigen or "TAA” refers to an antigen that is associated with or specific to a tumor.
  • TAAs examples include gplOO, MART and MAGE.
  • antibody is further intended to encompass digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or specified fragment or portion thereof, including single chain antibodies and fragments thereof.
  • binding fragments encompassed within the term "antigen binding portion" of an antibody include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH, domains; a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CH, domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH domain; and an isolated complementarity determining region (CDR).
  • Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH, domains
  • F(ab')2 fragment a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region
  • a Fd fragment consisting of the VH and CH, domains
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)).
  • scFv single chain Fv
  • Single chain antibodies are also intended to be encompassed within the term "fragment of an antibody.” Any of the above- noted antibody fragments are obtained using conventional techniques known to those of skill in the art, and the fragments are screened for binding specificity in the same manner as are intact antibodies. Such fragments can be produced by enzymatic cleavage, synthetic or recombinant techniques, as known in the art and/or as described herein. For example, an antibody fragment can be produced in a variety of truncated fo ⁇ ns using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site.
  • a combination gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and/or hinge region of the heavy chain.
  • the various portions of antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques.
  • the term "epitope" means a protein determinant capable of specific binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • antibody variant is intended to include antibodies produced in a species other than a mouse or an isotype of an antibody selected from the antibodies designated ACO-1 thorugh ACO-6.
  • antibody variant also includes antibodies containing post-translational modifications to the linear polypeptide sequence of the antibody or fragment. It further encompasses fully human antibodies.
  • antibody derivative is intended to encompass molecules that bind an epitope as defined above and which are modifications or derivatives of a native monoclonal antibody of this invention. Derivatives include, but are not limited to, for example, bispecific, multispecific, heterospecific, trispecific, tetraspecific, multispecific antibodies, diabodies, chimeric, recombinant and humanized.
  • bispecific molecule is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has two different binding specificities.
  • multispecific molecule or “heterospecific molecule” is intended to include any agent, e.g. a protein, peptide, or protein or peptide complex, which has more than two different binding specificities.
  • heteroantibodies refers to two or more antibodies, antibody binding fragments (e.g., Fab), derivatives thereof, or antigen binding regions linked together, at least two of which have different specificities.
  • human antibody as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • human antibody as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • human antibody refers to an antibody in which substantially every part of the protein (e.g., CDR, framework, C L , C H domains (e.g., C H ⁇ , C H2 , C H3 ), hinge, (V L , V H )) is substantially non-immunogenic in humans, with only minor sequence changes or variations.
  • antibodies designated primate monkey, baboon, chimpanzee, etc.
  • rodent mouse, rat, rabbit, guinea pig, hamster, and the like
  • other mammals designate such species, sub-genus, genus, sub-family, family specific antibodies.
  • chimeric antibodies include any combination of the above.
  • a human antibody is distinct from a chimeric or humanized antibody. It is pointed out that a human antibody can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes. Further, when a human antibody is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies.
  • an Fv can comprise a linker peptide, such as two to about eight glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain.
  • linker peptides are considered to be of human origin.
  • a human antibody is "derived from” a particular germline sequence if the antibody is obtained from a system using human immunoglobulin sequences, e.g., by immunizing a transgenic mouse carrying human immunoglobulin genes or by screening a human immunoglobulin gene library.
  • a human antibody that is "derived from” a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequence of human germline immunoglobulins.
  • a selected human antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the human antibody as being human when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences).
  • a human antibody may be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene.
  • a human antibody derived from a particular human germline sequence will display no more than 10 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene.
  • the human antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene.
  • the terms "monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • a "human monoclonal antibody” refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobulin sequences.
  • recombinant human antibody includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom, antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, antibodies isolated from a recombinant, combinatorial human antibody library, and antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences.
  • such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • “isotype” refers to the antibody class (e.g., IgM or IgGl) that is encoded by heavy chain constant region genes.
  • transgenic, nonhuman animal refers to a nonhuman animal having a genome comprising one or more human heavy and/or light chain transgenes or transchromosomes (either integrated or non-integrated into the animal's natural genomic DNA) and which is capable of expressing fully human antibodies.
  • a transgenic rat can have a human light chain transgene and either a human heavy chain transgene or human heavy chain transchromosome, such that the rat produces human anti-INF- ⁇ antibodies.
  • the human heavy chain transgene can be integrated into the chromosomal DNA of the rat, or the human heavy chain transgene can be maintained extrachromosomally.
  • Transgenic and transchromosomal animals are capable of producing multiple isotypes of human monoclonal antibodies to Alpha V (e.g., IgG, IgA and/or IgE) by undergoing V-D-J recombination and isotype switching.
  • the terms "major histocompatibility complex” or "MHC” refers to a complex of genes encoding cell-surface molecules that are required for antigen presentation to T cells and for rapid graft rejection. In humans, the MHC is also known as the "human leukocyte antigen" or "HLA” complex.
  • HLA human leukocyte antigen
  • the proteins encoded by the MHC are known as "MHC molecules" and are classified into class I and class II MHC molecules.
  • Class I MHC includes membrane heterodimeric proteins made up of an chain encoded in the MHC noncovalently linked with the /5 2 -microglobulin. Class I MHC molecules are expressed by nearly all nucleated cells and have been shown to function in antigen presentation to CD8 + T cells. Class I molecules include HLA-A, B, and C in humans. Class II MHC molecules also include membrane heterodimeric proteins consisting of noncovalently associated and ⁇ chains. Class II MHC molecules are known to function in CD4 + T cells and, in humans, include HLA-DP, -DQ, and DR. In a preferred embodiment, invention compositions and ligands can complex with MHC molecules of any HLA type.
  • antigen-presenting matrix intends a molecule or molecules which can present antigen in such a way that the antigen can be bound by a T-cell antigen receptor on the surface of a T cell.
  • An antigen-presenting matrix can be on the surface of an antigen-presenting cell (APC), on a vesicle preparation of an APC, or can be in the form of a synthetic matrix on a solid support such as a bead or a plate.
  • An example of a synthetic antigen-presenting matrix is purified MHC class I molecules complexed to ⁇ 2 -microglobulin, multimers of such purified MHC class I molecules, purified MHC Class II molecules, or functional portions thereof, attached to a solid support.
  • antigen presenting cells refers to a class of cells capable of presenting one or more antigens in the form of antigen-MHC complex recognizable by specific effector cells of the immune system, and thereby inducing an effective cellular immune response against the antigen or antigens being presented. While many types of cells may be capable of presenting antigens on their cell surface for T-cell recognition, only professional APCs have the capacity to present antigens in an efficient amount and further to activate T-cells for cytotoxic T-lymphocyte (CTL) responses.
  • CTL cytotoxic T-lymphocyte
  • APCs can be intact whole cells such as maerophages, B-cells and dendritic cells; or other molecules, naturally occurring or synthetic, such as purified MHC class I molecules complexed to ⁇ 2-microglobulin.
  • dendritic cells refers to a diverse population of morphologically similar cell types found in a variety of lymphoid and non-lymphoid tissues. (Steinman (1991) Ann. Rev. Immunol. 9:271-296). Dendritic cells constitute the most potent and preferred APCs in an organism.
  • dendritic cells are derived from bone marrow progenitor cells, circulate in small numbers in the peripheral blood and appear either as immature Langerhans' cells or terminally differentiated mature cells. While the dendritic cells can be differentiated from monocytes, they possess distinct phenotypes. For example, a particular differentiating marker, CD 14 antigen, is not found in dendritic cells but is possessed by monocytes. Also, mature dendritic cells are not phagocytic, whereas the monocytes are strongly phagocytosing cells. It has been shown that DCs provide all the signals necessary for T cell activation and proliferation.
  • APC recruitment factors include both intact, whole cells as well as other molecules that are capable of recruiting antigen presenting cells.
  • suitable APC recruitment factors include molecules such as interleukin 4 (IL-4), granulocyte macrophage colony stimulating factor (GM-CSF), Sepragel and macrophage inflammatory protein-3 -alpha (MIP3 ⁇ ). These are available from Immunex, Schering-Plough and R&D Systems (Minneapolis, MN). They also can be recombinantly produced using the methods disclosed in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY ((1987) supra). Peptides, proteins and compounds having the same biological activity as the above-noted factors are included within the scope of this invention.
  • immune effector cells refers to cells capable of binding an antigen and which mediate an immune response. These cells include, but are not limited to, T cells, B cells, monocytes, maerophages, NK cells and cytotoxic T lymphocytes (CTLs), for example CTL lines, CTL clones, and CTLs from tumor, inflammatory, or other infiltrates. Certain diseased tissue expresses specific antigens and CTLs specific for these antigens have been identified. For example, approximately 80% of melanomas express the antigen known as gplOO.
  • immune effector molecule refers to molecules capable of antigen-specific binding, and includes antibodies, T cell antigen receptors, and MHC Class I and Class II molecules.
  • a "naive" immune effector cell is an immune effector cell that has never been exposed to an antigen capable of activating that cell. Activation of naive immune effector cells requires both recognition of the peptide:MHC complex and the simultaneous delivery of a costimulatory signal by a professional APC in order to proliferate and differentiate into antigen-specific armed effector T cells.
  • a peptide of the invention binds to a ligand such as an antigenic determinant or epitope on an immune effector cell, e.g.
  • a ligand may be an antigen, peptide, protein or epitope of the invention.
  • Invention ligands may bind to a receptor on an antibody.
  • the peptides of the invention is about 4 to about 10 amino acids in length.
  • ⁇ Invention peptides may bind to a receptor on an MHC class I molecule.
  • the peptide of the invention is about 7 to about 12 amino acids in length.
  • Invention peptides may bind to a receptor on an MHC class II molecule.
  • the peptide of the invention is about 8 to about 20 amino acids long.
  • the term "educated, antigen-specific immune effector cell” is an immune effector cell as defined above, which has previously encountered an antigen. In contrast with its na ⁇ ve counterpart, activation of an educated, antigen-specific-immune effector cell does not require a costimulatory signal. Recognition of the peptide:MHC complex is sufficient. "Activated”, when used in reference to a T cell, implies that the cell is no longer in G 0 phase, and begins to produce one or more of cytotoxins, cytokines, and other related membrane-associated proteins characteristic of the cell type (e.g., CD8 + or CD4 + ), is capable of recognizing and binding any target cell that displays the particular antigen on its surface, and releasing its effector molecules.
  • Activated when used in reference to a T cell, implies that the cell is no longer in G 0 phase, and begins to produce one or more of cytotoxins, cytokines, and other related membrane-associated proteins characteristic of the cell type (e.g., CD8 + or CD
  • the term "recognized” intends that a peptide of the invention, is recognized and bound by its ligand, e.g., antibody, Tcell, or an immune effector cell wherein such binding initiates an effective immune response.
  • ligand e.g., antibody, Tcell, or an immune effector cell wherein such binding initiates an effective immune response.
  • cross-reactive is used to describe certain immunogenic properties of peptides which are functionally overlapping. More particularly, the immunogenic properties of a peptide and/or immune effector cells activated thereby are shared to a certain extent by other peptides.
  • cross-reactivity is manifested at multiple levels: (i) at the peptide level, the peptides can bind the TCR and activate CTLs; (ii) at the T cell level, i.e., peptides of the invention bind the TCR of and activate a population of T cells which can effectively target and lyse cells; and (iii) at the antibody level, e.g., "anti"-peptide antibodies can detect, recognize and bind the peptide and initiate effector mechanisms in an immune response.
  • the term "inducing an immune response in a subject” is a term understood in the art and intends that an increase of at least about 2-fold, more preferably at least about 5-fold, more preferably at least about 10-fold, more preferably at least about 100-fold, even more preferably at least about 500-fold, even more preferably at least about 1000-fold or more in an immune response to an antigen (or epitope) can be detected or measured, after introducing the antigen (or epitope) into the subject, relative to the immune response (if any) before introduction of the antigen (or epitope) into the subject.
  • An immune response to an antigen includes, but is not limited to, production of an antigen-specific (or epitope-specific) antibody, and production of an immune cell expressing on its surface a molecule which specifically binds to an antigen (or epitope).
  • Methods of determining whether an immune response to a given antigen (or epitope) has been induced are well known in the art.
  • antigen-specific antibody can be detected using any of a variety of immunoassays known in the art, including, but not limited to, ELISA, wherein, for example, binding of an antibody in a sample to an immobilized antigen (or epitope) is detected with a detectably-labeled second antibody (e.g., enzyme-labeled mouse anti-human Ig antibody).
  • ELISA electrospray-activated immunosorbent assay
  • a detectably-labeled second antibody e.g., enzyme-labeled mouse anti-human Ig antibody.
  • Co-stimulatory molecules are involved in the interaction between receptor-ligand pairs expressed on the surface of antigen presenting cells and T cells. Research accumulated over the past several years has demonstrated convincingly that resting T cells require at least two signals for induction of cytokine gene expression and proliferation. (Schwartz R.H. (1990) Science 248:1349-1356 and Jenkins M.K.
  • HSA heat stable antigen
  • Ii-CS chondroitin sulfate-modified MHC invariant chain
  • Ii-CS chondroitin sulfate-modified MHC invariant chain
  • Ii-CS chondroitin sulfate-modified MHC invariant chain
  • Ii-CS chondroitin sulfate-modified MHC invariant chain
  • Ii-CS intracellular adhesion molecule 1
  • IAM-1 Intrasion molecule 1
  • B7-1, and B7-2/B70 Schwartz R.H. (1992) Cell 71:1065-1068.
  • Co-stimulatory molecules mediate co-stimulatory signal(s), which are necessary, under normal physiological conditions, to achieve full activation of naive T cells.
  • One exemplary receptor-ligand pair is the B7 co-stimulatory molecule on the surface of APCs and its counter-receptor CD28 or CTLA-4 on T cells. (Freeman, et al. (1993) Science 262:909-911; Young, et al. (1992) J. Clin. Invest 90:229 and Nabavi, et al. (1992) Nature 360:266-268).
  • Other important co-stimulatory molecules are CD40, CD54, CD80, and CD86.
  • co-stimulatory molecule encompasses any single molecule or combination of molecules which, when acting together with a peptide/MHC complex bound by a TCR on the surface of a T cell, provides a co-stimulatory effect which achieves activation of the T cell that binds the peptide.
  • the term thus encompasses B7, or other co-stimulatory molecule(s) on an antigen-presenting matrix such as an APC, fragments thereof (alone, complexed with another molecule(s), or as part of a fusion protein) which, together with peptide/MHC complex, binds to a cognate ligand and results in activation of the T cell when the TCR on the surface of the T cell specifically binds the peptide.
  • Co-stimulatory molecules are commercially available from a variety of sources, including, for example, Beckman Coulter, Inc. (Fullerton, CA). It is intended, although not always explicitly stated, that molecules having similar biological activity as wild-type or purified co-stimulatory molecules (e.g. , recombinantly produced or muteins thereof) are intended to be used within the spirit and scope of the invention.
  • solid phase support or “solid support”, used interchangeably, is not limited to a specific type of support. Rather a large number of supports are available and are known to one of ordinary skill in the art.
  • Solid phase supports include silica gels, resins, derivatized plastic films, glass beads, glass slides, flasks, tissue culture flasks, cotton, plastic beads, alumina gels.
  • solid support also includes synthetic antigen-presenting matrices, cells, and liposomes.
  • a suitable solid phase support may be selected on the basis of desired end use and suitability for various protocols.
  • solid phase support may refer to resins such as polystyrene (e.g., PAM-resin obtained from Bachem Inc., Peninsula Laboratories, etc.), POLYHIPE ® resin (obtained from Aminotech, Canada), polyamdde resin (obtained from Peninsula Laboratories), polystyrene resin grafted with polyethylene glycol (TentaGel®, Rapp Polymere, Tubingen, Germany) or polydimethylacrylamide resin (obtained from Milligen/Biosearch, California).
  • Solid supports also include microchips and grids, on which cDNAs, oligonucleotides, peptides, antibodies or other molecules are fixed in arrays.
  • the surface of the grids may be composed of a wide variety of material including glass, plastic, silicon, gold, gelatin or nylon.
  • solid support tissue microarrays in which small cylinders of tissue are punched out of thousands of individual tumor specimens (from different tissues of hundreds of individuals in a study) and then probed with antibodies, RNA, etc. (Hoos A., et al. (2001) Am J. Pathol. 158:1245-51).
  • the components and compositions for use in the methods of this invention can also provided on microchips. For example, the use of the so-called
  • SELDI-MS method Surface-Enhanced Laser Desorption-Ionization & Mass Spectroscopy exposes samples to chips with biochemically characterized surfaces (containing molecules such as antibodies or receptors) followed by mass spectroscopy to visualize and identify the bound proteins.
  • biochemically characterized surfaces containing molecules such as antibodies or receptors
  • mass spectroscopy to visualize and identify the bound proteins.
  • immunomodulatory agent is a molecule, a macromolecular complex, or a cell that modulates an immune response and encompasses a peptide of the invention alone or in any of a variety of formulations described herein; a polypeptide comprising a peptide of the invention; a polynucleotide encoding a peptide or polypeptide of the invention; a peptide of the invention bound to a Class I or a Class II MHC molecule on an antigen-presenting matrix, including an APC and a synthetic antigen-presenting matrix (in the presence or absence of co-stimulatory molecule(s)); a peptide of the invention covalently or non-covalently complexed to another molecule(s) or macromolecular structure; and an educated, antigen-specific immune effector cell which is specific for a peptide of the
  • modulate an immune response includes inducing (increasing, eliciting) an immune response; and reducing (suppressing) an immune response.
  • An immunomodulatory method is one that modulates an immune response in a subject.
  • cytokine refers to any one of the numerous factors that exert a variety of effects on cells, for example, inducing growth or proliferation.
  • Non-limiting examples of cytokines which may be used alone or in combination in the practice of the present invention include, interleukin-2 (IL-2), stem cell factor (SCF), interleukin 3 (IL-3), interleukin 6 (IL-6), interleukin 12 (IL-12), G-CSF, granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin- 1 alpha (IL- 1 ⁇ ), interleukin- 11 (IL- 11), MIP- 11 , leukemia inhibitory factor (LIF), c-kit ligand, thrombopoietin (TPO) and flt3 ligand.
  • the present invention also includes culture conditions in which one or more cytokine is specifically excluded from the medium. Cytokines are commercially available from several vendors such as, for example, Genzyme Corp. (Framingham, MA), Genentech (South San Francisco, CA), Amgen (Thousand Oaks, CA), R&D
  • polynucleotide and “oligonucleotide” are used interchangeably, and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Polynucleotides can have any three-dimensional structure, and may perform any function, known or unknown.
  • polynucleotides a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers.
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs.
  • modifications to the nucleotide structure can be imparted before or after assembly of the polymer.
  • the sequence of nucleotides can be interrupted by non-nucleotide components.
  • a polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component.
  • the term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
  • a polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for guanine when the polynucleotide is RNA.
  • polynucleotide sequence is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
  • a “gene” refers to a polynucleotide containing at least one open reading frame (ORF) that is capable of encoding a particular polypeptide or protein after being transcribed and translated. Any of the polynucleotides sequences described herein may be used to identify larger fragments or full-length coding sequences of the gene with which they are associated. Methods of isolating larger fragment sequences are known to those of skill in the art.
  • a “gene product” or alternatively a “gene expression product” refers to the amino acid (e.g., peptide or polypeptide) generated when a gene is transcribed and translated.
  • polypeptide is used interchangeably with the term “protein” and in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs, or peptidomimetics.
  • the subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g., ester, ether, etc.
  • amino acid refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics.
  • a peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short.
  • the peptide is commonly called a polypeptide or a protein.
  • the term "peptide” is used in its broadest sense to refer to a compound of two or more subunit amino acids.
  • the subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g. ester, ether, etc.
  • amino acid refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers.
  • a peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein.
  • genetically modified means containing and/or expressing a foreign gene or nucleic acid sequence which in turn, modifies the genotype or phenotype of the cell or its progeny.
  • the term also refers to any enhancement, addition, deletion or disruption to a cell's endogenous nucleotides.
  • expression refers to the process by which polynucleotides are transcribed into mRNA and translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA, if an appropriate eukaryotic host is selected.
  • a bacterial expression vector includes a promoter such as the lac promoter and for transcription initiation the Shine-Dalgarno sequence and the start codon AUG (Sambrook et al. (1989) supra).
  • an eukaryotic expression vector includes a heterologous or homologous promoter for RNA polymerase II, a downstream polyadenylation signal, the start codon AUG, and a termination codon for detachment of the ribosome.
  • Such vectors can be obtained commercially or assembled by the sequences described by methods known in the art.
  • Under transcriptional control is a term understood in the art and indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operatively linked to an element which contributes to the initiation of, or promotes, transcription. "Operatively linked” refers to a juxtaposition wherein the elements are in an arrangement allowing them to function.
  • a “gene delivery vehicle” is defined as any molecule that can carry inserted polynucleotides into a host cell.
  • liposomes examples include liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; recombinant yeast cells or yeast spheroplasts; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • Gene delivery are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene") into a host cell, irrespective of the method used for the introduction.
  • exogenous polynucleotide sometimes referred to as a "transgene”
  • Such methods include a variety of techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfection, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation, "gene gun” delivery and various other techniques used for the introduction of polynucleotides).
  • the introduced polynucleotide may be stably or transiently maintained in the host cell.
  • Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome.
  • a number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein.
  • a "viral vector” is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro.
  • viral vectors examples include retroviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like.
  • Alphavirus vectors such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. (See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Zaks, et al. (1999) Nat. Med. 7:823-827).
  • a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene.
  • retroviral mediated gene transfer or “retroviral transduction” carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome.
  • the virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell.
  • retroviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism.
  • Retro viruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell.
  • the integrated DNA form is called a pro virus.
  • a vector construct refers to the polynucleotide comprising the viral genome or part thereof, and a transgene.
  • Ads adenoviruses
  • Ads are a relatively well characterized, homogenous group of viruses, including over 50 serotypes. See, e.g., WO 95/27071. Ads are easy to grow and do not require integration into the host cell genome. Recombinant
  • Ad-derived vectors particularly those that reduce the potential for recombination and generation of wild-type virus, have also been constructed.
  • Wild-type AAV has high infectivity and specificity integrating into the host cell's genome.
  • Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are known in the art.
  • Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available.
  • consensus ribosome binding sites can be inserted immediately 5' of the start codon to enhance expression.
  • Gene delivery vehicles also include several non- viral vectors, including DNA/liposome complexes, and targeted viral protein-DNA complexes. Liposomes that also comprise a targeting antibody or fragment thereof can be used in the methods of this invention.
  • the nucleic acid or proteins of this invention can be conjugated to antibodies or binding fragments thereof which bind cell surface antigens, e.g., TCR, CD3 or CD4.
  • “Hybridization” refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner.
  • the complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these.
  • a hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
  • stringent hybridization conditions include: incubation temperatures of about 25°C to about 37°C; hybridization buffer concentrations of about 6 X SSC to about 10 X SSC; formamide concentrations of about 0% to about 25%; and wash solutions of about 6 X SSC.
  • Examples of moderate hybridization conditions include: incubation temperatures of about 40°C to about 50°C; buffer concentrations of about 9 X SSC to about 2 X SSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5 X SSC to about 2 X SSC.
  • Examples of high stringency conditions include: incubation temperatures of about 55°C to about 68°C; buffer concentrations of about 1 X SSC to about 0.1 X SSC; formamide concentrations of about 55% to about 75%; and wash solutions of about 1 X SSC, 0.1 X SSC, or deionized water.
  • hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash incubation times are about 1, 2, or 15 minutes.
  • a "probe" when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially present in a sample of interest by hybridizing with the target.
  • a probe will comprise a label or a means by which a label can be attached, either before or subsequent to the hybridization reaction.
  • Suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes.
  • a “primer” is a short polynucleotide, generally with a free 3' -OH group that binds to a target or "template” potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target.
  • a “polymerase chain reaction” (“PCR”) is a reaction in which replicate copies are made of a target polynucleotide using a "pair of primers” or a “set of primers” consisting of an "upstream” and a “downstream” primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme. Methods for PCR are known in the art, and taught, for example in “PCR: A PRACTICAL APPROACH” (M. MacPherson et al, IRL Press at Oxford University Press
  • a primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook et al., supra.
  • An expression “database” denotes a set of stored data that represent a collection of sequences, which in turn represent a collection of biological reference materials.
  • cDNAs refers to complementary DNA that are mRNA molecules present in a cell or organism and made into cDNA with an enzyme such as reverse transcriptase.
  • a “cDNA library” is a collection of all of the mRNA molecules present in a cell or organism, all turned into cDNA molecules with the enzyme reverse transcriptase, then inserted into “vectors” (other DNA molecules that can continue to replicate after addition of foreign DNA).
  • vectors for libraries include bacteriophage (also known as “phage"), viruses that infect bacteria, for example, lambda phage.
  • the library can then be probed for the specific cDNA (and thus mRNA) of interest.
  • expression refers to the process by which polynucleotides are transcribed into mRNA and/or the process by which the transcribed mRNA is subsequently being translated into peptides, polypeptides, or proteins.
  • expression may include splicing of the mRNA in a eukaryotic cell.
  • "Differentially expressed” as applied to a gene refers to the differential production of the mRNA transcribed and/or translated from the gene or the protein product encoded by the gene.
  • a differentially expressed gene may be overexpressed or underexpressed as compared to the expression level of a normal or control cell. In one aspect, it refers to a differential that is 2.5 times, preferably 5 times, or preferably 10 times higher or lower than the expression level detected in a control sample.
  • the term "differentially expressed” also refers to nucleotide sequences in a cell or tissue which are expressed where silent in a control cell or not expressed where expressed in a control cell.
  • a polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) has a certain percentage (for example, 80%, 85%, 90%, or 95%) of "sequence identity" to another sequence means that, when aligned, that percentage of bases (or amino acids) are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY ((1987) supra Supplement 30, section 7.7.18, Table 7.7.1).
  • default parameters are used for alignment.
  • the alignment program is BLAST, using default parameters.
  • In vivo gene delivery, gene transfer, gene therapy and the like as used herein, are terms referring to the introduction of a vector comprising an exogenous polynucleotide directly into the body of an organism, such as a human or non-human mammal, whereby the exogenous polynucleotide is introduced to a cell of such organism in vivo.
  • isolated means separated from constituents, cellular and otherwise, in which the polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, are normally associated with in nature.
  • an isolated polynucleotide is one that is separated from the 5' and 3' sequences with which it is normally associated in the chromosome.
  • a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof does not require "isolation" to distinguish it from its naturally occurring counterpart.
  • a "concentrated”, “separated” or “diluted” polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof is distinguishable from its naturally occurring counterpart in that the concentration or number of molecules per volume is greater than “concentrated” or less than “separated” than that of its naturally occurring counterpart.
  • a non-naturally occurring polynucleotide is provided as a separate embodiment from the isolated naturally occurring polynucleotide.
  • a protein produced in a bacterial cell is provided as a separate embodiment from the naturally occurring protein isolated from a eukaryotic cell in which it is produced in nature.
  • "Host cell,” “target cell” or “recipient cell” are intended to include any individual cell or cell culture which can be or have been recipients for vectors or the inco ⁇ oration of exogenous nucleic acid molecules, polynucleotides and/or proteins.
  • the cells may be prokaryotic or eukaryotic, and include but are not limited to bacterial cells, yeast cells, animal cells, and mammalian cells, e.g., murine, rat, simian or human.
  • a "subject” is a vertebrate, in one aspect a mammal, and in another aspect a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets.
  • a “control” is an alternative subject or sample used in an experiment for comparison pu ⁇ ose.
  • a control can be "positive” or “negative”.
  • the pu ⁇ ose of the experiment is to determine a correlation of an altered expression level of a gene with a particular type of cancer
  • generally positive control is used (a subject or a sample from a subject, carrying such alteration and exhibiting syndromes characteristic of that disease), and a negative control (a subject or a sample from a subject lacking the altered expression and clinical syndrome of that disease).
  • cancer “neoplasm,” and “tumor,” used interchangeably and in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism.
  • Primary cancer cells that is, cells obtained from near the site of malignant transformation
  • the definition of a cancer cell includes not only a primary cancer cell, but also any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and cell lines derived from cancer cells.
  • a "clinically detectable" tumor is one that is detectable on the basis of tumor mass; e.g., by such procedures as CAT scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation. Biochemical or immunologic findings alone may be insufficient to meet this definition.
  • a neoplasm is an abnormal mass or colony of cells produced by a relatively autonomous new growth of tissue. Most neoplasms arise from the clonal expansion of a single cell that has undergone neoplastic transformation. The transformation of a normal to a neoplastic cell can be caused by a chemical, physical, or biological agent (or event) that directly and irreversibly alters the cell genome.
  • Neoplastic cells are characterized by the loss of some specialized functions and the acquisition of new biological properties, foremost, the property of relatively autonomous (uncontrolled) growth. Neoplastic cells pass on their heritable biological characteristics to progeny cells.
  • a malignant neoplasm manifests a greater degree of autonomy, is capable of invasion and metastatic spread, may be resistant to treatment, and may cause death.
  • a benign neoplasm has a lesser degree of autonomy, is usually not invasive, does not metastasize, and generally produces no great harm if treated adequately.
  • Cancer is a generic term for malignant neoplasms. Anaplasia is a characteristic property of cancer cells and denotes a lack of normal structural and functional characteristics (undifferentiation).
  • a tumor is literally a swelling of any type, such as an inflammatory or other swelling, but modern usage generally denotes a neoplasm.
  • the suffix "-oma” means tumor and usually denotes a benign neoplasm, as in fibroma, lipoma, and so forth, but sometimes implies a malignant neoplasm, as with so-called melanoma, hepatoma, and seminoma, or even a non-neoplastic lesion, such as a hematoma, granuloma, or hamartoma.
  • -blastoma denotes a neoplasm of embryonic cells, such as neuroblastoma of the adrenal or retinoblastoma of the eye.
  • Histogenesis is the origin of a tissue and is a method of classifying neoplasms on the basis of the tissue cell of origin.
  • Adenomas are benign neoplasms of glandular epithelium.
  • Carcinomas are malignant tumors of epithelium.
  • Sarcomas are malignant tumors of mesenchymal tissues.
  • One system to classify neoplasia utilizes biological (clinical) behavior, whether benign or malignant, and the histogenesis, the tissue or cell of origin of the neoplasm as determined by histologic and cytologic examination.
  • Neoplasms may originate in almost any tissue containing cells capable of mitotic division.
  • the histogenetic classification of neoplasms is based upon the tissue (or cell) of origin as determined by histologic and cytologic examination. "Suppressing" tumor growth indicates a growth state that is curtailed compared to growth without contact with educated, antigen-specific immune effector cells described herein. Tumor cell growth can be assessed by any means known in the art, including, but not limited to, measuring tumor size, determining whether tumor cells are proliferating using a 3 H-thymidine inco ⁇ oration assay, or counting tumor cells.
  • “Suppressing” tumor cell growth means any or all of the following states: slowing, delaying, and “suppressing” tumor growth indicates a growth state that is curtailed when stopping tumor growth, as well as tumor shrinkage.
  • the term “culturing” refers to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (mo ⁇ hologically, genetically, or phenotypically) to the parent cell. By “expanded” is meant any proliferation or division of cells.
  • composition is intended to mean a combination of active agent and another carrier, e.g., compound or composition, inert (for example, a detectable agent or label) or active, such as an adjuvant, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant or the like.
  • carrier e.g., compound or composition
  • inert for example, a detectable agent or label
  • active such as an adjuvant, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant or the like.
  • Carriers also include pharmaceutical excipients and additives proteins, peptides, amino acids, lipids, and carbohydrates (e.g., sugars, including monosaccharides, di-, tri-, tetra-, and oligosaccharides; derivatized sugars such as alditols, aldonic acids, esterified sugars and the like; and polysaccharides or sugar polymers), which can be present singly or in combination, comprising alone or in combination 1-99.99% by weight or volume.
  • Exemplary protein excipients include serum albumin such as human seram albumin (HSA), recombinant human albumin (rHA), gelatin, casein, and the like.
  • amino acid/antibody components which can also function in a buffering capacity, include alanine, glycine, arginine, betaine, histidine, glutamic acid, aspartic acid, cysteine, lysine, leucine, isoleucine, valine, methionine, phenylalanine, aspartame, and the like.
  • Carbohydrate excipients are also intended within the scope of this invention, examples of which include but are not limited to monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffmose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol) and myoinositol.
  • monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like
  • disaccharides such as lactose, sucrose,
  • the term carrier further includes a buffer or a pH adjusting agent; typically, the buffer is a salt prepared from an organic acid or base.
  • Representative buffers include organic acid salts such as salts of citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid, or phthalic acid; Tris, tromethamine hydrochloride, or phosphate buffers.
  • Additional carriers include polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl-. quadrature.
  • the term "pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • compositions also can include stabilizers and preservatives and any of the above noted carriers with the additional provisio that they be acceptable for use in vivo.
  • carriers stabilizers and adjuvants
  • a "pharmaceutical composition” is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
  • An "effective amount” is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages.
  • the present invention provides novel polypeptide compounds and related compositions for therapeutic, prophylactic and diagnostic use.
  • this invention provides polypeptides which were heretofore not known to be highly immunogenic.
  • the polypeptides of the invention are useful in therapeutic, prophylactic, and diagnostic applications.
  • polynucleotides encoding the peptides of the invention gene delivery vehicles comprising these polynucleotides and host cells comprising these polynucleotides.
  • the invention provides methods for inducing an immune response in a subject by delivering to the subject an effective amount of the peptides of the invention in the context of an MHC molecule.
  • the peptides of the invention are also useful as immunogens to generate antibodies that specifically recognize and bind to these peptides and cross-react with related peptides.
  • the peptides of the invention can also be used in recombinant methods to construct antibodies that specifically recognize and bind to these peptides.
  • the antibodies so generated are further useful in therapeutic and diagnostic applications.
  • the invention further provides immune effector cells raised in vivo or in vitro in the presence and at the expense of an antigen presenting cell (APC) that presents the peptides of the invention in the context of an MHC molecule and a method of adoptive immunotherapy comprising administering an effective amount of these immune effector cells to a subject.
  • APC antigen presenting cell
  • compositions of the invention comprise at least one, peptide of the invention.
  • such compositions may comprise at least one, or alternatively, two or more copies of a single peptide.
  • such compositions may comprise two or more peptides, wherein each peptide of said two or more peptides is distinct from all other peptides in the composition.
  • the two or more immunogenic peptides are covalently linked.
  • the proteins and polypeptides of this invention can be obtained by chemical synthesis using a commercially available automated peptide synthesizer such as those manufactured by Perkin Elmer/Applied Biosystems, Inc., Model 430A or 431 A, Foster City, CA, USA.
  • the synthesized protein or polypeptide can be precipitated and further purified, for example by high performance liquid chromatography (HPLC).
  • HPLC high performance liquid chromatography
  • this invention also provides a process for chemically synthesizing the proteins of this invention by providing the sequence of the protein and reagents, such as amino acids and enzymes and linking together the amino acids in the proper orientation and linear sequence.
  • the proteins and polypeptides can be obtained by recombinant methods known in the art using the host cell and vector systems described below.
  • amino acid refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics.
  • a peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein.
  • Peptides of the invention can include unnatural amino acids.
  • the peptides may comprise D-amino acids, a combination of D- and L-amino acids, and various "designer" amino acids (e.g., ⁇ -methyl amino acids, C- ⁇ -methyl amino acids, and N- ⁇ -methyl amino acids, etc.) to convey special properties to peptides.
  • various "designer" amino acids e.g., ⁇ -methyl amino acids, C- ⁇ -methyl amino acids, and N- ⁇ -methyl amino acids, etc.
  • peptides with ⁇ -helices ⁇ turns, ⁇ sheets, ⁇ -turns, and cyclic peptides can be generated.
  • ⁇ -helical secondary structure or random secondary structure is preferred.
  • subunits of peptides that confer useful chemical and structural properties will be used.
  • peptides comprising D-amino acids will be resistant to L-amino acid-specific proteases in vivo.
  • Modified compounds with D-amino acids may be synthesized with the amino acids aligned in reverse order to produce the peptides of the invention as retro-inverso peptides.
  • the present invention envisions preparing peptides that have better defined structural properties, and the use of peptidomimetics, and peptidomimetic bonds, such as ester bonds, to prepare peptides with novel properties.
  • a peptide may be generated that incorporates a reduced peptide bond, i. e.
  • Ri -CH 2 NH-R 2 where Ri, and R 2 are amino acid residues or sequences.
  • a reduced peptide bond may be introduced as a dipeptide subunit.
  • Such a molecule would be resistant to peptide bond hydrolysis, e.g., protease activity.
  • Such molecules would provide ligands with unique function and activity, such as extended half-lives in vivo due to resistance to metabolic breakdown, or protease activity.
  • NON-CLASSICAL AMINO ACIDS THAT INDUCE CONFORMATIONAL CONSTRAINTS The following non classical amino acids may be inco ⁇ orated in the peptides of the invention in order to introduce particular conformational motifs: l,2,3,4-tetrahydroisoquinoline-3-carboxylate (Kazamierski et al. (1991) J. Am. Chem. Soc.
  • histidine isoquinoline carboxylic acid Zechel et al. (1991) Int. J. Pep. Protein Res. 38(2):131-138); and HIC (histidine cyclic urea), (Dharanipragada et al. (1993) Int. J. Pep. Protein Res. 42(l):68-77) and ((1992) Acta. Cryst, Crystal Struc. Comm. 48(IV): 1239-1241).
  • LL-Acp LL-3-amino- 2- ⁇ ropenidone-6-carboxylic acid
  • ⁇ -turn inducing dipeptide analog Kemp et al. (1985) J. Org. Chem. 50:5834-5838
  • ⁇ -sheet inducing analogs Kemp et al. (1988) Tetrahedron Lett. 29:5081-5082
  • ⁇ -turn inducing analogs Kemp et al. (1988) Tetrahedron Lett.
  • a peptide of the invention can be covalently or non-covalently linked (complexed) to various other molecules, the nature of which may vary depending on the particular pu ⁇ ose.
  • a peptide of the invention can be covalently or non-covalently complexed to a macromolecular carrier, including, but not limited to, natural and synthetic polymers, proteins, polysaccharides, polypeptides (amino acids), polyvinyl alcohol, polyvinyl pyrrolidone, and lipids.
  • a peptide can be conjugated to a fatty acid, for introduction into a liposome. (See, U.S. Patent No. 5,837,249).
  • a peptide of the invention can be complexed covalently or non-covalently with a solid support, a variety of which are known in the art.
  • a peptide of the invention can be associated with an antigen-presenting matrix with or without co-stimulatory molecules, as described in more detail below.
  • protein carriers include, but are not limited to, superantigens, serum albumin, tetanus toxoid, ovalbumin, thyroglobulin, myoglobulin, and immunoglobulin.
  • Peptide-protein carrier polymers may be formed using conventional cross-linking agents such as carbodimides.
  • carbodimides examples include l-cyclohexyl-3-(2-mo ⁇ holinyl-(4-ethyl) carbodiimide (CMC), l-ethyl-3-(3-dimethyaminopropyl) carbodiimide (EDC) and 1 -ethyl-3 -(4-azonia-44-dimethylpentyl) carbodiimide.
  • CMC l-cyclohexyl-3-(2-mo ⁇ holinyl-(4-ethyl) carbodiimide
  • EDC l-ethyl-3-(3-dimethyaminopropyl) carbodiimide
  • 1 -ethyl-3 -(4-azonia-44-dimethylpentyl) carbodiimide examples include cyanogen bromide, glutaraldehyde and succinic anhydride.
  • any of a number of homo-bifunctional agents including a homo-bifunctional aldehyde, a homo-bifunctional epoxide, a homo-bifunctional imido-ester, a homo-bifunctional N-hydroxysuccinimide ester, a homo-bifunctional maleimide, a homo-bifunctional alkyl halide, a homo-bifunctional pyridyl disulfide, a homo-bifunctional aryl halide, a homo-bifunctional hydrazide, a homo-bifunctional diazonium derivative and a homo-bifunctional photoreactive compound may be used.
  • hetero-bifunctional compounds for example, compounds having an amine-reactive and a sulfhydryl-reactive group, compounds with an amine-reactive and a photoreactive group and compounds with a carbonyl-reactive and a sulfhydryl-reactive group.
  • homo-bifunctional cross-linking agents include the bifunctional N-hydroxysuccinimide esters dithiobis(succinimidy ⁇ propionate), disuccinimidyl suberate, and disuccinimidyl tartarate; the bifunctional imido-esters dimethyl adipimidate, dimethyl pimelimidate, and dimethyl suberimidate; the bifunctional sulfhydryl-reactive crosslinkers l,4-di-[3'-(2'-pyridyldithio) propion-amido]butane, bismaleimidohexane, and bis-N-maleimido-1, 8-octane; the bifunctional aryl halides l,5-difluoro-2,4-dinitrobenzene and
  • bifunctional photoreactive agents such as bis-[b-(4-azidosalicylamido)ethyl]disulfide
  • bifunctional aldehydes formaldehyde, malondialdehyde, succinaldehyde, glutaraldehyde, and adipaldehyde
  • a bifunctional epoxide such as 1,4-butaneodiol diglycidyl ether
  • the bifunctional hydrazides adipic acid dihydrazide, carbohydrazide, and succinic acid dihydrazide
  • the bifunctional diazoniums o-tolidine diazotized and bis-diazotized benzidine
  • the bifunctional alkylhalides NlN'-ethylene-bis(iodoacetamide), NlN'-hexamethylene-bis(iodoacetamide),
  • SMCC succinimidyl-4-(N-maleimidomethyl)cyclohexane- 1 -carboxylate
  • MBS m-maleimidobenzoyl-N-hydroxysuccinimide ester
  • SLAB N-succinimidyl(4-iodoacteyl)aminobenzoate
  • SMPB succinimidyl-4-(p-maleimidophenyl)butyrate
  • GMBS N-( ⁇ -maleimidobutyryloxy)succinimide ester
  • MPBH 4-(4-N-maleimidopohenyl) butyric acid hydrazide
  • M2C2H 4-(N-maleimidomethyl) cyclohexane-1-carboxyl-hydrazide
  • SMPT succin
  • Cross-linking may be accomplished by coupling a carbonyl group to an amine group or to a hydrazide group by reductive animation.
  • Peptides of the invention also may be formulated as non-covalent attachment of monomers through ionic, adso ⁇ tive, or biospecific interactions. Complexes of peptides with highly positively o negatively charged molecules may be done through salt bridge formation under low ionic strength environments, such as in deionized water. Large complexes can be created using charged polymers such as poly-(L-glutamic acid) or poly-(L-lysine) which contain numerous negative and positive charges, respectively.
  • peptides may be non-covalently linked through the use of biospecific interactions between other molecules. For instance, utilization of the strong affinity of biotin for proteins such as avidin or streptavidin or their derivatives could be used to form peptide complexes. These biotin-binding proteins contain four binding sites that can interact with biotin in solution or be covalently attached to another molecule. (Wilchek (1988) Anal. Biochem. 171:1-32).
  • Peptides can be modified to possess biotin groups using common biotinylation reagents such as the N-hydroxysuccinimidyl ester of D-biotin (NHS-biotin) which reacts with available amine groups on the protein. Biotinylated peptides then can be incubated with avidin or streptavidin to create large complexes. The molecular mass of such polymers can be regulated through careful control of the molar ratio of biotinylated peptide to avidin or streptavidin. Also provided by this application are the peptides and polypeptides described herein conjugated to a detectable agent for use in the diagnostic methods.
  • detectably labeled peptides and polypeptides can be bound to a column and used for the detection and purification of antibodies. They also are useful as immunogens for the production of antibodies, as described below.
  • the peptides of this invention also can be combined with various liquid phase carriers, such as sterile or aqueous solutions, pharmaceutically acceptable carriers, suspensions and emulsions.
  • non-aqueous solvents include propyl ethylene glycol, polyethylene glycol and vegetable oils.
  • the carriers also can include an adjuvant that is useful to non-specifically augment a specific immune response. A skilled artisan can easily determine whether an adjuvant is required and select one.
  • suitable adjuvants include, but are not limited to, Freund's Complete and Incomplete, mineral salts and polynucleotides.
  • POLYNUCLEOTIDES This invention further provides polynucleotides encoding polypeptides comprising one or more of the sequences of shown in Table 1, and the complements of these polynucleotides.
  • a "variant polynucleotide” means alternate polynucleotides sequence ' s to those shown in Table 1 (due to the degeneracy of the genetic code) that code for the same polypeptide.
  • polynucleotide encompasses DNA, RNA and nucleic acid mimetics.
  • this invention also provides the anti-sense polynucleotide stand, e.g. antisense RNA to the sequences or their complements.
  • antisense polynucleotide stand e.g. antisense RNA to the sequences or their complements.
  • the polynucleotides of this invention can be replicated using PCR.
  • PCR technology is the subject matter of United States Patent Nos. 4,683,195; 4,800,159; 4,754,065; and 4,683,202 and described in PCR: THE POLYMERASE CHAIN REACTION (Mullis, et al.
  • this invention also provides a process for obtaining the polynucleotides of this invention by providing the linear sequence of the polynucleotide, appropriate primer molecules, chemicals such as enzymes and instructions for their replication and chemically replicating or linking the nucleotides in the proper orientation to obtain the polynucleotides. In a separate embodiment, these polynucleotides are further isolated.
  • RNA can be obtained by first inserting a DNA polynucleotide into a suitable host cell.
  • the DNA can be inserted by any appropriate method, e.g., by the use of an appropriate gene delivery vehicle (e.g., liposome, plasmid or vector) or by electroporation.
  • RNA When the cell replicates and the DNA is transcribed into RNA; the RNA can then be isolated using methods known to those of skill in the art, for example, as set forth in Sambrook et al. (1989) supra. For instance, mRNA can be isolated using various lytic enzymes or chemical solutions according to the procedures set forth in Sambrook, et al. (1989) supra or extracted by nucleic-acid-binding resins following the accompanying instructions provided by manufactures.
  • Polynucleotides having at least 4 contiguous nucleotides, and more preferably at least 5 or 6 contiguous nucleotides and most preferably at least 10 contiguous nucleotides, and exhibiting sequence complementarity or homology to the polynucleotides encoding the peptides shown in Table 1 find utility as hybridization probes. It is known in the art that a "perfectly matched" probe is not needed for a specific hybridization. Minor changes in probe sequence achieved by substitution, deletion or insertion of a small number of bases do not affect the hybridization specificity. In general, as much as 20% base-pair mismatch (when optimally aligned) can be tolerated.
  • a probe useful for detecting the aforementioned mRNA is at least about 80% identical to the homologous region of comparable size contained in the previously identified sequences which correspond to previously characterized genes. More preferably, the probe is 85% identical to the corresponding gene sequence after alignment of the homologous region; even more preferably, it exhibits 90% identity.
  • These probes can be used in radioassays (e.g. Southern and Northern blot analysis) to detect or monitor various cells or tissue containing these cells.
  • the probes also can be attached to a solid support or an array such as a chip for use in high throughput screening assays for the detection of expression of the gene corresponding to one or more polynucleotide(s) of this invention.
  • this invention also provides at least one probe as defined above and/or the complement of one of these sequences, attached to a solid support for use in high throughput screens.
  • the polynucleotides of the present invention also can serve as primers for the detection of genes or gene transcripts that are expressed in APC, for example, to confirm transduction of the polynucleotides into host cells.
  • amplification means any method employing a primer-dependent polymerase capable of replicating a target sequence with reasonable fidelity. Amplification may be carried out by natural or recombinant DNA-polymerases such as T7 DNA polymerase, Klenow fragment of E. coli DNA polymerase, and reverse transcriptase.
  • a preferred length of the primer is the same as that identified for probes, above.
  • the invention further provides the isolated polynucleotide operatively linked to a promoter of RNA transcription, as well as other regulatory sequences for replication and/or transient or stable expression of the DNA or RNA.
  • a promoter of RNA transcription as well as other regulatory sequences for replication and/or transient or stable expression of the DNA or RNA.
  • the term "operatively linked” means positioned in such a manner that the promoter will direct transcription of RNA off the DNA molecule. Examples of such promoters are SP6, T4 and T7.
  • cell-specific promoters are used for cell-specific expression of the inserted polynucleotide.
  • Vectors which contain a promoter or a promoter/enhancer, with termination codons and selectable marker sequences, as well as a cloning site into which an inserted piece of DNA can be operatively linked to that promoter are known in the art and commercially available.
  • GENE EXPRESSION TECHNOLOGY Goeddel ed., Academic Press, Inc. (1991)
  • VECTORS ESSENTIAL DATA SERIES (Gacesa and Ramji, eds., John Wiley & Sons, N.Y. (1994)), which contains maps, functional properties, commercial suppliers and a reference to GenEMBL accession numbers for various suitable vectors.
  • these vectors are capable of transcribing RNA in vitro or in vivo.
  • Expression vectors containing these nucleic acids are useful to obtain host vector systems to produce proteins and polypeptides. It is implied that these expression vectors must be replicable in the host organisms either as episomes or as an integral part of the chromosomal DNA.
  • Suitable expression vectors include plasmids, viral vectors, including adenovirases, adeno-associated viruses, retroviruses, cosmids, etc.
  • Adenoviral vectors are useful for introducing genes into tissues in vivo because of their high levels of expression and efficient transformation of cells both in vitro and in vivo.
  • a suitable host cell e.g., a prokaryotic or a eukaryotic cell and the host cell replicates
  • the protein can be recombinantly produced.
  • suitable host cells will depend on the vector and can include mammalian cells, animal cells, human cells, simian cells, insect cells, yeast cells, and bacterial cells constructed using known methods. See Sambrook, et al. (1989) supra.
  • the nucleic acid can be inserted into the host cell by methods known in the art such as transformation for bacterial cells; transfection using calcium phosphate precipitation for mammalian cells; DEAE-dextran; electroporation; or microinjection. See Sambrook, et al. (1989) supra for this methodology.
  • this invention also provides a host cell, e.g. a mammalian cell, an animal cell (rat or mouse), a human cell, or a prokaryotic cell such as a bacterial cell, containing a polynucleotide encoding a protein or polypeptide or antibody.
  • the present invention also provides delivery vehicles suitable for delivery of a polynucleotide of the invention into cells (whether in vivo, ex vivo, or in vitro).
  • a polynucleotide of the invention can be contained within a cloning or expression vector.
  • These vectors (especially expression vectors) can in turn be manipulated to assume any of a number of forms which may, for example, facilitate delivery to and/or entry into a cell.
  • a pharmaceutically acceptable vector is preferred, such as a replication-incompetent retroviral or adenoviral vector.
  • compositions containing the nucleic acids of this invention can be further modified for transient or stable expression of the inserted polynucleotide.
  • pharmaceutically acceptable vector includes, but is not limited to, a vector or delivery vehicle having the ability to selectively target and introduce the nucleic acid into dividing cells.
  • An example of such a vector is a "replication-incompetent" vector defined by its inability to produce viral proteins, precluding spread of the vector in the infected host cell.
  • An example of a replication-incompetent retroviral vector is LNL6 (Miller, A.D. et al. (1989) BioTechniques 7:980-990).
  • the cells may be used to induce an immune response in a subject in the methods described herein.
  • the host cells are antigen presenting cells, they can be used to expand a population of immune effector cells such as tumor infiltrating lymphocytes which in turn are useful in adoptive immunotherapies.
  • ANTIBODIES Also provided by this invention is an antibody capable of specifically forming a complex with the polypeptides of this invention.
  • the term "antibody” includes polyclonai antibodies and monoclonal antibodies. The antibodies include, but are not limited to mouse, rat, and rabbit or human antibodies. The antibodies are useful to identify and purify polypeptides and APCs expressing the polypeptides.
  • the monoclonal antibodies of this invention can be biologically produced by introducing protein or a fragment thereof into an animal, e.g., a mouse or a rabbit.
  • the antibody producing cells in the animal are isolated and fused with myeloma cells or hetero-myeloma cells to produce hybrid cells or hybridomas. Accordingly, the hybridoma cells producing the monoclonal antibodies of this invention also are provided.
  • one of skill in the art can produce and screen the hybridoma cells and antibodies of this invention for antibodies having the ability to bind the proteins or polypeptides. If a monoclonal antibody being tested binds with the protein or polypeptide, then the antibody being tested and the antibodies provided by the hybridomas of this invention are equivalent. It also is possible to determine without undue experimentation, whether an antibody has the same specificity as the monoclonal antibody of this invention by determining whether the antibody being tested prevents a monoclonal antibody of this invention from binding the protein or polypeptide with which the monoclonal antibody is normally reactive.
  • the antibody being tested competes with the monoclonal antibody of the invention as shown by a decrease in binding by the monoclonal antibody of this invention, then it is likely that the two antibodies bind to the same or a closely related epitope.
  • the term "antibody” also is intended to include antibodies of all isotypes.
  • Particular isotypes of a monoclonal antibody can be prepared either directly by selecting from the initial fusion, or prepared secondarily, from a parental hybridoma secreting a monoclonal antibody of different isotype by using the sib selection technique to isolate class switch variants using the procedure described in Steplewski, et al. (1985) Proc. Natl. Acad. Sci. USA 82:8653 or Spira, et al. (1984J J. Immunol. Meth. 74:307.
  • This invention also provides biological active fragments of the polyclonai and monoclonal antibodies described above. These "antibody fragments" retain some ability to selectively bind with its antigen or immunogen.
  • Such antibody fragments can include, but are not limited to: (1) Fab, (2) Fab', (3) F(ab') 2 , (4) Fv, and (5) SCA
  • the antibodies of this invention are monoclonal antibodies, although in certain aspects, polyclonai antibodies can be utilized. They also can be immunogenic and functional fragments, antibody derivatives or antibody variants. They can be chimeric, humanized, or totally human.
  • a functional fragment of an antibody includes but is not limited to Fab, Fab', Fab2, Fab'2, and single chain variable regions.
  • Antibodies can be produced in cell culture, in phage, or in various animals, including but not limited to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats, monkeys, chimpanzees, apes, etc. So long as the fragment or derivative retains specificity of binding as the antibodies of this invention it can be used.
  • Antibodies can be tested for specificity of binding by comparing binding to appropriate antigen to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody binds to the appropriate antigen at least 2, 5, 7, and preferably 10 times more than to irrelevant antigen or antigen mixture then it is considered to be specific. Specific assays for determining specificity are described infra.
  • a hybridoma is produced by fusing a suitable immortal cell line (e.g., a myeloma cell line such as, but not limited to, Sp2/0, Sp2/0-AG14, NSO, NS1, NS2, AE-1, L.5, >243, P3X63Ag8.653, Sp2 SA3, Sp2 MAI, Sp2 SSI, Sp2 SA5, U397, MLA 144, ACT IV, MOLT4, DA-1, JURKAT, WEHI, K-562, COS, RAJI, NIH 3T3, HL-60, MLA 144, NAMAIWA, NEURO 2A, CHO, PerC.6, YB2/O) or the like, or heteromyelomas, fusion products thereof, or any cell or fusion cell derived therefrom, or any other suitable cell line as known in the art (see,
  • Antibody producing cells can also be obtained from the peripheral blood or, preferably the spleen or lymph nodes, of humans or other suitable animals that have been immunized with the antigen of interest. Any other suitable host cell can also be used for expressing-heterologous or endogenous nucleic acid encoding an antibody, specified fragment or variant thereof, of the present invention.
  • the fused cells (hybridomas) or recombinant cells can be isolated using selective culture conditions or other suitable known methods, and cloned by limiting dilution or cell sorting, or other known methods.
  • Suitable methods of producing or isolating antibodies of the requisite specificity can be used, including, but not limited to, methods that select recombinant antibody from a peptide or protein library (e.g. , but not limited to, a bacteriophage, ribosome, oligonucleotide, RNA, cDNA, or the like, display library; e.g., as available from various commercial vendors such as Cambridge Antibody Technologies (Cambridgeshire, UK), Mo ⁇ hoSys (Martinsreid/Planegg, Del), Biovation (Aberdeen, Scotland, UK) Biolnvent (Lund, Sweden), using methods known in the art. (See U.S. Pat. Nos.
  • Antibody variants of the present invention can also be prepared using delivering a polynucleotide encoding an antibody of this invention to a suitable host such as to provide transgenic animals or mammals, such as goats, cows, horses, sheep, and the like, that produce such antibodies in their milk. These methods are known in the art and are described for example in U.S. Pat.
  • antibody variant includes post-translational modification to linear polypeptide sequence of the antibody or fragment.
  • U.S. Patent No. 6,602,684 Bl describes a method for the generation of modified glycol-forms of antibodies, including whole antibody molecules, antibody fragments, or fusion proteins that include a region equivalent to the Fc region of an immunoglobulin, having enhanced Fc-mediated cellular toxicity, and glycoproteins so generated.
  • Antibody variants also can be prepared by delivering a polynucleotide of this invention to provide transgenic plants and cultured plant cells (e.g., but not limited to tobacco, maize, and duckweed) that produce such antibodies, specified portions or variants in the plant parts or in cells cultured therefrom.
  • transgenic plants and cultured plant cells e.g., but not limited to tobacco, maize, and duckweed
  • transgenic plants and cultured plant cells e.g., but not limited to tobacco, maize, and duckweed
  • transgenic plants and cultured plant cells e.g., but not limited to tobacco, maize, and duckweed
  • transgenic plants and cultured plant cells e.g., but not limited to tobacco, maize, and duckweed
  • Antibody variants have also been produced in large amounts from transgenic plant seeds including antibody fragments, such as single chain antibodies (scFv's), including tobacco seeds and potato tubers.
  • scFv's single chain antibodies
  • tobacco seeds and potato tubers See, e.g., Conrad et al.(1998) Plant Mol. Biol. 38:101- 109 and reference cited therein.
  • antibodies of the present invention can also be produced using transgenic plants, according to know methods.
  • Antibody derivatives can be produced, for example, by adding exogenous sequences to modify immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic. Generally part or all of the non-human or human CDR sequences are maintained while the non-human sequences of the variable and constant regions are replaced with human or other amino acids. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Humanization or engineering of antibodies of the present invention can be performed using any known method, such as but not limited to those described in U.S. Pat. Nos.
  • Fully human antibody sequences are made in a transgenic mouse which has been engineered to express human heavy and light chain antibody genes. Multiple strains of such transgenic mice have been made which can produce different classes of antibodies. B cells from transgenic mice which are producing a desirable antibody can be fused to make hybridoma cell lines for continuous production of the desired antibody.
  • Human monoclonal antibodies can also be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a numan heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • the antibodies of this invention also can be modified to create chimeric antibodies. Chimeric antibodies are those in which the various domains of the antibodies' heavy and light chains are coded for by DNA from more than one species. (See, e.g., U.S.
  • antibody derivative also includes “diabodies” which are small antibody fragments with two antigen-binding sites, wherein fragments comprise a heavy chain variable domain (V H ) connected to a light chain variable domain (V L ) in the same polypeptide chain (VH V L ).
  • V H heavy chain variable domain
  • V L light chain variable domain
  • linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Patent No. 6,632,926 to Chen et al. which discloses antibody variants that have one or more amino acids inserted into a hypervariable region of the parent antibody and a binding affinity for a target antigen which is at least about two fold stronger than the binding affinity of the parent antibody for the antigen.
  • the term "antibody derivative” further includes “linear antibodies”. The procedure for making the is known in the art and described in Zapata et al. (1995) Protein Eng. 8(10):1057-1062. Briefly, these antibodies comprise a pair of tandem Fd segments (V H -C H 1-VH -C H 1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.
  • the antibodies of this invention can be recovered and purified from recombinant cell cultures by known methods including, but not limited to, protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography (“HPLC”) can also be used for purification.
  • Antibodies of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells, or alternatively from a prokaryotic cells as described above.
  • antibodies can be labeled with a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like.
  • a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like.
  • Such labeled antibodies can be used for diagnostic techniques, either in vivo, or in an isolated test sample.
  • Antibodies can also be conjugated, for example, to a pharmaceutical agent, such as chemotherapeutic drag or a toxin. They can be linked to a cytokine, to a ligand, to another antibody.
  • Suitable agents for coupling to antibodies to achieve an anti-tumor effect include cytokines, such as interleukin 2 (IL-2) and Tumor Necrosis Factor (TNF); photosensitizers, for use in photodynamic therapy, including aluminum (III) phthalocyanine tetrasulfonate, hematopo ⁇ hyrin, and phthalocyanine; radionuclides, such as iodine-131 ( 131 I), yttrium-90 ( 90 Y), bismuth-212 ( 212 Bi), bismuth-213 ( 213 Bi), technetium-99m ( 99m Tc), rhenium-186 ( 186 Re), and rhenium- 188 ( 188 Re); antibiotics, such as doxorabicin, adriamycin, daunorubicin, methotrexate, daunomycin, neocarzinostatin, and carboplatin; bacterial, plant, and other toxins, such as dip
  • preparations containing antibodies covalently linked to organic molecules they can be prepared using suitable methods, such as by reaction with one or more modifying agents.
  • modifying agents refers to a suitable organic group (e.g., hydrophilic polymer, a fatty acid, a fatty acid ester) that comprises an activating group. Specific examples of these are provided supra.
  • An "activating group” is a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group.
  • electrophilic groups such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl esters (NHS), and the like.
  • Activating groups that can react with thiols include, for example, maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB- thiol), and the like.
  • An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form phosphoramidate or phosphorimide linkages.
  • Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., BIOCONJUGATE TECHNIQUES, Academic Press: San Diego, Calif. (1996)).
  • An activating group can be bonded directly to the organic group (e.g., hydrophilic polymer, fatty acid, fatty acid ester), or through a linker moiety, for example a divalent -C ⁇ group wherein one or more carbon atoms can be replaced by a heteroatom such as oxygen, nitrogen or sulfur.
  • Suitable linker moieties include, for example, tetraethylene glycol.
  • Modifying agents that comprise a linker moiety can be produced, for example, by reacting a mono-Boc-alkyldiamine (e.g., mono-Boc-ethylenediamine, mono-Boc- diaminohexane) with a fatty acid in the presence of l-ethyl-3-(3- dimethylaminopro ⁇ yl)carbodiimide (EDC) to form an amide bond between the free amine and the fatty acid carboxylate.
  • a mono-Boc-alkyldiamine e.g., mono-Boc-ethylenediamine, mono-Boc- diaminohexane
  • EDC l-ethyl-3-(3- dimethylaminopro ⁇ yl)carbodiimide
  • the Boc protecting group can be removed from the product by treatment with trifluoroacetic acid (TFA) to expose a primary amine that can be coupled to another carboxylate as described, or can be reacted with maleic anhydride and the resulting product cyclized to produce an activated maleimido derivative of the fatty acid.
  • TFA trifluoroacetic acid
  • the modified antibodies of the invention can be produced by reacting a human antibody or antigen-binding fragment with a modifying agent.
  • the organic moieties can be bonded to the antibody in a non-site specific manner by employing an amine-reactive modifying agent, for example, an NHS ester of PEG.
  • Modified human antibodies or antigen-binding fragments can also be prepared by reducing disulfide bonds (e.g., intra-chain disulfide bonds) of an antibody or antigen-binding fragment. The reduced antibody or antigen-binding fragment can then be reacted with a thiol-reactive modifying agent to produce the modified antibody of the invention.
  • Modified human antibodies and antigen- binding fragments comprising an organic moiety that is bonded to specific sites of an antibody of the present invention can be prepared using suitable methods, such as reverse proteolysis. (See generally, Hermanson, G. T., BIOCONJUGATE TECHNIQUES, Academic Press: San Diego, Calif. (1996)).
  • compositions containing the antibodies, fragments thereof or cell lines which produce the antibodies are encompassed by this invention.
  • these compositions are to be used pharmaceutically, they are combined with a pharmaceutically acceptable carrier.
  • the present invention provides a method of inducing an immune response comprising delivering the compounds and compositions of the invention in the context of an MHC molecule.
  • the polypeptides of this invention can be pulsed into antigen presenting cells using the methods described herein.
  • Antigen-presenting cells include, but are not limited to dendritic cells (DCs), monocytes/macrophages, B lymphocytes or other cell type(s) expressing the necessary MHC/co-stimulatory molecules.
  • DCs which are the most potent, preferred APCs.
  • host cells containing the polypeptides or proteins are further provided.
  • the antibodies and compositions can be delivered by any suitable means and with any suitable formulation. Accordingly, a formulation comprising an antibody of this invention is further provided herein.
  • the formulation can further comprise one or more preservative or stabilizer such as phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (e.g., hexahydrate), alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent.
  • preservative or stabilizer such as phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (e.g., hexahydrate), alky
  • Any suitable concentration or mixture can be used as known in the art, such as 0.001-5%, or any range or value therein, such as, but not limited to 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05, 0.09, 0.1, 0.2, 0.3, O.4., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.3, 4.5, 4.6, 4.7, 4.8, 4.9, or any range or value therein.
  • Non-limiting examples include, no preservative, 0.1-2% m-cresol (e.g., 0.2, 0.3. 0.4, 0.5, 0.9, 1.0%), 0.1- 3% benzyl alcohol (e.g., 0.5, 0.9, 1.1., 1.5, 1.9, 2.0, 2.5%), 0.001-0.5% thimerosal (e.g., 0.005, 0.01), 0.001-2.0% phenol (e.g., 0.05, 0.25, 0.28, 0.5, 0.9, 1.0%), 0.0005-1.0% alkylparaben(s) (e.g., 0.00075, 0.0009, 0.001, 0.002, 0.005, 0.0075, 0.009, 0.01, 0.02, 0.05, 0.075, 0.09, 0.1, 0.2, 0.3, 0.5, 0.75, 0.9, and 1.0%).
  • 0.1-2% m-cresol e.g., 0.2, 0.3. 0.4, 0.5, 0.9, 1.0
  • This invention also provides an article of manufacture, comprising packaging material and at least one vial comprising a solution of at least a peptide, polynucleotide or antibody as of this invention with the prescribed buffers and/or preservatives, optionally in an aqueous diluent, wherein said packaging material comprises a label that indicates that such solution can be held over a period of 1, 2, 3, 4, 5, 6, 9, 12, 18, 20, 24, 30, 36,40, 48, 54, 60, 66, 72 hours or greater.
  • the invention further comprises an article of manufacture, comprising packaging material, a first vial comprising at least one lyophiiized composition of this invention and a second vial comprising an aqueous diluent of prescribed buffer or preservative, wherein said packaging material comprises a label that instructs a patient to reconstitute the antibody in the aqueous diluent to form a solution that can be held over a period of twenty- four hours or greater.
  • the range antibody includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 ⁇ g/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal .
  • the formulations of the present invention can be prepared by a process which comprises mixing at least one antibody of this invention and a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous diluent.
  • a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous
  • aqueous diluent is carried out using conventional dissolution and mixing procedures. For example, a measured amount of at least one antibody in buffered solution is combined with the desired preservative in a buffered solution in quantities sufficient to provide the antibody and preservative at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art, e.g., the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that can be optimized for the concentration and means of administration used.
  • compositions and formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophiiized antibody that is reconstituted with a second vial containing the aqueous diluent.
  • Either a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available.
  • Recognized devices comprising these single vial systems include those pen-injector devices for delivery of a solution such as BD Pens, BD Autojectore, Humaject.RTM.' NovoPen.RTM., B-D.RTM.Pen, AutoPen.RTM., and OptiPen.RTM., GenotropinPen.RTM., Genotronorm Pen.RTM., Humatro Pen.RTM., Reco- Pen.RTM., Roferon Pen.RTM., Biojector.RTM., iject.RTM., J-tip Needle-Free Injector.RTM., Intraject.RTM., Medi-Ject.RTM., e.g., as made or developed by Becton Dickensen (Franklin Lakes, NJ. available at bectondickenson.com),
  • Isolated host cells which present the polypeptides of this invention in the context of MHC molecules are further useful to expand and isolate a population of educated, antigen-specific immune effector cells.
  • the immune effector cells e.g., cytotoxic T lymphocytes
  • the immune effector cells are produced by culturing na ⁇ ve immune effector cells with antigen-presenting cells which present the polypeptides in the context of MHC molecules on the surface of the APCs.
  • the population can be purified using methods known in the art, e.g., FACS analysis or ficoll gradient.
  • the methods to generate and culture the immune effector cells as well as the populations produced thereby also are the inventor's contribution and invention.
  • Pharmaceutical compositions comprising the cells and pharmaceutically acceptable carriers are useful in adoptive immunotherapy.
  • the immune effector cells Prior to administration in vivo, the immune effector cells are screened in vitro for their ability to lyse cells.
  • the immune effector cells and/or the APCs are genetically modified.
  • genes coding for co-stimulatory molecules and/or stimulatory cytokines can be inserted prior to, concurrent to or subsequent to expansion of the immune effector cells.
  • This invention also provides methods of inducing an immune response in a subject, comprising administering to the subject an effective amount of the polypeptides or immunogenic portion thereof under the conditions that induce an immune response to the polypeptide and induce a cytolytic Tcell response.
  • the polypeptides can be administered in formulations or as polynucleotides encoding the polypeptides.
  • the polynucleotides can be administered in a gene delivery vehicle or by inserting into a host cell which in turn recombinantly transcribes, translates and processed the encoded polypeptide.
  • Isolated host cells containing the polynucleotides of this invention in a pharmaceutically acceptable carrier can therefore combined with appropriate and effective amount of an adjuvant, cytokine or co-stimulatory molecule for an effective vaccine regimen.
  • the host cell is an APC such as a dendritic cell.
  • the host cell can be further modified by inserting of a polynucleotide coding for an effective amount of either or both a cytokine and/or a co-stimulatory molecule.
  • the methods of this invention can be further modified by co-administering an effective amount of a cytokine or co-stimulatory molecule to the subject.
  • the polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g.., poly-His), or to enhance binding of the polypeptide to a solid support.
  • a polypeptide can be conjugated to an immunoglobulin Fc region.
  • an antibody of the invention may be coupled to a bridging compound coupled to a solid support.
  • the bridging compound which is designed to link the solid support and the antibody may be hydrazide, Protein A, glutaraldehyde, carbodiimide, or lysine.
  • the solid support employed is e.g., a polymer or it may be a matrix coated with a polymer.
  • the matrix may be of any suitable solid material, e.g. glass, paper or plastic.
  • the polymer may be a plastic, cellulose such as specially treated paper. nitrocellulose paper or cyanogenbromide-activated paper.
  • suitable plastics are latex, a polystyrene, polyvinylchloride, polyurethane, polyacrylamide, polyvinylacetate and any suitable copolymer thereof.
  • silicone polymers include siloxane.
  • the solid support may be in the form of a tray, a plate such as a mitrotiter plate, e.g. a thin layer or, preferably, strip, film, glass slide, threads, solid particles such as beads, including Protein A-coated bacteria, or paper.
  • the antibody of the invention may be used in an assay for the identification and/or quantification of at least a form and/or a part of said polypeptide present in a sample.
  • the identification and/or quantification performed by the use according to the present invention may be any identification and/or quantification of a protein comprising any of the polypeptides of the invention.
  • both a qualitative and a quantitative determination of any protein comprising any of the polypeptides of the invention may be obtained according to the use of the present invention.
  • the identification and/or quantification may be performed for both a scientific, a clinical and an industrial pu ⁇ ose. As will be further described below, it is especially important in clinical routine to identify or quantify the amount of any protein comprising any of the polypeptides of the invention.
  • the antibody used in the method of the invention is a monoclonal antibody as this generally provides a higher precision and accuracy of the assay, at the same time possibly requiring less time to perform. Furthermore, a mixture of two or more different monoclonal antibodies may be employed to increase the detection limit and sensitivity of the test.
  • the monoclonal antibody may be obtained by methods described herein or as known in the art. Antibodies possessing high avidity may be selected for catching techniques.
  • the antibody used in the present method is preferably in substantially pure form (purified according to suitable techniques or by the methods of the invention, see below) in order to improve the precision and/or accuracy of the assays of the invention.
  • the invention relates to a diagnostic agent comprising an antibody capable of detecting and/or quantitating any of the ligands of the invention.
  • the antibody is provided with a label for the detection of bound antibody or, alternatively (such as in a double antibody assay), a combination of labelled and unlabelled antibody may be employed.
  • the substance used as label may be selected from any substance which is in itself detectable or which may be reacted with another substance to produce a detectable product.
  • the label may be selected from radioactive isotopes, enzymes, chromophores, fluorescent or chemiluminescent substances, and complexing agents.
  • these neoplasia are diagnosed by evaluating a biological sample obtained from the animal or patient and determining the level of one or more of the above polypeptides, relative to a predetermined cut-off value.
  • suitable "biological samples” include ovarian tissue, blood, stool, sera, urine and/or secretions.
  • the antibody used in the present method may be in substantially pure form in order to improve the precision and/or accuracy of the assays of the invention.
  • Functional equivalents to the monoclonal antibody that is, compounds or other ligands that bind to proteins comprising the peptides of the invention and which may inhibit the binding of the antibody to these proteins are also provided by this invention.
  • Another method according to the invention is a method for detecting or quantifying complexes of antibody and the peptides of the invention in a sample, comprising using an antibody capable of binding to one of the peptides of the invention together with an antibody which detects antibody-peptides complexes.
  • Another method according to the invention is a method for immunohistochemical detection of any of the peptides of the invention in tissue sections, using, as the detecting antibody, any monoclonal antibody which reacts with one of the peptides of the invention, or any polyclonai antibody which reacts with at least one of the polypeptides of the invention.
  • the monoclonal antibodies according to the invention may also be used in a method for targeting a diagnostic agent to a cell that expresses a peptide of the invention on the surface, the method comprising administering, to a mammal, in particular a human, in particular a mammal suffering from cancer or suspected to suffer from cancer, the diagnostic agent bound to a monoclonal antibody of the invention.
  • the diagnostic agent may be a radioactive substance, such as technetium.
  • Several embodiments of this invention include but are not limited to: 1) use as a catching antibody when immobilized as a catching antibody in a sandwich ELISA; 2) when used as a biotin-labelled detecting antibody in a sandwich
  • ELISA is capable of detecting one of the peptides of the invention; 3) when used in an ELISA, is capable of binding to an immobilized peptides of the invention; 5) in a radioimmunoprecipitation assay precipitates one of the peptides of the invention; or 8) is capable of binding to one of the peptides of the invention in tissue sections, including paraffine-embedded tissue sections, and thereby being useful for immunohistochemical detection of one of the peptides of the invention.
  • tissue sections including paraffine-embedded tissue sections
  • invention methods comprise detecting the differential expression of one or more of the peptides of the invention in a sample isolated from a cell or tissue, wherein the presence and/or amount of the protein is indicative of the neoplastic condition of cell or tissue.
  • a related neoplasia is one in which the expression or expression of the protein serves as a marker for the neoplastic phenotype.
  • Samples of cells or tissue can be provided free form or attached to a solid support and can be isolated from a tissue culture, commercially available cell line, from a patient biopsy or as in the case of use of the method for tissue imaging, in vivo.
  • Method No. 1 Normal or patient donor PBMC are exposed to the invention peptides either individually or as a cocktail under the appropriate reaction conditions to generate a mixed population of reactive cytotoxic T cells. These cells are tested for their ability to react with tumor cells lines known to express the antigen. This ensures that the peptide-educated CTL recognize the epitope in its native state.
  • These tumor-reactive T cells can be used as a bulk population or individual tumor-reactive clones can be derived by standard limiting dilution analysis, in order to determine whether a particular patient's tumor expresses the unknown antigen.
  • Some of the techniques that can be employed include: 1. One can use tumor cells derived from patient tumor biopsy in order to stimulate the invention peptide-educated CTL and measure the secretion of cytokines associated with CTL antigen recognition such as IL-2 or INF- ⁇ can be measured. Standard techniques such as ELISPOT, ELISA, or intracellular cytokine staining can be employed. Cytokine release indicates that the patient's tumor cells express the unnamed antigen, implying potential benefit to that patient by successful vaccination with the peptides. 2.
  • Single cell suspensions of patient tumor cells derived from biopsy or resected tumor can be reacted with the peptide-educated CTL of the present invention in a standard 51Cr-release assay in order to measure patient tumor lysis directly. Lysis indicates that the patient's tumor cells express the unnamed antigen, implying potential benefit to that patient by successful vaccination with the peptides.
  • Method No. 2 Peptide-educated clonal populations of CTL of the present invention can be used to construct recombinant soluble T cell receptor (TCR) protein with specificity for the native epitope corresponding to the unnamed antigen. Multimerization of the TCR proteins results in a reagent that will bind to the HLA-A2 molecule in the presence of the specific epitope recognized by the TCR. Therefore, this reagent is useful to test patient tumor cells derived from biopsy or tumor resection for the presence of the unnamed native epitope. A positive result indicates that the patient's tumor expresses the unnamed antigen implying potential benefit to the patient by successful vaccination with the peptides.
  • TCR soluble T cell receptor
  • Phage display technology has been successfully employed to identify recombinant antibodies capable of binding specifically to distinct MHC/peptide complexes (See, Denkberg, G. et al., (2002) PNAS 99(14):9421-9426). These monoclonal antibodies are useful to detect the presence of particular epitopes presented in the context of MHC molecules on the surface of tumor cells. This approach is useful to generate monoclonal antibodies against invention peptide-bound HLA-A2 molecules. These antibodies can be tested for specificity with tumor cell lines known to express or not express the antigen. Patient tumor derived from biopsy or tumor resection are tested for the presence of the native epitope/MHC surface complex by measuring antibody binding.
  • the method is practiced by detecting and/or quantifying mRNA encoding one of the peptides of the invention by hybridization or PCR. Modification of current technology enables this method, e.g., detecting is by probing said sample with a probe or primer that specifically hybridizes under conditions of moderate or highly stringent conditions with said mRNA. In one aspect, the probe or primer is detectably labeled.
  • RT-PCR techniques allows for quantitation of mRNA expression.
  • PCR is also useful to detect mutations in the nucleic acid sequences encoding any of the peptides of the invention.
  • suitable probes include but are not limited to a sequence selected from the group consisting of SEQ ID NOs: 1 to 43 as shown in Table 1, and complements thereof; a nucleic acid sequence encoding a peptide selected from the group consisting of SEQ ID NOs: 2 to 44 as shown in Table 1, and complements thereof; and a probe or primer complementary to a sequence encoding a protein comprising at least 9 consecutive residues of an amino acid sequence recited in the group consisting of SEQ ID NOs: 2 to 44, shown in Table 1, and complements thereof.
  • the peptides of the invention are detected and/or quantified by probing the sample peptides with an agent that specifically recognizes and binds one or more of the amino acid sequences shown in Table 1.
  • Antibodies, and antigen binding fragments thereof, that specifically recognize or bind to the peptides of the invention are examples of agents.
  • an antibody is a polyclonai or monoclonal antibody, which may or may not be detectably labeled.
  • modifications to antibodies and antigen binding fragments are known in the art and included for the pu ⁇ ose of this invention. Examples are described herein.
  • Suitable monoclonal antibodies for use in this aspect are prepared from an animal immunized with a peptide selected from the group consisting of those shown in Table 1.
  • an antigen binding fragment include a biologically active immunoglobulin variable domain isolated from an antibody prepared from an animal immunized with a peptide selected from the group consisting of those shown in Table 1.
  • the method utilizing antibodies can be practiced in vitro or in vivo, using application of well known methods as described herein.
  • the agent is a cell that binds to a protein comprising a peptides of the invention, such as an immune effector cell raised in the presence and at the expense of a peptide selected from the group consisting of those shown in Table 1.
  • differential expression of any of the peptides of the invention can be detected and/or quantified by determining the identity and expression level of mRNA by expression analysis and comparing the sequences and amount of mRNA to sequences and amount of expression in a normal control cell or tissue, using methods such as expression analysis, e.g., SAGE (United States Pat. No. 5,695,937) and expression arrays.
  • expression analysis e.g., SAGE (United States Pat. No. 5,695,937) and expression arrays.
  • detection and analysis of the presence in a sample of the amount of any of the peptides of the invention is repeated to monitor disease progression, response to a therapeutic regimen, or disease recurrence in a subject:
  • the detecting/monitoring method as described herein, is performed on one or more sample(s) isolated at one or more times time points subsequent to a prior (control) sample and comparing the amount of protein detected at the subsequent points in time to the amount previously detected in a prior (control) sample.
  • the present invention provides a screen to identify agents which bind to any of the peptides of the invention, or which bind to proteins comprising any of the peptides of the invention.
  • test compounds may be labeled with a detectable agent, e.g., tritium, and incubated with any of the peptides of the invention. At the end of the incubation period, the incubation solution is treated with a washing solution to remove unbound test compounds. After washing, the amount of labeled test compound remaining is measured and compared to a control sample to determine whether a test compound or compound is capable of specific binding.
  • a detectable agent e.g., tritium
  • the invention provides a screen for agents that inhibit the binding of an antibody or a protein or peptide comprising any of the peptides of the invention to a ligand by contacting a sample comprising the protein or peptide and its ligand under conditions and in the presence of a test agent and detecting any binding between the protein or peptide and its ligand, the absence of binding being indicative of an agent that modulates the binding of the protein or peptide to its binding partner.
  • the invention is useful to screen for agents that enhance the binding of a protein or peptide comprising any of the peptides of the invention to its ligand by contacting a sample comprising the protein or peptide and the ligand under conditions and in the presence of a test agent and detecting and comparing any binding between the protein or peptide and its ligand with a control sample that does not contain the test agent. Enhancement of binding over the control sample is a positive indication that the test agent enhances the binding of the protein or peptide to its ligand. Modulation (increase or inhibit) of binding includes a variation in avidity as well as affinity.
  • one such assay comprises contacting a mammalian cell with an invention composition and the "test compound” and [ 3 H ]thymidine, under conditions where the mammalian cell would normally proliferate.
  • a control assay may be performed without the "test compound” and compared to the amount of cell proliferation in the presence of the "test compound” to determine if the compound stimulates or inhibits proliferation.
  • [ 3 H ] thymidine uptake can be measured by liquid scintillation chromatography which measures the inco ⁇ oration of the label. Agonists and antagonist compounds may be identified using this assay.
  • a mammalian cell or membrane preparation expressing a binding site or determinant for a polypeptide or peptide of the invention is incubated with a labeled polypeptide or peptide of the invention in the presence of the "test compound".
  • the ability of the "test compound” to enhance or block the interaction of the polypeptide or peptide with its binding site can be measured.
  • the response of a known second messenger system following the interaction of the "test compound” and the binding site is measured and the ability of the "test compound” to bind the site or determinant and elicit a second messenger response can be measured to determine if the "test compound” is a potential agonist or antagonist.
  • These assays can be used as diagnostic or prognostic markers.
  • the invention includes a method of identifying compounds which bind to a polypeptide, peptide, antibody, host cell or immune effector cell of the invention comprising: (a) incubating a candidate test compound with a composition described herein of the invention; and (b) determining if binding has occurred.
  • the invention further comprises a method of identifying agonists and/or antagonists comprising: (a) incubating a candidate test compound with a composition described herein of the invention; (b) assaying a biological activity; and (c) determining if said biological activity has been altered.
  • the kit contains at least one agent that specifically recognizes and binds to a protein or peptide comprising any of the peptides of the invention and a detection reagent which can support a reporter group.
  • a detection reagent which can support a reporter group.
  • antibodies When antibodies are used as the agent, they can be in solution or immobilized on a solid support, such as nitrocellulose, glass, latex or a plastic.
  • detection reagents include but are not limited to anti-immunoglobulin, protein G, protein A, or lectin. Reporter groups can be provided in the kit, but are not necessarily supplied.
  • kits will contain one or more of binding agents described above, e.g., a probe, a primer, an antibody or immune effector cell specific for any of the peptides of the invention, or an agent that specifically recognizes and binds to the specific antibody, e.g., antibodies raised and isolated to specifically recognize and bind a peptide of the invention. Instructions for use can also be provided.
  • binding agents such as antibodies or fragments thereof, that are capable of detecting neoplastic cells or tissues, or alternatively, for monitoring disease progression in an animal or patient having a neoplastic condition related to the expression of a peptide or protein comprising any of the peptides of the invention, e.g, ovarian cancer.
  • Binding agents of the present invention may generally be prepared using methods known to those of ordinary skill in the art, including the representative procedures described herein. Binding agents are capable of differentiating between patients with and without a neoplastic condition associated with the expression and or overexpression of a peptide or protein comprising any of the peptides of the invention, using the representative assays described herein.
  • antibodies or other binding agents raised against any of the peptides of the invention will generate a signal indicating the presence of primary or metastatic cancer in patients afflicted with the disease, and will generate a negative signal indicating the absence of the disease in individuals without primary or metastatic disease.
  • the representative assays described below, such as the two-antibody sandwich assay, can be employed for evaluating the ability of a binding agent to detect ovarian neoplasia.
  • the ability of a polypeptide prepared as described herein to generate antibodies capable of detecting neoplasia can be evaluated by raising one or more antibodies against the polypeptide and determining the ability of such antibodies to detect neoplasia in animals or patients. In one aspect, this determination is made by assaying biological samples from animals or patients with and without primary or metastatic neoplasia for the presence of a polypeptide that binds to the generated antibodies. Such test assays may be performed, for example, using a representative procedure described below. Polypeptide specific antibodies raised the polypeptides identified in Table 1, or a fragment thereof, can be used alone or in combination to improve sensitivity.
  • binding agent in the context of this invention, is any agent (such as a compound or a cell) that binds to a polypeptide as described above.
  • binding refers to a noncovalent association between two separate molecules (each of which may be free (i.e., in solution) or present on the surface of a cell or a solid support), such that a "complex" is formed.
  • the complex can be free or immobilized (either covalently or noncovalently) on a support material.
  • binding constant is the value obtained when the concentration of the complex is divided by the product of the component concentrations. Binding constants are determined using methods known to those of ordinary skill in the art. Any agent that satisfies the above requirements may be a binding agent.
  • a binding agent may be a ribosome with or without a peptide component, an RNA molecule, a host cell expressing epitope, an immune effector cell as described herein or a peptide.
  • the binding partner is an antibody, or a fragment thereof.
  • the antibodies can be polyclonai or monoclonal which include, but are not limited to single chain, chimeric, CDR-grafted or humanized. Methods for generating these antibodies are known to those of skill in the art.
  • This invention also provides compositions containing any of the above-mentioned proteins, polypeptides, polynucleotides, vectors, cells, antibodies and fragments thereof, and an acceptable solid or liquid carrier. When the compositions are used pharmaceutically, they are combined with a
  • compositions for diagnostic and therapeutic use. These compositions also can be used for the preparation of medicaments for the diagnosis and treatment of diseases such as cancer.
  • the following materials and methods are intended to illustrate, but not limit this invention and to illustrate how to make and use the inventions described above.
  • isolated polypeptides of the present invention can be synthesized using an appropriate solid state synthetic procedure.
  • SOLID PHASE PEPTIDE SYNTHESIS Freemantle, San Francisco, Calif. (1968)
  • a preferred method is the Merrifield process. (See, Merrifield (1967) Recent Progress in Hormone Res. 23:451).
  • the antigenic activity of these peptides may conveniently be tested using, for example, the assays as described herein.
  • an isolated polypeptide of the invention may be purified by standard methods including chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification.
  • chromatography e.g., ion exchange, affinity, and sizing column chromatography
  • affinity tags such as hexa-His (Invitrogen), Maltose binding domain (New England Biolabs), influenza coat sequence (Kolodziej, et al. (1991) Meth. Enzymol.
  • Isolated immunogens can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance, and x-ray crystallography. Also included within the scope of the invention are immunogenic peptides that are differentially modified during or after translation, e.g., by phosphorylation, glycosylation, cross-linking, acylation, proteolytic cleavage, linkage to an antibody molecule, membrane molecule or other ligand. (Ferguson, et al. (1988) Ann. Rev. Biochem. 57:285-320).
  • the APC are precommitted or mature dendritic cells which can be isolated from the white blood cell fraction of a mammal, such as a murine, simian or a human (See, e.g., WO 96/23060).
  • the white blood cell fraction can be from the peripheral blood of the mammal.
  • This method includes the following steps: (a) providing a white blood cell fraction obtained from a mammalian source by methods known in the art such as leukophoresis; (b) separating the white blood cell fraction of step (a) into four or more subtractions by countercurrent centrifugal elutriation; (c) stimulating conversion of monocytes in one or more fractions from step (b) to dendritic cells by contacting the cells with calcium ionophore, GM-CSF and IL-13 or GM-CSF and IL-4, (d) identifying the dendritic cell-enriched fraction from step (c); and (e) collecting the enriched fraction of step (d), preferably at about 4°C.
  • the white blood cell fraction can be treated with calcium ionophore in the presence of other cytokines, such as recombinant (rh) rhIL-12, rhGM-CSF, or rhIL-4.
  • the cells of the white blood cell fraction can be washed in buffer and suspended in CaATVlg "1 ⁇ free media prior to the separating step.
  • the white blood cell fraction can be obtained by leukapheresis.
  • the dendritic cells can be identified by the presence of at least one of the following markers: HLA-DR, HLA-DQ, or B7.
  • CD3, CD14, CD16, 56, 57, and CD 19, 20 Monoclonal antibodies specific to these cell surface markers are commercially available. More specifically, the method requires collecting an enriched collection of white cells and platelets from leukapheresis that is then further fractionated by countercurrent centrifugal elutriation (CCE) (Abrahamsen, T.G. et al. (1991) J. Clin. Apheresis. 6:48-53). Cell samples are placed in a special elutriation rotor. The rotor is then spun at a constant speed of, for example, 3000 ⁇ m.
  • CCE countercurrent centrifugal elutriation
  • pressurized air is used to control the flow rate of cells.
  • Cells in the elutriator are subjected to simultaneous centrifugation and a washout stream of buffer that is constantly increasing in flow rate. This results in fractional cell separations based largely but not exclusively on differences in cell size.
  • Quality control of APC and more specifically DC collection and confirmation of their successful activation in culture is dependent upon a simultaneous multi-color FACS analysis technique which monitors both monocytes and the dendritic cell subpopulation as well as possible contaminant T lymphocytes.
  • DCs do not express the following markers: CD3 (T cell); CD14 (monocyte); CD16, 56, 57 (NK/LAK cells); CD19, 20 (B cells).
  • CD3 T cell
  • CD14 monocyte
  • CD16 56, 57
  • CD19 20
  • DCs do express large quantities of HLA-DR, significant HLA-DQ and B7.2 (but little or no B7.1) at the time they are circulating in the blood (in addition they express Leu M7 and M9, myeloid markers which are also expressed by monocytes and neutrophils).
  • PI propridium iodide
  • Additional markers can be substituted for additional analysis: Color #1 : CD3 alone, CD 14 alone, etc.; Leu M7 or Leu M9; anti-Class I, etc. Color #2: HLA-DQ, B7.1, B7.2, CD25 (IL2r), ICAM, LFA-3, etc.
  • FACS analysis at the time of collection is to confirm that the DCs are enriched in the expected fractions, to monitor neutrophil contamination, and to make sure that appropriate markers are expressed.
  • This rapid bulk collection of enriched DCs from human peripheral blood, suitable for clinical applications is absolutely dependent on the analytic FACS technique described above for quality control. If need be, mature DCs can be immediately separated from monocytes at this point by fluorescent sorting for "cocktail negative" cells.
  • the monocytes themselves are still capable of differentiating into DCs or functional DC-like cells in culture.
  • the DC rich/monocyte APC fractions (usually 150 through 190) can be pooled and cryopreserved for future use, or immediately placed in short term culture.
  • others have reported a method for upregulating (activating) dendritic cells and converting monocytes to an activated dendritic cell phenotype. This method involves the addition of calcium ionophore to the culture media convert monocytes into activated dendritic cells.
  • cytokines include but are not limited to purified or recombinant ("rh") rhGM-CSF, rhIL-2, and rhIL-4. Each cytokine when given alone is inadequate for optimal upregulation.
  • the peptides can be delivered to antigen-presenting cells as protein/peptide or in the form of cDNA encoding the protein/peptide.
  • Antigen-presenting cells can consist of dendritic cells (DCs), monocytes/macrophages, B lymphocytes or other cell type(s) expressing the necessary MHC/co- stimulatory molecules.
  • DCs dendritic cells
  • monocytes/macrophages monocytes/macrophages
  • B lymphocytes or other cell type(s) expressing the necessary MHC/co- stimulatory molecules.
  • the methods described below focus primarily on DCs which are the most potent, preferred APCs. Pulsing is accomplished in vitro/ex vivo by exposing APCs to the antigenic protein or peptide(s) of this invention.
  • the protein or peptide(s) are added to , APCs at a concentration of 1-10 ⁇ m for approximately 3 hours.
  • Pulsed APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery.
  • Protein/peptide antigen can also be delivered in vivo with adjuvant via the intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery.
  • Foster antigen Presenting Cells are particularly useful as target cells. Foster
  • APCs are derived from the human cell line 174xCEM.T2, referred to as T2, which contains a mutation in its antigen processing pathway that restricts the association of endogenous peptides with cell surface MHC class I molecules.
  • T2 contains a mutation in its antigen processing pathway that restricts the association of endogenous peptides with cell surface MHC class I molecules.
  • T2 cells Exogenous peptide added to the culture medium binds to these MHC molecules provided that the peptide contains the allele-specific binding motif.
  • These T2 cells are referred to herein as "foster" APCs. They can be used in conjunction with this invention to present antigen(s). Transduction of T2 cells with specific recombinant MHC alleles allows for redirection of the MHC restriction profile. Libraries tailored to the recombinant allele will be preferentially presented by them because the anchor residues will prevent efficient binding to the endogenous allele. High level expression of MHC molecules makes the APC more visible to the CTLs.
  • MHC allele of interest in T2 cells using a powerful transcriptional promoter results in a more reactive APC (most likely due to a higher concentration of reactive MHC-peptide complexes on the cell surface).
  • various immunoassays can be employed, including but not limited to, competitive and non-competitive assay systems using technologies such as radioimmunoassays, ELISA, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays, Western Blot, precipitation reactions, agglutination assays, immunofluorescence assays, protein A assays, electrophoretic assays, and the like. Binding may also be detected using antibody capture assays, antigen capture assays, and multiple-antibody sandwich assays. Methods for detecting binding in such assays are known in the art.
  • the present invention makes use of these APCs to stimulate production of an enriched population of antigen- specific immune effector cells.
  • the antigen-specific immune effector cells are expanded at the expense of the APCs, which die in the culture.
  • the process by which naive immune effector cells become educated by other cells is described essentially in Coulie (1997) Molec. Med. Today 3:261-268.
  • the APCs prepared as described above are mixed with naive immune effector cells.
  • the cells may be cultured in the presence of a cytokine, for example IL2.
  • the culture conditions are such that the antigen-specific immune effector cells expand (i.e., proliferate) at a much higher rate than the APCs. Multiple infusions of APCs and optional cytokines can be performed to further expand the population of antigen-specific cells.
  • the immune effector cells are T cells.
  • the immune effector cells can be genetically modified by transduction with a transgene coding for example, IL-2, IL-11 or IL-13. Methods for introducing transgenes in vitro, ex vivo and in vivo are well known in the art. (See Sambrook, et al. (1989) supra).
  • Vectors Useful in Genetic Modifications are accomplished by introducing a vector containing a polypeptide or transgene encoding a heterologous or an altered immunogen.
  • a variety of different gene transfer vectors including viral as well as non-viral systems can be used.
  • Viral vectors useful in the genetic modifications of this invention include, but are not limited to adenovirus, adeno-associated virus vectors, retroviral vectors and adeno-retroviral chimeric vectors.
  • APC and immune effector cells can be modified using the methods described below or by any other appropriate method known in the art.
  • Adenovirus and adeno-associated virus vectors useful in the genetic modifications of this invention may be produced according to methods already taught in the art. (See, e.g., Karisson, et al. (1986) EMBO J. 5:2377; Carter (1992) /
  • a transgene of interest can be cloned and expressed in cells infected with the replication deficient adenovirus.
  • adenoviras-based gene transfer does not result in integration of the transgene into the host genome (less than 0.1% adenovirus-mediated transfections result in transgene inco ⁇ oration into host DNA), and therefore is not stable, adenoviral vectors can be propagated in high titer and transfect non-replicating cells.
  • Human 293 cells which are human embryonic kidney cells transformed with adenovirus E1A/E1B genes, typify useful permissive cell lines.
  • adenovirus vectors and other viral vectors which could be used in the methods of the present invention include the following: Horwitz M.S. ADENOVLRIDAE AND THEIR REPLICATION, in Fields B, et al. (eds.) VIROLOGY, Vol. 2, Raven Press New York, pp. 1679-1721 (1990); Graham F., et al. pp. 109-128 in METHODS IN MOLECULAR BIOLOGY, Vol. 7: GENE TRANSFER AND EXPRESSION PROTOCOLS, Murray, E. (ed.) Humana Press, Clifton, NJ.
  • WO 94/26914 24 November 1994; Perricaudet M., et al. WO 95/02697 (26 January 1995); Zhang W., et al. WO 95/25071 (12 October 1995).
  • adenovirus plasmids are also available from commercial sources, including, e.g., Microbix Biosystems of Toronto, Ontario (see, e.g., Microbix Product Information Sheet: Plasmids for Adenovirus Vector Construction, 1996). See also, the papers by Vile, et al. (1997) Nature Biotechnology 15:840-841; and Feng, et al.
  • APCs can be transduced with viral vectors encoding a relevant polypeptides.
  • the most common viral vectors include recombinant poxviruses such as vaccinia and fowlpox virus (Bronte, et al. (1997) Proc. Natl. Acad. Sci. USA 94:3183-3188; Kim, et al. (1997) J. Immunother. 20:276-286) and, preferentially, adenovirus (Arthur, et al. (1997) J. Immunol. 159:1393-1403; Wan, et al. (1997) Human Gene Therapy 8:1355-1363; Huang, et al. (1995) J. Virol. 69:2257-2263). Retrovirus also may be used for transduction of human APCs (Marin, et al. (1996) J.
  • Transduced APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery.
  • In vivo transduction of DCs, or other APCs can be accomplished by administration of Ad (or other viral vectors) via different routes including intravenous, intramuscular, intranasal, intraperitoneal or cutaneous delivery.
  • Ad or other viral vectors
  • the preferred method is cutaneous delivery of Ad vector at multiple sites using a total dose of approximately lxl0 10 -lx 10 12 i.u.
  • Levels of in vivo transduction can be roughly assessed by co-staining with antibodies directed against APC marker(s) and the TAA being expressed.
  • the staining procedure can be carried out on biopsy samples from the site of administration or on cells from draining lymph nodes or other organs where APCs (in particular DCs) may have migrated. (Condon, et al. (1996) Nature Med. 2:1122-1128 and Wan, et al. (1997) Hum. Gene Ther. 8: 1355-1363).
  • the amount of antigen being expressed at the site of injection or in other organs where transduced APCs may have migrated can be evaluated by ELISA on tissue homogenates.
  • DCs can also be transduced in vitro/ex vivo by non- viral gene delivery methods such as electroporation, calcium phosphate precipitation or cationic lipid/plasmid DNA complexes.
  • Transduced APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery.
  • In vivo transduction of DCs, or other APCs can potentially be accomplished by administration of cationic lipid/plasmid DNA complexes delivered via the intravenous, intramuscular, intranasal, intraperitoneal or cutaneous route of administration.
  • Adoptive Immunotherapy and Vaccines The expanded populations of antigen-specific immune effector cells of the present invention also find use in adoptive immunotherapy regimes and as vaccines.
  • Adoptive immunotherapy methods involve, in one aspect, administering to a subject a substantially pure population of educated, antigen-specific immune effector cells made by culturing naive immune effector cells with APCs as described above.
  • the APCs are dendritic cells.
  • the adoptive immunotherapy methods described herein are autologous.
  • the APCs are made using parental cells isolated from a single subject.
  • the expanded population also employs T cells isolated from that subject.
  • the expanded population of antigen-specific cells is administered to the same patient.
  • APCs or immune effector cells are administered with an effective amount of a stimulatory cytokine, such as IL-2 or a co-stimulatory molecule.
  • a stimulatory cytokine such as IL-2 or a co-stimulatory molecule.
  • the agents identified herein as effective for their intended pu ⁇ ose can be administered to subjects having tumors previously determined to be reactive with any of the peptides of the invention as well as or in addition to individuals susceptible to or at risk of developing such tumors. Methods for identifying these individuals are described, infra.
  • the agent When the agent is administered to a subject such as a mouse, a rat or a human patient, the agent can be added to a pharmaceutically acceptable carrier and systemically or topically administered to the subject. To determine patients that can be beneficially treated, a tumor regression can be assayed.
  • Therapeutic amounts can be empirically determined and will vary with the pathology being treated, the subject being treated and the efficacy and toxicity of the therapy. Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the pu ⁇ ose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be found below.
  • agents and compositions of the present invention can be used in the manufacture of medicaments and for the treatment of humans and other animals by administration in accordance with conventional procedures, such as an active ingredient in pharmaceutical compositions. More particularly, an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including nasal, topical (including transdermal, aerosol, buccal and sublingual), parental (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It will also be appreciated that the preferred route will vary with the condition and age of the recipient, and the disease being treated.
  • the preceding discussion and examples are intended merely to illustrate the art. As is apparent to one of skill in the art, various modifications can be made to the above without departing from the spirit and scope of this invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention provides novel compositions and methods for the therapy, diagnosis, and prognosis of cancer. In one aspect, this invention is a peptide wherein the sequence of the peptide is represented by the peptide shown in Table 1. The peptides can be combined with a carrier such as a pharmaceutically acceptable carrier. Further provided are polynucleotides encoding the peptides of the inventions. The polynucleotides can be combined with a carrier such as a pharmaceutically acceptable carrier. Also provide are gene delivery vehicles and/or host cells comprising these polynucleotides. Therapeutic, diagnostic and prognostic methods using these compounds are also provided.

Description

EMMUNOGENIC PEPTIDES
CROSS-REFERENCE TO RELATED APPLICATIONS The subject application claims the benefit under 35 U.S.C. § 119(e) of U.S. Application Serial No. 60/510,630, filed October 10, 2003, the contents of which are hereby incorporated by reference into the present disclosure.
TECHNICAL FIELD The invention relates to compounds useful in therapeutic, diagnostic and screening methods for human cancers and related malignancies.
BACKGROUND OF THE INVENTION Therapeutic cancer vaccines seek to activate a patient's own immune system to kill cancer cells. Antigen-specific T cell activity has provided a venue for developing novel strategies for anti-cancer vaccines based on the observation that antigenic epitopes presented by molecules of the Major Histocompatibility Complex (MHC) play a central role in the establishment, maintenance and execution ofthe mammalian immune response. Antigen-specific cytotoxic T lymphocytes (CTLs) that recognize certain cancer antigens have been used to attack cells expressing these antigens to reject established tumors. Initial attempts to use vaccine-based strategies involved immunization with tumor cells that were provided as lysate, or as irradiated products and tumor proteins, and were either mixed with adjuvant or genetically modified to increase immunogenicity. (See Yee and Greenbert (June 2002) Nature Reviews/Cancer 2:409-419, and reference cited therein). The later identification of T-cell defined "tumor" antigens led to the use of peptide based vaccines. Anti-tumor T cells are localized within various cancer patient's tissues, including in the blood (where they can be found in the peripheral blood mononuclear cell fraction), in primary and secondary lymphoid tissue, e.g., the spleen, in ascites fluid in ovarian cancer patients (tumor associated lymphocytes or TALs) or within the tumor itself (tumor infiltrating lymphocytes or TILs). Of these, TILs have been the most useful in the identification of tumor antigens and tumor antigen-derived peptides recognized by T cells. In vitro studies have shown that the ability of a particular peptide to function as a T cell epitope requires that it binds effectively to the antigen presenting domain of an MHC molecule and also that it displays an appropriate set of amino acids that can be specifically recognized by a T cell receptor molecule. Thus, to date the development of peptide-based vaccines and therapeutics has relied either on the direct characterization of peptides associated with class I molecules, or alternatively on the identification of putative epitopes by inspection of the primary structures of proteins for motif sequences. (See, e.g., Deroo and Muller, (2001) Comb. Chem. & High Throughput Screening 4:75-110). The underlying assumption of the structure-activity relationship (SAR) for cancer vaccines has been that peptides that bind with high affinity to their restricting class I molecule will be the most immunogenic and will actually be generated in vivo by antigen processing. (Ochoa-Garay, et al. (1997) Mol. Immun. 34(3):273-281). Thus, in vitro screening of candidate target antigens (immunogens) have focused on optimization of binding to class I MHC (defined as the antigenicity of the peptide) on the belief that highly antigenic compounds will be immunogenic (defined as the ability to induce T cells that crossreact with the original antigen). Autologous dendritic cells engineered to express "tumor" antigens with the goal of augmenting the antigen-specific T-cells responses have also been tried. Alternatively, TILs manipulated to specifically recognize, bind and lyse cancer cells have been used as a means of adoptive T-cell therapy. (Yee and Greenbert (June 2002) supra). Conventional methods to generate TILs involve mincing tumor biopsy tissue and culturing the cell suspension in vitro in the presence of the T cell growth factor interleukin-2 (IL-2). Over a period of several days, the combination of the tumor cells and IL-2 can stimulate the proliferation of tumor specific T cells at the expense of tumor cells. In this way, the T cell population is expanded. The T cells derived from the first expansion are subsequently mixed with either mitomycin C-treated or irradiated tumor cells and cultured in vitro with IL-2 to promote further proliferation and enrichment of tumor reactive T cells. After several rounds of in vitro expansion, a potent anti-tumor T cell population can be recovered and used to identify tumor antigens via conventional but tedious expression cloning methodology. (Kawakani Y., et al. (1994) Proc. Natl. Acad. Sci. USA 91(9):3515-3519). Unfortunately to date, these immotherapeutic strategies, while initially promising, have not provided a cancer vaccine. Dissis and Cheever report that many newly defined tumor antigens are self proteins (e.g., MAGE, MART, gplOO, and tyrosinase) and therefore will not induce an effective immune response. (Dissi and Cheever (1998) Critical Rev. Immunol. 18:37-45). These therapeutic failures are believed to be the result of immune tolerance which reduces the ability of cancer vaccines to trigger a robust and sustained immune response against cancer cells. In other words, cancer cells and therefore therapeutics based on cancer-specific peptides, are not typically seen by a patient's immune system as being non-self or foreign (i.e., different from normal cells) and thus, do not activate the immune system in the same way a "foreign" protein can. Those of skill in the art define tolerance as a state of immunologic unresponsiveness to antigens, whether self or foreign. A recent publication explains tolerance as resulting in the journal "from one of three inhibitory influences on T and B cells: (1) clonal deletion, in which antigenic recognition leads to the activation-induced death of specific lymphocytes; (2) clonal anergy, in which lymphocytes are not killed but are rendered unresponsive to the recognized antigen; and (3) T cell-mediated suppression, in which regulatory T cells actively inhibit an immune response to an antigen. Several factors help determine which of those responses will occur. Immature lymphocytes are more susceptible to induction of tolerance than are mature lymphocytes. Tolerance can be induced in immature lymphocytes either centrally or in the periphery. Central tolerance is acquired when immature lymphocytes encounter antigens in the organs that generate these cells: the thymus (T cells) and the bone marrow (B cells)." (Scientific American, infra). T cells recognize antigens that have been processed into peptides and presented in a complex with MHC molecules (self-MHC-peptide complexes). Consequently, immature T cells must be screened for their ability to recognize self-MHC. This screening takes place in the thymus gland. T cells bearing receptors that recognize self-MHC are subjected to the processes of positive and negative selection. Positive selection occurs when T cells bearing receptors with a moderate affinity for self-MHC-peptide complexes receive survival and maturation signals after receptor ligation. Once these cells mature, they are exported to the periphery. Negative selection occurs when T cells bearing receptors with a high affinity for self-MHC-peptide complexes undergo activation-induced death. The thymus gland is capable of presenting many self-antigens that are normally expressed outside of the thymus or during restricted developmental stages. This allows the elimination of most T cells bearing high-affinity receptors for self-MHC-peptide complexes and plays a major role in preventing autoimmunity in peripheral organs and thus explains the failure of recent therapeutic approaches. (See Ochoa-Garay, et al. (1997) Mol. Immunol. 34(3):273-281). Because positive selection allows the maturation of T cells bearing receptors capable of low-affinity interactions with self-MHC-peptide complexes, potentially self-reactive T cells are normally found in peripheral lymphoid organs. Peripheral tolerance prevents these cells from inducing autoimmune disease. Peripheral tolerance is most often caused by the failure of T cells bearing low-affinity receptors to recognize self-antigen in the periphery. In this situation, the potentially self-reactive T cell is not activated and remains functionally naive. These cells are functional, however, as is shown by the fact that they can be activated by immunization with self-antigen delivered in the presence of immune adjuvants. Failure to respond to self-antigen may simply reflect a receptor-binding affinity that is below the threshold for T cell activation. A second mechanism of peripheral tolerance involves the elimination of self-reactive T cells by apoptosis. This process is analogous to clonal deletion in the thymus. Whether peripheral deletion plays an important role in tolerance to self-antigens is not known. A third mechanism of peripheral tolerance involves the acquisition of anergy after ligation of the T cell receptor complex. This antigen-nonresponsive state can be induced in several distinct ways. The most extensively characterized mechanism of anergy induction occurs when the T cell receptor is ligated in the absence of costimulation. After a T cell has bound with an antigen, the cell requires a so-called second signal delivered by one or more costimulatory molecules to be primed for an immune response. T cells express several surface molecules that can transmit this second signal. These costimulatory receptors are engaged by ligands expressed on the surface of APCs. T cells that are activated in the absence of costimulation acquire defects in the transcriptional control pathways that allow the production of IL-2, an important T cell autocrine growth factor. In vitro anergy can often be overcome by supplying exogenous IL-2 to anergic T cells. Costimulatory signals can be delivered to T cells by soluble factors or cell-surface molecules expressed on APCs. The most potent costimulatory signals are delivered when CD28, CD 154, or both are ligated on the surface of T cells. The ligand for CD 154 is CD40, a protein expressed on the surface of activated B cells, DCs, and maerophages. The ligands for CD28 (B7-1 (CD80), B7-2 (CD86), and related proteins) are expressed on the surface of APCs, such as DCs, monocytes, and B cells. Their expression is induced when APCs are activated in the course of microbial infection. This property heightens the immune response in the setting of perceived danger. B7-1 and B7-2 have overlapping immunostimulatory roles: mice lacking either protein are only partially deficient in generating an immune response to foreign antigen. Ligation of CD28 induces the expression of CTLA-4, a structurally related protein that turns off activated T cells. By this mechanism, the activated T cell initiates a program that will ensure its elimination at the conclusion of the immune response. Compared with CD28, CTLA-4 has a higher affinity for B7-1 and B7-2. CD28-B7 interactions may play a role in promoting tolerance to self-antigens. CD28-B7 interactions are required for the maturation of a distinct class of regulatory T cells that help maintain peripheral tolerance. These regulatory T cells (Tr) are included in a subpopulation (5%-15%) of peripheral blood CD4+ T cells that express CD25, a subunit of the IL-2 receptor. The selective removal of CD4+ and CD25+ T cells from BALB/c mice results in the development of T cell-mediated autoimmune thyroiditis, gastritis, and diabetes. The CD4+ and CD25+ Tr cells that mature in the thymus gland bear receptors that have an intermediate affinity for self-MHC-peptide complexes. In the periphery, antigen exposure confers the ability to suppress the activation of CD4+and CD25+ T cells in an antigen-independent, cell contact-dependent manner. Although these cells secrete IL-10, a potent anti-inflammatory cytokine, their suppressive activity is cytokine independent. CD4+ and CD25+ T cells can suppress graft versus host disease in allotransplants and they can prevent autoimmune disease in several different animal models. Consequently, these cells probably play an essential role in maintaining peripheral tolerance to self-antigens." (WebMD Scientific American Medicine (samed.com/sam/forms/index/htm; ISSN: 0194-9063; Section 6: Immunology/ Alergy). Thus, current research is focused on how to direct the immune system to attack cancer cells as if they were foreign. One approach has been to stimulate cancer killing cytotoxic T cells (CTLS) using a potent immunostimulant chemical, together with delivery of a protein expressed at high levels on tumors and molecules that activate the helper T cells. An alternative approach utilizes T cell epitopes derived from native antigenic polypeptides mutated by the introduction of a single amino or multiple acids substitutions to alter sequence. (Valmori, et al. (2000) J. Immunol 164(2).T 125-1131). This work has focused on improving antigenicity of the epitopes, with the belief that an increase in antigenicity will increase immunogenicity in vivo. (Hickling (1998) Exp. Rev. Mol. Med. ISSN 1462-3994 ermm.cbu.cam.ac.uk). Reports of these studies have shown that while peptide affinity can sometimes be related to immunogenicity, the immunogenicity of a peptide cannot always be predicted from its affinity for MHC class I molecules or the presence of class I MHC binding motifs. (Ochoa-Garay, et al. (1997) supra; Deroo, et al. (2001) Combinatorial Chem. & High Throughput Screening 4:75-100; Purbhoo, et al. (1998) Proc. Natl. Acad. Sci. (95):4527-4532). In sum, other factors unrelated to MHC:peptide affinity such as CTL precursor frequency, solubility in water, cross-presentation and peptide stability, play a role in raising the immune response. (Ochoa-Garay, et al. (1997) supra; Yee and Greenbert (June 2002) supra; and Kurts C. (2000) J. Mol. Med. 78:326-332). For all these reasons, one would not expect non-mutated self-antigens to be immunogenic (i.e., capable of initiating an immune response). Thus, there exists a continuing need to identify immunogens, immunogenic epitopes and other biomarkers associated with cancer and to develop new materials and kits to aid in the early detection, therapy and monitoring of related cancers. The present invention satisfies this need and provides related advantages as well.
DISCLOSURE OF THE INVENTION The present invention provides novel compositions and methods for the therapy, diagnosis, and prognosis of cancer. In one aspect, this invention is a peptide wherein the sequence of the peptide is represented by the group comprising SEQ ID NOs: 2 through 44 (shown in Table 1, infra). The peptides can be combined with a carrier such as a pharmaceutically acceptable carrier. Further provided are polynucleotides encoding the peptides of the invention, for example the nucleic acid sequences provided in SEQ ID NOs: 1 through 43, complements and variants thereof. The polynucleotides can be combined with a carrier such as a pharmaceutically available carrier. Also provided are gene delivery vehicles and/or host cells comprising these polynucleotides. Further provided are compositions comprising at least one of the biological materials represented by SEQ ID NOS. 1 through 44 and a carrier, such as a pharmaceutically acceptable carrier.
TABLE 1
Figure imgf000009_0001
Further provided by this invention is a host cell comprising at least one peptide and/or polynucleotide encoding said peptide, wherein said peptide comprises a sequence selected from SEQ ID NOs: 2 through 44. In one aspect, the host cell is an antigen presenting cell, e.g., a dendritic cell. Compositions comprising such host cells and a carrier such as a pharmaceutically acceptable carrier, are also provided. In addition, the invention provides methods for inducing an immune response in a subject by delivering to the subject the peptides of the invention, and delivering these in the context of an MHC molecule. The peptides of the invention are also useful to generate antibodies that specifically recognize and bind to these polypeptides. These antibodies are further useful for immunotherapy when administered to a subject. The invention also provides immune effector cells raised in vivo or in vitro in the presence and at the expense of an antigen presenting cell that presents one or more polyρeptide(s) of the invention in the context of an MHC molecule and a method of adoptive immunotherapy comprising administering an effective amount of these immune effector cells to a subject. Further provided by this invention is a method for inducing an immune response in a subject by delivering to the subject a composition comprising an effective amount of at least one polypeptide shown in Table 1. The present invention additionally provides methods and compositions for detecting, diagnosing, prognosing and monitoring the progress of cancers and malignancies and kits for use in said methods. Further provided are methods for screening to identify agonists and antagonists of cancer antigens associated with cancers and malignancies.
MODES OF CARRYING OUT THE INVENTION Throughout this disclosure, various publications, patents and published patent specifications are referenced by an identifying citation. The disclosures of these publications, patents and published patent specifications are hereby incoφorated by reference into the present disclosure to more fully describe the state of the art to which this invention pertains. The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. These methods are described in the following publications. See, e.g., Sambrook, et al. MOLECULAR CLONING: A LABORATORY MANUAL, 2nd edition (1989); CURRENT PROTOCOLS IN MOLECULAR BIOLOGY (F. M. Ausubel, et al. eds. (1987)); the series METHODS IN ENZYMOLOGY (Academic Press, Inc.); 005/037854
PCR: A PRACTICAL APPROACH (M. MacPherson, et al. IRL Press at Oxford University Press (1991)); PCR 2: A PRACTICAL APPROACH (M.J. MacPherson, B.D. Hames and G.R. Taylor eds. (1995)); ANTIBODIES, A LABORATORY MANUAL (Hariow and Lane eds. (1988)); USING ANTIBODIES, A LABORATORY MANUAL (Hariow and Lane eds. ( 1999)); and ANIMAL CELL CULTURE (R.I. Freshney ed. (1987)).
Definitions As used herein, certain terms have the following defined meanings. As used in the specification and claims, the singular form "a," "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a cell" includes a plurality of cells, including mixtures thereof. As used herein, the term "comprising" is intended to mean that the compositions and methods include the recited elements, but do not exclude others. "Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. "Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention. Embodiments defined by each of these transition terms are within the scope of this invention. A "native" or "natural" or "wild-type" antigen is a polypeptide, protein or a fragment which contains an epitope, which has been isolated from a natural biological source, and which can specifically bind to an antigen receptor, in particular a T cell antigen receptor (TCR), in a subject. An "antigen" is a substance that is antigenic. "Antigenic" refers to the ability of a peptide to bind to its ligand. As an example, the antigenicity of a peptide or epitope is determined by its ability to bind to a T cell. A "self-antigen" also referred to herein as a native or wild-type antigen is an antigenic peptide that induces little or no immune response in the subject due to self-tolerance to the antigen. An example of a self-antigen is the melanoma specific antigen gplOO. Any substance that can elicit an immune response is said to be
"immunogenic" and is referred to as an "immunogen". All immunogens are antigens, however, not all antigens are immunogenic. An immune response of this invention can be humoral (via antibody activity) or cell-mediated (via T cell activation). The term "ligand" as used herein refers to any molecule that binds to a specific site on another molecule. In other words, the ligand confers the specificity of the protein in a reaction with an immune effector cell. It is the ligand site within the protein that combines directly with the complementary binding site on the immune effector cell. The term "tumor associated antigen" or "TAA" refers to an antigen that is associated with or specific to a tumor. Examples of known TAAs include gplOO, MART and MAGE. The term "antibody" is further intended to encompass digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or specified fragment or portion thereof, including single chain antibodies and fragments thereof. Examples of binding fragments encompassed within the term "antigen binding portion" of an antibody include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH, domains; a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CH, domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH domain; and an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)). Bird et al. (1988) Science 242:423-426 and Huston et al. (1988) Proc. Natl. Acad Sci. USA 85:5879-5883. Single chain antibodies are also intended to be encompassed within the term "fragment of an antibody." Any of the above- noted antibody fragments are obtained using conventional techniques known to those of skill in the art, and the fragments are screened for binding specificity in the same manner as are intact antibodies. Such fragments can be produced by enzymatic cleavage, synthetic or recombinant techniques, as known in the art and/or as described herein. For example, an antibody fragment can be produced in a variety of truncated foπns using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a combination gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and/or hinge region of the heavy chain. The various portions of antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques. The term "epitope" means a protein determinant capable of specific binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents. The term "antibody" variant is intended to include antibodies produced in a species other than a mouse or an isotype of an antibody selected from the antibodies designated ACO-1 thorugh ACO-6. The term "antibody variant" also includes antibodies containing post-translational modifications to the linear polypeptide sequence of the antibody or fragment. It further encompasses fully human antibodies. The term "antibody derivative" is intended to encompass molecules that bind an epitope as defined above and which are modifications or derivatives of a native monoclonal antibody of this invention. Derivatives include, but are not limited to, for example, bispecific, multispecific, heterospecific, trispecific, tetraspecific, multispecific antibodies, diabodies, chimeric, recombinant and humanized. The term "bispecific molecule" is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has two different binding specificities. The term "multispecific molecule" or "heterospecific molecule" is intended to include any agent, e.g. a protein, peptide, or protein or peptide complex, which has more than two different binding specificities. The term "heteroantibodies" refers to two or more antibodies, antibody binding fragments (e.g., Fab), derivatives thereof, or antigen binding regions linked together, at least two of which have different specificities. The term "human antibody", as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Thus, as used herein, the term "human antibody" refers to an antibody in which substantially every part of the protein (e.g., CDR, framework, CL, CH domains (e.g., CHι, CH2, CH3), hinge, (VL, VH)) is substantially non-immunogenic in humans, with only minor sequence changes or variations. Similarly, antibodies designated primate (monkey, baboon, chimpanzee, etc.), rodent (mouse, rat, rabbit, guinea pig, hamster, and the like) and other mammals designate such species, sub-genus, genus, sub-family, family specific antibodies. Further, chimeric antibodies include any combination of the above. Such changes or variations optionally and preferably retain or reduce the immunogenicity in humans or other species relative to non-modified antibodies. Thus, a human antibody is distinct from a chimeric or humanized antibody. It is pointed out that a human antibody can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes. Further, when a human antibody is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies. For example, an Fv can comprise a linker peptide, such as two to about eight glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain. Such linker peptides are considered to be of human origin. As used herein, a human antibody is "derived from" a particular germline sequence if the antibody is obtained from a system using human immunoglobulin sequences, e.g., by immunizing a transgenic mouse carrying human immunoglobulin genes or by screening a human immunoglobulin gene library. A human antibody that is "derived from" a human germline immunoglobulin sequence can be identified as such by comparing the amino acid sequence of the human antibody to the amino acid sequence of human germline immunoglobulins. A selected human antibody typically is at least 90% identical in amino acids sequence to an amino acid sequence encoded by a human germline immunoglobulin gene and contains amino acid residues that identify the human antibody as being human when compared to the germline immunoglobulin amino acid sequences of other species (e.g., murine germline sequences). In certain cases, a human antibody may be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to the amino acid sequence encoded by the germline immunoglobulin gene. Typically, a human antibody derived from a particular human germline sequence will display no more than 10 amino acid differences from the amino acid sequence encoded by the human germline immunoglobulin gene. In certain cases, the human antibody may display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the amino acid sequence encoded by the germline immunoglobulin gene. The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. A "human monoclonal antibody" refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobulin sequences. The term "recombinant human antibody", as used herein, includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom, antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma, antibodies isolated from a recombinant, combinatorial human antibody library, and antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. As used herein, "isotype" refers to the antibody class (e.g., IgM or IgGl) that is encoded by heavy chain constant region genes. The terms "transgenic, nonhuman animal" refers to a nonhuman animal having a genome comprising one or more human heavy and/or light chain transgenes or transchromosomes (either integrated or non-integrated into the animal's natural genomic DNA) and which is capable of expressing fully human antibodies. For example, a transgenic rat can have a human light chain transgene and either a human heavy chain transgene or human heavy chain transchromosome, such that the rat produces human anti-INF-α antibodies. The human heavy chain transgene can be integrated into the chromosomal DNA of the rat, or the human heavy chain transgene can be maintained extrachromosomally. Transgenic and transchromosomal animals are capable of producing multiple isotypes of human monoclonal antibodies to Alpha V (e.g., IgG, IgA and/or IgE) by undergoing V-D-J recombination and isotype switching. The terms "major histocompatibility complex" or "MHC" refers to a complex of genes encoding cell-surface molecules that are required for antigen presentation to T cells and for rapid graft rejection. In humans, the MHC is also known as the "human leukocyte antigen" or "HLA" complex. The proteins encoded by the MHC are known as "MHC molecules" and are classified into class I and class II MHC molecules. Class I MHC includes membrane heterodimeric proteins made up of an chain encoded in the MHC noncovalently linked with the /52-microglobulin. Class I MHC molecules are expressed by nearly all nucleated cells and have been shown to function in antigen presentation to CD8+ T cells. Class I molecules include HLA-A, B, and C in humans. Class II MHC molecules also include membrane heterodimeric proteins consisting of noncovalently associated and β chains. Class II MHC molecules are known to function in CD4+ T cells and, in humans, include HLA-DP, -DQ, and DR. In a preferred embodiment, invention compositions and ligands can complex with MHC molecules of any HLA type. Those of skill in the art are familiar with the serotypes and genotypes of the HLA. (See bimas.dcrt.nih.gov/cgi-bin/molbio/hla coefficient viewing page; Rammensee H.G., et al, MHC LIGANDS AND PEPTIDE MOTIFS (1997) Chapman & Hall Publishers; Schreuder G.M. Th. et al. The HLA Dictionary (1999) Tissue Antigens 54:409-437). The term "antigen-presenting matrix", as used herein, intends a molecule or molecules which can present antigen in such a way that the antigen can be bound by a T-cell antigen receptor on the surface of a T cell. An antigen-presenting matrix can be on the surface of an antigen-presenting cell (APC), on a vesicle preparation of an APC, or can be in the form of a synthetic matrix on a solid support such as a bead or a plate. An example of a synthetic antigen-presenting matrix is purified MHC class I molecules complexed to β2-microglobulin, multimers of such purified MHC class I molecules, purified MHC Class II molecules, or functional portions thereof, attached to a solid support. The term "antigen presenting cells (APC)" refers to a class of cells capable of presenting one or more antigens in the form of antigen-MHC complex recognizable by specific effector cells of the immune system, and thereby inducing an effective cellular immune response against the antigen or antigens being presented. While many types of cells may be capable of presenting antigens on their cell surface for T-cell recognition, only professional APCs have the capacity to present antigens in an efficient amount and further to activate T-cells for cytotoxic T-lymphocyte (CTL) responses. APCs can be intact whole cells such as maerophages, B-cells and dendritic cells; or other molecules, naturally occurring or synthetic, such as purified MHC class I molecules complexed to β2-microglobulin. The term "dendritic cells (DC)" refers to a diverse population of morphologically similar cell types found in a variety of lymphoid and non-lymphoid tissues. (Steinman (1991) Ann. Rev. Immunol. 9:271-296). Dendritic cells constitute the most potent and preferred APCs in an organism. A subset, if not all, of dendritic cells are derived from bone marrow progenitor cells, circulate in small numbers in the peripheral blood and appear either as immature Langerhans' cells or terminally differentiated mature cells. While the dendritic cells can be differentiated from monocytes, they possess distinct phenotypes. For example, a particular differentiating marker, CD 14 antigen, is not found in dendritic cells but is possessed by monocytes. Also, mature dendritic cells are not phagocytic, whereas the monocytes are strongly phagocytosing cells. It has been shown that DCs provide all the signals necessary for T cell activation and proliferation. The term "antigen presenting cell recruitment factors" or "APC recruitment factors" include both intact, whole cells as well as other molecules that are capable of recruiting antigen presenting cells. Examples of suitable APC recruitment factors include molecules such as interleukin 4 (IL-4), granulocyte macrophage colony stimulating factor (GM-CSF), Sepragel and macrophage inflammatory protein-3 -alpha (MIP3α). These are available from Immunex, Schering-Plough and R&D Systems (Minneapolis, MN). They also can be recombinantly produced using the methods disclosed in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY ((1987) supra). Peptides, proteins and compounds having the same biological activity as the above-noted factors are included within the scope of this invention. The term "immune effector cells" refers to cells capable of binding an antigen and which mediate an immune response. These cells include, but are not limited to, T cells, B cells, monocytes, maerophages, NK cells and cytotoxic T lymphocytes (CTLs), for example CTL lines, CTL clones, and CTLs from tumor, inflammatory, or other infiltrates. Certain diseased tissue expresses specific antigens and CTLs specific for these antigens have been identified. For example, approximately 80% of melanomas express the antigen known as gplOO. The term "immune effector molecule" as used herein, refers to molecules capable of antigen-specific binding, and includes antibodies, T cell antigen receptors, and MHC Class I and Class II molecules. A "naive" immune effector cell is an immune effector cell that has never been exposed to an antigen capable of activating that cell. Activation of naive immune effector cells requires both recognition of the peptide:MHC complex and the simultaneous delivery of a costimulatory signal by a professional APC in order to proliferate and differentiate into antigen-specific armed effector T cells. , In one embodiment, a peptide of the invention binds to a ligand such as an antigenic determinant or epitope on an immune effector cell, e.g. an antibody or a T cell receptor (TCR). A ligand may be an antigen, peptide, protein or epitope of the invention. Invention ligands may bind to a receptor on an antibody. In one embodiment, the peptides of the invention is about 4 to about 10 amino acids in length. \ Invention peptides may bind to a receptor on an MHC class I molecule. In one embodiment, the peptide of the invention is about 7 to about 12 amino acids in length. Invention peptides may bind to a receptor on an MHC class II molecule. In one embodiment, the peptide of the invention is about 8 to about 20 amino acids long. As used herein, the term "educated, antigen-specific immune effector cell", is an immune effector cell as defined above, which has previously encountered an antigen. In contrast with its naϊve counterpart, activation of an educated, antigen-specific-immune effector cell does not require a costimulatory signal. Recognition of the peptide:MHC complex is sufficient. "Activated", when used in reference to a T cell, implies that the cell is no longer in G0 phase, and begins to produce one or more of cytotoxins, cytokines, and other related membrane-associated proteins characteristic of the cell type (e.g., CD8+ or CD4+), is capable of recognizing and binding any target cell that displays the particular antigen on its surface, and releasing its effector molecules. In the context of the present invention, the term "recognized" intends that a peptide of the invention, is recognized and bound by its ligand, e.g., antibody, Tcell, or an immune effector cell wherein such binding initiates an effective immune response. The term "cross-reactive" is used to describe certain immunogenic properties of peptides which are functionally overlapping. More particularly, the immunogenic properties of a peptide and/or immune effector cells activated thereby are shared to a certain extent by other peptides. For purposes of this invention, cross-reactivity is manifested at multiple levels: (i) at the peptide level, the peptides can bind the TCR and activate CTLs; (ii) at the T cell level, i.e., peptides of the invention bind the TCR of and activate a population of T cells which can effectively target and lyse cells; and (iii) at the antibody level, e.g., "anti"-peptide antibodies can detect, recognize and bind the peptide and initiate effector mechanisms in an immune response. As used herein, the term "inducing an immune response in a subject" is a term understood in the art and intends that an increase of at least about 2-fold, more preferably at least about 5-fold, more preferably at least about 10-fold, more preferably at least about 100-fold, even more preferably at least about 500-fold, even more preferably at least about 1000-fold or more in an immune response to an antigen (or epitope) can be detected or measured, after introducing the antigen (or epitope) into the subject, relative to the immune response (if any) before introduction of the antigen (or epitope) into the subject. An immune response to an antigen (or epitope), includes, but is not limited to, production of an antigen-specific (or epitope-specific) antibody, and production of an immune cell expressing on its surface a molecule which specifically binds to an antigen (or epitope). Methods of determining whether an immune response to a given antigen (or epitope) has been induced are well known in the art. For example, antigen-specific antibody can be detected using any of a variety of immunoassays known in the art, including, but not limited to, ELISA, wherein, for example, binding of an antibody in a sample to an immobilized antigen (or epitope) is detected with a detectably-labeled second antibody (e.g., enzyme-labeled mouse anti-human Ig antibody). "Co-stimulatory molecules" are involved in the interaction between receptor-ligand pairs expressed on the surface of antigen presenting cells and T cells. Research accumulated over the past several years has demonstrated convincingly that resting T cells require at least two signals for induction of cytokine gene expression and proliferation. (Schwartz R.H. (1990) Science 248:1349-1356 and Jenkins M.K. (1992) Immunol. Today 13:69-73). One signal, the one that confers specificity, can be produced by interaction of the TCR/CD3 complex with an appropriate MHC/peptide complex. The second signal is not antigen specific and is termed the "co-stimulatory" signal. This signal was originally defined as an activity provided by bone-marrow-derived accessory cells such as maerophages and dendritic cells, the so called "professional" APCs. Several molecules have been shown to enhance co-stimulatory activity. These are heat stable antigen (HSA) (Liu Y, et al. (1992) J. Exp. Med. 175:437-445), chondroitin sulfate-modified MHC invariant chain (Ii-CS) (Naujokas M.F., et al. (1993) Cell 74:257-268), intracellular adhesion molecule 1 (ICAM-1) (Van Seventer G.A. (1990) J. Immunol. 144:4579-4586), B7-1, and B7-2/B70 (Schwartz R.H. (1992) Cell 71:1065-1068). These molecules each appear to assist co-stimulation by interacting with their cognate ligands on the T cells.
Co-stimulatory molecules mediate co-stimulatory signal(s), which are necessary, under normal physiological conditions, to achieve full activation of naive T cells. One exemplary receptor-ligand pair is the B7 co-stimulatory molecule on the surface of APCs and its counter-receptor CD28 or CTLA-4 on T cells. (Freeman, et al. (1993) Science 262:909-911; Young, et al. (1992) J. Clin. Invest 90:229 and Nabavi, et al. (1992) Nature 360:266-268). Other important co-stimulatory molecules are CD40, CD54, CD80, and CD86. The term "co-stimulatory molecule" encompasses any single molecule or combination of molecules which, when acting together with a peptide/MHC complex bound by a TCR on the surface of a T cell, provides a co-stimulatory effect which achieves activation of the T cell that binds the peptide. The term thus encompasses B7, or other co-stimulatory molecule(s) on an antigen-presenting matrix such as an APC, fragments thereof (alone, complexed with another molecule(s), or as part of a fusion protein) which, together with peptide/MHC complex, binds to a cognate ligand and results in activation of the T cell when the TCR on the surface of the T cell specifically binds the peptide. Co-stimulatory molecules are commercially available from a variety of sources, including, for example, Beckman Coulter, Inc. (Fullerton, CA). It is intended, although not always explicitly stated, that molecules having similar biological activity as wild-type or purified co-stimulatory molecules (e.g. , recombinantly produced or muteins thereof) are intended to be used within the spirit and scope of the invention. As used herein, "solid phase support" or "solid support", used interchangeably, is not limited to a specific type of support. Rather a large number of supports are available and are known to one of ordinary skill in the art. Solid phase supports include silica gels, resins, derivatized plastic films, glass beads, glass slides, flasks, tissue culture flasks, cotton, plastic beads, alumina gels. As used herein, "solid support" also includes synthetic antigen-presenting matrices, cells, and liposomes. A suitable solid phase support may be selected on the basis of desired end use and suitability for various protocols. For example, for peptide synthesis, solid phase support may refer to resins such as polystyrene (e.g., PAM-resin obtained from Bachem Inc., Peninsula Laboratories, etc.), POLYHIPE ® resin (obtained from Aminotech, Canada), polyamdde resin (obtained from Peninsula Laboratories), polystyrene resin grafted with polyethylene glycol (TentaGel®, Rapp Polymere, Tubingen, Germany) or polydimethylacrylamide resin (obtained from Milligen/Biosearch, California). Solid supports also include microchips and grids, on which cDNAs, oligonucleotides, peptides, antibodies or other molecules are fixed in arrays. The surface of the grids may be composed of a wide variety of material including glass, plastic, silicon, gold, gelatin or nylon. (See, e.g., Lockhart (2000) Nature 405:827-836 and Srinivas (2001) Clin. Chem., 47: 1901-1911). Also included within the term " solid support" are tissue microarrays in which small cylinders of tissue are punched out of thousands of individual tumor specimens (from different tissues of hundreds of individuals in a study) and then probed with antibodies, RNA, etc. (Hoos A., et al. (2001) Am J. Pathol. 158:1245-51). The components and compositions for use in the methods of this invention can also provided on microchips. For example, the use of the so-called
SELDI-MS method (Surface-Enhanced Laser Desorption-Ionization & Mass Spectroscopy) exposes samples to chips with biochemically characterized surfaces (containing molecules such as antibodies or receptors) followed by mass spectroscopy to visualize and identify the bound proteins. For a review of this recently available technology see Srivinas P. et al. (2001) Clinical Chemistry 47(10):1901-1911, and references cited therein such as De Wildt R.M.T., et al. (2000) Nat. Biotech 18:989-94; Arenkov P., et al. (2000) Anal. Biochem. 278:123-31; Haab B.B., et al. (2001) Genome Biol. 2:1-13; and Cahill D.J. (2001) J. Immunol. Methods 250:81-91. The term "immunomodulatory agent", as used herein, is a molecule, a macromolecular complex, or a cell that modulates an immune response and encompasses a peptide of the invention alone or in any of a variety of formulations described herein; a polypeptide comprising a peptide of the invention; a polynucleotide encoding a peptide or polypeptide of the invention; a peptide of the invention bound to a Class I or a Class II MHC molecule on an antigen-presenting matrix, including an APC and a synthetic antigen-presenting matrix (in the presence or absence of co-stimulatory molecule(s)); a peptide of the invention covalently or non-covalently complexed to another molecule(s) or macromolecular structure; and an educated, antigen-specific immune effector cell which is specific for a peptide of the invention. The term "modulate an immune response" includes inducing (increasing, eliciting) an immune response; and reducing (suppressing) an immune response. An immunomodulatory method (or protocol) is one that modulates an immune response in a subject. As used herein, the term "cytokine" refers to any one of the numerous factors that exert a variety of effects on cells, for example, inducing growth or proliferation. Non-limiting examples of cytokines which may be used alone or in combination in the practice of the present invention include, interleukin-2 (IL-2), stem cell factor (SCF), interleukin 3 (IL-3), interleukin 6 (IL-6), interleukin 12 (IL-12), G-CSF, granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin- 1 alpha (IL- 1 α), interleukin- 11 (IL- 11), MIP- 11 , leukemia inhibitory factor (LIF), c-kit ligand, thrombopoietin (TPO) and flt3 ligand. The present invention also includes culture conditions in which one or more cytokine is specifically excluded from the medium. Cytokines are commercially available from several vendors such as, for example, Genzyme Corp. (Framingham, MA), Genentech (South San Francisco, CA), Amgen (Thousand Oaks, CA), R&D
Systems (Minneapolis, MN) and Immunex (Seattle, WA). It is intended, although not always explicitly stated, that molecules having similar biological activity as wild-type or purified cytokines (e.g., recombinantly produced or muteins thereof) are intended to be used within the spirit and scope of the invention. All numerical designations, e.g., pH, temperature, time, concentration, and molecular weight, including ranges, are approximations which are varied ( + ) or ( - ) by increments of 0.1. It is to be understood, although not always explicitly stated that all numerical designations are preceded by the term "about". It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art. The terms "polynucleotide" and "oligonucleotide" are used interchangeably, and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Polynucleotides can have any three-dimensional structure, and may perform any function, known or unknown. The following are non-limiting examples of polynucleotides: a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polymer. The sequence of nucleotides can be interrupted by non-nucleotide components. A polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component. The term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any embodiment of this invention that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form. A polynucleotide is composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); thymine (T); and uracil (U) for guanine when the polynucleotide is RNA. Thus, the term "polynucleotide sequence" is the alphabetical representation of a polynucleotide molecule. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching. A "gene" refers to a polynucleotide containing at least one open reading frame (ORF) that is capable of encoding a particular polypeptide or protein after being transcribed and translated. Any of the polynucleotides sequences described herein may be used to identify larger fragments or full-length coding sequences of the gene with which they are associated. Methods of isolating larger fragment sequences are known to those of skill in the art. A "gene product" or alternatively a "gene expression product" refers to the amino acid (e.g., peptide or polypeptide) generated when a gene is transcribed and translated. The term "polypeptide" is used interchangeably with the term "protein" and in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g., ester, ether, etc. As used herein the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. The term "peptide" is used in its broadest sense to refer to a compound of two or more subunit amino acids. The subunits may be linked by peptide bonds. In another embodiment, the subunit may be linked by other bonds, e.g. ester, ether, etc. As used herein the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. The term "genetically modified" means containing and/or expressing a foreign gene or nucleic acid sequence which in turn, modifies the genotype or phenotype of the cell or its progeny. The term also refers to any enhancement, addition, deletion or disruption to a cell's endogenous nucleotides. As used herein, "expression" refers to the process by which polynucleotides are transcribed into mRNA and translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA, if an appropriate eukaryotic host is selected. Regulatory elements required for expression include promoter sequences to bind RNA polymerase and transcription initiation sequences for ribosome binding. For example, a bacterial expression vector includes a promoter such as the lac promoter and for transcription initiation the Shine-Dalgarno sequence and the start codon AUG (Sambrook et al. (1989) supra). Similarly, an eukaryotic expression vector includes a heterologous or homologous promoter for RNA polymerase II, a downstream polyadenylation signal, the start codon AUG, and a termination codon for detachment of the ribosome. Such vectors can be obtained commercially or assembled by the sequences described by methods known in the art. "Under transcriptional control" is a term understood in the art and indicates that transcription of a polynucleotide sequence, usually a DNA sequence, depends on its being operatively linked to an element which contributes to the initiation of, or promotes, transcription. "Operatively linked" refers to a juxtaposition wherein the elements are in an arrangement allowing them to function. A "gene delivery vehicle" is defined as any molecule that can carry inserted polynucleotides into a host cell. Examples of gene delivery vehicles are liposomes, biocompatible polymers, including natural polymers and synthetic polymers; lipoproteins; polypeptides; polysaccharides; lipopolysaccharides; artificial viral envelopes; recombinant yeast cells or yeast spheroplasts; metal particles; and bacteria, or viruses, such as baculovirus, adenovirus and retrovirus, bacteriophage, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression. "Gene delivery," "gene transfer," and the like as used herein, are terms referring to the introduction of an exogenous polynucleotide (sometimes referred to as a "transgene") into a host cell, irrespective of the method used for the introduction. Such methods include a variety of techniques such as vector-mediated gene transfer (by, e.g., viral infection/transfection, or various other protein-based or lipid-based gene delivery complexes) as well as techniques facilitating the delivery of "naked" polynucleotides (such as electroporation, "gene gun" delivery and various other techniques used for the introduction of polynucleotides). The introduced polynucleotide may be stably or transiently maintained in the host cell. Stable maintenance typically requires that the introduced polynucleotide either contains an origin of replication compatible with the host cell or integrates into a replicon of the host cell such as an extrachromosomal replicon (e.g., a plasmid) or a nuclear or mitochondrial chromosome. A number of vectors are known to be capable of mediating transfer of genes to mammalian cells, as is known in the art and described herein. A "viral vector" is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro. Examples of viral vectors include retroviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like. Alphavirus vectors, such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. (See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Zaks, et al. (1999) Nat. Med. 7:823-827). In aspects where gene transfer is mediated by a retroviral vector, a vector construct refers to the polynucleotide comprising the retroviral genome or part thereof, and a therapeutic gene. As used herein, "retroviral mediated gene transfer" or "retroviral transduction" carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome. The virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell. As used herein, retroviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. Retro viruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell. The integrated DNA form is called a pro virus. In aspects where gene transfer is mediated by a DNA viral vector, such as an adenovirus (Ad) or adeno-associated virus (AAV), a vector construct refers to the polynucleotide comprising the viral genome or part thereof, and a transgene. Adenoviruses (Ads) are a relatively well characterized, homogenous group of viruses, including over 50 serotypes. See, e.g., WO 95/27071. Ads are easy to grow and do not require integration into the host cell genome. Recombinant
Ad-derived vectors, particularly those that reduce the potential for recombination and generation of wild-type virus, have also been constructed. (See, WO 95/00655 and WO 95/11984). Wild-type AAV has high infectivity and specificity integrating into the host cell's genome. (See, Hermonat and Muzyczka (1984) Proc. Natl. Acad. Sci. USA 81:6466-6470 and Lebkowski, et al. (1988) Mol. Cell. Biol. 8:3988-3996). Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are known in the art. Such vectors are capable of transcribing RNA in vitro or in vivo, and are commercially available.. In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5' and/or 3' untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5' of the start codon to enhance expression. Gene delivery vehicles also include several non- viral vectors, including DNA/liposome complexes, and targeted viral protein-DNA complexes. Liposomes that also comprise a targeting antibody or fragment thereof can be used in the methods of this invention. To enhance delivery to a cell, the nucleic acid or proteins of this invention can be conjugated to antibodies or binding fragments thereof which bind cell surface antigens, e.g., TCR, CD3 or CD4. "Hybridization" refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme. Examples of stringent hybridization conditions include: incubation temperatures of about 25°C to about 37°C; hybridization buffer concentrations of about 6 X SSC to about 10 X SSC; formamide concentrations of about 0% to about 25%; and wash solutions of about 6 X SSC. Examples of moderate hybridization conditions include: incubation temperatures of about 40°C to about 50°C; buffer concentrations of about 9 X SSC to about 2 X SSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5 X SSC to about 2 X SSC. Examples of high stringency conditions include: incubation temperatures of about 55°C to about 68°C; buffer concentrations of about 1 X SSC to about 0.1 X SSC; formamide concentrations of about 55% to about 75%; and wash solutions of about 1 X SSC, 0.1 X SSC, or deionized water. In general, hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash incubation times are about 1, 2, or 15 minutes. SSC is 0.15 M NaCI and 15 mM citrate buffer. It is understood that equivalents of SSC using other buffer systems can be employed. A "probe" when used in the context of polynucleotide manipulation refers to an oligonucleotide that is provided as a reagent to detect a target potentially present in a sample of interest by hybridizing with the target. Usually, a probe will comprise a label or a means by which a label can be attached, either before or subsequent to the hybridization reaction. Suitable labels include, but are not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. A "primer" is a short polynucleotide, generally with a free 3' -OH group that binds to a target or "template" potentially present in a sample of interest by hybridizing with the target, and thereafter promoting polymerization of a polynucleotide complementary to the target. A "polymerase chain reaction" ("PCR") is a reaction in which replicate copies are made of a target polynucleotide using a "pair of primers" or a "set of primers" consisting of an "upstream" and a "downstream" primer, and a catalyst of polymerization, such as a DNA polymerase, and typically a thermally-stable polymerase enzyme. Methods for PCR are known in the art, and taught, for example in "PCR: A PRACTICAL APPROACH" (M. MacPherson et al, IRL Press at Oxford University Press
(1991)). All processes of producing replicate copies of a polynucleotide, such as PCR or gene cloning, are collectively referred to herein as "replication." A primer can also be used as a probe in hybridization reactions, such as Southern or Northern blot analyses. Sambrook et al., supra. An expression "database" denotes a set of stored data that represent a collection of sequences, which in turn represent a collection of biological reference materials. The term "cDNAs" refers to complementary DNA that are mRNA molecules present in a cell or organism and made into cDNA with an enzyme such as reverse transcriptase. A "cDNA library" is a collection of all of the mRNA molecules present in a cell or organism, all turned into cDNA molecules with the enzyme reverse transcriptase, then inserted into "vectors" (other DNA molecules that can continue to replicate after addition of foreign DNA). Exemplary vectors for libraries include bacteriophage (also known as "phage"), viruses that infect bacteria, for example, lambda phage. The library can then be probed for the specific cDNA (and thus mRNA) of interest. As used herein, "expression" refers to the process by which polynucleotides are transcribed into mRNA and/or the process by which the transcribed mRNA is subsequently being translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell. "Differentially expressed" as applied to a gene, refers to the differential production of the mRNA transcribed and/or translated from the gene or the protein product encoded by the gene. A differentially expressed gene may be overexpressed or underexpressed as compared to the expression level of a normal or control cell. In one aspect, it refers to a differential that is 2.5 times, preferably 5 times, or preferably 10 times higher or lower than the expression level detected in a control sample. The term "differentially expressed" also refers to nucleotide sequences in a cell or tissue which are expressed where silent in a control cell or not expressed where expressed in a control cell. A polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) has a certain percentage (for example, 80%, 85%, 90%, or 95%) of "sequence identity" to another sequence means that, when aligned, that percentage of bases (or amino acids) are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY ((1987) supra Supplement 30, section 7.7.18, Table 7.7.1). In one aspect, default parameters, e.g., are used for alignment. In another aspect, the alignment program is BLAST, using default parameters. BLASTN and BLASTP, using the following default parameters: Genetic code = standard; filter = none; strand = both; cutoff = 60; expect = 10; Matrix = BLOSUM62; Descriptions = 50 sequences; sort by = HIGH SCORE; Databases = non-redundant, GenBank + EMBL + DDBJ + PDB + GenBank CDS translations + SwissProtein + SPupdate + PIR. Details of these programs can be found at the following Internet address: www.ncbi.nlm.nih.gov/cgi-bin/BLAST. "In vivo" gene delivery, gene transfer, gene therapy and the like as used herein, are terms referring to the introduction of a vector comprising an exogenous polynucleotide directly into the body of an organism, such as a human or non-human mammal, whereby the exogenous polynucleotide is introduced to a cell of such organism in vivo. The term "isolated" means separated from constituents, cellular and otherwise, in which the polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, are normally associated with in nature. For example, with respect to a polynucleotide, an isolated polynucleotide is one that is separated from the 5' and 3' sequences with which it is normally associated in the chromosome. As is apparent to those of skill in the art, a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, does not require "isolation" to distinguish it from its naturally occurring counterpart. In addition, a "concentrated", "separated" or "diluted" polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, is distinguishable from its naturally occurring counterpart in that the concentration or number of molecules per volume is greater than "concentrated" or less than "separated" than that of its naturally occurring counterpart. A polynucleotide, peptide, polypeptide, protein, antibody, or fragments thereof, which differs from the naturally occurring counteφart in its primary sequence or for example, by its glycosylation pattern, need not be present in its isolated form since it is distinguishable from its naturally occurring counteφart by its primary sequence, or alternatively, by another characteristic such as glycosylation pattern. Although not explicitly stated for each of the inventions disclosed herein, it is to be understood that all of the above embodiments for each of the compositions disclosed below and under the appropriate conditions, are provided by this invention. Thus, a non-naturally occurring polynucleotide is provided as a separate embodiment from the isolated naturally occurring polynucleotide. A protein produced in a bacterial cell is provided as a separate embodiment from the naturally occurring protein isolated from a eukaryotic cell in which it is produced in nature. "Host cell," "target cell" or "recipient cell" are intended to include any individual cell or cell culture which can be or have been recipients for vectors or the incoφoration of exogenous nucleic acid molecules, polynucleotides and/or proteins. It also is intended to include progeny of a single cell, and the progeny may not necessarily be completely identical (in moφhology or in genomic or total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. The cells may be prokaryotic or eukaryotic, and include but are not limited to bacterial cells, yeast cells, animal cells, and mammalian cells, e.g., murine, rat, simian or human. A "subject" is a vertebrate, in one aspect a mammal, and in another aspect a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. A "control" is an alternative subject or sample used in an experiment for comparison puφose. A control can be "positive" or "negative". For example, where the puφose of the experiment is to determine a correlation of an altered expression level of a gene with a particular type of cancer, generally positive control is used (a subject or a sample from a subject, carrying such alteration and exhibiting syndromes characteristic of that disease), and a negative control (a subject or a sample from a subject lacking the altered expression and clinical syndrome of that disease). The terms "cancer," "neoplasm," and "tumor," used interchangeably and in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism. Primary cancer cells (that is, cells obtained from near the site of malignant transformation) can be readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination. The definition of a cancer cell, as used herein, includes not only a primary cancer cell, but also any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and cell lines derived from cancer cells. When referring to a type of cancer that normally manifests as a solid tumor, a "clinically detectable" tumor is one that is detectable on the basis of tumor mass; e.g., by such procedures as CAT scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation. Biochemical or immunologic findings alone may be insufficient to meet this definition. A neoplasm is an abnormal mass or colony of cells produced by a relatively autonomous new growth of tissue. Most neoplasms arise from the clonal expansion of a single cell that has undergone neoplastic transformation. The transformation of a normal to a neoplastic cell can be caused by a chemical, physical, or biological agent (or event) that directly and irreversibly alters the cell genome. Neoplastic cells are characterized by the loss of some specialized functions and the acquisition of new biological properties, foremost, the property of relatively autonomous (uncontrolled) growth. Neoplastic cells pass on their heritable biological characteristics to progeny cells. The past, present, and future predicted biological behavior, or clinical course, of a neoplasm is further classified as benign or malignant, a distinction of great importance in diagnosis, treatment, and prognosis. A malignant neoplasm manifests a greater degree of autonomy, is capable of invasion and metastatic spread, may be resistant to treatment, and may cause death. A benign neoplasm has a lesser degree of autonomy, is usually not invasive, does not metastasize, and generally produces no great harm if treated adequately. Cancer is a generic term for malignant neoplasms. Anaplasia is a characteristic property of cancer cells and denotes a lack of normal structural and functional characteristics (undifferentiation). A tumor is literally a swelling of any type, such as an inflammatory or other swelling, but modern usage generally denotes a neoplasm. The suffix "-oma" means tumor and usually denotes a benign neoplasm, as in fibroma, lipoma, and so forth, but sometimes implies a malignant neoplasm, as with so-called melanoma, hepatoma, and seminoma, or even a non-neoplastic lesion, such as a hematoma, granuloma, or hamartoma. The suffix "-blastoma" denotes a neoplasm of embryonic cells, such as neuroblastoma of the adrenal or retinoblastoma of the eye. Histogenesis is the origin of a tissue and is a method of classifying neoplasms on the basis of the tissue cell of origin. Adenomas are benign neoplasms of glandular epithelium. Carcinomas are malignant tumors of epithelium. Sarcomas are malignant tumors of mesenchymal tissues. One system to classify neoplasia utilizes biological (clinical) behavior, whether benign or malignant, and the histogenesis, the tissue or cell of origin of the neoplasm as determined by histologic and cytologic examination. Neoplasms may originate in almost any tissue containing cells capable of mitotic division. The histogenetic classification of neoplasms is based upon the tissue (or cell) of origin as determined by histologic and cytologic examination. "Suppressing" tumor growth indicates a growth state that is curtailed compared to growth without contact with educated, antigen-specific immune effector cells described herein. Tumor cell growth can be assessed by any means known in the art, including, but not limited to, measuring tumor size, determining whether tumor cells are proliferating using a 3H-thymidine incoφoration assay, or counting tumor cells. "Suppressing" tumor cell growth means any or all of the following states: slowing, delaying, and "suppressing" tumor growth indicates a growth state that is curtailed when stopping tumor growth, as well as tumor shrinkage. The term "culturing" refers to the in vitro propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (moφhologically, genetically, or phenotypically) to the parent cell. By "expanded" is meant any proliferation or division of cells. A "composition" is intended to mean a combination of active agent and another carrier, e.g., compound or composition, inert (for example, a detectable agent or label) or active, such as an adjuvant, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant or the like. Carriers also include pharmaceutical excipients and additives proteins, peptides, amino acids, lipids, and carbohydrates (e.g., sugars, including monosaccharides, di-, tri-, tetra-, and oligosaccharides; derivatized sugars such as alditols, aldonic acids, esterified sugars and the like; and polysaccharides or sugar polymers), which can be present singly or in combination, comprising alone or in combination 1-99.99% by weight or volume. Exemplary protein excipients include serum albumin such as human seram albumin (HSA), recombinant human albumin (rHA), gelatin, casein, and the like. Representative amino acid/antibody components, which can also function in a buffering capacity, include alanine, glycine, arginine, betaine, histidine, glutamic acid, aspartic acid, cysteine, lysine, leucine, isoleucine, valine, methionine, phenylalanine, aspartame, and the like. Carbohydrate excipients are also intended within the scope of this invention, examples of which include but are not limited to monosaccharides such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffmose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol) and myoinositol. The term carrier further includes a buffer or a pH adjusting agent; typically, the buffer is a salt prepared from an organic acid or base. Representative buffers include organic acid salts such as salts of citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid, or phthalic acid; Tris, tromethamine hydrochloride, or phosphate buffers. Additional carriers include polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl-. quadrature. - cyclodextrin), polyethylene glycols, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antistatic agents, surfactants (e.g., polysorbates such as "TWEEN 20" and "TWEEN 80"), lipids (e.g., phospholipids, fatty acids), steroids (e.g., cholesterol), and chelating agents (e.g., EDTA). As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents. The compositions also can include stabilizers and preservatives and any of the above noted carriers with the additional provisio that they be acceptable for use in vivo. For examples of carriers, stabilizers and adjuvants, see Martin REMINGTON'S PHARM. SCI., 15th Ed. (Mack Publ. Co., Easton (1975) and Williams & Williams, (1995), and in the "PHYSICIAN'S DESK REFERENCE", 52nd ed., Medical Economics, Montvale, NJ. (1998). A "pharmaceutical composition" is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo. An "effective amount" is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages.
POLYPEPTIDES AND PROTEINS The present invention provides novel polypeptide compounds and related compositions for therapeutic, prophylactic and diagnostic use. In a first aspect, this invention provides polypeptides which were heretofore not known to be highly immunogenic. The polypeptides of the invention are useful in therapeutic, prophylactic, and diagnostic applications. Further provided are polynucleotides encoding the peptides of the invention, gene delivery vehicles comprising these polynucleotides and host cells comprising these polynucleotides. In addition, the invention provides methods for inducing an immune response in a subject by delivering to the subject an effective amount of the peptides of the invention in the context of an MHC molecule. The peptides of the invention are also useful as immunogens to generate antibodies that specifically recognize and bind to these peptides and cross-react with related peptides. The peptides of the invention can also be used in recombinant methods to construct antibodies that specifically recognize and bind to these peptides. The antibodies so generated are further useful in therapeutic and diagnostic applications. The invention further provides immune effector cells raised in vivo or in vitro in the presence and at the expense of an antigen presenting cell (APC) that presents the peptides of the invention in the context of an MHC molecule and a method of adoptive immunotherapy comprising administering an effective amount of these immune effector cells to a subject. This invention further provides compositions which are useful as components of anti-cancer vaccines and to expand immune effector cells attack and lyse cancer cells. In one embodiment, compositions of the invention comprise at least one, peptide of the invention. In one aspect, such compositions may comprise at least one, or alternatively, two or more copies of a single peptide. In another aspect, such compositions may comprise two or more peptides, wherein each peptide of said two or more peptides is distinct from all other peptides in the composition. In one embodiment, the two or more immunogenic peptides are covalently linked. Further provided by this invention are multimers (concatemers) of a peptide of the invention, optionally including intervening amino acid sequences. The proteins and polypeptides of this invention can be obtained by chemical synthesis using a commercially available automated peptide synthesizer such as those manufactured by Perkin Elmer/Applied Biosystems, Inc., Model 430A or 431 A, Foster City, CA, USA. The synthesized protein or polypeptide can be precipitated and further purified, for example by high performance liquid chromatography (HPLC). Accordingly, this invention also provides a process for chemically synthesizing the proteins of this invention by providing the sequence of the protein and reagents, such as amino acids and enzymes and linking together the amino acids in the proper orientation and linear sequence. Alternatively, the proteins and polypeptides can be obtained by recombinant methods known in the art using the host cell and vector systems described below.
PEPTIDE ANALOGUES It is well know to those skilled in the art that the peptides of the invention may be prepared as peptide analogues. As used herein the term "amino acid" refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics. A peptide of three or more amino acids is commonly called an oligopeptide if the peptide chain is short. If the peptide chain is long, the peptide is commonly called a polypeptide or a protein. Peptides of the invention can include unnatural amino acids. Thus, the peptides may comprise D-amino acids, a combination of D- and L-amino acids, and various "designer" amino acids (e.g., β-methyl amino acids, C-α-methyl amino acids, and N-α-methyl amino acids, etc.) to convey special properties to peptides. Additionally, by assigning specific amino acids at specific coupling steps in the synthesis, peptides with α-helices β turns, β sheets, γ-turns, and cyclic peptides can be generated. Generally, it is believed that α-helical secondary structure or random secondary structure is preferred. hi a further embodiment, subunits of peptides that confer useful chemical and structural properties will be used. For example, peptides comprising D-amino acids will be resistant to L-amino acid-specific proteases in vivo. Modified compounds with D-amino acids may be synthesized with the amino acids aligned in reverse order to produce the peptides of the invention as retro-inverso peptides. In addition, the present invention envisions preparing peptides that have better defined structural properties, and the use of peptidomimetics, and peptidomimetic bonds, such as ester bonds, to prepare peptides with novel properties. In another embodiment, a peptide may be generated that incorporates a reduced peptide bond, i. e. , Ri -CH2NH-R2, where Ri, and R2 are amino acid residues or sequences. A reduced peptide bond may be introduced as a dipeptide subunit. Such a molecule would be resistant to peptide bond hydrolysis, e.g., protease activity. Such molecules would provide ligands with unique function and activity, such as extended half-lives in vivo due to resistance to metabolic breakdown, or protease activity.
NON-CLASSICAL AMINO ACIDS THAT INDUCE CONFORMATIONAL CONSTRAINTS. The following non classical amino acids may be incoφorated in the peptides of the invention in order to introduce particular conformational motifs: l,2,3,4-tetrahydroisoquinoline-3-carboxylate (Kazamierski et al. (1991) J. Am. Chem. Soc. 113:2275-2283); (2S,3S)-methyl-ρhenylalanine, (2S,3R)- methyl- phenylalanine, (2R,3S)-methyl-phenylalanine and (2R,3R)-methyl-phenylalanine (Kazmierski and Hraby (1991) Tetrahedron Lett. 32(41): 5769-5772); 2- aminotetrahydronaphthalene-2- carboxylic acid (Landis (1989) Ph.D. Thesis, University of Arizona); hydroxy- 1, 2,3, 4-tetrahydroisoquinoline-3-carboxylate (Miyake et al. (1984) J. Takeda Res. Labs. 43:53-76) histidine isoquinoline carboxylic acid (Zechel et al. (1991) Int. J. Pep. Protein Res. 38(2):131-138); and HIC (histidine cyclic urea), (Dharanipragada et al. (1993) Int. J. Pep. Protein Res. 42(l):68-77) and ((1992) Acta. Cryst, Crystal Struc. Comm. 48(IV): 1239-1241). The following amino acid analogs and peptidomimetics may be incoφorated into a peptide to induce or favor specific secondary structures: LL-Acp (LL-3-amino- 2-ρropenidone-6-carboxylic acid), a β-turn inducing dipeptide analog (Kemp et al. (1985) J. Org. Chem. 50:5834-5838); β-sheet inducing analogs (Kemp et al. (1988) Tetrahedron Lett. 29:5081-5082); β-turn inducing analogs (Kemp et al. (1988) Tetrahedron Lett. 29:5057-5060); α-helix inducing analogs (Kemp et al. (1988) Tetrahedron Lett. 29:4935-4938); γ-turn inducing analogs (Kemp et al. (1989) J. Org. Chem. 54:109:115); analogs provided by the following references: Nagai and Sato (1985) Tetrahedron Lett. 26:647-650; and DiMaio et al. (1989) J. Chem. Soc. Perkin Trans, p. 1687; a Gly-Ala turn analog (Kahn et al. (1989) Tetrahedron Lett. 30:2317); amide bond isostere (Jones et al. (1988) Tetrahedron Lett. 29:5875-5880); tretrazol (Zabrocki et al. (1988) J. Am. Chem. Soc. 110:587S-5880); DTC (Samanen et al.
(1990) Int. J. Protein Pep. Res. 35:501 :509); and analogs taught in Olson et al. (1990) J. Am. Chem. Sci. 112:323-333 and Garvey et al. (1990) J. Org. Chem. 55(3):936- 940. Conformationally restricted mimetics of beta turns and beta bulges, and peptides containing them, are described in U.S. Patent No. 5,440,013, issued August 8, 1995 to Kahn. A peptide of the invention can be used in a variety of formulations, which may vary depending on the intended use. A peptide of the invention can be covalently or non-covalently linked (complexed) to various other molecules, the nature of which may vary depending on the particular puφose. For example, a peptide of the invention can be covalently or non-covalently complexed to a macromolecular carrier, including, but not limited to, natural and synthetic polymers, proteins, polysaccharides, polypeptides (amino acids), polyvinyl alcohol, polyvinyl pyrrolidone, and lipids. A peptide can be conjugated to a fatty acid, for introduction into a liposome. (See, U.S. Patent No. 5,837,249). A peptide of the invention can be complexed covalently or non-covalently with a solid support, a variety of which are known in the art. A peptide of the invention can be associated with an antigen-presenting matrix with or without co-stimulatory molecules, as described in more detail below. Examples of protein carriers include, but are not limited to, superantigens, serum albumin, tetanus toxoid, ovalbumin, thyroglobulin, myoglobulin, and immunoglobulin. Peptide-protein carrier polymers may be formed using conventional cross-linking agents such as carbodimides. Examples of carbodimides are l-cyclohexyl-3-(2-moφholinyl-(4-ethyl) carbodiimide (CMC), l-ethyl-3-(3-dimethyaminopropyl) carbodiimide (EDC) and 1 -ethyl-3 -(4-azonia-44-dimethylpentyl) carbodiimide. Examples of other suitable cross-linking agents are cyanogen bromide, glutaraldehyde and succinic anhydride. In general, any of a number of homo-bifunctional agents including a homo-bifunctional aldehyde, a homo-bifunctional epoxide, a homo-bifunctional imido-ester, a homo-bifunctional N-hydroxysuccinimide ester, a homo-bifunctional maleimide, a homo-bifunctional alkyl halide, a homo-bifunctional pyridyl disulfide, a homo-bifunctional aryl halide, a homo-bifunctional hydrazide, a homo-bifunctional diazonium derivative and a homo-bifunctional photoreactive compound may be used. Also included are hetero-bifunctional compounds, for example, compounds having an amine-reactive and a sulfhydryl-reactive group, compounds with an amine-reactive and a photoreactive group and compounds with a carbonyl-reactive and a sulfhydryl-reactive group. Specific examples of such homo-bifunctional cross-linking agents include the bifunctional N-hydroxysuccinimide esters dithiobis(succinimidyιpropionate), disuccinimidyl suberate, and disuccinimidyl tartarate; the bifunctional imido-esters dimethyl adipimidate, dimethyl pimelimidate, and dimethyl suberimidate; the bifunctional sulfhydryl-reactive crosslinkers l,4-di-[3'-(2'-pyridyldithio) propion-amido]butane, bismaleimidohexane, and bis-N-maleimido-1, 8-octane; the bifunctional aryl halides l,5-difluoro-2,4-dinitrobenzene and
4,4'-difluoro-3,3'-dinitrophenylsulfone; bifunctional photoreactive agents such as bis-[b-(4-azidosalicylamido)ethyl]disulfide; the bifunctional aldehydes formaldehyde, malondialdehyde, succinaldehyde, glutaraldehyde, and adipaldehyde; a bifunctional epoxide such as 1,4-butaneodiol diglycidyl ether; the bifunctional hydrazides adipic acid dihydrazide, carbohydrazide, and succinic acid dihydrazide; the bifunctional diazoniums o-tolidine, diazotized and bis-diazotized benzidine; the bifunctional alkylhalides NlN'-ethylene-bis(iodoacetamide), NlN'-hexamethylene-bis(iodoacetamide),
NlN'-undecamethylene-bis(iodoacetamide), as well as benzylhalides and halomustards, such as ala'-diiodo-p-xylene sulfonic acid and tri(2-chloroethyl)amine, respectively. Examples of common hetero-bifunctional cross-linking agents that may be used to effect the conjugation of proteins to peptides include, but are not limited to, SMCC (succinimidyl-4-(N-maleimidomethyl)cyclohexane- 1 -carboxylate), MBS (m-maleimidobenzoyl-N-hydroxysuccinimide ester), SLAB (N-succinimidyl(4-iodoacteyl)aminobenzoate), SMPB (succinimidyl-4-(p-maleimidophenyl)butyrate), GMBS (N-(γ-maleimidobutyryloxy)succinimide ester), MPBH (4-(4-N-maleimidopohenyl) butyric acid hydrazide), M2C2H (4-(N-maleimidomethyl) cyclohexane-1-carboxyl-hydrazide), SMPT (succinimidyloxycarbonyl-a-methyl-a-(2-pyridyldithio)toluene), and SPDP (N-succinimidyl 3-(2-pyridyldithio)propionate). Cross-linking may be accomplished by coupling a carbonyl group to an amine group or to a hydrazide group by reductive animation. Peptides of the invention also may be formulated as non-covalent attachment of monomers through ionic, adsoφtive, or biospecific interactions. Complexes of peptides with highly positively o negatively charged molecules may be done through salt bridge formation under low ionic strength environments, such as in deionized water. Large complexes can be created using charged polymers such as poly-(L-glutamic acid) or poly-(L-lysine) which contain numerous negative and positive charges, respectively. Adsoφtion of peptides may be done to surfaces such as microparticle latex beads or to other hydrophobic polymers, forming non-covalently associated peptide-superantigen complexes effectively mimicking cross-linked or chemically polymerized protein. Finally, peptides may be non-covalently linked through the use of biospecific interactions between other molecules. For instance, utilization of the strong affinity of biotin for proteins such as avidin or streptavidin or their derivatives could be used to form peptide complexes. These biotin-binding proteins contain four binding sites that can interact with biotin in solution or be covalently attached to another molecule. (Wilchek (1988) Anal. Biochem. 171:1-32). Peptides can be modified to possess biotin groups using common biotinylation reagents such as the N-hydroxysuccinimidyl ester of D-biotin (NHS-biotin) which reacts with available amine groups on the protein. Biotinylated peptides then can be incubated with avidin or streptavidin to create large complexes. The molecular mass of such polymers can be regulated through careful control of the molar ratio of biotinylated peptide to avidin or streptavidin. Also provided by this application are the peptides and polypeptides described herein conjugated to a detectable agent for use in the diagnostic methods. For example, detectably labeled peptides and polypeptides can be bound to a column and used for the detection and purification of antibodies. They also are useful as immunogens for the production of antibodies, as described below. The peptides of this invention also can be combined with various liquid phase carriers, such as sterile or aqueous solutions, pharmaceutically acceptable carriers, suspensions and emulsions. Examples of non-aqueous solvents include propyl ethylene glycol, polyethylene glycol and vegetable oils. When used to prepare antibodies, the carriers also can include an adjuvant that is useful to non-specifically augment a specific immune response. A skilled artisan can easily determine whether an adjuvant is required and select one. However, for the puφose of illustration only, suitable adjuvants include, but are not limited to, Freund's Complete and Incomplete, mineral salts and polynucleotides. POLYNUCLEOTIDES This invention further provides polynucleotides encoding polypeptides comprising one or more of the sequences of shown in Table 1, and the complements of these polynucleotides. A "variant polynucleotide" means alternate polynucleotides sequence's to those shown in Table 1 (due to the degeneracy of the genetic code) that code for the same polypeptide. As used herein, the term "polynucleotide" encompasses DNA, RNA and nucleic acid mimetics. In addition to these polynucleotides, or their complements, this invention also provides the anti-sense polynucleotide stand, e.g. antisense RNA to the sequences or their complements. One can obtain an antisense RNA using the sequences provided in Table 1 a reverse translation computer program, and the methodology described in Van der Krol, et al. (1988) BioTechniquas 6:958. The polynucleotides of this invention can be replicated using PCR. PCR technology is the subject matter of United States Patent Nos. 4,683,195; 4,800,159; 4,754,065; and 4,683,202 and described in PCR: THE POLYMERASE CHAIN REACTION (Mullis, et al. eds, Birkhauser Press, Boston (1994) and references cited therein). Alternatively, one of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to replicate the DNA. Accordingly, this invention also provides a process for obtaining the polynucleotides of this invention by providing the linear sequence of the polynucleotide, appropriate primer molecules, chemicals such as enzymes and instructions for their replication and chemically replicating or linking the nucleotides in the proper orientation to obtain the polynucleotides. In a separate embodiment, these polynucleotides are further isolated. Still further, one of skill in the art can insert the polynucleotide into a suitable replication vector and insert the vector into a suitable host cell (prokaryotic or eukaryotic) for replication and amplification. The DNA so amplified can be isolated from the cell by methods known to those of skill in the art. A process for obtaining polynucleotides by this method is further provided herein as well as the polynucleotides so obtained. RNA can be obtained by first inserting a DNA polynucleotide into a suitable host cell. The DNA can be inserted by any appropriate method, e.g., by the use of an appropriate gene delivery vehicle (e.g., liposome, plasmid or vector) or by electroporation. When the cell replicates and the DNA is transcribed into RNA; the RNA can then be isolated using methods known to those of skill in the art, for example, as set forth in Sambrook et al. (1989) supra. For instance, mRNA can be isolated using various lytic enzymes or chemical solutions according to the procedures set forth in Sambrook, et al. (1989) supra or extracted by nucleic-acid-binding resins following the accompanying instructions provided by manufactures. Polynucleotides having at least 4 contiguous nucleotides, and more preferably at least 5 or 6 contiguous nucleotides and most preferably at least 10 contiguous nucleotides, and exhibiting sequence complementarity or homology to the polynucleotides encoding the peptides shown in Table 1 , find utility as hybridization probes. It is known in the art that a "perfectly matched" probe is not needed for a specific hybridization. Minor changes in probe sequence achieved by substitution, deletion or insertion of a small number of bases do not affect the hybridization specificity. In general, as much as 20% base-pair mismatch (when optimally aligned) can be tolerated. Preferably, a probe useful for detecting the aforementioned mRNA is at least about 80% identical to the homologous region of comparable size contained in the previously identified sequences which correspond to previously characterized genes. More preferably, the probe is 85% identical to the corresponding gene sequence after alignment of the homologous region; even more preferably, it exhibits 90% identity. These probes can be used in radioassays (e.g. Southern and Northern blot analysis) to detect or monitor various cells or tissue containing these cells. The probes also can be attached to a solid support or an array such as a chip for use in high throughput screening assays for the detection of expression of the gene corresponding to one or more polynucleotide(s) of this invention. Accordingly, this invention also provides at least one probe as defined above and/or the complement of one of these sequences, attached to a solid support for use in high throughput screens. The polynucleotides of the present invention also can serve as primers for the detection of genes or gene transcripts that are expressed in APC, for example, to confirm transduction of the polynucleotides into host cells. In this context, amplification means any method employing a primer-dependent polymerase capable of replicating a target sequence with reasonable fidelity. Amplification may be carried out by natural or recombinant DNA-polymerases such as T7 DNA polymerase, Klenow fragment of E. coli DNA polymerase, and reverse transcriptase. A preferred length of the primer is the same as that identified for probes, above. The invention further provides the isolated polynucleotide operatively linked to a promoter of RNA transcription, as well as other regulatory sequences for replication and/or transient or stable expression of the DNA or RNA. As used herein, the term "operatively linked" means positioned in such a manner that the promoter will direct transcription of RNA off the DNA molecule. Examples of such promoters are SP6, T4 and T7. In certain embodiments, cell-specific promoters are used for cell-specific expression of the inserted polynucleotide. Vectors which contain a promoter or a promoter/enhancer, with termination codons and selectable marker sequences, as well as a cloning site into which an inserted piece of DNA can be operatively linked to that promoter are known in the art and commercially available. For general methodology and cloning strategies, see GENE EXPRESSION TECHNOLOGY (Goeddel ed., Academic Press, Inc. (1991)) and references cited therein and VECTORS: ESSENTIAL DATA SERIES (Gacesa and Ramji, eds., John Wiley & Sons, N.Y. (1994)), which contains maps, functional properties, commercial suppliers and a reference to GenEMBL accession numbers for various suitable vectors. In one aspect, these vectors are capable of transcribing RNA in vitro or in vivo. Expression vectors containing these nucleic acids are useful to obtain host vector systems to produce proteins and polypeptides. It is implied that these expression vectors must be replicable in the host organisms either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include plasmids, viral vectors, including adenovirases, adeno-associated viruses, retroviruses, cosmids, etc. Adenoviral vectors are useful for introducing genes into tissues in vivo because of their high levels of expression and efficient transformation of cells both in vitro and in vivo. When a nucleic acid is inserted into a suitable host cell, e.g., a prokaryotic or a eukaryotic cell and the host cell replicates, the protein can be recombinantly produced. Suitable host cells will depend on the vector and can include mammalian cells, animal cells, human cells, simian cells, insect cells, yeast cells, and bacterial cells constructed using known methods. See Sambrook, et al. (1989) supra. In addition to the use of viral vector for insertion of exogenous nucleic acid into cells, the nucleic acid can be inserted into the host cell by methods known in the art such as transformation for bacterial cells; transfection using calcium phosphate precipitation for mammalian cells; DEAE-dextran; electroporation; or microinjection. See Sambrook, et al. (1989) supra for this methodology. Thus, this invention also provides a host cell, e.g. a mammalian cell, an animal cell (rat or mouse), a human cell, or a prokaryotic cell such as a bacterial cell, containing a polynucleotide encoding a protein or polypeptide or antibody. The present invention also provides delivery vehicles suitable for delivery of a polynucleotide of the invention into cells (whether in vivo, ex vivo, or in vitro). A polynucleotide of the invention can be contained within a cloning or expression vector. These vectors (especially expression vectors) can in turn be manipulated to assume any of a number of forms which may, for example, facilitate delivery to and/or entry into a cell. When the vectors are used for gene therapy in vivo or ex vivo, a pharmaceutically acceptable vector is preferred, such as a replication-incompetent retroviral or adenoviral vector. Pharmaceutically acceptable vectors containing the nucleic acids of this invention can be further modified for transient or stable expression of the inserted polynucleotide. As used herein, the term "pharmaceutically acceptable vector" includes, but is not limited to, a vector or delivery vehicle having the ability to selectively target and introduce the nucleic acid into dividing cells. An example of such a vector is a "replication-incompetent" vector defined by its inability to produce viral proteins, precluding spread of the vector in the infected host cell. An example of a replication-incompetent retroviral vector is LNL6 (Miller, A.D. et al. (1989) BioTechniques 7:980-990). The methodology of using replication-incompetent retroviruses for retroviral-mediated gene transfer of gene markers has been established. (Correll, et al. (1989) Proc. Natl. Acad. Sci. USA 86:8912; Bordignon, (1989) Proc. Natl. Acad. Sci. USA 86:8912-52; Culver, K. (1991) Proc. Natl. Acad. Sci. USA 88:3155; and Rill, D.R. (1991) Blood 79(10):2694-2700). These isolated host cells containing the polynucleotides of this invention are useful for the recombinant replication of the polynucleotides and for the recombinant production of peptides. Alternatively, the cells may be used to induce an immune response in a subject in the methods described herein. When the host cells are antigen presenting cells, they can be used to expand a population of immune effector cells such as tumor infiltrating lymphocytes which in turn are useful in adoptive immunotherapies. ANTIBODIES Also provided by this invention is an antibody capable of specifically forming a complex with the polypeptides of this invention. The term "antibody" includes polyclonai antibodies and monoclonal antibodies. The antibodies include, but are not limited to mouse, rat, and rabbit or human antibodies. The antibodies are useful to identify and purify polypeptides and APCs expressing the polypeptides. Laboratory methods for producing polyclonai antibodies and monoclonal antibodies, as well as deducing their corresponding nucleic acid sequences, are known in the art, see Hariow and Lane (1988) and (1999) supra and Sambrook, et al. (1989) supra. The monoclonal antibodies of this invention can be biologically produced by introducing protein or a fragment thereof into an animal, e.g., a mouse or a rabbit. The antibody producing cells in the animal are isolated and fused with myeloma cells or hetero-myeloma cells to produce hybrid cells or hybridomas. Accordingly, the hybridoma cells producing the monoclonal antibodies of this invention also are provided. Thus, using the protein or fragment thereof, and art-recognized methods, one of skill in the art can produce and screen the hybridoma cells and antibodies of this invention for antibodies having the ability to bind the proteins or polypeptides. If a monoclonal antibody being tested binds with the protein or polypeptide, then the antibody being tested and the antibodies provided by the hybridomas of this invention are equivalent. It also is possible to determine without undue experimentation, whether an antibody has the same specificity as the monoclonal antibody of this invention by determining whether the antibody being tested prevents a monoclonal antibody of this invention from binding the protein or polypeptide with which the monoclonal antibody is normally reactive. If the antibody being tested competes with the monoclonal antibody of the invention as shown by a decrease in binding by the monoclonal antibody of this invention, then it is likely that the two antibodies bind to the same or a closely related epitope. Alternatively, one can pre-incubate the monoclonal antibody of this invention with a protein with which it is normally reactive, and determine if the monoclonal antibody being tested is inhibited in its ability to bind the antigen. If the monoclonal antibody being tested is inhibited then, in all likelihood, it has the same, or a closely related, epitopic specificity as the monoclonal antibody of this invention. The term "antibody" also is intended to include antibodies of all isotypes. Particular isotypes of a monoclonal antibody can be prepared either directly by selecting from the initial fusion, or prepared secondarily, from a parental hybridoma secreting a monoclonal antibody of different isotype by using the sib selection technique to isolate class switch variants using the procedure described in Steplewski, et al. (1985) Proc. Natl. Acad. Sci. USA 82:8653 or Spira, et al. (1984J J. Immunol. Meth. 74:307. This invention also provides biological active fragments of the polyclonai and monoclonal antibodies described above. These "antibody fragments" retain some ability to selectively bind with its antigen or immunogen. Such antibody fragments can include, but are not limited to: (1) Fab, (2) Fab', (3) F(ab')2, (4) Fv, and (5) SCA The antibodies of this invention are monoclonal antibodies, although in certain aspects, polyclonai antibodies can be utilized. They also can be immunogenic and functional fragments, antibody derivatives or antibody variants. They can be chimeric, humanized, or totally human. A functional fragment of an antibody includes but is not limited to Fab, Fab', Fab2, Fab'2, and single chain variable regions. Antibodies can be produced in cell culture, in phage, or in various animals, including but not limited to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats, monkeys, chimpanzees, apes, etc. So long as the fragment or derivative retains specificity of binding as the antibodies of this invention it can be used. Antibodies can be tested for specificity of binding by comparing binding to appropriate antigen to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody binds to the appropriate antigen at least 2, 5, 7, and preferably 10 times more than to irrelevant antigen or antigen mixture then it is considered to be specific. Specific assays for determining specificity are described infra. The monoclonal antibodies of the invention can be generated using conventional hybridoma techniques known in the art and well-described in the literature. For example, a hybridoma is produced by fusing a suitable immortal cell line (e.g., a myeloma cell line such as, but not limited to, Sp2/0, Sp2/0-AG14, NSO, NS1, NS2, AE-1, L.5, >243, P3X63Ag8.653, Sp2 SA3, Sp2 MAI, Sp2 SSI, Sp2 SA5, U397, MLA 144, ACT IV, MOLT4, DA-1, JURKAT, WEHI, K-562, COS, RAJI, NIH 3T3, HL-60, MLA 144, NAMAIWA, NEURO 2A, CHO, PerC.6, YB2/O) or the like, or heteromyelomas, fusion products thereof, or any cell or fusion cell derived therefrom, or any other suitable cell line as known in the art (see, e.g., www.atcc.org, www.lifetech.com., and the like), with antibody producing cells, such as, but not limited to, isolated or cloned spleen, peripheral blood, lymph, tonsil, or other immune or B cell containing cells, or any other cells expressing heavy or light chain constant or variable or framework or CDR sequences, either as endogenous or heterologous nucleic acid, as recombinant or endogenous, viral, bacterial, algal, prokaryotic, amphibian, insect, reptilian, fish, mammalian, rodent, equine; ovine, goat, sheep, primate, eukaryotic, genomic DNA, cDNA, rDNA, mitochondrial DNA or RNA, chloroplast DNA or RNA, hnRNA, mRNA, tRNA, single, double or triple stranded, hybridized, and the like or any combination thereof. Antibody producing cells can also be obtained from the peripheral blood or, preferably the spleen or lymph nodes, of humans or other suitable animals that have been immunized with the antigen of interest. Any other suitable host cell can also be used for expressing-heterologous or endogenous nucleic acid encoding an antibody, specified fragment or variant thereof, of the present invention. The fused cells (hybridomas) or recombinant cells can be isolated using selective culture conditions or other suitable known methods, and cloned by limiting dilution or cell sorting, or other known methods. Other suitable methods of producing or isolating antibodies of the requisite specificity can be used, including, but not limited to, methods that select recombinant antibody from a peptide or protein library (e.g. , but not limited to, a bacteriophage, ribosome, oligonucleotide, RNA, cDNA, or the like, display library; e.g., as available from various commercial vendors such as Cambridge Antibody Technologies (Cambridgeshire, UK), MoφhoSys (Martinsreid/Planegg, Del), Biovation (Aberdeen, Scotland, UK) Biolnvent (Lund, Sweden), using methods known in the art. (See U.S. Pat. Nos. 4,704,692; 5,723,323; 5,763,192; 5,814,476; 5,817,483; 5,824,514; 5,976,862). Alternative methods rely upon immunization of transgenic animals (e.g., SCID mice, Nguyen et al. (1977) Microbiol. Immunol. 41:901-907 (1997); Sandhu et al., (1996) Crit. Rev. Biotechnol. 16:95-118; Eren et al. (1998) Immunol. 93:154-161) that are capable of producing a repertoire of human antibodies, as known in the art and/or as described herein. Such techniques, include, but are not limited to, ribosome display (Hanes et al. (1997) Proc. Natl. Acad. Sci. USA, 94:4937-4942; Hanes et al., (1998) Proc. Natl. Acad. Sci. USA, 95:14130-14135); single cell antibody producing technologies (e.g., selected lymphocyte antibody method ("SLAM") (U.S. Pat. No. 5,627,052, Wen et al. (1987) J. Immunol. 17:887-892; Babcook et al., Proc. Natl. Acad. Sci. USA (1996) 93:7843-7848); gel microdroplet and flow cytometry (Powell et al. (1990) Biotechnol. 8:333-337; One Cell Systems, (Cambridge, Mass).; Gray et al. (1995) J. Imm. Meth. 182: 155-163; Kenny et al. (1995) Bio/Technol. 13:787-790); B-cell selection (Steenbakkers et al. (1994) Molec. Biol. Reports 19:125-134 (1994). Antibody variants of the present invention can also be prepared using delivering a polynucleotide encoding an antibody of this invention to a suitable host such as to provide transgenic animals or mammals, such as goats, cows, horses, sheep, and the like, that produce such antibodies in their milk. These methods are known in the art and are described for example in U.S. Pat. Nos. 5,827,690; 5,849,992; 4,873,316; 5,849,992; 5,994,616; 5,565,362; and 5,304,489. The term "antibody variant" includes post-translational modification to linear polypeptide sequence of the antibody or fragment. For example, U.S. Patent No. 6,602,684 Bl describes a method for the generation of modified glycol-forms of antibodies, including whole antibody molecules, antibody fragments, or fusion proteins that include a region equivalent to the Fc region of an immunoglobulin, having enhanced Fc-mediated cellular toxicity, and glycoproteins so generated. Antibody variants also can be prepared by delivering a polynucleotide of this invention to provide transgenic plants and cultured plant cells (e.g., but not limited to tobacco, maize, and duckweed) that produce such antibodies, specified portions or variants in the plant parts or in cells cultured therefrom. For example, Cramer et al. (1999) Curr. Top. Microbol. Immunol. 240:95-118 and references cited therein, describe the production of transgenic tobacco leaves expressing large amounts of recombinant proteins, e.g., using an inducible promoter. Transgenic maize have been used to express mammalian proteins at commercial production levels, with biological activities equivalent to those produced in other recombinant systems or purified from natural sources. (See, e.g., Hood et al., Adv. Exp. Med. Biol. (1999) 464:127-147 and references cited therein). Antibody variants have also been produced in large amounts from transgenic plant seeds including antibody fragments, such as single chain antibodies (scFv's), including tobacco seeds and potato tubers. (See, e.g., Conrad et al.(1998) Plant Mol. Biol. 38:101- 109 and reference cited therein.) Thus, antibodies of the present invention can also be produced using transgenic plants, according to know methods. Antibody derivatives can be produced, for example, by adding exogenous sequences to modify immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic. Generally part or all of the non-human or human CDR sequences are maintained while the non-human sequences of the variable and constant regions are replaced with human or other amino acids. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Humanization or engineering of antibodies of the present invention can be performed using any known method, such as but not limited to those described in U.S. Pat. Nos. 5,723,323, 5,976,862, 5,824,514, 5,817,483, 5,814,476, 5,763,192, 5,723,323, 5,766,886, 5,714,352, 6,204,023, 6,180,370, 5,693,762, 5,530,101, 5,585,089, 5,225,539; and 4,816,567. Techniques for making partially to fully human antibodies are known in the art and any such techniques can be used. According to one embodiment, fully human antibody sequences are made in a transgenic mouse which has been engineered to express human heavy and light chain antibody genes. Multiple strains of such transgenic mice have been made which can produce different classes of antibodies. B cells from transgenic mice which are producing a desirable antibody can be fused to make hybridoma cell lines for continuous production of the desired antibody. (See Russel, N.D. et al. (2000) Infection and Immunity April 2000:1820-1826; Gallo, M. L. et al. (2000) European J. of Immun. 30:534-540; Green, L. L. (1999) J. of Immun. Methods 231:11-23; Yang, X-D et al. (1999A) J. of Leukocyte Biology 66:401-410; Yang, X-D (1999B) Cancer Research 59(6): 1236-1243; Jakobovits, A. (1998) Advanced Drug Delivery
Reviews 31:33-42; Green, L. and Jakobovits, A. (1998) J. Exp. Med. 188(3):483- 495; Jakobovits, A. (1998) Exp. Opin. Invest. Drugs 7(4):607-614; Tsuda, H. et al. (1997) Genomics 42:413-421; Sherman-Gold, R. (1997). Genetic Engineering News 17(14); Mendez, M. et al. (1997) Nature Genetics 15:146-156; Jakobovits, A. (1996) WEIR'S HANDBOOK OF EXPERIMENTAL IMMUNOLOGY, THE INTEGRATED IMMUNE SYSTEM VOL. IV, 194.1-194.7; Jakobovits, A. (1995) Current Opinion in Biotechnology 6:561-566; Mendez, M. et al. (1995) Genomics 26:294-307; Jakobovits, A. (1994) Current Biology 4(8):761-763; Arbones, M. et al. (1994) Immunity l(4):247-260; Jakobovits, A. (1993) Nature 362(6417):255-258; Jakobovits, A. et al. (1993) Proc. Natl. Acad. Sci. USA 90(6):2551-2555; Kucherlapati, et al. U.S. Patent No. 6,075,181.) Human monoclonal antibodies can also be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a numan heavy chain transgene and a light chain transgene fused to an immortalized cell. The antibodies of this invention also can be modified to create chimeric antibodies. Chimeric antibodies are those in which the various domains of the antibodies' heavy and light chains are coded for by DNA from more than one species. (See, e.g., U.S. Patent No.: 4,816,567.) The term "antibody derivative" also includes "diabodies" which are small antibody fragments with two antigen-binding sites, wherein fragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the same polypeptide chain (VH VL). (See EP 404,097; WO 93/11161; and Hollinger et al., (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448.) By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. (See also, U.S. Patent No. 6,632,926 to Chen et al. which discloses antibody variants that have one or more amino acids inserted into a hypervariable region of the parent antibody and a binding affinity for a target antigen which is at least about two fold stronger than the binding affinity of the parent antibody for the antigen.) The term "antibody derivative" further includes "linear antibodies". The procedure for making the is known in the art and described in Zapata et al. (1995) Protein Eng. 8(10):1057-1062. Briefly, these antibodies comprise a pair of tandem Fd segments (VH -CH 1-VH -CH1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific. The antibodies of this invention can be recovered and purified from recombinant cell cultures by known methods including, but not limited to, protein A purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography ("HPLC") can also be used for purification. Antibodies of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells, or alternatively from a prokaryotic cells as described above. In some aspects of this invention, it will be useful to detectably or therapeutically label the antibody. Methods for conjugating antibodies to these agents are l iown in the art. For the puφose of illustration only, antibodies can be labeled with a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like. Such labeled antibodies can be used for diagnostic techniques, either in vivo, or in an isolated test sample. Antibodies can also be conjugated, for example, to a pharmaceutical agent, such as chemotherapeutic drag or a toxin. They can be linked to a cytokine, to a ligand, to another antibody. Suitable agents for coupling to antibodies to achieve an anti-tumor effect include cytokines, such as interleukin 2 (IL-2) and Tumor Necrosis Factor (TNF); photosensitizers, for use in photodynamic therapy, including aluminum (III) phthalocyanine tetrasulfonate, hematopoφhyrin, and phthalocyanine; radionuclides, such as iodine-131 (131I), yttrium-90 (90Y), bismuth-212 (212Bi), bismuth-213 (213Bi), technetium-99m (99mTc), rhenium-186 (186Re), and rhenium- 188 (188Re); antibiotics, such as doxorabicin, adriamycin, daunorubicin, methotrexate, daunomycin, neocarzinostatin, and carboplatin; bacterial, plant, and other toxins, such as diphtheria toxin, pseudomonas exotoxin A, staphylococcal enterotoxin A, abrin-A toxin, ricin A (deglycosylated ricin A and native ricin A), TGF-alpha toxin, cytotoxin from Chinese cobra (naja naja atra), and gelonin (a plant toxin); ribosome inactivating proteins from plants, bacteria and fungi, such as restrictocin (a ribosome inactivating protein produced by Aspergillus restrictus), saporin (a ribosome inactivating protein from Saponaria officinalis), and RNase; tyrosine kinase inhibitors; ly207702 (a difluorinated purine nucleoside); liposomes containing anti cystic agents (e.g., antisense oligonucleotides, plasmids which encode for toxins, methotrexate, etc.); and other antibodies or antibody fragments, such as F(ab). With respect to preparations containing antibodies covalently linked to organic molecules, they can be prepared using suitable methods, such as by reaction with one or more modifying agents. Examples of such include modifying and activating groups. A "modifying agent" as the term is used herein, refers to a suitable organic group (e.g., hydrophilic polymer, a fatty acid, a fatty acid ester) that comprises an activating group. Specific examples of these are provided supra. An "activating group" is a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group. Examples of such are electrophilic groups such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl esters (NHS), and the like. Activating groups that can react with thiols include, for example, maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB- thiol), and the like. An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form phosphoramidate or phosphorimide linkages. Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., BIOCONJUGATE TECHNIQUES, Academic Press: San Diego, Calif. (1996)). An activating group can be bonded directly to the organic group (e.g., hydrophilic polymer, fatty acid, fatty acid ester), or through a linker moiety, for example a divalent -C^ group wherein one or more carbon atoms can be replaced by a heteroatom such as oxygen, nitrogen or sulfur. Suitable linker moieties include, for example, tetraethylene glycol. Modifying agents that comprise a linker moiety can be produced, for example, by reacting a mono-Boc-alkyldiamine (e.g., mono-Boc-ethylenediamine, mono-Boc- diaminohexane) with a fatty acid in the presence of l-ethyl-3-(3- dimethylaminoproρyl)carbodiimide (EDC) to form an amide bond between the free amine and the fatty acid carboxylate. The Boc protecting group can be removed from the product by treatment with trifluoroacetic acid (TFA) to expose a primary amine that can be coupled to another carboxylate as described, or can be reacted with maleic anhydride and the resulting product cyclized to produce an activated maleimido derivative of the fatty acid. The modified antibodies of the invention can be produced by reacting a human antibody or antigen-binding fragment with a modifying agent. For example, the organic moieties can be bonded to the antibody in a non-site specific manner by employing an amine-reactive modifying agent, for example, an NHS ester of PEG. Modified human antibodies or antigen-binding fragments can also be prepared by reducing disulfide bonds (e.g., intra-chain disulfide bonds) of an antibody or antigen-binding fragment. The reduced antibody or antigen-binding fragment can then be reacted with a thiol-reactive modifying agent to produce the modified antibody of the invention. Modified human antibodies and antigen- binding fragments comprising an organic moiety that is bonded to specific sites of an antibody of the present invention can be prepared using suitable methods, such as reverse proteolysis. (See generally, Hermanson, G. T., BIOCONJUGATE TECHNIQUES, Academic Press: San Diego, Calif. (1996)). Compositions containing the antibodies, fragments thereof or cell lines which produce the antibodies, are encompassed by this invention. When these compositions are to be used pharmaceutically, they are combined with a pharmaceutically acceptable carrier. In another embodiment the present invention provides a method of inducing an immune response comprising delivering the compounds and compositions of the invention in the context of an MHC molecule. Thus, the polypeptides of this invention can be pulsed into antigen presenting cells using the methods described herein. Antigen-presenting cells, include, but are not limited to dendritic cells (DCs), monocytes/macrophages, B lymphocytes or other cell type(s) expressing the necessary MHC/co-stimulatory molecules. The methods described below focus primarily on DCs which are the most potent, preferred APCs. These host cells containing the polypeptides or proteins are further provided. The antibodies and compositions can be delivered by any suitable means and with any suitable formulation. Accordingly, a formulation comprising an antibody of this invention is further provided herein. The formulation can further comprise one or more preservative or stabilizer such as phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (e.g., hexahydrate), alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent. Any suitable concentration or mixture can be used as known in the art, such as 0.001-5%, or any range or value therein, such as, but not limited to 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05, 0.09, 0.1, 0.2, 0.3, O.4., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.3, 4.5, 4.6, 4.7, 4.8, 4.9, or any range or value therein. Non-limiting examples include, no preservative, 0.1-2% m-cresol (e.g., 0.2, 0.3. 0.4, 0.5, 0.9, 1.0%), 0.1- 3% benzyl alcohol (e.g., 0.5, 0.9, 1.1., 1.5, 1.9, 2.0, 2.5%), 0.001-0.5% thimerosal (e.g., 0.005, 0.01), 0.001-2.0% phenol (e.g., 0.05, 0.25, 0.28, 0.5, 0.9, 1.0%), 0.0005-1.0% alkylparaben(s) (e.g., 0.00075, 0.0009, 0.001, 0.002, 0.005, 0.0075, 0.009, 0.01, 0.02, 0.05, 0.075, 0.09, 0.1, 0.2, 0.3, 0.5, 0.75, 0.9, and 1.0%). COMPOSITIONS, KITS AND USES THEREFOR This invention also provides an article of manufacture, comprising packaging material and at least one vial comprising a solution of at least a peptide, polynucleotide or antibody as of this invention with the prescribed buffers and/or preservatives, optionally in an aqueous diluent, wherein said packaging material comprises a label that indicates that such solution can be held over a period of 1, 2, 3, 4, 5, 6, 9, 12, 18, 20, 24, 30, 36,40, 48, 54, 60, 66, 72 hours or greater. The invention further comprises an article of manufacture, comprising packaging material, a first vial comprising at least one lyophiiized composition of this invention and a second vial comprising an aqueous diluent of prescribed buffer or preservative, wherein said packaging material comprises a label that instructs a patient to reconstitute the antibody in the aqueous diluent to form a solution that can be held over a period of twenty- four hours or greater. The range antibody includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 μg/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal . patch, pulmonary, transmucosal, or osmotic or micro pump methods. The formulations of the present invention can be prepared by a process which comprises mixing at least one antibody of this invention and a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous diluent. Mixing of the antibody and preservative in an aqueous diluent is carried out using conventional dissolution and mixing procedures. For example, a measured amount of at least one antibody in buffered solution is combined with the desired preservative in a buffered solution in quantities sufficient to provide the antibody and preservative at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art, e.g., the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that can be optimized for the concentration and means of administration used. The compositions and formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophiiized antibody that is reconstituted with a second vial containing the aqueous diluent. Either a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available. Recognized devices comprising these single vial systems include those pen-injector devices for delivery of a solution such as BD Pens, BD Autojectore, Humaject.RTM.' NovoPen.RTM., B-D.RTM.Pen, AutoPen.RTM., and OptiPen.RTM., GenotropinPen.RTM., Genotronorm Pen.RTM., Humatro Pen.RTM., Reco- Pen.RTM., Roferon Pen.RTM., Biojector.RTM., iject.RTM., J-tip Needle-Free Injector.RTM., Intraject.RTM., Medi-Ject.RTM., e.g., as made or developed by Becton Dickensen (Franklin Lakes, NJ. available at bectondickenson.com),
Disetronic (Burgdorf, Switzerland, available at disetronic.com; Bioject, Portland, Oregon (available at bioject.com); National Medical Products, Weston Medical (Peterborough, UK, available at weston-medical.com), Medi-Ject Coφ (Minneapolis, Minn., available at mediject.com). Isolated host cells which present the polypeptides of this invention in the context of MHC molecules are further useful to expand and isolate a population of educated, antigen-specific immune effector cells. The immune effector cells, e.g., cytotoxic T lymphocytes, are produced by culturing naϊve immune effector cells with antigen-presenting cells which present the polypeptides in the context of MHC molecules on the surface of the APCs. The population can be purified using methods known in the art, e.g., FACS analysis or ficoll gradient. The methods to generate and culture the immune effector cells as well as the populations produced thereby also are the inventor's contribution and invention. Pharmaceutical compositions comprising the cells and pharmaceutically acceptable carriers are useful in adoptive immunotherapy. Prior to administration in vivo, the immune effector cells are screened in vitro for their ability to lyse cells. In one embodiment, the immune effector cells and/or the APCs are genetically modified. Using standard gene transfer, genes coding for co-stimulatory molecules and/or stimulatory cytokines can be inserted prior to, concurrent to or subsequent to expansion of the immune effector cells. This invention also provides methods of inducing an immune response in a subject, comprising administering to the subject an effective amount of the polypeptides or immunogenic portion thereof under the conditions that induce an immune response to the polypeptide and induce a cytolytic Tcell response. The polypeptides can be administered in formulations or as polynucleotides encoding the polypeptides. The polynucleotides can be administered in a gene delivery vehicle or by inserting into a host cell which in turn recombinantly transcribes, translates and processed the encoded polypeptide. Isolated host cells containing the polynucleotides of this invention in a pharmaceutically acceptable carrier can therefore combined with appropriate and effective amount of an adjuvant, cytokine or co-stimulatory molecule for an effective vaccine regimen. In one embodiment, the host cell is an APC such as a dendritic cell. The host cell can be further modified by inserting of a polynucleotide coding for an effective amount of either or both a cytokine and/or a co-stimulatory molecule. The methods of this invention can be further modified by co-administering an effective amount of a cytokine or co-stimulatory molecule to the subject. The polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g.., poly-His), or to enhance binding of the polypeptide to a solid support. For example, a polypeptide can be conjugated to an immunoglobulin Fc region. In an embodiment of the invention an antibody of the invention may be coupled to a bridging compound coupled to a solid support. The bridging compound, which is designed to link the solid support and the antibody may be hydrazide, Protein A, glutaraldehyde, carbodiimide, or lysine. The solid support employed is e.g., a polymer or it may be a matrix coated with a polymer. The matrix may be of any suitable solid material, e.g. glass, paper or plastic. The polymer may be a plastic, cellulose such as specially treated paper. nitrocellulose paper or cyanogenbromide-activated paper. Examples of suitable plastics are latex, a polystyrene, polyvinylchloride, polyurethane, polyacrylamide, polyvinylacetate and any suitable copolymer thereof. Examples of silicone polymers include siloxane. The solid support may be in the form of a tray, a plate such as a mitrotiter plate, e.g. a thin layer or, preferably, strip, film, glass slide, threads, solid particles such as beads, including Protein A-coated bacteria, or paper. The antibody of the invention may be used in an assay for the identification and/or quantification of at least a form and/or a part of said polypeptide present in a sample. The identification and/or quantification performed by the use according to the present invention may be any identification and/or quantification of a protein comprising any of the polypeptides of the invention. Thus, both a qualitative and a quantitative determination of any protein comprising any of the polypeptides of the invention may be obtained according to the use of the present invention. The identification and/or quantification may be performed for both a scientific, a clinical and an industrial puφose. As will be further described below, it is especially important in clinical routine to identify or quantify the amount of any protein comprising any of the polypeptides of the invention. h one aspect, the antibody used in the method of the invention is a monoclonal antibody as this generally provides a higher precision and accuracy of the assay, at the same time possibly requiring less time to perform. Furthermore, a mixture of two or more different monoclonal antibodies may be employed to increase the detection limit and sensitivity of the test. The monoclonal antibody may be obtained by methods described herein or as known in the art. Antibodies possessing high avidity may be selected for catching techniques. The antibody used in the present method is preferably in substantially pure form (purified according to suitable techniques or by the methods of the invention, see below) in order to improve the precision and/or accuracy of the assays of the invention. In a further aspect, the invention relates to a diagnostic agent comprising an antibody capable of detecting and/or quantitating any of the ligands of the invention. For most assay uses it preferred that the antibody is provided with a label for the detection of bound antibody or, alternatively (such as in a double antibody assay), a combination of labelled and unlabelled antibody may be employed. The substance used as label may be selected from any substance which is in itself detectable or which may be reacted with another substance to produce a detectable product. Thus, the label may be selected from radioactive isotopes, enzymes, chromophores, fluorescent or chemiluminescent substances, and complexing agents. In one embodiment, these neoplasia are diagnosed by evaluating a biological sample obtained from the animal or patient and determining the level of one or more of the above polypeptides, relative to a predetermined cut-off value. As used herein, suitable "biological samples" include ovarian tissue, blood, stool, sera, urine and/or secretions. The antibody used in the present method may be in substantially pure form in order to improve the precision and/or accuracy of the assays of the invention. Functional equivalents to the monoclonal antibody, that is, compounds or other ligands that bind to proteins comprising the peptides of the invention and which may inhibit the binding of the antibody to these proteins are also provided by this invention. Another method according to the invention is a method for detecting or quantifying complexes of antibody and the peptides of the invention in a sample, comprising using an antibody capable of binding to one of the peptides of the invention together with an antibody which detects antibody-peptides complexes. Another method according to the invention is a method for immunohistochemical detection of any of the peptides of the invention in tissue sections, using, as the detecting antibody, any monoclonal antibody which reacts with one of the peptides of the invention, or any polyclonai antibody which reacts with at least one of the polypeptides of the invention. The monoclonal antibodies according to the invention may also be used in a method for targeting a diagnostic agent to a cell that expresses a peptide of the invention on the surface, the method comprising administering, to a mammal, in particular a human, in particular a mammal suffering from cancer or suspected to suffer from cancer, the diagnostic agent bound to a monoclonal antibody of the invention. The diagnostic agent may be a radioactive substance, such as technetium. Several embodiments of this invention include but are not limited to: 1) use as a catching antibody when immobilized as a catching antibody in a sandwich ELISA; 2) when used as a biotin-labelled detecting antibody in a sandwich
ELISA is capable of detecting one of the peptides of the invention; 3) when used in an ELISA, is capable of binding to an immobilized peptides of the invention; 5) in a radioimmunoprecipitation assay precipitates one of the peptides of the invention; or 8) is capable of binding to one of the peptides of the invention in tissue sections, including paraffine-embedded tissue sections, and thereby being useful for immunohistochemical detection of one of the peptides of the invention. Further provided by the present invention are methods for aiding in the detecting, diagnosing, prognosing, and monitoring the progression, course, or stage of cancers or malignancies in subjects afflicted therewith. These invention methods comprise detecting the differential expression of one or more of the peptides of the invention in a sample isolated from a cell or tissue, wherein the presence and/or amount of the protein is indicative of the neoplastic condition of cell or tissue. A related neoplasia is one in which the expression or expression of the protein serves as a marker for the neoplastic phenotype. Samples of cells or tissue can be provided free form or attached to a solid support and can be isolated from a tissue culture, commercially available cell line, from a patient biopsy or as in the case of use of the method for tissue imaging, in vivo. Three methods are provided to identify subjects suitably treated by administration of a peptide, polypeptide, antibody, host cell or immune effector cell of the invention : Method No. 1: Normal or patient donor PBMC are exposed to the invention peptides either individually or as a cocktail under the appropriate reaction conditions to generate a mixed population of reactive cytotoxic T cells. These cells are tested for their ability to react with tumor cells lines known to express the antigen. This ensures that the peptide-educated CTL recognize the epitope in its native state. These tumor-reactive T cells can be used as a bulk population or individual tumor-reactive clones can be derived by standard limiting dilution analysis, in order to determine whether a particular patient's tumor expresses the unknown antigen. Some of the techniques that can be employed include: 1. One can use tumor cells derived from patient tumor biopsy in order to stimulate the invention peptide-educated CTL and measure the secretion of cytokines associated with CTL antigen recognition such as IL-2 or INF-α can be measured. Standard techniques such as ELISPOT, ELISA, or intracellular cytokine staining can be employed. Cytokine release indicates that the patient's tumor cells express the unnamed antigen, implying potential benefit to that patient by successful vaccination with the peptides. 2. Single cell suspensions of patient tumor cells derived from biopsy or resected tumor can be reacted with the peptide-educated CTL of the present invention in a standard 51Cr-release assay in order to measure patient tumor lysis directly. Lysis indicates that the patient's tumor cells express the unnamed antigen, implying potential benefit to that patient by successful vaccination with the peptides.
Method No. 2: Peptide-educated clonal populations of CTL of the present invention can be used to construct recombinant soluble T cell receptor (TCR) protein with specificity for the native epitope corresponding to the unnamed antigen. Multimerization of the TCR proteins results in a reagent that will bind to the HLA-A2 molecule in the presence of the specific epitope recognized by the TCR. Therefore, this reagent is useful to test patient tumor cells derived from biopsy or tumor resection for the presence of the unnamed native epitope. A positive result indicates that the patient's tumor expresses the unnamed antigen implying potential benefit to the patient by successful vaccination with the peptides.
Method No. 3: Phage display technology has been successfully employed to identify recombinant antibodies capable of binding specifically to distinct MHC/peptide complexes (See, Denkberg, G. et al., (2002) PNAS 99(14):9421-9426). These monoclonal antibodies are useful to detect the presence of particular epitopes presented in the context of MHC molecules on the surface of tumor cells. This approach is useful to generate monoclonal antibodies against invention peptide-bound HLA-A2 molecules. These antibodies can be tested for specificity with tumor cell lines known to express or not express the antigen. Patient tumor derived from biopsy or tumor resection are tested for the presence of the native epitope/MHC surface complex by measuring antibody binding. A positive result indicates that the patient's tumor expresses the unnamed antigen implying potential benefit to the patient by successful vaccination with the peptides. In another aspect, the method is practiced by detecting and/or quantifying mRNA encoding one of the peptides of the invention by hybridization or PCR. Modification of current technology enables this method, e.g., detecting is by probing said sample with a probe or primer that specifically hybridizes under conditions of moderate or highly stringent conditions with said mRNA. In one aspect, the probe or primer is detectably labeled. Application of RT-PCR techniques allows for quantitation of mRNA expression. Application of PCR is also useful to detect mutations in the nucleic acid sequences encoding any of the peptides of the invention. Examples of suitable probes include but are not limited to a sequence selected from the group consisting of SEQ ID NOs: 1 to 43 as shown in Table 1, and complements thereof; a nucleic acid sequence encoding a peptide selected from the group consisting of SEQ ID NOs: 2 to 44 as shown in Table 1, and complements thereof; and a probe or primer complementary to a sequence encoding a protein comprising at least 9 consecutive residues of an amino acid sequence recited in the group consisting of SEQ ID NOs: 2 to 44, shown in Table 1, and complements thereof. In another aspect, the peptides of the invention are detected and/or quantified by probing the sample peptides with an agent that specifically recognizes and binds one or more of the amino acid sequences shown in Table 1. Antibodies, and antigen binding fragments thereof, that specifically recognize or bind to the peptides of the invention are examples of agents. For the puφose of this invention, an antibody is a polyclonai or monoclonal antibody, which may or may not be detectably labeled. Various modifications to antibodies and antigen binding fragments are known in the art and included for the puφose of this invention. Examples are described herein. Suitable monoclonal antibodies for use in this aspect are prepared from an animal immunized with a peptide selected from the group consisting of those shown in Table 1. Examples of an antigen binding fragment include a biologically active immunoglobulin variable domain isolated from an antibody prepared from an animal immunized with a peptide selected from the group consisting of those shown in Table 1. The method utilizing antibodies can be practiced in vitro or in vivo, using application of well known methods as described herein. In another aspect, the agent is a cell that binds to a protein comprising a peptides of the invention, such as an immune effector cell raised in the presence and at the expense of a peptide selected from the group consisting of those shown in Table 1. Alternatively, differential expression of any of the peptides of the invention can be detected and/or quantified by determining the identity and expression level of mRNA by expression analysis and comparing the sequences and amount of mRNA to sequences and amount of expression in a normal control cell or tissue, using methods such as expression analysis, e.g., SAGE (United States Pat. No. 5,695,937) and expression arrays. In another embodiment, detection and analysis of the presence in a sample of the amount of any of the peptides of the invention is repeated to monitor disease progression, response to a therapeutic regimen, or disease recurrence in a subject: In this aspect, the detecting/monitoring method, as described herein, is performed on one or more sample(s) isolated at one or more times time points subsequent to a prior (control) sample and comparing the amount of protein detected at the subsequent points in time to the amount previously detected in a prior (control) sample. Additionally, the present invention provides a screen to identify agents which bind to any of the peptides of the invention, or which bind to proteins comprising any of the peptides of the invention. In one embodiment, test compounds may be labeled with a detectable agent, e.g., tritium, and incubated with any of the peptides of the invention. At the end of the incubation period, the incubation solution is treated with a washing solution to remove unbound test compounds. After washing, the amount of labeled test compound remaining is measured and compared to a control sample to determine whether a test compound or compound is capable of specific binding. In a variation of this embodiment, the peptides of the invention are labeled instead of the test compounds. In another aspect, the invention provides a screen for agents that inhibit the binding of an antibody or a protein or peptide comprising any of the peptides of the invention to a ligand by contacting a sample comprising the protein or peptide and its ligand under conditions and in the presence of a test agent and detecting any binding between the protein or peptide and its ligand, the absence of binding being indicative of an agent that modulates the binding of the protein or peptide to its binding partner. Alternatively, the invention is useful to screen for agents that enhance the binding of a protein or peptide comprising any of the peptides of the invention to its ligand by contacting a sample comprising the protein or peptide and the ligand under conditions and in the presence of a test agent and detecting and comparing any binding between the protein or peptide and its ligand with a control sample that does not contain the test agent. Enhancement of binding over the control sample is a positive indication that the test agent enhances the binding of the protein or peptide to its ligand. Modulation (increase or inhibit) of binding includes a variation in avidity as well as affinity. For example, one such assay comprises contacting a mammalian cell with an invention composition and the "test compound" and [ 3 H ]thymidine, under conditions where the mammalian cell would normally proliferate. A control assay may be performed without the "test compound" and compared to the amount of cell proliferation in the presence of the "test compound" to determine if the compound stimulates or inhibits proliferation. [ 3 H ] thymidine uptake can be measured by liquid scintillation chromatography which measures the incoφoration of the label. Agonists and antagonist compounds may be identified using this assay. In an alternative example, a mammalian cell or membrane preparation expressing a binding site or determinant for a polypeptide or peptide of the invention is incubated with a labeled polypeptide or peptide of the invention in the presence of the "test compound". The ability of the "test compound" to enhance or block the interaction of the polypeptide or peptide with its binding site, can be measured. Further the response of a known second messenger system following the interaction of the "test compound" and the binding site is measured and the ability of the "test compound" to bind the site or determinant and elicit a second messenger response can be measured to determine if the "test compound" is a potential agonist or antagonist. These assays can be used as diagnostic or prognostic markers. The molecules discovered using these screening assays can be used to treat disease or to bring about a particular result in a patient by activating or inhibiting the polypeptide or molecule. Thus, the invention includes a method of identifying compounds which bind to a polypeptide, peptide, antibody, host cell or immune effector cell of the invention comprising: (a) incubating a candidate test compound with a composition described herein of the invention; and (b) determining if binding has occurred. The invention further comprises a method of identifying agonists and/or antagonists comprising: (a) incubating a candidate test compound with a composition described herein of the invention; (b) assaying a biological activity; and (c) determining if said biological activity has been altered. Diagnostic kits to practice the methods described herein are also provided. In one aspect, the kit contains at least one agent that specifically recognizes and binds to a protein or peptide comprising any of the peptides of the invention and a detection reagent which can support a reporter group. When antibodies are used as the agent, they can be in solution or immobilized on a solid support, such as nitrocellulose, glass, latex or a plastic. Examples of detection reagents include but are not limited to anti-immunoglobulin, protein G, protein A, or lectin. Reporter groups can be provided in the kit, but are not necessarily supplied. Examples of reporter groups suitable for use in the methods include but are not limited to radioisotopes, fluorescent groups, luminescent groups, enzymes, biotin and dye particles. The kits will contain one or more of binding agents described above, e.g., a probe, a primer, an antibody or immune effector cell specific for any of the peptides of the invention, or an agent that specifically recognizes and binds to the specific antibody, e.g., antibodies raised and isolated to specifically recognize and bind a peptide of the invention. Instructions for use can also be provided. The polypeptides of the present invention, can be used to generate binding agents, such as antibodies or fragments thereof, that are capable of detecting neoplastic cells or tissues, or alternatively, for monitoring disease progression in an animal or patient having a neoplastic condition related to the expression of a peptide or protein comprising any of the peptides of the invention, e.g, ovarian cancer. Binding agents of the present invention may generally be prepared using methods known to those of ordinary skill in the art, including the representative procedures described herein. Binding agents are capable of differentiating between patients with and without a neoplastic condition associated with the expression and or overexpression of a peptide or protein comprising any of the peptides of the invention, using the representative assays described herein. In other words, antibodies or other binding agents raised against any of the peptides of the invention will generate a signal indicating the presence of primary or metastatic cancer in patients afflicted with the disease, and will generate a negative signal indicating the absence of the disease in individuals without primary or metastatic disease. Suitable portions of a protein comprising any of the peptides of the invention, as well as the polypeptides shown in Table 1 that are able to generate a binding agent that indicates the presence of ovarian neoplasia in substantially all (t'.e., at least about 80%, and preferably at least about 90%) of the patients for which such neoplasia would be indicated and that indicates the absence of ovarian neoplasia in substantially all of those samples that would be negative when tested with full length protein. The representative assays described below, such as the two-antibody sandwich assay, can be employed for evaluating the ability of a binding agent to detect ovarian neoplasia. The ability of a polypeptide prepared as described herein to generate antibodies capable of detecting neoplasia can be evaluated by raising one or more antibodies against the polypeptide and determining the ability of such antibodies to detect neoplasia in animals or patients. In one aspect, this determination is made by assaying biological samples from animals or patients with and without primary or metastatic neoplasia for the presence of a polypeptide that binds to the generated antibodies. Such test assays may be performed, for example, using a representative procedure described below. Polypeptide specific antibodies raised the polypeptides identified in Table 1, or a fragment thereof, can be used alone or in combination to improve sensitivity. The level of a protein or proteins comprising any of the peptides of the invention can be evaluated using any binding agent specific for any of the ligands of the invention. A "binding agent," in the context of this invention, is any agent (such as a compound or a cell) that binds to a polypeptide as described above. As used herein, "binding" refers to a noncovalent association between two separate molecules (each of which may be free (i.e., in solution) or present on the surface of a cell or a solid support), such that a "complex" is formed. The complex can be free or immobilized (either covalently or noncovalently) on a support material. The ability to bind can be evaluated by determining a binding constant for the formation of the complex. The binding constant is the value obtained when the concentration of the complex is divided by the product of the component concentrations. Binding constants are determined using methods known to those of ordinary skill in the art. Any agent that satisfies the above requirements may be a binding agent. For example, a binding agent may be a ribosome with or without a peptide component, an RNA molecule, a host cell expressing epitope, an immune effector cell as described herein or a peptide. In a preferred embodiment, the binding partner is an antibody, or a fragment thereof. The antibodies can be polyclonai or monoclonal which include, but are not limited to single chain, chimeric, CDR-grafted or humanized. Methods for generating these antibodies are known to those of skill in the art. This invention also provides compositions containing any of the above-mentioned proteins, polypeptides, polynucleotides, vectors, cells, antibodies and fragments thereof, and an acceptable solid or liquid carrier. When the compositions are used pharmaceutically, they are combined with a
"pharmaceutically acceptable carrier" for diagnostic and therapeutic use. These compositions also can be used for the preparation of medicaments for the diagnosis and treatment of diseases such as cancer. The following materials and methods are intended to illustrate, but not limit this invention and to illustrate how to make and use the inventions described above.
Materials and Methods
Production of the Polypeptides of the Invention Most preferably, isolated polypeptides of the present invention can be synthesized using an appropriate solid state synthetic procedure. (Steward and Young, SOLID PHASE PEPTIDE SYNTHESIS, Freemantle, San Francisco, Calif. (1968)). A preferred method is the Merrifield process. (See, Merrifield (1967) Recent Progress in Hormone Res. 23:451). The antigenic activity of these peptides may conveniently be tested using, for example, the assays as described herein. Once an isolated polypeptide of the invention is obtained, it may be purified by standard methods including chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. For irnmuno-affinity chromatography, an epitope may be isolated by binding it to an affinity column comprising antibodies that were raised against that peptide, or a related peptide of the invention, and were affixed to a stationary support. Alternatively, affinity tags such as hexa-His (Invitrogen), Maltose binding domain (New England Biolabs), influenza coat sequence (Kolodziej, et al. (1991) Meth. Enzymol. 194:508-509), and glutathione- S-transferase can be attached to the peptides of the invention to allow easy purification by passage over an appropriate affinity column. Isolated immunogens can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance, and x-ray crystallography. Also included within the scope of the invention are immunogenic peptides that are differentially modified during or after translation, e.g., by phosphorylation, glycosylation, cross-linking, acylation, proteolytic cleavage, linkage to an antibody molecule, membrane molecule or other ligand. (Ferguson, et al. (1988) Ann. Rev. Biochem. 57:285-320).
Isolation. Culturing and Expansion of APCs, Including Dendritic Cells The following is a brief description of two fundamental approaches for the isolation of APC. These approaches involve (1) isolating bone marrow precursor cells (CD34+) from blood and stimulating them to differentiate into APC; or (2) collecting the precommitted APCs from peripheral blood. In the first approach, the patient must be treated with cytokines such as GM-CSF to boost the number of circulating CD34+ stem cells in the peripheral blood. The second approach for isolating APCs is to collect the relatively large numbers of precommitted APCs already circulating in the blood. Previous techniques for isolating committed APCs from human peripheral blood have involved combinations of physical procedures such as metrizamide gradients and adherence/non-adherence steps (Freudenthal P.S., et al. (1990) Proc. Natl. Acad. Sci. USA 87:7698-7702); Percoll gradient separations (Mehta-Damani, et al. (1994) J. Immunol. 153:996-1003); and fluorescence activated cell sorting techniques (Thomas R., et al. (1993) J. Immunol. 151:6840-6852). One technique for separating large numbers of cells from one another is known as countercurrent centrifugal elutriation (CCE). In this technique, cells are subject to simultaneous centrifugation and a washout stream of buffer that is constantly increasing in flow rate. The constantly increasing countercurrent flow of buffer leads to fractional cell separations that are largely based on cell size. In one aspect of the invention, the APC are precommitted or mature dendritic cells which can be isolated from the white blood cell fraction of a mammal, such as a murine, simian or a human (See, e.g., WO 96/23060). The white blood cell fraction can be from the peripheral blood of the mammal. This method includes the following steps: (a) providing a white blood cell fraction obtained from a mammalian source by methods known in the art such as leukophoresis; (b) separating the white blood cell fraction of step (a) into four or more subtractions by countercurrent centrifugal elutriation; (c) stimulating conversion of monocytes in one or more fractions from step (b) to dendritic cells by contacting the cells with calcium ionophore, GM-CSF and IL-13 or GM-CSF and IL-4, (d) identifying the dendritic cell-enriched fraction from step (c); and (e) collecting the enriched fraction of step (d), preferably at about 4°C. One way to identify the dendritic cell-enriched fraction is by fluorescence-activated cell sorting. The white blood cell fraction can be treated with calcium ionophore in the presence of other cytokines, such as recombinant (rh) rhIL-12, rhGM-CSF, or rhIL-4. The cells of the white blood cell fraction can be washed in buffer and suspended in CaATVlg"1^ free media prior to the separating step. The white blood cell fraction can be obtained by leukapheresis. The dendritic cells can be identified by the presence of at least one of the following markers: HLA-DR, HLA-DQ, or B7. 2, and the simultaneous absence of the following markers: CD3, CD14, CD16, 56, 57, and CD 19, 20. Monoclonal antibodies specific to these cell surface markers are commercially available. More specifically, the method requires collecting an enriched collection of white cells and platelets from leukapheresis that is then further fractionated by countercurrent centrifugal elutriation (CCE) (Abrahamsen, T.G. et al. (1991) J. Clin. Apheresis. 6:48-53). Cell samples are placed in a special elutriation rotor. The rotor is then spun at a constant speed of, for example, 3000 φm. Once the rotor has reached the desired speed, pressurized air is used to control the flow rate of cells. Cells in the elutriator are subjected to simultaneous centrifugation and a washout stream of buffer that is constantly increasing in flow rate. This results in fractional cell separations based largely but not exclusively on differences in cell size. Quality control of APC and more specifically DC collection and confirmation of their successful activation in culture is dependent upon a simultaneous multi-color FACS analysis technique which monitors both monocytes and the dendritic cell subpopulation as well as possible contaminant T lymphocytes. It is based upon the fact that DCs do not express the following markers: CD3 (T cell); CD14 (monocyte); CD16, 56, 57 (NK/LAK cells); CD19, 20 (B cells). At the same time, DCs do express large quantities of HLA-DR, significant HLA-DQ and B7.2 (but little or no B7.1) at the time they are circulating in the blood (in addition they express Leu M7 and M9, myeloid markers which are also expressed by monocytes and neutrophils). When combined with a third color reagent for analysis of dead cells, propridium iodide (PI), it is possible to make positive identification of all cell subpopulations (see Table 2):
TABLE 2 FACS analysis of fresh peripheral cell subpopulations
Figure imgf000077_0001
Additional markers can be substituted for additional analysis: Color #1 : CD3 alone, CD 14 alone, etc.; Leu M7 or Leu M9; anti-Class I, etc. Color #2: HLA-DQ, B7.1, B7.2, CD25 (IL2r), ICAM, LFA-3, etc. The goal of FACS analysis at the time of collection is to confirm that the DCs are enriched in the expected fractions, to monitor neutrophil contamination, and to make sure that appropriate markers are expressed. This rapid bulk collection of enriched DCs from human peripheral blood, suitable for clinical applications, is absolutely dependent on the analytic FACS technique described above for quality control. If need be, mature DCs can be immediately separated from monocytes at this point by fluorescent sorting for "cocktail negative" cells. It may not be necessary to routinely separate DCs from monocytes because, as will be detailed below, the monocytes themselves are still capable of differentiating into DCs or functional DC-like cells in culture. Once collected, the DC rich/monocyte APC fractions (usually 150 through 190) can be pooled and cryopreserved for future use, or immediately placed in short term culture. Alternatively, others have reported a method for upregulating (activating) dendritic cells and converting monocytes to an activated dendritic cell phenotype. This method involves the addition of calcium ionophore to the culture media convert monocytes into activated dendritic cells. Adding the calcium ionophore A23187, for example, at the beginning of a 24-48 hour culture period resulted in uniform activation and dendritic cell phenotypic conversion of the pooled "monocyte plus DC" fractions: characteristically, the activated population becomes uniformly CD 14 (Leu M3) negative, and upregulates HLA-DR, HLA-DQ, ICAM-1, B7.1, and B7.2. Furthermore, this activated bulk population functions as well on a small numbers basis as a further purified. Specific combination(s) of cytokines have been used successfully to amplify (or partially substitute) for the activation/conversion achieved with calcium ionophore: these cytokines include but are not limited to purified or recombinant ("rh") rhGM-CSF, rhIL-2, and rhIL-4. Each cytokine when given alone is inadequate for optimal upregulation.
Presentation of Antigen to the APC For puφoses of immunization, the peptides can be delivered to antigen-presenting cells as protein/peptide or in the form of cDNA encoding the protein/peptide. Antigen-presenting cells (APCs) can consist of dendritic cells (DCs), monocytes/macrophages, B lymphocytes or other cell type(s) expressing the necessary MHC/co- stimulatory molecules. The methods described below focus primarily on DCs which are the most potent, preferred APCs. Pulsing is accomplished in vitro/ex vivo by exposing APCs to the antigenic protein or peptide(s) of this invention. The protein or peptide(s) are added to , APCs at a concentration of 1-10 μm for approximately 3 hours. Pulsed APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery. Protein/peptide antigen can also be delivered in vivo with adjuvant via the intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery. Paglia, et al. (1996) J. Exp. Med. 183:317-322 has shown that APC incubated with whole protein in vitro were recognized by MHC class I-restricted CTLs, and that immunization of animals with these APCs led to the development of antigen- specific CTLs in vivo. In addition, several different techniques have been described which lead to the expression of antigen in the cytosol of APCs, such as DCs. These include (1) the introduction into the APCs of RNA isolated from tumor cells, (2) infection of APCs with recombinant vectors to induce endogenous expression of antigen, and (3) introduction of tumor antigen into the DC cytosol using liposomes. (See Boczkowski, D. et al. (1996) J. Exp. Med. 184:465-472; Rouse, et al. (1994) J. Virol. 68:5685-5689; and Nair, et al. (1992) J. Exp. Med. 175:609-612).
Foster Antigen Presenting Cells Foster antigen presenting cells are particularly useful as target cells. Foster
APCs are derived from the human cell line 174xCEM.T2, referred to as T2, which contains a mutation in its antigen processing pathway that restricts the association of endogenous peptides with cell surface MHC class I molecules. (Zweerink, et al. (1993) J. Immunol. 150:1763-1771). This is due to a large homozygous deletion in the MHC class II region encompassing the genes TAPl, TAP2, LMPl, and LMP2, which are required for antigen presentation to MHC class 1 -restricted CD8+ CTLs. In effect, only "empty" MHC class I molecules are presented on the surface of these cells. Exogenous peptide added to the culture medium binds to these MHC molecules provided that the peptide contains the allele-specific binding motif. These T2 cells are referred to herein as "foster" APCs. They can be used in conjunction with this invention to present antigen(s). Transduction of T2 cells with specific recombinant MHC alleles allows for redirection of the MHC restriction profile. Libraries tailored to the recombinant allele will be preferentially presented by them because the anchor residues will prevent efficient binding to the endogenous allele. High level expression of MHC molecules makes the APC more visible to the CTLs. Expressing the MHC allele of interest in T2 cells using a powerful transcriptional promoter (e.g., the CMV promoter) results in a more reactive APC (most likely due to a higher concentration of reactive MHC-peptide complexes on the cell surface). For example, to determine the ability of a polypeptide, peptide, variant or analog, to compete with another peptide for binding to an antibody, various immunoassays can be employed, including but not limited to, competitive and non-competitive assay systems using technologies such as radioimmunoassays, ELISA, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays, Western Blot, precipitation reactions, agglutination assays, immunofluorescence assays, protein A assays, electrophoretic assays, and the like. Binding may also be detected using antibody capture assays, antigen capture assays, and multiple-antibody sandwich assays. Methods for detecting binding in such assays are known in the art.
Expansion of Immune Effector Cells The present invention makes use of these APCs to stimulate production of an enriched population of antigen- specific immune effector cells. The antigen-specific immune effector cells are expanded at the expense of the APCs, which die in the culture. The process by which naive immune effector cells become educated by other cells is described essentially in Coulie (1997) Molec. Med. Today 3:261-268. The APCs prepared as described above are mixed with naive immune effector cells. Preferably, the cells may be cultured in the presence of a cytokine, for example IL2. Because dendritic cells secrete potent immunostimulatory cytokines, such as IL12, it may not be necessary to add supplemental cytokines during the first and successive rounds of expansion. In any event, the culture conditions are such that the antigen-specific immune effector cells expand (i.e., proliferate) at a much higher rate than the APCs. Multiple infusions of APCs and optional cytokines can be performed to further expand the population of antigen-specific cells. In one embodiment, the immune effector cells are T cells. In a separate embodiment, the immune effector cells can be genetically modified by transduction with a transgene coding for example, IL-2, IL-11 or IL-13. Methods for introducing transgenes in vitro, ex vivo and in vivo are well known in the art. (See Sambrook, et al. (1989) supra).
Vectors Useful in Genetic Modifications In general, genetic modifications of cells employed in the present invention are accomplished by introducing a vector containing a polypeptide or transgene encoding a heterologous or an altered immunogen. A variety of different gene transfer vectors, including viral as well as non-viral systems can be used. Viral vectors useful in the genetic modifications of this invention include, but are not limited to adenovirus, adeno-associated virus vectors, retroviral vectors and adeno-retroviral chimeric vectors. APC and immune effector cells can be modified using the methods described below or by any other appropriate method known in the art.
Construction of Recombinant Adenoviral Vectors or Adeno- Associated Virus Vectors Adenovirus and adeno-associated virus vectors useful in the genetic modifications of this invention may be produced according to methods already taught in the art. (See, e.g., Karisson, et al. (1986) EMBO J. 5:2377; Carter (1992) /
Curr. Op. Biotechnol. 3:533-539; Muzcyzka (1992) Current Top. Microbiol. Immunol. 158:97-129; GENE TARGETING: A PRACTICAL APPROACH (1992) ed. A. L. Joyner, Oxford University Press, NY). Several different approaches are feasible. Preferred is the helper-independent replication deficient human adenovirus system. The recombinant adenoviral vectors based on the human adenovirus 5 (Virology 163:614-617 (1988)) are missing essential early genes from the adenoviral genome (usually El A El B), and are therefore unable to replicate unless grown in permissive cell lines that provide the missing gene products in trans. In place of the missing adenoviral genomic sequences, a transgene of interest can be cloned and expressed in cells infected with the replication deficient adenovirus. Although adenoviras-based gene transfer does not result in integration of the transgene into the host genome (less than 0.1% adenovirus-mediated transfections result in transgene incoφoration into host DNA), and therefore is not stable, adenoviral vectors can be propagated in high titer and transfect non-replicating cells. Human 293 cells, which are human embryonic kidney cells transformed with adenovirus E1A/E1B genes, typify useful permissive cell lines. However, other cell lines which allow replication-deficient adenoviral vectors to propagate therein can be used, including HeLa cells. Additional references describing adenovirus vectors and other viral vectors which could be used in the methods of the present invention include the following: Horwitz M.S. ADENOVLRIDAE AND THEIR REPLICATION, in Fields B, et al. (eds.) VIROLOGY, Vol. 2, Raven Press New York, pp. 1679-1721 (1990); Graham F., et al. pp. 109-128 in METHODS IN MOLECULAR BIOLOGY, Vol. 7: GENE TRANSFER AND EXPRESSION PROTOCOLS, Murray, E. (ed.) Humana Press, Clifton, NJ. (1991); Miller N, et al. (1995) FASEB J. 9:190-199; Schreier H. (1994) Pharmaceutica Acta Helvetiae 68:145-159; Schneider and French (1993) Circulation 88:1937-1942; Curiel D.T., et al.(1992) Hum. Gene Ther. 3:147-154; Graham, F.L. et al. WO 95/00655 (5 January 1995); Falck-Pedersen E.S. WO 95/16772 (22 June 1995); Denefle P., et al. WO 95/23867 (8 September 1995); Haddada H., et al. WO 94/26914 (24 November 1994); Perricaudet M., et al. WO 95/02697 (26 January 1995); Zhang W., et al. WO 95/25071 (12 October 1995). A variety of adenovirus plasmids are also available from commercial sources, including, e.g., Microbix Biosystems of Toronto, Ontario (see, e.g., Microbix Product Information Sheet: Plasmids for Adenovirus Vector Construction, 1996). See also, the papers by Vile, et al. (1997) Nature Biotechnology 15:840-841; and Feng, et al. (1997) Nature Biotechnology 15:866-870, describing the construction and use of adeno-retroviral chimeric vectors that can be employed for genetic modifications. Additional references describing AAV vectors that could be used in the methods of the present invention include the following: Carter B. HANDBOOK OF PARVOVIRUSES, Vol. I, pp. 169-228, 1990; Berns, VIROLOGY, pp. 1743-1764 (Raven Press 1990); Carter B. (1992) Curr. Opin. Biotechnol. 3:533-539; MuzyczkaN. (1992) Current Topics in Micro, and Immunol, 158:92-129; Flotte T.R., et al. (1992) Am. J. Respir. Cell Mol. Biol. 7:349-356; Chatterjee, et al. (1995) Ann. NY Acad. Sci. 770:79-90; Flotte T.R., et al. WO 95/13365 (18 May 1995); Trempe J.P, et al. WO 95/13392 (18 May 1995); Kotin R.(1994) Hum. Gene Ther. 5:793-801; Flotte T.R., et al. (1995) Gene Therapy 2:357-362; Allen J.M., WO 96/17947 (13 June 1996); and Du, et al. (1996) Gene Therapy 3:254-261. APCs can be transduced with viral vectors encoding a relevant polypeptides. The most common viral vectors include recombinant poxviruses such as vaccinia and fowlpox virus (Bronte, et al. (1997) Proc. Natl. Acad. Sci. USA 94:3183-3188; Kim, et al. (1997) J. Immunother. 20:276-286) and, preferentially, adenovirus (Arthur, et al. (1997) J. Immunol. 159:1393-1403; Wan, et al. (1997) Human Gene Therapy 8:1355-1363; Huang, et al. (1995) J. Virol. 69:2257-2263). Retrovirus also may be used for transduction of human APCs (Marin, et al. (1996) J. Virol. 70:2957-2962). In vitro/ex vivo, exposure of human DCs to adenovirus (Ad) vector at a multiplicity of infection (MOI) of 500 for 16-24 h in a minimal volume of seram-free medium reliably gives rise to transgene expression in 90-100% of DCs. The efficiency of transduction of DCs or other APCs can be assessed by immunofluorescence using fluorescent antibodies specific for the tumor antigen being expressed. (Kim, et al. (1997) J. Immunother. 20:276-286). Alternatively, the antibodies can be conjugated to an enzyme (e.g., HRP) giving rise to a colored product upon reaction with the substrate. The actual amount of antigenic polypeptides being expressed by the APCs can be evaluated by ELISA. Transduced APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery. In vivo transduction of DCs, or other APCs, can be accomplished by administration of Ad (or other viral vectors) via different routes including intravenous, intramuscular, intranasal, intraperitoneal or cutaneous delivery. The preferred method is cutaneous delivery of Ad vector at multiple sites using a total dose of approximately lxl010-lx 1012 i.u. Levels of in vivo transduction can be roughly assessed by co-staining with antibodies directed against APC marker(s) and the TAA being expressed. The staining procedure can be carried out on biopsy samples from the site of administration or on cells from draining lymph nodes or other organs where APCs (in particular DCs) may have migrated. (Condon, et al. (1996) Nature Med. 2:1122-1128 and Wan, et al. (1997) Hum. Gene Ther. 8: 1355-1363). The amount of antigen being expressed at the site of injection or in other organs where transduced APCs may have migrated can be evaluated by ELISA on tissue homogenates. Although viral gene delivery is more efficient, DCs can also be transduced in vitro/ex vivo by non- viral gene delivery methods such as electroporation, calcium phosphate precipitation or cationic lipid/plasmid DNA complexes. (Arthur, et al. (1997) Cancer Gene Ther. 4:17-25). Transduced APCs can subsequently be administered to the host via an intravenous, subcutaneous, intranasal, intramuscular or intraperitoneal route of delivery. In vivo transduction of DCs, or other APCs, can potentially be accomplished by administration of cationic lipid/plasmid DNA complexes delivered via the intravenous, intramuscular, intranasal, intraperitoneal or cutaneous route of administration. Gene gun delivery or injection of naked plasmid DNA into the skin also leads to transduction of DCs. (Condon, et al. (1996) Nature Med. 2: 1122-1128; Raz, et al. (1994) Proc. Natl. Acad. Sci. USA 91:9519-9523). Intramuscular delivery of plasmid DNA may also be used for immunization. (Rosato, et al. (1997) Hμm. Gene Ther. 8:1451-1458.) The transduction efficiency and levels of transgene expression can be assessed as described above for viral vectors.
Adoptive Immunotherapy and Vaccines The expanded populations of antigen-specific immune effector cells of the present invention also find use in adoptive immunotherapy regimes and as vaccines. Adoptive immunotherapy methods involve, in one aspect, administering to a subject a substantially pure population of educated, antigen-specific immune effector cells made by culturing naive immune effector cells with APCs as described above. Preferably, the APCs are dendritic cells. In one embodiment, the adoptive immunotherapy methods described herein are autologous. In this case, the APCs are made using parental cells isolated from a single subject. The expanded population also employs T cells isolated from that subject. Finally, the expanded population of antigen-specific cells is administered to the same patient. In a further embodiment, APCs or immune effector cells are administered with an effective amount of a stimulatory cytokine, such as IL-2 or a co-stimulatory molecule. The agents identified herein as effective for their intended puφose can be administered to subjects having tumors previously determined to be reactive with any of the peptides of the invention as well as or in addition to individuals susceptible to or at risk of developing such tumors. Methods for identifying these individuals are described, infra. When the agent is administered to a subject such as a mouse, a rat or a human patient, the agent can be added to a pharmaceutically acceptable carrier and systemically or topically administered to the subject. To determine patients that can be beneficially treated, a tumor regression can be assayed. Therapeutic amounts can be empirically determined and will vary with the pathology being treated, the subject being treated and the efficacy and toxicity of the therapy. Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the puφose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents can be found below. The agents and compositions of the present invention can be used in the manufacture of medicaments and for the treatment of humans and other animals by administration in accordance with conventional procedures, such as an active ingredient in pharmaceutical compositions. More particularly, an agent of the present invention also referred to herein as the active ingredient, may be administered for therapy by any suitable route including nasal, topical (including transdermal, aerosol, buccal and sublingual), parental (including subcutaneous, intramuscular, intravenous and intradermal) and pulmonary. It will also be appreciated that the preferred route will vary with the condition and age of the recipient, and the disease being treated. The preceding discussion and examples are intended merely to illustrate the art. As is apparent to one of skill in the art, various modifications can be made to the above without departing from the spirit and scope of this invention.

Claims

CLAIMS "What is claimed is: 1. An isolated polynucleotide encoding a polypeptide selected from the group consisting of SEQ ID NOS: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42 and 44.
2. The isolated polynucleotide of claim 1 , fxirther comprising a carrier.
3. The isolated polynucleotide of claim 1 and a pharmaceutically acceptable carrier.
4. A host cell comprising a polynucleotide of claim 1.
5. The host cell of claim 4, wherein the host cell is an antigen presenting cell and the peptide is presented in the context of an MHC molecule.
6. The host cell of claim 5, wherein the antigen presenting cell is a dendritic cell.
7. A composition comprising the host cell of any one of claims 4 to 6, and a carrier.
8. A composition comprising the host cell of claim 4 and a pharmaceutically acceptable carrier.
9. An isolated polypeptide selected from the group consisting of SEQ ID NOS.: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42 and 44.
10. A host cell comprising the polypeptide of claim 9.
11. A composition comprising the polypeptide of claim 9 and a carrier.
12. A composition comprising the host cell of claim 10 and a carrier.
13. An antibody that specifically recognizes and binds the polypeptide of claim 9.
14. A composition comprising the antibody of claim 13 and a carrier.
15. An immune effector cell raised in the presence and at the expense of a host cell of claim 5.
16. A method for eliciting a cytolytic response in a subject comprising administering to said subject an effective amount of a composition of any one of claims 3, 8, 11, 12, or 14.
17. A composition comprising at least one immunogenic ligand, wherein said immunogenic ligand is individually characterized by an ability to elicit an immune response against the same native ligand, and wherein said immunogenic ligand is selected from the group consisting of SEQ ID NOS.: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42 and 44.
18. The composition of claim 1 , further comprising an immunogenic portion of SEQ ID NO: 18.
19. The composition of claim 18, further comprising a carrier.
20. The composition of claim 19, wherein the carrier is a pharmaceutically acceptable carrier.
21. A host cell comprising at least one immunogenic ligand, wherein said immunogenic ligand is individually characterized by an ability to elicit an immune response against the same native ligand, and wherein said immunogenic ligand is selected from the group consisting of SEQ ID NOs.: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42 and 44.
22. The host cell of claim 21 , wherein the host cell is an antigen presenting cell and the immunogenic ligands are presented on the surface of the cell.
23. The host cell of claim 22, wherein the antigen presenting cell is a dendritic cell.
24. A composition comprising the host cell of any of claims 21 to 23 and a carrier.
25. The composition of claim 24, wherein the carrier is a pharmaceutically acceptable carrier.
26. A method for inducing an immune response in a subject, comprising delivering to the subject a composition comprising an effective amount of at least one immunogenic ligand, wherein each of said immunogenic ligands is characterized by an ability to elicit an immune response against the same native ligand, and wherein said immunogenic ligand is selected from the group consisting of SEQ ID NOs.: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42 and 44.
PCT/US2004/033241 2003-10-10 2004-10-08 Immunogenic peptides WO2005037854A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US51063003P 2003-10-10 2003-10-10
US60/510,630 2003-10-10

Publications (2)

Publication Number Publication Date
WO2005037854A2 true WO2005037854A2 (en) 2005-04-28
WO2005037854A3 WO2005037854A3 (en) 2005-10-27

Family

ID=34465141

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/033241 WO2005037854A2 (en) 2003-10-10 2004-10-08 Immunogenic peptides

Country Status (1)

Country Link
WO (1) WO2005037854A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050059056A1 (en) * 2003-07-28 2005-03-17 Nicolette Charles A. Antigenic gp100 (154-162) compounds for therapy and diagnosis and methods for using same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050059056A1 (en) * 2003-07-28 2005-03-17 Nicolette Charles A. Antigenic gp100 (154-162) compounds for therapy and diagnosis and methods for using same

Also Published As

Publication number Publication date
WO2005037854A3 (en) 2005-10-27

Similar Documents

Publication Publication Date Title
JP2007518399A (en) Compositions and methods for diagnosing and treating lung cancer
JP2008527998A (en) GITR antibody for diagnosis of NSCLC
US20070065889A1 (en) Compositions and methods to diagnose and treat lung cancer
US6737062B2 (en) Immunogenic compositions
US20030147866A1 (en) Novel BGP compounds for therapy and diagnosis and methods for using same
JP2004505607A (en) Derivatives of breast cancer antigen HER-2 for therapeutic use
USRE44024E1 (en) Isolation and characterization of ECA1, a gene overexpressed in endometroid carcinomas of ovary and endometrium
US20020122791A1 (en) Antigenic CK-18 compounds for therapy and diagnosis and methods for using same
US20060014668A1 (en) Antigenic gp 100 compounds for therapy and diagnosis and methods of using same
WO2005037854A2 (en) Immunogenic peptides
US20050059056A1 (en) Antigenic gp100 (154-162) compounds for therapy and diagnosis and methods for using same
US20020197243A1 (en) Novel p53BP2 compounds for therapy and diagnosis and methods for using same
US6861408B2 (en) Therapeutic anti-melanoma compounds
WO2006078780A2 (en) Rdc1 antibodies for the diagnosis of nsclc
US20050196860A1 (en) Novel EPS8 compounds for therapy and diagnosis and methods for using same
US20020182218A1 (en) Therapeutic anti-melanoma compounds
WO2003050307A1 (en) Compounds for therapy and diagnosis and methods for using same
US20020187494A1 (en) PAR-3 compounds for therapy and diagnosis and methods for using same
US20030013075A1 (en) MART-1 compounds for therapy and diagnosis and methods for using same
WO2003070162A2 (en) Par-3 compounds for therapy and diagnosis and methods for using same
US20020155471A1 (en) Compounds for therapy and diagnosis and methods for using same
US20040138135A1 (en) Therapeutic compounds for ovarian cancer
WO2003050543A1 (en) Diagnosis of cancer using eif3 as a marker
WO2003064693A1 (en) Mart-1 compounds for therapy and diagnosis and methods for using same
CA2403684A1 (en) Therapeutic anti-melanoma compounds

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase