MX2008003464A - Identification of tumor-associated antigens for diagnosis and therapy - Google Patents

Identification of tumor-associated antigens for diagnosis and therapy

Info

Publication number
MX2008003464A
MX2008003464A MX/A/2008/003464A MX2008003464A MX2008003464A MX 2008003464 A MX2008003464 A MX 2008003464A MX 2008003464 A MX2008003464 A MX 2008003464A MX 2008003464 A MX2008003464 A MX 2008003464A
Authority
MX
Mexico
Prior art keywords
nucleic acid
tumor
associated antigen
expression
antibody
Prior art date
Application number
MX/A/2008/003464A
Other languages
Spanish (es)
Inventor
Sahin Ugur
Tureci Ozlem
Koslowski Michael
Usener Dirk
Original Assignee
Ganymed Pharmaceuticals Ag
Johannes Gutenberguniversitaet Mainz Vertreten Durch Den Praesidenten
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 Ganymed Pharmaceuticals Ag, Johannes Gutenberguniversitaet Mainz Vertreten Durch Den Praesidenten filed Critical Ganymed Pharmaceuticals Ag
Publication of MX2008003464A publication Critical patent/MX2008003464A/en

Links

Abstract

The invention relates to genetic products the expression of which is associated with cancer diseases. The invention also relates to the therapy and diagnosis of diseases in which the genetic products are expressed or aberrantly expressed, in particular cancer diseases.

Description

IDENTIFIED! * TUMOR-ASSOCIATED ANTIGENS FOR DIAGNOSIS AND THERAPY FIELD OF THE INVENTION Despite interdisciplinary approaches and the exhaustive use of classical therapeutic procedures, cancers still remain among the causes that lead to death. The most recent therapeutic concepts are aimed at incorporating the patient's immune system into the general therapeutic concept by using recombinant tumor vaccines and other specific measures such as antibody therapy. A prerequisite for the success of this strategy is the recognition of tumor-specific or tumor-associated antigens or epitopes by the patient's immune system whose effector functions will be improved in intervention. The tumor cells differ biologically and substantially from their non-malignant cells. These differences are due to the genetic alterations acquired during the development of the tumor and result, inter alia, also in the formation of altered molecular structures, qualitatively or quantitatively in the cancer cells. Tumor-associated structures of this type that are recognized by the host-specific immune system that The tumor is housed as tumor-associated antigens. The specific recognition of tumor-associated antigens, involves cellular and humoral mechanisms, which are two functionally interconnected units: the CD4 + and CD8 + T lymphocytes, recognize the processed antigens, presented in the molecules of the MHC (main histocompatibility complex) class I and class II, respectively, while B lymphocytes produce circulating antibody molecules that bind directly to unprocessed antigens. The potential clinical-therapeutic importance of tumor-associated antigens results from the fact that the recognition of antigens in neoplastic cells by the immune system leads to the initiation of cytotoxic effector mechanisms and, in the presence of helper T lymphocytes, it can cause the elimination of cancer cells (Pardoll, Nat. Med. 4: 525-31, 1998). Therefore, a central purpose of tumor immunology is to molecularly define these structures. The molecular nature of these antigens has been enigmatic for a long time. Only after the development of suitable cloning techniques, it has been possible to select libraries for tumor cDNA expression systematically for tumor-associated antigens, by analyzing the white structures of cytotoxic T lymphocytes (CTL, for its acronym in English) (van der Bruggen et al., Science 254: 1643-7, 1991) or by using autoantibodies in circulation (Sahin et al., Curr. Opin. Immunol., 9: 709- 16, 1997) as test solutions. For this purpose, libraries for cDNA expression were prepared from freshly prepared tumor tissue and expressed recombinantly as proteins in suitable systems. Immunoeffectors isolated from patients, namely CTL clones with tumor-specific lysis patterns, or circulating antibodies were used to clone the respective antigens.
BACKGROUND OF THE INVENTION In recent years, a multiplicity of antigens in various neoplasms have been defined by these procedures. However, the test solutions used for the identification of antigens in classical methods are immunoeffectors (circulating antibodies or CTL clones) from patients who usually already have advanced cancer. Several data indicate that tumors can lead, for example, to the tolerization and anergization of T lymphocytes and that, during the course of the disease, especially those specificities that could cause an effective immune recognition are they lose the immuno-effector repertoire. Current studies in patients have not yet produced any solid evidence of a real action from what was previously found and tumor-associated antigens are used. Therefore, it can not be ruled out that the proteins that evoke spontaneous immune responses are the undue target structures. An objective of the present invention is to provide target structures for a diagnosis and therapy of cancers. This objective is achieved by the subject matter of the claims. According to the invention, genes are identified, which are expressed selectively or aberrantly in tumor cells and in this way, provide tumor-associated antigens. These genes and / or their genetic products and / or the derivatives and / or fragments thereof, are useful as target structures for therapeutic and diagnostic procedures. The tumor-associated antigens, identified according to the invention, have an amino acid sequence encoded by a nucleic acid that is selected from the group consisting of a nucleic acid comprising (a) a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67 of the sequence listing, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid that degenerates with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a) , (b) or (c). In a preferred embodiment, a tumor-associated antigen, identified according to the invention has an amino acid sequence encoded by a nucleic acid that is selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67 of the sequence listing. In a preferred embodiment, a tumor-associated antigen, identified according to the invention, comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31 , 36, 40, 42, 46, 50-60, 63, 68, and 69 of the sequence listing, a portion or derivative thereof. The present invention is generally related to the use of tumor-associated antigens, identified according to the invention or to the parts or derivatives thereof, of nucleic acids encoding tumor-associated antigens, identified according to the invention or portions or derivatives thereof or acids nucleic acids directed against the coding nucleic acids, antibodies or T lymphocytes, directed against the tumor-associated antigens, identified according to the invention or portions or derivatives thereof and / or the host cells that express the tumor-associated antigens, identified according to the invention or portions or derivatives thereof for therapy, prophylaxis, diagnosis and / or monitoring of noplastic diseases. This may also involve the use of a combination of two or more of these antigens, nucleic acids, antibodies, T lymphocytes and / or host cells, in a mode also in combination with tumor-associated antigens, other than those identified in accordance with the invention. invention, the nucleic acids encoding the same or the nucleic acids directed against the coding nucleic acids, the antibodies or T lymphocytes directed against the tumor-associated antigens, and / or the host cells expressing the tumor-associated antigens. In those embodiments of the invention which relates to the use of antibodies directed against tumor-associated antigens, identified according to the invention or portions or derivatives thereof, T lymphocyte receptors directed against tumor antigens may also be used. -sociated, identified from according to the invention or portions or derivatives thereof, optionally in a complex with MHC molecules. Especially suitable for therapy, prophylaxis, diagnosis and / or monitoring is a portion of the tumor-associated antigens identified according to the invention, which corresponds to the non-transmembrane portion, in particular the extracellular portion of the tumor-associated antigens or understands them Therefore, according to the invention, a portion of the tumor-associated antigens will be identified according to the invention which corresponds to the non-transmembrane portion, in particular, the extracellular portion of the tumor-associated antigens or comprised of the same, or a portion corresponding to the nucleic acids encoding the tumor-associated antigens identified according to the invention, is preferred for therapy, prophylaxis, diagnosis and / or monitoring. Similarly, the use of antibodies directed against a portion of the tumor-associated antigens identified according to the invention, which corresponds to the non-transmembrane portion, in particular the extracellular portion of the tumor-associated antigens, is preferred. or they are comprised of them. Preferred diseases for therapy, prophylaxis and / or diagnosis are those in which one or more of the tumor-associated antigens identified according to the invention are selectively expressed or expressed abnormally. In addition, the invention relates to nucleic acids and proteins or peptides, which result from altered splicing (splice variants) of known genes or altered translation using alternative open reading frames. In this aspect, the invention relates to nucleic acids comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 28 and 49 of the sequence listing. In addition, in this aspect, the invention relates to proteins or peptides from the group consisting of SEQ ID NOs: 29 and 50 of the sequence listing. The altered splicing of a gene results in an altered transcription sequence (splicing variant). The translation of a splice variant in the region of its altered sequence results in an altered protein, which may be distinctly different in structure and functions from the original protein. The tumor-associated splice variants can produce tumor-associated transcripts and proteins / associated tumor antigens. These can be used as molecular markers, both for the detection of tumor cells and for the therapeutic target of tumors. The detection of tumor cells in a sample from a patient, can be carried out according to the invention, for example, after the extraction of nucleic acids by PCR amplification with variant-specific splicing oligonucleotides. In accordance with the invention, all systems for sequence-dependent detection are suitable for detection. These are, apart from PCR, for example, microplate / gene microarray systems, Northern transfer, RNSA protection analysis (RDA) and others. All detection systems have in common that the detection is based on a specific hybridization with at least one splice variant-specific nucleic acid sequence. However, tumor cells can also be detected according to the invention, by antibodies that recognize an epitope encoded by the splice variant. Antibodies can be prepared by using immunization peptides that are specific for the splice variant. Suitable for immunization are in particular amino acid sequences which are distinctly different from the variants of the gene product, which occur or are preferably produced in healthy cells. The detection of tumor cells with antibodies can be carried out on a sample isolated from the patient or as imaging with antibodies administered intravenously. In addition to the utility for diagnosis, splice variants having new or altered epitopes are attractive targets for immunotherapy, since these epitopes can be used for the assignment of antibodies or T lymphocytes, as described herein. In passive immunotherapy, antibodies or T lymphocytes, which recognize variant-specific splice epitopes are adoptively transferred here. As in the case of other antigens, the antibodies can also be generated by using standard technologies with the use of polypeptides that include these epitopes. Alternatively, it is possible to use nucleic acids coding for peptides containing the epitopes for immunization. Various techniques for in vitro or in vivo generation of epitope-specific T lymphocytes are known and described in detail (eg, Kessler JH, et al., 2001, Sahin et al., 1997) and are also based on the use of peptides containing the variant-specific splice epitopes or the nucleic acids encoding the peptides. Peptides that contain variant-specific splice epitopes or nucleic acids that code for peptides can also be used as substances pharmaceutically active in active immunotherapy (eg, vaccination, vaccine therapy).
SUMMARY OF THE INVENTION In one aspect, the invention relates to a pharmaceutical composition, comprising an agent that recognizes a tumor-associated antigen identified according to the invention or a nucleic acid that encodes the tumor-associated antigen and that preferably is selective for cells that have expression or abnormal expression of a tumor-associated antigen identified according to the invention. In a further aspect, the invention relates to a pharmaceutical composition comprising an agent that (I) inhibits the expression or activity of a tumor-associated antigen identified in accordance with the invention, and / or (II) has activity for inhibition of tumors or tumor destruction and is selective for cells that express or abnormally express a tumor-associated antigen identified according to the invention, and / or (III) when administered, selectively increases the amount of complexes between a HC molecule and a tumor-associated antigen identified according to the invention or a portion thereof, such as a peptide epitope. In particular modalities, the agent can induce the induction of cell death, the reduction in the cell growth, damage to the cell membrane or secretion of cytosines and preferably has an activity for tumor inhibition. In one embodiment, the agent is an anti-sense nucleic acid that hybridizes selectively with the nucleic acid encoding the tumor-associated antigen. In a further embodiment, the agent is a siRNA preferably comprising a sense RNA strand and an antisense RNA strand, wherein the sense and antisense strands of RNA form a duplex RNA, and wherein the strand of sense RNA comprises a nucleotide sequence virtually identical to a target sequence between about 19 to about 25 contiguous nucleotides in a nucleic acid encoding the tumor-associated antigen, preferably the mRNA encoding the tumor-associated antigen. In a further embodiment, the agent is an antibody that selectively binds to the tumor-associated antigen, in particular a complement-activated or toxin-conjugated antibody that selectively binds to the tumor-associated antigen. In a preferred embodiment, the antibody that selectively binds to the tumor-associated antigen, is coupled to a therapeutically useful substance and / or selects the natural or artificial effector mechanisms for cellular expression that abnormally expresses the tumor-associated antigen. In an additional mode, the agent is a cytotoxic T lymphocyte that recognizes the tumor-associated antigen or a portion thereof bound by a HC molecule in a cell and smooth the cells marked in this way. In a further embodiment, the agent is an auxiliary T lymphocyte that enhances the effector functions of other cells that specifically recognize the tumor-associated antigen or a portion thereof. In a further embodiment, the agent comprises two or more agents which, each recognize different tumor-associated antigens and / or inhibit the expression or activity of different tumor-associated antigens, and / or have activity for tumor inhibition or tumor destruction and are selective for cells that express or abnormally express different tumor-associated antigens, and / or when administered, selectively increase the amount of complexes between the MHC molecules and different tumor-associated antigens or portions thereof, wherein at least one of the different tumor-associated antigens is a tumor-associated antigen identified according to the invention. Preferably, a tumor-associated antigen, selectively limited to tumors, serves as a marker to select effector mechanisms for this specific location. The invention includes embodiments wherein the agent itself has no ability to inhibit the activity of an antigen tumor-associated or an activity for tumor inhibition or tumor destruction although it provides this effect, in particular when selecting effector mechanisms, in particular those that have cellular damage potential, towards a specific location, in particular a tumor or tumor cells. The activity of a tumor-associated antigen identified according to the invention can be any activity of a protein or a peptide. In one embodiment, this activity is an enzymatic activity. According to the invention, the phrase "inhibit the expression or activity" includes a complete or almost complete inhibition of the expression or activity and a reduction in expression or activity. The agent which, when administered, selectively increases the amount of complexes between an MHC molecule and a tumor-associated antigen identified according to the invention or a portion thereof comprises one or more components selected from the group consisting of (i) the tumor-associated antigen or a portion thereof, (ii) a nucleic acid encoding a tumor-associated antigen or a portion thereof, (iii) a host cell expressing the tumor-associated antigen or a portion thereof, and (iv) isolated complexes between the peptide epitopes from the tumor-associated antigen and a molecule of MHC. The invention further relates to a pharmaceutical composition comprising, one or more components selected from the group consisting of (i) a tumor-associated antigen identified according to the invention or a portion thereof, (ii) a nucleic acid encoding for a tumor-associated antigen identified according to the invention or a portion thereof, (iii) an antibody that binds to a tumor-associated antigen identified according to the invention or to a portion thereof, (iv) a nucleic acid antisense that hybridizes specifically with a nucleic acid encoding a tumor-associated antigen identified according to the invention, (v) an siRNA directed against a nucleic acid encoding a tumor-associated antigen identified according to the invention, ( vi) a host cell expressing a tumor-associated antigen identified according to the invention or a portion thereof, and (vii) isolated complexes between a tumor-associated tumor identified according to the invention or a portion thereof and an MHC molecule. In one embodiment, a nucleic acid encoding a tumor-associated antigen, identified according to the invention or a portion thereof is present in the pharmaceutical composition in an expression vector and is functionally links to a promoter. In a further embodiment, a nucleic acid encoding a tumor-associated antigen identified according to the invention or a portion thereof is present in the pharmaceutical composition in a virus as will be described further below. In a further embodiment, a host cell present in a pharmaceutical composition of the invention secretes the tumor-associated antigen or portion thereof, is expressed on the surface and preferably additionally expresses an MHC molecule, which binds to the tumor antigen -associated or the portion thereof. In one embodiment, the host cell endogenously expresses the MHC molecule. In a further embodiment, the host cell expresses the MHC molecule and / or the tumor-associated antigen or portion thereof in a recombinant manner. The host cell, preferably is non-proliferative. In a preferred embodiment, the host cell is a cell for presentation of antigens, in particular a dendritic cell, a monocyte or a macrophage. In a further embodiment, an antibody present in a pharmaceutical composition of the invention is a monoclonal antibody. In additional embodiments, the antibody is a chimeric or humanized antibody, a fragment of a natural antibody or an antibody synthetic. The antibody can be coupled to a therapeutically or diagnostically useful agent also referred to herein as a therapeutic or diagnostic agent. An antisense nucleic acid, present in a pharmaceutical composition of the invention, can comprise a sequence of 6-50, particularly 10- 30, 15-30 and 20-30, contiguous nucleotides of nucleic acid encoding the tumor-associated antigen identified according to the invention. In the further embodiments, a tumor-associated antigen or a portion thereof, provided by a pharmaceutical composition of the invention, either directly or via the expression of a nucleic acid, binds to MHC molecules on the surface of the cells, the binding preferably causes a cytolytic response and / or the induction of cytokine release. In particular embodiments of the siRNA that is assigned to the nucleic acid according to SEQ ID NO: 1, the sense RNA strand has the sequence of SEQ ID NO: 70 and the antisense RNA strand has the sequence of SEQ ID NO: 1. NO: 71, or the sense RNA strand has the sequence of SEQ ID NO: 72 and the antisense RNA strand has the sequence of SEQ ID NO: 73. A pharmaceutical composition of the invention, can comprise a carrier and / or an adjuvant pharmaceutically compatible A pharmaceutical composition of the invention is preferably used for the treatment or prevention of a disease characterized by the selective expression or abnormal expression of a tumor-associated antigen. In a preferred embodiment, the disease is a neoplastic disease, preferably cancer. In a preferred embodiment, the pharmaceutical composition of the invention is in the form of a vaccine that can be used therapeutically or prophylactically. This vaccine preferably comprises a tumor-associated antigen, identified according to the invention or a portion thereof, and / or a nucleic acid encoding a tumor-associated antigen, identified according to the invention or a portion thereof. In particular embodiments, the nucleic acid is present in a virus or host cell. The invention also relates to methods for the treatment or prevention, diagnosis or monitoring, that is, to determine the regression, progression, course and / or onset of a disease characterized by the expression or abnormal expression of one or more antigens. tumor-associated, identified according to the invention, preferably a neoplastic disease, in particular, cancer. In one modality, the treatment or prevention, comprises administering a pharmaceutical composition of the invention. Diagnostic methods and / or methods for monitoring in accordance with the invention are generally related to the detection and / or determination of the amount of one or more parameters selected from the group consisting of (i) a nucleic acid, which encodes for a tumor-associated antigen identified according to the invention, or a portion thereof, (ii) a tumor-associated antigen identified according to the invention, or a portion thereof (iii) an antibody against a tumor antigen- associated identified according to the invention or a portion thereof, and (iv) T lymphocytes, preferably cytotoxic or T helper lymphocytes, which are specific for a tumor-associated antigen identified according to the invention or a portion thereof and / or a complex between the tumor-associated antigen or a portion thereof and an MHC molecule, in a biological sample isolated from a patient, preferably from a patient having the disease , that you suspect that you have or fall ill with the disease or that you have a potential for the disease. The means for carrying out the detection and / or determination of the quantity are described herein and will be evident to someone with experience. Preferably, the presence of the nucleic acid, the tumor-associated antigen or the portion thereof, the antibody and / or the T lymphocytes and / or an amount of the nucleic acid, the tumor-associated antigen or the portion thereof, the antibody and / or the T lymphocytes, which is increased in comparison with a patient if the disease is indicative of the presence of the disease or a potential for the development of the disease. The methods for diagnosis and / or monitoring of the invention also include the modalities wherein by detecting or determining the amount of the nucleic acid, the tumor-associated antigen or the portion thereof, the antibody and / or the T lymphocytes. , it is possible that the metastatic behavior of the disease is assessed and / or predicted, where, preferably, the presence of the nucleic acid, the tumor-associated antigen or the portion thereof, the antibody and / or the T lymphocytes and / or an amount of the nucleic acid, the tumor-associated antigen or the portion thereof, the antibody and / or the T lymphocytes that are increased in comparison with a patient without the disease or without a metastasis of the disease is indicative of a metastatic behavior of the disease or a potential for a metastatic behavior of the disease. In particular embodiments, the detection or determination of the amount comprises: (i) contacting a biological sample with an agent that binds specifically to the nucleic acid encoding the tumor-associated antigen or portion thereof, the tumor-associated antigen or portion thereof, the antibody or portion thereof or the T lymphocytes, and (ii) detecting the formation or determination of the amount of a complex between the agent and the nucleic acid or portion thereof, the tumor-associated antigen or portion thereof, the antibody or portion thereof, or the T lymphocytes. In one embodiment, the disease is characterized by the expression of the abnormal expression of two or more different tumor-associated antigens and a detection or determination of the amount, comprises a detection or determination of the amount of two or more nucleic acids encoding the two or more different tumor antigens -associated or parts thereof, of two or more different tumor-associated antigens or parts thereof, of two or more antibodies that bind to the two or more different antigens tumor-associated antigens or parts thereof and / or two or more T lymphocytes specific for two or more different tumor-associated antigens or portions thereof, or complexes thereof with MHC molecules. In a further embodiment, the biological sample isolated from the patient is compared to a comparable normal biological sample. The methods of supervision according to the invention preferably comprises a detection and / or determination of the amount of one or more of the parameters mentioned above in a first sample at a first point in time and in an additional sample at a second point in time, wherein the course of the disease is determined by comparing the two samples. According to the invention, the detection of a nucleic acid or a portion thereof or the determination of the amount of a nucleic acid or a part thereof, can be carried out using an oligo- or polynucleotide assay solution, which hybridizes specifically with the nucleic acid or portion thereof or can be carried out by selective amplification of the nucleic acid or portion thereof, eg, by means of PCR amplification. In one embodiment, the test solution of oligo- or polynucleotides comprises a sequence of 6-50, in particular 10-30, 15-30 and 20-30, contiguous nucleotides of the nucleic acid. In particular embodiments, the tumor-associated antigen or portion thereof to be detected or the amount thereof that will be determined in the methods of the present invention is present intracellularly on the cell surface or in a complex with an MHC molecule. According to the invention, the detection of a A tumor-associated antigen or a portion thereof or the determination of the amount of a tumor-associated antigen or a portion thereof, can be carried out using an antibody that binds specifically to the tumor-associated antigen or portion thereof. According to the invention, the detection of an antibody or the determination of the amount of an antibody, can be carried out using a protein or peptide that specifically binds to the antibody. According to the invention, the detection or determination of the number of T lymphocytes, which are specific for a tumor-associated antigen or a portion thereof and / or a complex thereof with an MHC molecule, can be carried out using a cell that presents the complex between the tumor-associated antigen or the portion thereof and an MHC molecule. The T lymphocytes can additionally be detected by detecting their proliferation, their production of cytokines, and their cytotoxic activity activated by specific stimulation with a complex of an MHC molecule and a tumor-associated antigen or a portion thereof. The T lymphocytes can also be detected with the aid of a recombinant MHC molecule or a complex of two or more MHC molecules loaded with immunogenic fragments of one or more tumor-associated antigens.
An agent that is used for the detection or determination of the amount in the methods of the invention such as an assay solution of oligo- or polynucleotides, an antibody, a protein or peptide or a cell of preference is labeled in a detectable manner, in particular by a detectable label such as a radioactive label or an enzyme label. In a particular aspect, the invention relates to a method for the treatment, prevention, diagnosis or monitoring of a disease characterized by the expression or abnormal expression of a tumor-associated antigen, identified according to the invention, the method comprises administering a antibody that binds to the tumor-associated antigen or to a portion thereof and that is coupled to a therapeutic or diagnostic agent. The antibody can be a monoclonal antibody. In additional embodiments, the antibody is a chimeric or humanized antibody or a fragment of a natural antibody. In certain embodiments, the methods of the invention for diagnosis or monitoring of a disease characterized by the expression or abnormal expression of a tumor-associated antigen identified according to the invention are performed with a biological sample that contains or is suspected to contain cells. tumor in dissemination or metastatic tumor cells. These biological samples include, for example, blood, serum, bone marrow, sputum, bronchial aspirate, and / or bronchial lavage. In a particular aspect, the invention relates to a method for treating a patient having a disease characterized by the expression or abnormal expression of a tumor-associated antigen identified according to the invention, the method comprising: (i) providing a sample containing immunoreactive cells, obtained either from the patient or from another individual of the same species, in particular a healthy individual, or an individual of a different species, (ii) contacting the sample with a host cell expressing the tumor-associated antigen or a portion thereof, under conditions that favor the production of cytolytic T lymphocytes against the tumor-associated antigen or a portion thereof, and (iii) introduce the cytolytic T lymphocytes in the patient in a suitable amount to lyse cells that express the tumor-associated antigen or a portion thereof. In one embodiment, the method includes cloning the T lymphocyte receptor of the obtained cytolytic T lymphocytes and transferring the nucleic acid encoding the T lymphocyte receptor to the T lymphocytes, whether obtained from the patient or from another individual of the T cell. same species, in particular a healthy individual, or an individual of a different species, the T lymphocytes in this way receive the desired specificity and, like (iii), can be introduced into the patient. In one embodiment, the host cell endogenously expresses an MHC molecule. In a further embodiment, the host cell recombinantly expresses an MHC molecule and / or the tumor-associated antigen or portion thereof. Preferably, the host cell presents the tumor-associated antigen or the portion thereof by the MHC molecules on its surface. The host cell, preferably is non-proliferative. In a preferred embodiment, the host cell is a cell for the presentation of antigens, in particular a dendritic cell, a monocyte or a macrophage. The invention also relates to a method for the treatment of a disease characterized by the expression or abnormal expression of a tumor-associated antigen identified according to the invention, the method comprising: (i) identifying cells from the patient expressing abnormal amounts of the tumor-associated antigen, (ii) isolating a sample from the cells, (iii) culturing the cells, and (iv) introducing the cells into the patient in a suitable amount to activate an immune response to the cells.
The present invention further relates to a nucleic acid selected from the group consisting of (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17 , 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) ) or (c). The invention further relates to a nucleic acid, which encodes a protein or polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31, 36, 40, 42, 46, 50-60, 63, 68, and 69, a portion or derivative thereof. In a further aspect, the invention relates to a recombinant nucleic acid molecule, in particular a DNA or RNA molecule, comprising a nucleic acid of the invention. The invention also relates to host cells that contain a nucleic acid or a recombinant nucleic acid molecule of the invention. The host cell may also comprise a nucleic acid encoding an MHC molecule. In one modality, the host cell, endogenously expresses the MHC molecule. In a further embodiment, the host cell co-synthesizes the MHC molecule and / or the nucleic acid or the recombinant nucleic acid molecule of the invention or a portion thereof. Preferably, the host cell is not proliferative. In a preferred embodiment, the host cell is a cell for presentation of antigens, in particular a dendritic cell, a monocyte or a macrophage. In a further embodiment, the invention relates to oligonucleotides which hybridize with a nucleic acid identified according to the invention and which can be used as genetic test solutions or as "antisense" molecules. Nucleic acid molecules in the form of oligonucleotide primers or competent test solutions, which hybridize with a nucleic acid identified according to the invention or portions thereof, can be used for the search for nucleic acids that are homologous to the acid nucleic acid identified according to the invention, for example, by PCR amplification, Southern and Northern hybridization. Hybridization can be carried out under mild stringent conditions, more preferably, under medium stringent conditions and most preferably under high stringent conditions.
In a further aspect, the invention relates to a protein or peptide that is encoded by a nucleic acid selected from the group consisting of: (a) a nucleic acid, comprising a nucleic acid sequence selected from the group consisting of SEQ. ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b ) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid that degenerates with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c). In a preferred embodiment, the invention relates to a protein or peptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31, 36 , 40, 42, 46, 50-60, 63, 68, and 69, a portion or derivative thereof. In a further aspect, the invention relates to an immunogenic fragment of a tumor-associated antigen, identified according to the invention. The fragment preferably binds to an MHC molecule or an antibody, preferably to a human HLA receptor or a human antibody. According to the invention, a fragment preferably comprises a sequence of at least 6, in particular at least 8, at least 10, at least 12, at least 15, at least 20, at least 30 or at least 50, amino acids. In this aspect the invention relates, in particular, to a peptide having or comprising a sequence selected from the group consisting of SEQ ID NOs: 51-60, 68 and 69 of the sequence listing, a portion or derivative thereof . In a further aspect, the invention relates to an agent that binds to a tumor-associated antigen, identified according to the invention or to a portion thereof. In a preferred embodiment, the agent is a protein or peptide, in particular an antibody, a T lymphocyte receptor or an HC molecule. In additional embodiments, the antibody is a monoclonal, chimeric, or humanized antibody, an antibody produced by recombinant techniques, or a fragment of an antibody. In a preferred embodiment, the invention relates to an antibody that selectively binds to a complex of: (i) a tumor-associated antigen, identified therefrom according to the invention or a portion thereof and (ii) a molecule of MHC for which the tumor-associated antigen identified according to the invention or the portion thereof binds, with the antibody without binding to (i) or (ii) alone. In particular, the invention relates to a agent, in particular an antibody that specifically binds to a peptide having or comprising a sequence selected from the group consisting of SEQ ID NOs: 51-60, 68, and 69 of the sequence listing, a portion or derivative thereof . According to the invention, the term "binding" is preferably related to a specific binding. "Specific binding" means that an agent such as an antibody binds more potently to a target such as an epitope for which it is specific as compared to binding to another target. An agent binds more powerfully to a first target compared to a second target, if it joins the first target with a dissociation constant (KD) that is less than the dissociation constant for the second White. Preferably, the dissociation constant (KD) for the target to which the agent specifically binds is greater than 10 times, preferably greater than 20 times, more preferably more than 50 times, even more preferably more than 100 times, 200 times, 500 times or 1000 times less than the dissociation constant (KD) for the target to which the agent does not specifically bind. These specific antibodies, for example, can be obtained by immunization, using the peptides mentioned above.
The invention further relates to a conjugate between an agent of the invention that binds to a tumor-associated antigen identified according to the invention or to a portion thereof or an antibody of the invention and a therapeutic or diagnostic agent. In one embodiment, the therapeutic or diagnostic agent is a toxin. In a further aspect, the invention relates to an apparatus for detecting the expression or abnormal expression of a tumor-associated antigen identified according to the invention, the kit comprises agents for detecting or determining the amount (i) of the nucleic acid encoding for the tumor-associated antigen or a portion thereof, (ii) for tumor-associated antigen or a portion thereof, (iii) the antibodies that bind to the tumor-associated antigen or to a portion thereof, and / or (iv) ) T lymphocytes that are specific for the tumor-associated antigen or a part thereof or a complex thereof with an MHC molecule. In one embodiment, the nucleic acid detection agents or portion thereof are nucleic acid molecules for selective amplification of the nucleic acid, comprising, in particular, a sequence of 6-50, in particular 10-30, 15- 30 and 20-30, contiguous nucleotides of the nucleic acid.
BRIEF DESCRIPTION OF THE FIGURES The present invention is described in detail by the following figures and examples, which are used only for purposes of illustration and are not intended to be limiting. According to the description and examples, the additional modalities that are likewise included in the invention are accessible to the expert.
Figures 1A-1C show expression of ISC-468 RNAX. Fig. 1 (A). Investigations by RT-PCR with ISC-468-specific primers did not show significant expression within all tested normal tissues except the placenta. Fig. 1 (B). Expression of ISC-468 mRNA in head and neck, liver, kidney and colon carcinomas. Fig. 1 (C). Expression of ISC-468 mRNA in breast, ovarian and stomach carcinomas.
Figure 2. Quantitative PCR analysis of the expression of ISC-468 mRNA in normal control tissues and breast cancers Real-time PCR investigation with ISC-468-specific primers showed the expression of selective mRNA in testes, placenta, stomach and PBMC normal, and in all biopsies of breast carcinoma.
Figures 3A-3B show the expression ISC-507 specific in normal testis and prostate carcinoma. The analysis by RT-PCR with primers and gene-specific ISC-507 shows the amplification of the cDNA exclusively in normal testes Fig. 3 (A) and in biopsies of prostate carcinoma Fig. 3 (B).
Figure 4. Quantitative expression of ISC-507 Quantitative RT-PCR with ISC-507-specific primers shows selective expression in samples of testis, lymph nodes and prostate and samples of prostate cancer.
Figures 5A-5D show the expression ISC-466 in normal testes and various tumor samples. RT-PCR analysis with ISC-466-specific primers shows no expression within normal tissues except the placenta Fig. 5 (A), although there is expression in biopsies of head and neck carcinoma and in renal carcinoma biopsies Fig. 5 (B). Different expression was also detected in breast and lung carcinoma cell lines, as well as in ovarian carcinoma cell lines, Fig. 5 (C) and Fig. 5 (D).
Figure 6. Expression of ISC-518 mRNA The analysis by RT-PCR with ISC-518-specific primers showed no expression within normal tissues except the testes.
Figure 7. Quantitative expression of ISC-518 Quantitative RT-PCR showed high and selective expression in normal testes and in a pool of liver carcinoma.
Figures 8A-8D show the expression of ISC-477 in normal and tumor tissues Investigations by RT-PCR with ISC-477-specific primers, showed selective expression in placenta and normal ovarian tissue Fig. 8 (A) and high expression in carcinomas stomachs investigated Fig. 8 (B), breast, colon and lung carcinomas Fig. 8 (C), as well as in ovarian and pancreatic carcinoma samples Fig.8 (D).
Figures 9A-9C show the expression of ISC-489 mRNA Investigations by RT-PCR with ISC-489-specific primers, showed a selective expression in placental control tissue and additionally diverse Expression levels in lung carcinoma samples Fig. 9 (A), Fig. 9 (C), stomach carcinomas Fig. 9 (B), Fig. 9 (C), head and neck tumors Fig. 9 (C) and Liver carcinoma samples Fig. 9 (C).
Figures 10A-10D show the expression of ISC-461 in normal testes and various tumor samples Investigations by RT-PCR with ISC-461-specific primers showed selective expression in control tissue of placenta and additionally various expression levels in carcinomas of breast and melanomas Fig. 10 (B), as well as in breast carcinoma, lung carcinoma and melanoma cell lines Fig. 10 (C) and ovarian carcinoma cell lines Fig. 10 (D).
Figures 11A-11B show the expression of ISC-465 mRNA in samples derived from placenta and cancer. Investigations by RT-PCR with ISC-465-specific primers showed selective expression in placenta Fig. 11 (A) and in some cell lines derived from breast cancer, melanoma, lung cancer or stomach cancer Fig. 11 (B).
Figures 12A-12B show quantitative expression of Mem-030 Fig. 12 (A) Quantitative RT-PCR with em-030-specific primers showed significant overexpression in all head and neck carcinoma samples investigated. The following normal tissues were analyzed: bladder, brain, bone marrow, cervix, colon, duodenum, heart, lung, lymph nodes, breast, muscle, ovary, PBMC, PBMC-activated, placenta, prostate, retina, spleen, stomach, testes , thymus and tonsil. Fig. 12 (B). Prevalence of Mem-030 in esophageal, hepatic, uterine carcinomas and melanoma-derived tissues.
Figures 13A-13B show the quantitative expression of Mem-055 Quantitative RT-PCR with primers Mem-055-specific, shows high and selective expression in normal control tissues and a significant overexpression in tissues derived from stomach and lung cancer Fig. 13 (A). Mem-055, is also over-expressed in hepatic carcinomas, ovarian carcinomas and breast cancer samples Fig. 13 (B).
Figures 14A-14B show the expression of RNA, Mem-062 The analysis by means of RT-PCR with primers Mem-064-specific, shows the selective expression in testes and a weak expression in tissues derived from lung cancer Fig. 14 (A). Strong levels of significant expression of Mem-064 transcripts were detectable in various ovarian tumors Fig. 14 (B).
Figures 15A-15B show the expression Mem-068 specifies in normal testicles and renal cell carcinomas. The analysis by means of RT-PCR with gene-specific primers Mem-068, shows an amplification of the cDNA in normal testes, weak in placenta Fig. 15 ( A), in renal cell carcinomas and in stomach cancers Fig. 15 (B).
Figures 16A-16B show the expression Mem-071 in normal testicles and various tumor samples. RT-PCR analysis with Mem-071-specific primers show no expression within normal tissues except testes Fig. 16 (A). Different expression was also detected in samples of renal cell carcinoma and in stomach cancers Fig. 16 (B).
Figures 17A-17B show the expression of the ANm Mem-072 Analysis by RT-PCR with primers Mem-072-specific, did not show expression within normal tissues Fig. 17 (A) or significant expression in various lung cancer samples Fig. 17 (A) and Fig. 17 ( B).
Figures 18A-18B show the expression Mem-106 in normal and tumor tissues Investigations by RT-PCR with Mem-106-specific primers, showed no expression within normal tissues, except in testicles Fig. 18 (A) and were investigated. high expression in ovarian and prostatic carcinomas, as well as melanomas and colon cancer cell lines Fig. 18 (B).
Figures 19A-19C show mRNA expression Mem-131 Investigations by RT-PCR with Mem-131-specific primers did not show significant expression within all normal tissues tested except expression of PBMC mRNA. Mem-131 activated in breast and lung carcinoma. The expression of Mem-131 mRNA in lung and ovarian carcinomas.
Figures 20A-20B show mRNA expression ISC-468 Fig. 20 (A) RT-PCR and Fig. 20 (B) Real-time PCR, investigation with ISC-468-specific primers, showed selective mRNA expression in normal testes, placenta, and in 80% of breast carcinoma biopsies.
Figures 21A-21C show the immunofluorescence analysis of the expression ISC-468 Fig. 21 (A) the specificity of anti-ISC-468 antibodies, was confirmed by staining transfected cells in ISC-468-eGFP. Fig. 21 (B) staining of MeOH-fixed cells either transfected with ISC-468-specific RNAi duplexes, or control duplexes without slowing. Fig. 21 (C) by staining non-fixed cells either transfected with the duplexes of ISC-468-specific RNAi, or duplexes without slowing.
Figures 22A-22D show the immunohistochemical analysis of the expression ISC-468 No expression was detected in normal breast tissue Fig. 22 (A) lOOx, Fig. 22 (B) 200x. In contrast, strong staining of homogeneous membranes was observed in breast carcinoma specimens Fig. 22 (C) lOOx, Fig. 22 (D) 200x.
Figure 23. RNAi-induced disarming of the expression of ISC-468 mRNA Transfection of the cells with the duplexes of the ISC-468-specific siRNA, resulted in the unsetting of the different expression of the ISC-468 mRNA, in comparison with the control cells.
Figure 24. Cell proliferation analysis Transfection of cells with the duplexes of the ISC-468-specific siRNA resulted in distinct impairment of cell proliferation as compared to control cells.
Figures 25A-25B show the cell cycle analysis Transfection of cells with the ISC-468-specific duplex siRNA, resulted in the arrest of Gl / S in breast carcinoma cells Fig. 25 (A) MCF-7 and Fig. 25 (B) BT-549 compared to control cells.
Figure 26. AKT Phosphorylation Transfection of cells with the ISC-468-specific siRNA duplexes resulted in different impairment of AKT phosphorylation compared to control cells.
Figure 27. Inhibition of Proliferation Delivered by Antibodies Incubation of MCF-7 breast carcinoma cells with ISC-468-specific antibodies resulted in reduced proliferation compared to cells incubated with an irrelevant control antibody.
Figures 28A-28C show the analysis of cell proliferation Transfection of cells with the siRNA duplexes ISC-468-specific, resulted in deterioration other than Fig. 28 (A) chemotaxis, Fig. 28 (B) chemokinesis, and Fig. 28 (C) invasion compared to control cells.
Figure 29. Estrogen receptor correlation The expression levels of ISC-468 mRNA in breast carcinoma sample correlates with the estrogen receptor status. Median, 10th and 90th percentiles are shown with error bars.
Figure 30. Treatment with 17 -estradiol. Expression of ISC-468 mRNA was induced by treatment of the estrogen receptor-positive breast cancer cell line MCF-7 with 17 -estradiol. 100 mn. No induction was observed in the MDA-MB-231 cell line of the estrogen receptor.
DETAILED DESCRIPTION OF THE INVENTION According to the invention, a "reference" such as a reference sample or reference organism can be used to correlate and compare the results obtained in the methods of the invention from a test sample or organism test, that is, a patient. Typically, the reference organism is a healthy organism, in particular an organism that does not suffer from cancer. A "reference value" can be determined from a reference empirically by measuring a sufficiently large number of references. Preferably, the reference value is determined by measuring at least 2, preferably at least 3, preferably at least 5, preferably at least 8, preferably at least 12, preferably at least 20, preferably at least 30 , preferably at least 50, or preferentially at least 100 references. "Derivative" of a nucleic acid, means according to the invention, that individual or multiple such as at least 2, at least 4, or at least 6 and of preference up to 3, up to 4, up to 5, up to 6, up to 10, up to 15, or up to 20, substitutions, deletions and / or additions of nucleotides are present in the nucleic acid. In addition, the term "derivative" also comprises the chemical derivation of a nucleic acid in a nucleotide base, on sugar or on phosphate. The term "derivative" also comprises nucleic acids containing nucleotides and nucleotide analogs that do not occur in nature. According to the invention, a nucleic acid is preferably deoxyribonucleic acid (DNA) or ribonucleic acid (RNA). The nucleic acids comprise according to the invention DNA, cDNA, genomic mRNA, recombinantly produced and chemically synthesized molecules. According to the invention, a nucleic acid can be present as a single chain or double chain molecule and circularly enclosed, linearly or covalently. As used herein, the term "RNA" means a molecule that comprises at least one ribonucleotide residue. By "ribonucleotide", we mean a nucleotide with a hydroxyl group at the 2 'position of a beta-D-ribofuranose entity. The term includes double-stranded RNA, RNA single chain, isolated RNA such as, partially purified RNA, Essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA, which differs from RNA that occurs in nature by the addition, deletion, substitution and / or alteration of one or more nucleotides. These alterations may include the addition of non-nucleotide material, such as, to the end or ends of an RNA or internally, for example, to one or more nucleotides of the RNA. The nucleotides in the RNA molecules can also comprise non-standard nucleotides, such as nucleotides that do not occur in nature or chemically synthesized nucleosides or deoxynucleotides. These altered RNAs can be referred to as analogs or RNA analogues that occur in nature. The nucleic acids described according to the invention have preferably been isolated. The term "isolated nucleic acid", means according to the invention, that the nucleic acid was (i) amplified in vitro, for example, by polymerase chain reaction (PCR), (ii) produced recombinantly by cloning, (iii) purified, for example, by excision and gel electrophoretic fractionation or (iv) synthesized, for example, by chemical synthesis. An isolated nucleic acid is a nucleic acid that is available for manipulation by recombinant DNA techniques. A nucleic acid is "complementary" to another nucleic acid if the two sequences are able to hybridize and form a stable duplex with another, with hybridization that is preferably carried out under conditions that allow for specific hybridization between polynucleotides (stringent conditions) . Rigorous conditions are described, for example, in Molecular Cloning: A Laboratory Manual, J. Sambrook et al., Editors, 2nd Edition, Cold Spring Harbor Laboratory press, Cold Spring Harbor, New York, 1989 or Current Protocols in Molecular Biology, FM Ausubel et al., Editors, John iley & Sons, Inc., New York and refer, for example, to hybridization at 65 ° C in hybridization buffer (3.5 x SSC, 0.02% Ficoll, 0.02% polyvinylpyrrolidone, 0.02% bovine serum albumin, NaH2P04 2.5 mm (pH 7), 0.5% SDS, EDTA 2 mm). SSC is 0.15 M sodium chloride / 0.15 M sodium citrate, pH 7. After hybridization, the membrane to which the DNA has been transferred is washed, for example, in 2 x SSC at room temperature and then in 0.1- 0.5 x SSC / 0.1 x SDS at temperatures up to 68 ° C. According to the invention, the complementary nucleic acids have at least 40%, in particular at least 50%, at least 60%, at least 70%, at least 80%, at least less 90% and preferably at least 95%, at least 98% or at least 99%, identical nucleotides. The term "percent identity" is intended to denote a percentage of nucleotides of amino acid residues that are identical between the two sequences to be compared, obtained after the best alignment, this percentage will be simply statistical and the differences between the two sequences will be randomly distributed and about its total length. Sequence comparisons between two nucleotide or amino acid sequences will be carried out conventionally by comparing these sequences after they have been optimally aligned, the comparison will be carried out by segments or by means of a "comparison window" to identify and compare the local regions of the similarity of sequences. The optimal alignment of the sequences for comparison can be produced, also manually, by means of the local homology algorithm of Smith and Waterman, 1981, Ads App. Math. 2, 482, by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443, by means of the similarity search method of Pearson and Lipman, 1988, Proc. Nati Acad. Sci. USA 85, 2444, or by means of computerized programs that use these algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N and TFASTA in isconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, Wis.). The percentage identity is calculated by determining the numof identical positions between the two sequences to be compared, dividing their numby the numof positions compared and multiplying the result obtained by 100 to obtain the percentage identity between these two sequences. The nucleic acids encoding tumor-associated antigens can, according to the invention, be present alone or in combination with other nucleic acids, in particular, heterologous nucleic acids. In preferred embodiments, a nucleic acid is functionally linked to the sequences for expression control or regulatory sequences that can be homologous or heterologous with respect to the nucleic acid. A coding sequence and a regulatory sequence are linked "functionally" to each other, if they are covalently linked together, so that the expression or transcription of the coding sequence is carried out under the control or under the influence of the regulatory sequence. If the coding sequence will be translated into a functional protein, then, with a regulatory sequence functionally linked to the coding sequence, the induction of the regulatory sequence results in the transcription of the coding sequence, without causing a reading frame in the coding sequence or coding sequence that is not capable of being translated into the desired protein or peptide. The term "sequence for expression control" or "regulatory sequence", comprises according to the invention, enhancers and other control elements that regulate the expression of a gene. In particular embodiments of the invention, the sequences for expression control can be regulated. The exact structure of the regulatory sequences may vary as a function of the species or cell type, although in general it comprises 5 'untranslated and 5' untranslated sequences that are involved in the initiation of transcription and translation, respectively, such like the TATA box, the auction sequence, the CAAT sequence, and the like. More specifically, the non-transcribed 5 'regulatory sequences comprise a promoter region that includes a promoter sequence for the transcriptional control of the functionally linked gene. Regulatory sequences may also comprise enhancer sequences or activating sequences in the 5 'direction. According to the invention, a nucleic acid may additionally be present in combination with another nucleic acid encoding a peptide that controls the secretion of the protein or peptide encoded by the nucleic acid from a host cell. According to the invention, a nucleic acid may also be present in combination with another nucleic acid encoding a peptide that causes the encoded protein or peptides to anneal to the cell membrane of the host cell or to be divided into categories into particular organelles of the cell. Similarly, a combination with a nucleic acid may represent a reporter gene or any "brand". In a preferred embodiment, a recombinant nucleic acid molecule according to the invention is a vector, when suitable with a promoter, which controls the expression of a nucleic acid, for example, a nucleic acid encoding a tumor-associated antigen identified according to the invention. The term "vector", as used herein, is used in its broadest sense and comprises any intermediary vehicle for a nucleic acid that facilitates nucleic acid, for example, being introduced into prokaryotic cells and / or eukaryotic cells and, when appropriate , is integrated into a genome. The vectors of this type of preference are explained and / or expressed in the cells. An intermediary vehicle can be adapted, for example, to be used in electroporation, in the bombardment with microprojectiles, in liposomal administration, in the transfer with the help of agrobacteria or in the insertion by viruses with DNA or RNA. The vectors comprise plasmids, phagemids, bacteriophages or viral genomes. The nucleic acids encoding a tumor-associated antigen identified according to the invention can be used for the transfection of host cells. Nucleic acids, herein, mean both recombinant DNA and RNA. Recombinant RNA can be prepared by in vitro transcription of a DNA template. In addition, it can be modified by stabilizing sequences, by caption and polyadenylation before application. According to the invention, the term "host cell" is related to any cell that can be transformed or transfected with an exogenous nucleic acid. The term "host cells", comprises according to the invention, prokaryotic cells (e.g. E. coli) or eukaryotes (e.g., dendritic cells, B lymphocytes, CHO cells, COS cells, K562 cells, yeast cells and insect) . Particular preference is given to mammalian cells such as cells from humans, mice, hamsters, pigs, goats, primates. The cells can be derived from a multiplicity of tissue types and comprise, cells primary and cellular lines. Specific examples include keratinocytes, peripheral blood leukocytes, bone marrow stem cells and embryonic stem cells. In additional embodiments, the host cell is a cell for presentation of antigens, in particular a dendritic cell, monocyte or a macrophage. A nucleic acid may be present in the host cell in the form of an individual copy or two or more copies and, in one embodiment, is expressed in the host cell. According to the invention, the term "expression" is used in its most general meaning and includes the production of RNA or RNA and protein. It also comprises the partial expression of nucleic acids. In addition, the expression can be carried out transiently or stably. Preferred systems for expression in mammalian cells include pcDNA3.1 and pRc / CV (Invitrogen, Carlsbad, CA), which contain a selectable marker such as, a gene that imparts resistance to G418 (and thus allows transfected cell lines stably selected) and cytomegalovirus enhancer-promoter (CMV) sequences. In those cases of the invention, in which an MHC molecule, presents a tumor-associated antigen or a portion thereof, an expression vector may also comprise a nucleic acid sequence encoding the MHC molecule. The nucleic acid sequence encoding the MHC molecule may be present in the same expression vector as the nucleic acid encoding the tumor-associated antigen or portion thereof, or both nucleic acids may be present in different vectors expression. In the latter case, the two expression vectors can be co-transfected into a cell. If a host cell does not express the tumor-associated antigen or the portion thereof, nor the MHC molecule, both nucleic acids encoding them can be transfected into the cell either on the same expression vector or on different vectors of expression. If the cell already expresses the MHC molecule, only the nucleic acid sequence encoding the tumor-associated antigen or portion thereof can be transfected into the cell. The invention also encompasses kits for amplification of a nucleic acid encoding a tumor-associated antigen. These kits comprise, for example, a pair of amplification primers that hybridize to the nucleic acid encoding the tumor-associated antigen. The primers preferably comprise a sequence of 6-50, in particular 10-30, preferably 15- and 20-30 of contiguous nucleotides of the nucleic acid and are not overlapping, in order to avoid the formation of primer dimer. One of the primers will hybridize to a nucleic acid strand encoding the tumor-associated antigen, and the other primer will anneal to the complementary strand in an arrangement that allows amplification of the nucleic acid encoding the tumor-associated antigen. "Antisense molecules" or "antisense nucleic acids" can be used to regulate, in particular reduce, the expression of a nucleic acid. The term "antisense molecule" or "antisense nucleic acid", refers according to the invention to an oligonucleotide that is an oligo-ribonucleotide, oligo-deoxyribonucleotide, modified oligo-ribonucleotide or modified oligo-deoxyribonucleotide and that hybridizes under physiological conditions for DNA comprising a particular gene or for the mRNA of the gene, thus inhibiting transcription of the gene and / or translation of the mRNA. According to the invention, an "antisense molecule" also comprises a construct that contains a nucleic acid or a portion thereof in reverse orientation with respect to its natural promoter. An antisense transcript of a nucleic acid or a portion thereof can form a duplex with the mRNA present in the nature specifying the enzyme and thus avoiding the accumulation of a translation of the mRNA in the active enzyme. Another possibility is the use of ribozyme for the inactivation of a nucleic acid. Preferred antisense oligonucleotides according to the invention have a sequence of 6-50, in particular 10-30, 15-30 and 20-30, of contiguous nucleotides of white nucleic acid and preferably are completely complementary to the target nucleic acid or with a portion of it. In preferred embodiments, the antisense oligonucleotide hybridizes to an N-terminal or 5 'site, such as a site for translation initiation, a site for transcription initiation or a promoter site. In additional embodiments, the antisense oligonucleotide hybridizes with a 3 'untranslated region or a splice site with mRNA. In one embodiment, an oligonucleotide of the invention consists of, ribonucleotides, deoxyribonucleotides or a combination thereof with the 5 'end of one nucleotide and the 3' end of another nucleotide that will be linked together by a phosphodiester linkage. These oligonucleotides can be synthesized in a conventional manner or produced recombinantly. In preferred embodiments, an oligonucleotide of the invention is a "modified oligonucleotide". In the present, the oligonucleotide can be modified in very different ways, without impairing its ability to bind to its target, to increase, for example, its stability or therapeutic efficacy. According to the invention, the term "modified oligonucleotide" means an oligonucleotide in which (i) at least two of its nucleotides are linked together by a synthetic internucleoside link (i.e., an internucleoside link that is not a linkage) phosphodiester) and / or (ii) a chemical group that is not generally found in nucleic acids is covalently linked to the oligonucleotide. Synthetic internucleoside linkages are phosphorothioates, alkyl phosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidates, carboxymethyl esters and peptides. The term "modified oligonucleotide" also comprises oligonucleotides, which have a covalently modified base and / or sugar. "Oligonucleotides" comprise, for example, oligonucleotides modified with sugar residues that are covalently attached to organic groups of low molecular weight other than the hydroxyl group at the 3 'position and a phosphate group at the 5' position. The modified oligonucleotides can comprise, for example, a ribose residue 2'-0- alkylated or other sugar instead of ribose, such as arabinose. It should be understood that all of the embodiments described above with respect to the oligonucleotides can also be applied to polynucleotides. By "low-interference RNA" or "siRNA", in the sense in which it is used herein, an isolated RNA molecule should be understood, preferably, greater than 10 nucleotides in length, more preferably, greater than 15 nucleotides in length, and most preferably 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length which is used to identify a target gene or mRNA that it will be degraded. A variation of 19-25 nucleotides is the most preferred size for siRNAs. The siRNA according to the invention can comprise partially purified RNA, substantially pure RNA, synthetic RNA, or recombinantly produced RNA, as well as altered RNA that differs from the RNA that occurs in nature, by addition, deletion, substitution and / or alteration of one or more nucleotides. These alterations may include the addition of non-nucleotide material, such as to the end or ends of the siRNA or to one or more internal nucleotides of the siRNA; the modifications that make the siRNA resistant to nuclease digestion (eg, the use of 2'-substituted ribonucleotides or modifications to the sugar-phosphate structure); or the substitution of one or more nucleotides in the siRNA with deoxyribonucleotides. In addition, the siRNA can be modified to increase the stability thereof as described above for the modified oligonucleotides, in particular by introducing one or more phosphorothioate linkages. One or both strands of the siRNA can also comprise a 3 'overhang. In the sense in which it is used herein, a "3 'overhang" refers to at least one odd nucleotide extending from the 3' end of an RNA chain. Thus, in one embodiment, the siRNA comprises at least one 3 'overhang between 1 to about 6 nucleotides (which includes ribonucleotides or deoxynucleotides) in length, preferably 1 to about 5 nucleotides in length, most preferably 1 to about 4 nucleotides in length, and particularly preferably between about 2 to about 4 nucleotides in length. In the embodiment in which both strands of the siRNA molecule comprise a 3 'overhang, the length of the protrusions may be the same or different for each strand. In a more preferred mode, the outgoing 3 ', is present in both strands of the siRNA, and is 2 nucleotides in length. For example, each strand of the siRNA of the invention can comprise the 3 'overhangs of dideoxythylamic acid ("TT") or diuridylic acid ("uu"). To enhance the stability of the siRNA, the 3 'overhangs can also be stabilized against degradation. In one embodiment, the projections are stabilized by including purine nucleotides, such as adenosine or guanosine nucleotides. Alternatively, the substitution of pyrimidine nucleotides by modified analogues, for example, the substitution of uridine nucleotides in the 3 'overhang with 2'-deoxythymidine, is tolerated and does not affect the efficiency of RNAi degradation. In particular, the absence of a 2'-hydroxyl in the 2'-deoxythymidine significantly improves the nuclease resistance of the 3 'overhang in tissue culture medium. The sense and antisense strands of the siRNA can comprise two complementary single-stranded RNA molecules or can comprise an individual molecule in which the two complementary portions are the base pairs and are covalently linked by a "hairpin" area. That is, the sense region and the antisense region can be connected covalently via a binding molecule. The binding molecule can be a polynucleotide or linker without nucleotide. Without wishing to be bound by any theory, it is believed that the hairpin area of the last type of the siRNA molecule is cleaved intracellularly by the "dicer" protein (or its equivalent) to form an siRNA of two RNA molecules of pairs of individual bases. In the sense in which it is used herein, "white mRNA" refers to an RNA molecule that is a target for sub-regulation. The siRNA can be expressed from the pol III expression vectors without a change in the assignment site, such as the expression of the RNAs from the pol III promoters, it is only believed that it will be efficient when the first nucleotide transcribed is a purine The siRNA according to the invention can be assigned to any extension of approximately 19-25 contiguous nucleotides in any of the target mRNA sequences (the "target sequence"). Techniques for selecting the target sequences for the siRNA are provided, for example, in Tuschl T. et al., "The siRNA User Guide", revised on October 11, 2002, the entire exposure thereof being incorporated herein as reference. "The siRNA User Guide", the global mesh is available on a website maintained by Dr. Thomas Tuschl, Laboratory of RNA Molecular Biology, Rockefeller University, New York, USA, and can be found by accessing the Rockefeller University website and searching with the keyword "ARNsi". In this way, the sense strand of the siRNA of the present invention comprises a nucleotide sequence virtually identical to any contiguous extent between about 19 to about 25 nucleotides in the target mRNA. In general, a target sequence on the target mRNA can be selected from a given cDNA sequence corresponding to the target mRNA, preferably starting from 50 to 100 nt in the 3 'direction (ie, in the 3' direction) of the start codon. However, the target sequence can be located in the 5'- or 3'-untranslated regions, or in the region near the start codon. The siRNA can be obtained using various techniques known to those with experience in this field. For example, the siRNA can be chemically synthesized or recombinantly produced using methods known in the art, such as, the in vitro Drosophila system described in United States application published 2002/0086356 by Tuschl et al. The total disclosure thereof is incorporated herein by reference.
Preferably, the siRNA is synthesized chemically using appropriately protected ribonucleoside phosphoramidites and a conventional DNA / RNA synthesizer. The siRNA can be synthesized as two complementary RNA molecules, separately, or as an individual molecule of RNA with two complementary regions. Alternatively, the siRNA can also be expressed from recombinant circular or linear DNA plasmids, using any suitable promoter. These embodiments are included in accordance with the present invention, when referring herein to the administration of siRNA or the incorporation of siRNA in the pharmaceutical compositions. Promoters suitable for the expression of the siRNA of the invention of a plasmid include, for example, the pol II promoter sequences of U6 or Hl RNA and the cytomegalovirus promoter. The selection of other suitable promoters is within the skill in the art. The recombinant plasmids of the invention may also comprise inducible or regulatable promoters for expression of the siRNA in a particular tissue or in a particular intracellular environment. The siRNA, expressed from recombinant plasmids can either be isolated from systems for expression of cultured cells by standard techniques, or it can be expressed intracellularly. The use of recombinant plasmids to deliver the siRNA to cells in vivo is discussed in more detail below. The siRNA can be expressed from a recombinant plasmid either as two RNA molecules, complementary, separately, or as an individual RNA molecule with two complementary regions. The selection of suitable plasmids for siRNA expression, methods for inserting nucleic acid sequences to express the siRNA in the plasmid, and methods for delivering the recombinant plasmid to the cells of interest, are within the skill in the art. The siRNA can also be expressed from recombinant viral vectors intracellularly in vivo. The recombinant viral vectors comprise sequences encoding the siRNA and any promoter suitable for the expression of the siRNA sequences. Recombinant viral vectors may also comprise inducible or regulatable promoters for the expression of siRNA in a particular tissue or in a particular intracellular environment. The siRNA can be expressed from a recombinant viral vector, either as two complementary RNA molecules, separately, or as an individual RNA molecule with two complementary regions.
The term "peptide" comprises oligo- and polypeptides and refers to substances comprising two or more, preferably 3 or more, preferably 4 or more, preferably 6 or more, preferably 8 or more, preferably 10 or more , preferably 13 or more, preferably 16 more, preferably 21 or more and even preferably 8, 10, 20, 30, 40 or 50, in particular 100 amino acids covalently linked by peptide bonds. The term "protein" refers to large peptides, preferably to peptides with more than 100 amino acid residues, although in general the terms "peptides" and "proteins" are synonymous and are used interchangeably herein. Preferably, the proteins and peptides described according to the invention have been isolated. The terms "isolated protein" or "isolated peptide" mean that the protein or peptide has been separated from its natural environment. An isolated protein or peptide may be in an essentially purified state. The term "essentially purified" means that the protein or peptide is virtually free of other substances with which it is associated in nature or in vivo. These proteins and peptides can be used, for example, to produce antibodies and in an immunological or diagnostic analysis or as therapeutics. The Proteins and peptides described according to the invention can be isolated from biological samples such as tissues or cell homogenates and can also be expressed recombinantly in a multiplicity of eukaryotic or expression systems. For the purposes of the present invention, the "derivatives" of a protein or peptide or an amino acid sequence comprise variants for amino acid insertion, variants for the deletion of amino acids and / or variants for amino acid substitution. The variants for amino acid insertion comprise amino- and / or carboxy-terminal fusions and also the insertions of one or two or more amino acids in a particular amino acid sequence. In the case of amino acid sequence variants having an insert, one or more amino acid residues are inserted at a particular site in an amino acid sequence, although random insertion with the appropriate selection of the resulting product is also possible. Variants for amino acid suppression are characterized by the deletion of one or more amino acids from the sequence. Variants for amino acid substitution are characterized by at least one residue in the sequence that will be deleted and another residue that will be inserted in its place. Preferably they are provided to the modifications that are in the positions in the amino acid sequence that are not conserved between homologous proteins or peptides and / or to replace the amino acids with others having similar properties such as, hydrophobicity, hydrophilicity, electronegativity, volume of the side chain and the like (conservative substitution). Conservative substitutions, for example, are related to the exchange of an amino acid with another amino acid later listed in the same group as the amino acid to be substituted: 1. small, non-polar or slightly polar aliphatic residues: Ala, Ser, Thr ( Pro, Gly), 2. negatively charged residues and their amides: Asn, Asp, Glu, Gln, 3. positively charged residues: His, Arg, Lys, 4. Large aliphatic, non-polar residues: Met, Leu, Lie, Val (Cys), 5. Large aromatic residues: Phe, Tyr, Trp.
Due to its particular part in the protein architecture, three residues are shown in parentheses. Gly is the only residue without a side chain and in this way imparts flexibility to the chain. Pro has an unusual geometry that greatly restricts the chain.
Cys can form a disulfide bridge. The amino acid variants described above can be easily prepared with the aid of techniques for synthesis of known peptides such as, for example, by solid phase synthesis (Merrifield, 1964) and similar methods or by manipulation of recombinant DNA. The manipulation of DNA sequences for the preparation of proteins and peptides having substitutions, insertions or deletions is described in detail in Sambrook et al. (1989), for example. According to the invention, the "derivatives" of proteins and peptides also comprise one or multiple substitutions, deletions and / or additions of any molecules associated with the protein or peptide, such as carbohydrates, lipids and / or proteins or peptides. The term "derivative" also extends to all functional chemical equivalents of proteins and peptides. According to the invention, a part or fragment of a tumor-associated antigen preferably has a functional property of the protein or peptide from which it has been derived. These functional properties include the interaction with antibodies, the interaction with other peptides or proteins, the binding selective nucleic acids and an enzymatic activity. A particular property is the ability to form a complex with MHC molecules and, when appropriate, generate an immune response, preferably by stimulation of cytotoxic cells or helper T lymphocytes. A part or fragment of a tumor-associated antigen of the invention preferably comprises a sequence of at least 6, in particular at least 8, at least 10, at least 12, at least 15, at least 20, at least 30 or minus 50, of consecutive amino acids of the tumor-associated antigen. A part or fragment of a tumor-associated antigen of the invention, preferably comprises a sequence of up to 8, in particular up to 10, up to 12, up to 15, up to 20, up to 30 or up to 55 consecutive amino acids of the tumor-associated antigen . A part or fragment of a tumor-associated antigen is preferably a part of the tumor-associated antigen that corresponds to the portion that is not transmembrane, in particular the extracellular portion of the antigen, or is comprised thereof. Preferred parts or fragments of a tumor-associated antigen according to the invention in particular are suitable for the stimulation of cytotoxic T lymphocytes in vivo but also for the production of expanded and stimulated T lymphocytes for ex vivo therapeutic adoptive transfer.
A part or a fragment of a nucleic acid encoding a tumor-associated antigen is related according to the invention to the part of the nucleic acid, which codes for at least the tumor-associated antigen and / or for a part or a tumor-associated antigen fragment as defined above. A part or fragment of a nucleic acid encoding a tumor-associated antigen, preferably is a portion of nucleic acid corresponding to an open reading frame. According to the invention, particular modalities are necessary to involve the provision of dominant "negative" proteins or peptides derived from tumor-associated antigens. A dominant protein or negative peptide is an inactive variant of protein or peptide, to interact with the cellular machinery, displace an active protein or peptide from its interaction with the cellular machinery or compete with the active protein or peptide, thereby reducing the effect of active protein. Antisera that contain specific antibodies that bind specifically to the target protein can be prepared by various standard processes: see, for example, "Monoclonal Antibodies: A Practical Approach" by Philip Shepherd, Christopher Dean ISBN 0-19-963722-9; "Antibodies: A Laboratory Manual" by Ed Harlow, David Lane, ISBN: 0879693142 and "Using Antibodies: A Laboratory Manual: Portable Protocol NO" by Edward Harlow, David Lane, Ed Harlow ISBN 0879695447. With this it is also possible to generate cognate antibodies and specific ones that recognize complex membrane proteins in their natural form (Azorsa et al., J. Immunol., Methods 229: 35-48, 1999; Anderson et al., J. Immunol., 143: 1899-1904, 1989; Gardsvoll; , J. Immunol, Methods 234: 107-116, 2000). This is relevant in particular for the preparation of antibodies to be used therapeutically, but also for many diagnostic applications. With respect to this, it is possible to immunize with the whole protein, with extracellular partial sequences, as well as with cells that express the white molecule in physiologically folded form. Monoclonal antibodies are traditionally prepared using hybridoma technology. (For technical details see: "Monoclonal Antibodies: A Practical Approach" by Philip Shepherd, Christopher Dean ISBN 0-19-963722-9; "Antibodies: A Laboratory Manual" by Ed Harlow, David Lane ISBN: 0879693142; "Using Antibodies: A Laboratory Manual: Portable Protocol NO "by Edward Harlow, David Lane, Ed Harlow ISBN: 0879695447.) It is known that only a small portion of a The antibody molecule, paratope, is involved in the binding of the antibody to its epitope (see, Clark, WR (1986), The Experimental Foundations of Modern Immunology, Wiley &Sons, Inc., New York; Roitt, I. ( 1991), Essential Immunology, 7th Edition, Blackwell Scientific Publications, Oxford). The pFc 'and Fe regions are, for example, effectors of the complementary cascade although they are not involved in the binding with antigens. An antibody from which the pFc 'region has been enzymatically removed or which has been produced without the pFc' region, referred to as the F (ab ') 2 fragment, carries both binding sites with antigens of a complete antibody. Similarly, an antibody from which the Fe region has been enzymatically removed or that has been produced without the Fe region, referred to as the Fab fragment, carries an antigen binding site of an intact antibody molecule. In addition, the Fab fragments consist of a light chain covalently linked to an antibody and part of the heavy chain of the antibody, designated as Fd. Fd fragments are the main determinants of antibody specificity (an individual Fd fragment can be associated with up to ten different light chains, without altering the specificity of the antibody) and Fd fragments, when isolated, maintain the ability to bind to an epitope. Located inside the union part with antigens of an antibody are the regions for complementary determination (CDR, for its acronym in English) that interact directly with the epitope of the antigen and structural regions (FR, for its acronym in English) that maintain the tertiary structure of paratope. Both the Fd fragment of the heavy chain and the light chain of IgG immunoglobulins contain four structural regions (FR1 to FR4) that are separated in each case by three regions for complementary determination (CDR1 to CDR3). The CDRs and, in particular, the CDR3 regions and, even more particularly, the CDR3 region of the heavy chain are responsible to a large extent for the specificity of antibodies. The non-CDR regions of a mammalian antibody are known to be capable of being replaced by similar regions of antibodies with the same or a different specificity, with the specificity for the epitope of the original antibody to be maintained. This makes possible the development of "humanized" antibodies in which the non-human CDRs are covalently linked to the FR regions and / or human Fc / pFc regions to produce a functional antibody. As another example, WO 92/04381, describes the production and use of humanized murine RSV antibodies in which at least part of the murine FR regions have been replaced with the FR regions of human origin. Antibodies of this type, including fragments of intact antibodies capable of binding antigens, are often referred to as "chimeric" antibodies. According to the invention, the term "antibody" also includes the F (ab ') 2 Fab, Fv, and Fd fragments of antibodies, chimeric antibodies in which the Fe and / or FR regions and / or CDRl and / or CDR2 and / or light chain CDR3 have been replaced with human or non-human homologous sequences, chimeric antibodies of the F (ab ') 2 fragment in which the FR and / or CDR1 and / or CDR2 and / or light chain CDR3 regions have replaced, with human or non-human homologous sequences, antibodies of the chimeric Fab fragment in which the FR and / or CDR1 and / or CDR2 and / or light chain CDR3 regions have been replaced with human or non-human homologous sequences, and chimeric Fd fragment antibodies in which the FR and / or CDR1 and / or CDR2 regions have been replaced with human or non-human homologous sequences. The term "antibody" also comprises "single chain" antibodies. The invention also comprises proteins and peptides that specifically bind to tumor-associated antigens. The binding substances of this type can be provide, for example, by degenerating peptide libraries that can be prepared simply in solution in an immobilized form or as libraries for phage display. Likewise, it is possible to prepare combinatorial libraries of peptides with one or more amino acids. Libraries of synthetic residues of peptoids and non-peptides can also be prepared. The antibodies can also be coupled to specific diagnostic substances to display cells and tissues that express the tumor-associated antigens. They can also be coupled to therapeutically useful substances. The substances for diagnosis include any label that functions to: (i) provide a detectable signal; (ii) interacting with a second mark to modify the detectable signal, provided by the first or second mark, for example FRET (Fluorescent Resonance Energy Transfer); (iii) affecting mobility, e.g., electrophoretic mobility, by loading, hydrophobicity, shape, or other physical parameters, or (iv) providing a capture entity, e.g., affinity, antibody / antigen, or ionic complexation. Suitable structures as a brand are, such as, fluorescent labels, luminescent labels, brands with chromophores, radioisotopic labels, isotopic labels, preferably stable isotopic labels, isobaric labels, enzymatic labels, particle labels, in particular, metal particle labels, magnetic particle labels, polymeric particle labels, small organic molecules such as, biotin, receptor ligands or binding molecules such as, proteins or cell adhesion lectins, sequences for marking comprising, nucleic acids and / or amino acid residues that can be detected by the use of binding agents, etc. The diagnostic substances comprise, in a non-limiting manner, barium sulfate, iocetamic acid, iopanoic acid, calcium ipodate, diatrizoate sodium, methylglucamine diatrizoate, metrizamide, sodium thyropanoate and radiodiagnosis, including positronic emitters such as fluorine. 18 and carbon-11, gamma emitters, such as iodine-123, technetium-99m, iodine-131 and indium-111, nuclides for nuclear magnetic resonance, such as fluorine and gadolinium. According to the invention, the term "therapeutically useful substance" means any molecule that can exert a therapeutic effect. According to the invention, a therapeutically useful substance is preferably selectively guided to a cell that expresses one or more tumor-associated antigens and includes anticancer agents, iodine-labeled compounds radioactive, toxins, cytostatic or cytolytic drugs, etc. Anticancer agents comprise, for example, aminoglutethimide, azathioprine, bleomycin sulfate, busulfan, carmustine, chlorambucil, cisplatin, cyclophosphamide, cyclosporine, cytarabidine, dacarbazine, dactinomycin, daunorubin, doxorubicin, taxol, etoposide, fluorouracil, interferon-a, lomustine, mercaptopurine, methotrexate, mitotane, procarbazine HC1, thioguanine, vinblastine sulfate and vincristine sulfate. Other anticancer agents are described, for example, in Goodman and Gilman, "The Pharmacological Basis of Therapeutics", 8th Edition, 1990, McGraw-Hill, Inc., in particular Chapter 52 (Antineoplastic Agents (Paul Calabresi and Bruce A. Chabner The toxins can be proteins such as, antiviral weed in bag, cholera toxins, pertussis toxin, ricin, gelonin, abrin, diphtheria exotoxin or Pseudomonas exotoxin.The toxin residues, can also be radionuclides for high-energy emission such as cobalt-60. The term "major histocompatibility complex" or "MHC" is related to a complex of genes present in all vertebrates.MHC proteins or molecules are involved in signaling between lymphocytes and cells for presentation of antigens in normal immune reactions by binding peptides and presenting them for recognition by T lymphocyte receptors (TCRs). MHC molecules bind to peptides within an intracellular processing compartment and present these peptides on the surface of cells for presentation of antigens for recognition by T lymphocytes. The human MHC region, also referred to as HLA, it is located on chromosome 6 and includes the class I and class II regions. In a preferred embodiment of all aspects of the invention, an MHC molecule is an HLA molecule. "Reduce" or "inhibit". In the sense in which they are used herein, they mean the ability to cause a general decrease, preferably of 20% or greater, of greater preference of 50% or greater, and most preferably of 75% or greater, in the level, for example, at the level of protein or mRNA compared to a reference sample (eg, a sample not treated with siRNA). This reduction or inhibition of RNA or protein expression can occur through the cleavage or degradation of the designated mRNA. Analyzes for protein expression or nucleic acid expression are known in the art and include, for example, ELISA, Western blot analysis for protein expression, and Northern blot or assays for RNase protection for RNA. The term "patient" means according to the invention a human being, a non-human primate or other animal, in particular a mammal such as for example, cow, horse, pig, sheep, goat, dog, cat or a rodent such like, a mouse and a rat. In a particularly preferred embodiment, the patient is a human being. "Abnormal expression" means according to the invention that the expression is altered, preferably increases, compared to the state in a healthy individual. According to the invention the term "increased" or "quantity increases", preferably refers to an increase of at least 10%, in particular at least 20%, at least 50% or at least 100%. The amount of a substance is also increased in a test sample such as a biological sample compared to a reference sample if it is detectable in the test sample although absent or not detectable in the reference sample. According to the invention, the term "disease" refers to any pathological state in which the tumor-associated antigens are expressed or expressed abnormally. The "abnormal expression" means according to the invention, that the expression is altered preferably increased in comparison with the state in a healthy guy An increase in expression refers to an increase of at least 10%, in particular at least 20%, at least 50% or at least 100%. In one embodiment, the tumor-associated antigen is expressed only in the tissue of a diseased individual, while the expression is repressed in a healthy individual. An example of this disease is cancer, wherein the term "cancer" according to the invention comprises, leukemias, seminomas, melanomas, teratomas, lymphomas, neuroblastomas, gliomas, rectal cancer, endometrial cancer, renal cancer, adrenal cancer, cancer thyroid, blood cancer, skin cancer, brain cancer, cervical cancer, intestinal cancer, liver cancer, colon cancer, stomach cancer, intestinal cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophageal cancer, cancer colorectal cancer, pancreatic cancer, cancer of the ear, nose and throat (ENT, for its acronym in English), breast cancer, prostate cancer, cancer of the uterus, ovarian cancer and lung cancer and their metastasis. Examples thereof are lung carcinomas, breast carcinomas, prostate carcinomas, colon carcinomas, renal cell carcinomas, cervical carcinomas, or metastases of the types of cancer or tumors described above. The term "cancer" according to the invention also comprises metastasis of cancer. By "tumor", it should be understood an abnormal group of cells or tissue that grows by rapid uncontrolled cell proliferation and continues to grow after the stimuli that initiated the new growth end. Tumors show a partial or complete lack of structural organization and functional coordination with normal tissue, and usually form a distinct mass of tissue, which may be either benign or malignant. By "metastasis", the spread of cancer cells from their original site to another part of the body must be understood. The formation of metastases is a very complex process and depends on the separation of the malignant cells from the primary tumor, the invasion of the extracellular matrix, the penetration of the endothelial basement membranes to enter the body cavity and blood vessels, and then, after being transported by blood, infiltration to target organs. Finally, the growth of a new tumor in the target site depends on angiogenesis. Tumor metastasis often occurs even after removal of the primary tumor because the cells or tumor components can remain and develop a metastatic potential. In one modality, the term "metastasis", according to the invention is related to "distant metastasis", which is related to a metastasis that is remote from the primary tumor and the regional lymph node system. According to the invention, a biological sample can be a tissue sample, including body fluids, and / or a cell sample and can be obtained in the conventional manner such as, by tissue biopsy, including puncture biopsy, and the extraction of blood, bronchial aspirate, sputum, urine, feces or other bodily fluids. According to the invention, the term "biological sample" also includes the fractions of biological samples. According to the invention, the term "immunoreactive cell" means a cell that can mature in an immune cell (such as, B lymphocyte, helper T lymphocyte, cytolytic T lymphocyte) with adequate stimulation. Immunoreactive cells comprise hematopoietic CD34 + hematopoietic cells, immature and mature T lymphocytes and immature and mature B lymphocytes. If a production of cytolytic cells or helper T lymphocytes that recognize a tumor-associated antigen is desired, the immunoreactive cell is contacted with a cell that expresses a tumor-associated antigen, under conditions that favor production, differentiation and / or selection. of cytolytic T lymphocytes and of auxiliary T lymphocytes. The differentiation of T lymphocyte precursors into a cytolytic T lymphocyte, when exposed to an antigen, is similar to the clonal selection of the immune system. The terms "T cell" and "T lymphocyte" are used interchangeably herein and include helper T cells and cytotoxic T lymphocytes comprising cytolytic T lymphocytes. Some therapeutic methods are based on a reaction of a patient's immune system, which results in a lysis of cells presenting antigens such as cancer cells that present one or more tumor-associated antigens. In this connection, for example, autologous cytotoxic T lymphocytes, specific for a complex of a tumor-associated antigen and an MHC molecule, are administered to a patient having a cellular abnormality. The production of these cytotoxic T lymphocytes in vitro is known. An example of a method for differentiating T lymphocytes can be found in WO-A-9633265. In general, a sample containing cells such as blood cells is removed from the patient and the cells are contacted with a cell that has the complex and which can cause the spread of cytotoxic T lymphocytes (e.g., dendritic cells). The white cell, can be a transfected cell such as, a COS cell. These transfected cells present the desired complex on their surface and, when placed in contact with the cytotoxic T lymphocytes, stimulate the propagation of the latter. Clonally expanded autologous cytotoxic T lymphocytes are then administered to the patient. In another method for selecting cytotoxic, antigen-specific T lymphocytes, the fluorogenic tetramers of the MHC class I peptide / molecule complexes are used to obtain specific clones of cytotoxic T lymphocytes (Altman et al., Science 274: 94- 96, 1996; Dunbar et al., Curr. Biol. 8: 413-416, 1998). The present invention also includes the therapeutic methods referred to as adoptive transfer (Greenberg, J., Immunol., 136 (5): 1917, 1986; Riddel et al., Science 257: 238, 1992; Lynch et al., Eur. J. Immunol., 21: 1403-1410, 1991; Kast et al., Cell 59: 603-614, 1989), wherein the cells presenting the desired complex (e.g., dendritic cells) are combined with the patient's cytotoxic T lymphocytes. which will be treated, resulting in a spread of specific cytotoxic T lymphocytes. The propagated cytotoxic T lymphocytes are then administered to a patient having a cellular abnormality characterized by particular abnormal cells presenting the specific complex. T lymphocytes Cytotoxic then lyses the abnormal cells, thereby achieving a desired therapeutic effect. In addition, cells that exhibit the desired complex (e.g., dendritic cells) can be combined with cytotoxic T lymphocytes from healthy individuals or other species (e.g., mouse) which may result in the spread of specific cytotoxic T lymphocytes. with high affinity. The high affinity T lymphocyte receptor of these specific propagated T lymphocytes can be cloned and optionally humanized to a different degree, and the T lymphocyte receptors thus obtained, then transduced via gene transfer, for example, using retroviral vectors, inside the T lymphocytes of patients. Adoptive transfer can then be carried out using these genetically altered T lymphocytes (Stanislawski et al., Nat Immunol 2: 962-70, 2001, Kessels et al., Nat Immunol., 2: 957-61, 2001). Adoptive transfer is not the only form of therapy that can be applied according to the invention. The cototóxicos T lymphocytes can also be generated in vivo, in a manner known per se. One method uses monoproliferative cells that express the complex. The cells used here will be those that usually express the complex, such as irradiated tumor cells or cells transfected with one or both genes necessary for the presentation of the complex (ie, the antigenic peptide and the presentation of the MHC molecule). Another preferred form is the introduction of the tumor-associated antigen, for example, in the form of recombinant RNA which can be introduced into the cells by liposomal transfer or by electroporation. The resulting cells present the complex of interest and are recognized by autologous cytotoxic T lymphocytes that then propagate. A similar effect can be achieved by combining the tumor-associated antigen or a fragment thereof with an adjuvant so that incorporation into antigen-presenting cells in vivo is possible. The tumor-associated antigen or a fragment thereof can be represented as a protein, as DNA (for example, within a vector) or as RNA. The tumor-associated antigen is processed to produce a participant peptide for the MHC molecule, while a fragment thereof can be presented without the need for an additional process. The latter is the case in particular, if they can be linked to the MHC molecules. Preferably, it is provided for the administration forms, in which the complete antigen is processed in vivo by a dendritic cell, since this can also produce responses of the helper T lymphocytes that are necessary for an effective immune response (Ossendorp et al., Immunol Lett 74: 75-9, 2000; Ossendorp et al., J. Exp. Med. 187: 693-702, 1998). In general, it is possible to administer an effective amount of the tumor-associated antigen to a patient by intradermal injection, for example. However, the injection can also be carried out intranodally in a lymph node (Aloy et al., Proc Nati Acad Sci USA 98: 3299-303, 2001). The pharmaceutical compositions and methods for treatment described, according to the invention can also be used for immunization or vaccination to therapeutically treat or prevent a disease described herein. According to the invention, the terms "immunization" or "vaccination", preferably are related to an increase or activation of an immune response to an antigen. It is possible to use animal models to test an immune effect in cancer by using a tumor-associated antigen or a nucleic acid encoding it. For example, human cancer cells can be introduced into a mouse to generate a tumor, and one or more nucleic acids encoding tumor-associated antigens can be administered. The effect on cancer cells (for example, the reduction in tumor size) can be measured as a measurement for the effectiveness of an immunization by nucleic acid. As part of the composition for an immunization or a vaccination, preferably one or more tumor-associated antigens or stimulatory fragments thereof are administered together with one or more adjuvants by inducing an immune response or to increase an immune response. An adjuvant is a substance that is incorporated into the antigen or is administered together with the latter and that enhances the immune response. Adjuvants can enhance the immune response by providing a deposit of antigens (extracellularly or in macrophages), by activating macrophages and / or by stimulating particular lymphocytes. Adjuvants are known and understood in a non-limiting lipid A form of monophosphoryl (MPL, SmithKline Beecham), saponins such as QS21 (SmithKline Beecham), DQS21 (SmithKline Beecham, WO 96/33739), QS7, QS17, QS18 and QS -L1 (So et al., Mol.Cell 7: 178-186, 1997), incomplete Freund's adjuvants, complete Freund's adjuvant, vitamin E, montanide, alum, CpG oligonucleotides (see, Kreig et al., Nature 374 : 546-9, 1995) and various water-in-oil emulsions prepared from biologically degradable oils such as squalene and / or tocopherol. Preferably, the peptides are administered in a mixture with DQS21 / PL. The ratio of DQS21 to MPL is typically from about 1:10 to 10: 1, preferably from about 1: 5 to 5: 1 and in particular about 1: 1. For administration to humans, a vaccine formulation typically contains DQS21 and MPL in a variation between approximately 1 μ? up to approximately 100 μ ?. Other substances that stimulate a patient's immune response can also be administered. It is possible, for example, to use cytokines in a vaccination, owing to their regulatory properties on lymphocytes. These cytokines comprise, for example interleukin-12 (IL-12) which was shown to increase the protective actions of vaccines (see, Science 268: 1432-1434, 1995), GM-CSF and IL-18. There are several compounds that enhance an immune response and that can therefore be used in a vaccination. The compounds comprise the co-stimulatory molecules provided in the form of proteins or nucleic acids such as, B7-1 and B7-2 (CD80 and CD86, respectively). The invention also provides administration of nucleic acids, proteins or peptides. The proteins and peptides can be administered in a manner known per se. In one embodiment, the nucleic acids are they administer by ex vivo methods, that is, by removing cells from a patient, by genetically modifying the cells to incorporate a tumor-associated antigen and by reintroducing the altered cells into the patient. This generally comprises the introduction of a functional copy of a gene into the cells of a patient in vitro and the reintroduction of the cells genetically altered in the patient. The functional copy of the gene is under the functional control of regulatory elements that allow the gene to be expressed in genetically altered cells. Transfection and transduction methods are known to the skilled person. The invention also provides for the delivery of nucleic acids in vivo by using vectors such as viruses and white-controlled liposomes. If reference is made according to the invention to the administration or incorporation into pharmaceutical compositions of nucleic acids, this includes the embodiments wherein the nucleic acid is present in these vectors. In a preferred embodiment, a virus or viral vector for administering a nucleic acid encoding a tumor-associated antigen, is selected from the group consisting of adenovirus, adenovirus associated, pox virus, including vaccinia virus, and pox virus. attenuated viruses, Semliki Forest virus, retroviruses, Sindbis virus and particles similar to the Ty virus. Particular preference is given to adenoviruses and retroviruses. Retroviruses typically have poor replication (ie, they are incapable of generating infectious particles). Methods for introducing nucleic acids into cells in vitro or in vivo comprise the transfection of calcium phosphate nucleic acid precipitates, the transfection of nucleic acids associated with DEAE, the transfection or infection with the above viruses carrying the nucleic acids of interest, the transfection supplied by liposomes, and the like. In particular modalities preference is given to the. direction of the nucleic acid towards particular cells. In these embodiments, a carrier is used to deliver a nucleic acid to a cell (e.g., a retrovirus or a liposome) may have a bound white control molecule. For example, a molecule such as an antibody specific for a surface membrane protein on the target cell or a ligand for a receptor on the target cell can be incorporated or bound to the nucleic acid carrier. Preferred antibodies comprise antibodies that selectively bind to the tumor-associated antigen. If the administration of a nucleic acid via liposomes is desired, the binding proteins to a surface membrane protein associated with endocytosis can be incorporate into the liposome formulation to make control and / or absorption of assignment possible. These proteins comprise capsid proteins or fragments thereof that are specific to a particular cell type, antibodies to proteins that are internalized, proteins that are directed to an intracellular site, and the like. The therapeutic compositions of the invention can be administered in pharmaceutically compatible preparations. These preparations may generally contain pharmaceutically compatible concentrations of salts, buffer substances, preservatives, carriers, substances for enhancement of suppressive immunity, such as, adjuvants, for example, CpG oligonucleotides, cytokines, chemokines, saponin, G -CSF and / or RNA and, when appropriate, other therapeutically active compounds. The therapeutically active compounds of the invention can be administered via any conventional route, including injection or infusion. The administration can be carried out, for example, orally, intravenously, intraperitoneally, intramuscularly, subcutaneously or transdermally. Preferably, the antibodies are administered therapeutically by means of a pulmonary aerosol. The antisense nucleic acids are preferably administered by administration slow intravenous The compositions of the invention are administered in effective amounts. An "effective amount" refers to the amount that a desired reaction achieves or a desired effect alone or together with additional doses. In the case of treatment of a particular disease or a particular condition characterized by the expression of one or more tumor-associated antigens, the preferred reaction is preferably related to the inhibition of the course of the disease. This includes retarding the progress of the disease and in particular interrupting or reversing the progress of the disease. The desired reaction in a treatment of a disease of a condition may also be delayed from the onset or prevention of the onset of the disease or condition. In accordance with the invention, a diagnosis or treatment of cancer may also include the diagnosis or treatment of cancer metastases that have already formed or will be formed. According to the invention, the term "treatment", comprises therapeutic and prophylactic treatment, ie prevention. An effective amount of a composition of the invention will depend on the condition to be treated, the severity of the disease, the patient's individual parameters, including age, physiological condition, size and weight, the duration of treatment, the type of an accompanying therapy (if present), the specific route of administration, and similar factors. The pharmaceutical compositions of the invention are preferably sterile and contain an effective amount of the therapeutically active substance to generate the desired reaction or the desired effect. The administered doses of the compositions of the invention may depend on various parameters such as the type of administration, the condition of the patient, the desired period of administration, etc. In case a reaction in a patient is insufficient with an initial dose, higher doses (or effectively higher doses reached by a different, more localized route of administration) can be used. In general, dosages of the tumor-associated antigen of 1 ng to 1 mg, preferably 10 ng to 100 μg, are formulated and administered for a treatment or to generate or increase an immune response. If the administration of nucleic acids (DNA and RNA) coding for the tumor-associated antigens is desired, dosages between 1 ng and 0.1 mg are formulated and administered. The pharmaceutical compositions of the invention are generally administered in pharmaceutically compatible amounts and in pharmaceutical compositions. compatible The term "pharmaceutically compatible", refers to a non-toxic material that does not interact with the action of the active component of the pharmaceutical composition. Preparations of this type can generally contain salts, buffer substances, preservatives, carriers and, when appropriate, other therapeutically active compounds. When used in medicine, the salts must be pharmaceutically compatible. However, salts that are not pharmaceutically compatible can be used to prepare pharmaceutically compatible salts and are included in the invention. The pharmacologically and pharmaceutically compatible salts of this type comprise in a non-limiting form those preparations of the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic acids, and the like. similar. The pharmaceutically compatible salts can also be prepared as alkali metal salts or alkaline earth metal salts, such as sodium salts, potassium salts or calcium salts. A pharmaceutical composition of the invention may comprise a pharmaceutically compatible carrier. According to the invention, the term "pharmaceutically compatible carrier" refers to one or more compatible solid or liquid packing materials, diluents or encapsulating substances, which are suitable for administration to human beings. The term "carrier" refers to an organic or inorganic, natural or synthetic or synthetic component in which the active component is combined in order to facilitate the application. The components of the pharmaceutical composition of the invention are generally such that there is no interaction that substantially impairs the desired pharmaceutical efficacy. The pharmaceutical compositions of the invention may contain suitable buffer substances such as acetic acid in a salt, citric acid in a salt, boric acid in a salt and phosphoric acid in a salt. The pharmaceutical compositions, when appropriate, may also contain suitable preservatives such as benzalkonium chloride, chlorobutanol, paraben and thimerosal. The pharmaceutical compositions are generally provided in a uniform dosage form and can be prepared in a manner known per se. The pharmaceutical compositions of the invention may be in the form of capsules, tablets, troches, solutions, suspensions, syrups, elixirs or in the form of an emulsion, for example.
Compositions suitable for parenteral administration, generally comprise a sterile aqueous non-aqueous preparation of the active compound, which is preferably isotonic for the blood of the container. Examples of compatible carriers and solvents are Ringer's solution and isotonic sodium chloride solution. In addition, sterile fatty oils are generally used as a solution or suspension medium.
E ploses: Materials and methods The techniques and methods mentioned herein are carried out in a manner known per se and are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY All methods that include the use of equipment and reagents are carried out according to the information of the manufacturers.
RNA extraction, preparation of the polyd (T) primed cDNA and conventional analyzes by RT-PCR. Total RNA was extracted from natural tissue material by using guanidium isothiocyanate as the chaotropic agent (Chomczynski &Sacchi, Anal. Bíochem. 162: 156-9, 1987). After extraction with phenol acid and precipitation with isopropanol, the RNA was dissolved in water treated with DEPC. The synthesis of the first cDNA chain from 4 μg of total RNA was carried out in 20 μ? of the reaction mixture by superscript II (Invitrogen), according to the manufacturer's information. The primer used was an dT oligonucleotide (18). The integrity and quality of the cDNA were verified and by amplification of p53 in a 30 cycle PCR ((SEQ ID NO: 33, 34)), hybridization temperature 67 ° C). A file of the first cDNA chain was prepared from several normal tissues and tumor entities. For expression studies, 0.5 μ? of these cDNAs were amplified in 30 μ? of the reaction mixture, using GOI-specific primers (see below) and 1 U of HotStarTaq DNA polymerase (Qiagen). Each reaction mixture contained 150μ ?? dNTPs, 0.3μp? of each primer and 3 μ? of 10 x reaction buffer. The primers were selected to be located in two different exons, and the elimination of the interference by contaminating genomic DNA as the reason for the false-positive results was confirmed by testing the non-inverse transcribed DNA as a template. After 15 minutes at 95 ° C, to activate the HotStarTaq DNA polymerase, 35 cycles of PCR were carried out (0.5 min at 94 ° C, 0.5 min. at the particular hybridization temperature, 0.5 min. at 72 ° C and final elongation at 72 ° C for 6 min.). 20 μ? of this reaction were fractionated and analyzed on agarose gel stained with ethidium bromide.
Preparation of randomized hexamer-primed cDNA and quantitative real-time PCR The expression of various genes was quantified by real-time PCR. The PCR products were detected using SYBR Green as the intercalating reporter dye. The fluorescence of the SYBR Green reporter was suppressed in solution and the dye is activated only after binding to double-stranded DNA fragments. The increase in SYBR Green fluorescence as a result of specific amplification using GOI-specific primers after each PCR cycle, was used for quantification. The expression of the target gene was quantified absolutely or in relation to the expression of a control gene with constant expression in the tissues that will be investigated. The expression was measured after the standardization of the samples against 18s RNA as the so-called initiation gene using the ?? - Ct method (PE Biosystems, USA). The reactions were carried out in duplicates and determined in triplicates. The QuantiTect SYBR Green PCR kit (Qiagen, Hilden) was used for according to the manufacturer's instructions. The cDNA was synthesized with random primers (Invitrogen) using the protocol described above. Each of the 5 μ? of the diluted cDNA was used in a total volume of 30 μ? for PCR: 300 nm sense primer, 300 nm antisense primer; initial denaturation at 95 ° C for 15 min .; 95 ° C for 30 sec .; reassociation for 30 sec .; 72 ° C for 30 sec .; 40 cycles. The sequences of the primers used are indicated in the respective examples. Cloning of sequence analysis The cloning of total lengths and gene fragments was carried out by conventional methods. To ensure the sequence, the corresponding antigens were amplified using the pfu polymerase test reading (Stratagene). After completion of the PCR, adenosine was ligated by means of HotStarTaq DNA polymerase to the ends of the amplicon in order to clone the fragments according to the manufacturer's instructions in the TOPO-TA vector. The sequencing was carried out by a commercial service. The sequences were analyzed using programs and algorithms for conventional prediction.
Cell proliferation analysis 24 hours after transfection with the siRNA duplexes 1 x 104 cells were cultured in medium supplemented with varying concentrations of FCS for 48 hours. Proliferation was analyzed by measuring the incorporation of BrdU into the newly synthesized DNA strands using the DELFIA cell proliferation kit (Perkin Elmer) according to the manufacturer's instructions in a Wallac Victor2 multi-brand counter (Perkin Elmer).
Analysis and apoptosis of the cell cycle The cells were cultured in medium supplemented with FCS in varying concentrations, harvested after 48 hours and stained with propidium iodide before analysis of the fluocitometric DNA content. The apoptotic cells and the cells in the S / G2 / phases of the cell cycle were quantified using the CellQuest software (Becton Dickinson).
Cell migration Cell migration analyzes were conducted in transverse cavity chambers with membranes with pore 8.0 μ? T? (BD Biosciences) with cells cultured in medium without serum for 12 hours before the experiments. For siRNA experiments the cells were transferred to serum-free conditions 24 hours after transfection with the siRNA duplexes as described above. 4 x 104 cells in 400 μ? of culture medium without serum were added to the upper chamber. The lower chambers contained 800 μ? of culture medium supplemented with either FCS, PDGF-BB (Sigma-Aldrich) or SDF-la / CXCL12 (R &D Systems) as chemoattractants. After 24 hours, the cells that had migrated to the lower side of the membrane were fixed in ice-cooled methanol; the membranes were cut, placed on microscope slides and mounted with Hoechst (Dako) for fluorescence microscopy. Cells in five random visual fields (magnification of 100x) were counted for each membrane. All the experiments were performed in triplicate. The effects on cell chemokinesis were analyzed using the same experimental procedure with (i) without chemoattractant added to the upper and lower chamber and (ii) with chemoattractant added to the upper and lower chamber.
In Vivo Invasion Analysis In vivo invasion analyzes were conducted in transverse cavity chambers with 8.0 μm pore membranes (BD Biosciences) with cells cultured in serum-free medium for 12 hours before experiments. The upper chambers were prepared with 100 μ? of Matrigel (BD Biosciences) diluted to 1 mg / ml in medium without serum. The chambers were incubated at 37 ° C for 5 hours for gelation. For the siRNA experiments, the cells were transferred to serum-free conditions 24 hours after transfection with the siRNA duplexes as described above. 1 x 105 cells in 400 μ? of culture medium without serum were added to the upper chamber. The lower chambers contained 800 μ? of culture medium supplemented with FCS as a chemoattractant. After 24 hours, the invaded cells on the lower side of the membrane were fixed in ice-cooled methanol; the membranes were cut, placed on microscope slides and mounted with Hoechst (Dako) for fluorescence microscopy. Cells in five random visual fields (magnification of 100x) were counted for each membrane. All the experiments were performed in triplicate.Example 1: Identification of ISC-468 as the target of therapeutic and diagnostic cancer ISC-468 (SEQ ID NO: 1) codes for a protein of 212 amino acids (SEQ ID NO: 2) and with a molecular weight of 23.6 kDa It has been previously described as the placenta-specific protein expressed during pregnancy (Fant et al., Mol Reprod Dev. 63: 430-6, 2002). The protein was predicted to have a cleavable signal peptide of aa 1-23, followed by a short putative transmembrane domain (aa 25-47) as analyzed by bioinformatic tools (TMpred, SOUSI). It was predicted that the remaining protein will be extracellular and therefore can be used according to the invention as a target structure for monoclonal antibodies. In accordance with the invention, a gene-specific primer pair (SEQ ID NO: 3, 4) for ISC-468 was used in RT-PCR analysis to amplify the cDNA derived from a definitive panel of normal and tumor tissues. As expected, the placenta was confirmed as the only healthy tissue expressing this gene (Figures 1A-1C). However, no significant expression was observed in any other normal organ tissue. More surprisingly, when cancer specimens were investigated, high and significant expression levels were found in different tumor types, including colon, pancreatic, esophageal, stomach, lung, breast, ovarian, head and neck, renal, prosthetic carcinomas. and hepatic (Figures 1A-1C and 2, as well as Table 1). Quantitative real-time RT-PCR analyzes of the expression ISC-468 in 60 breast carcinoma samples revealed that 80% of all samples expressed significant levels of ISC-468 (Figures 20A and 20B).
Table 1: Expression of ISC-468 in normal and tumor tissues The selective and high expression of ISC-468 transcripts in tumors was not previously known and can be used according to the invention for methods of molecular diagnostics such as, RT-PCR to detect the spread of tumor cells in serum and bone marrow and to detect metastases in other tissues. This molecule can also be used as a specific target for therapeutic approaches. The following peptides, inter alia, were selected for the production of ISC-468-specific antibodies according to the invention: SEQ ID NO: 58, 59, 60, 68, 69, 2. The specificity of the antibodies was confirmed by analysis of immunofluorescence of cells transfected with ISC-468-eGFP (Figure 21A). The subcellular localization of ISC-468 in breast carcinoma cell lines that are endogenously expressed MCF-7 and BT-549 was analyzed by immunofluorescence analysis. Staining of cells either fixed with eOH (Figure 21B) or non-fixed (Figure 21C), revealed that ISC-468 is located in the plasma membranes of the expression cells. The specificity of the staining was confirmed by the RNAi-induced disassembly of the expression of ISC-468, resulting in the loss of plasma membrane staining. In addition, ISC-468-specific antibodies were used for immunohistochemical analysis of ISC-468 expression in clinical samples of normal breast and breast carcinomas. The expression of ISC-468 was not detectable in specimens of normal breast (Figures 22A, 22B). In contrast, breast carcinoma specimens showed strong and homogeneous expression of ISC-468 (Figures 22C, 22D). The signals were accentuated in the plasma membrane of the cancer cells of expression, confirming that ISC-468 is a membrane protein expressed selectively in cancer cells. The extracellular domains of ISC-468 can be used according to the invention as the target structure for immunodiagnosis and therapy by means of monoclonal antibodies. In addition, ISC-468 can be used according to the invention as a vaccine (RNA, DNA, protein, peptides) to induce tumor-specific immune responses (immune responses provided by T and B lymphocytes). Disarmed RNAi-induced expression of ISC-468 was achieved by transfection of cells with the siRNA duplexes that specifically target the ISC-468 mRNA (SEQ ID NOs: 70-73). Transfection of breast carcinoma cell lines MCF-7 and BT-549 that are expressed endogenously, resulted in a stable and specific reduction of expression of ISC-468 mRNA (Figure 23). In order to understand the physiological function of the expression of ISC-468, several analyzes were carried out In vitro cell based on RNAi. Transfection of breast carcinoma MCF-7 and BT-549 cell lines with the siRNA duplexes resulted in a distinct reduction of cell proliferation compared to the respective controls, as analyzed in a proliferation analysis based on in BrdU (Figure 24). The cell cycle analysis based on FACS showed that the abrogation of cell proliferation resulted from a Gl / S arrest (Figures 25A, 25B). Additionally, it could be shown that the disarmed induced RNAi of ISC-468 profoundly affected the AKT signaling pathway in cancer cells that are endogenously expressed by the inhibition of AKT phosphorylation (Figure 26). In addition, the proliferation of MCF-7 cells was attenuated when the cells were incubated with specific ISC-468 antibodies generated against the ISC-468-specific peptides (SEQ ID NO: 68, 69) compared to an irrelevant control antibody (FIG. 27). These results indicate that ISC-468 is a decisive factor for the proliferation of cancer cells probably by supplying the activation induced by the growth factor of the signaling pathway AKT and others. The ISC-468 itself could represent a receptor, a co-receptor or a membrane-bound companion for growth factors, chemokines or other substances.
In addition, the impact of the expression ISC-468 on the migratory capacity of cancer cells was analyzed. Disarmed RNAi induced expression of ISC-468 in MCF-7 and BT-549 cell lines of breast carcinoma resulted in different deterioration of chemotaxis, chemokinesis and cell invasion, as assessed by migration analysis in transverse cavities (Figures 28A, 28B, 28C). Chemotaxis, chemokinesis and invasion are decisive factors for the metastasis of cancer cells to other organs. Therefore, the expression of ISC-468 in cancer cells could be a positive factor for the metastasis of cancer cells. In breast carcinomas, it could be shown that the expression of ISC-468 correlates with the estrogen receptor status of the tumor. Quantitative real-time RT-PCR analysis of ISC-468 expression in 60 breast carcinoma samples revealed that estrogen-positive breast carcinomas showed significantly higher levels of ISC-468 expression than recipient tumors -negatives (Figure 29). Accordingly, the expression of ISC-468 could be induced in the positive estrogen-receptor breast carcinoma cell line MCF-7 by treatment with 17p-estradiol (Figure 30).
Example 2: Identification of ISC-507 as target of therapeutic and diagnostic cancer ISC-507 (SEQ ID NO: 5), codes for a 754 aa protein (SEQ ID NO: 6), with a molecular weight of 85.6 kDa . ISC-507 is a member of a family of zinc binding proteins with disintegrin metalloprotease activities that can function as adhesion proteins and / or endopeptidases. It has already been described that members of this family are involved in several biological processes, including fertilization, neurogenesis, muscle development, and immune response (Seáis et al., Genes Dev. 17 (1): 7-30 , 2003) ISC-507 has a transmembrane domain (aa 671-687), a large N-terminal extracellular region and short C-terminal cytoplasmic region. The expression of ISC-507 has been reported to be restricted specifically to the mammalian epididymis, the small gland adjacent to the testicle that is decisively involved in sperm maturation. According to the literature, ISC-507 is transferred from the epididymis to the sperm surface and redistributed in the sperm head during the acrosome reaction (Adachi et al., Mol Reprod Dev. 64: 414-21, 2003) .
Investigations by RT-PCR with ISC-507-specific primers (SEQ ID NO: 7, 8) confirmed the selective expression in the testes and the absence of ISC-507 from any other normal tissue (Table 2, Figures 3A-3B) , except for the weak expression in tissues derived from prostate and lymph nodes (Table 2, Figure 4). However, and more surprisingly, the expression of ISC-507 was observed in a significant number of prostate cancers (Figures 3A-3B, 4). This protein had not been reported before to be involved in cancer.
Table 2: Expression of ISC-507 normal and tumor tissues Tumor type Expression The absence of toxicity in relevant normal tissues and the frequent and significant expression of ISC-507 in prosthetic cancers make this protein according to the invention, a valuable marker for diagnosis and therapy. This includes, according to the invention, the detection of disseminated tumor cells in serum, bone marrow, urine, and the detection of metastases in other organs by means of RT-PCR. In addition, the extracellular domains of ISC-507 can be used according to the invention as a target structure for immunodiagnosis and therapy by means of monoclonal antibodies. Furthermore, ISC-507 can be used according to the invention as a vaccine (RNA, DNA, protein, peptides) for inducing tumor-specific immune responses (immune responses provided by T and B lymphocytes). Antibodies to detect ISC-507, could be produced with the following peptides and proteins: SEQ ID NO: 51, 52, 53, 54, 55, 6, 56 and 57.
In accordance with the invention, an antibody that binds to ISC-507, could be useful for therapeutic or diagnostic purposes.
Example 3: Identification of ISC-466 as target of therapeutic cancer and for diagnosis ISC-466 (SEQ ID NO: 9) codes for a 426 aa protein (SEQ ID NO: 10) with a molecular weight of 48.2 kDa. It belongs to the family of pregnancy-specific glycoproteins. Human pregnancy-specific glycoproteins (PSGs) are a group of molecules that are produced primarily by placental syncytiotrophoblasts during pregnancy and are part of the immunoglobulin superfamily (Beauchemin et al., Exp Cell Res. 252 (2): 243-9, 1999). As another PSG, it has also been reported that ISC-466 will be restricted to the placenta. According to the invention, a gene-specific primer pair (SEQ ID NO: 11, 12) for ISC-466 was used in analysis by RT-PCR to amplify cDNA derived from a broad panel of normal and tumor tissues. Analysis by RT-PCR revealed the expression of ISC-466 transcripts in normal placenta, and a weak expression in thymus and ovary (Table 3, Figure 5A). Not detected, no significant expression in any other normal organic tissue. More surprisingly, when cancer cell lines were investigated, high and significant expression levels were found in several of the tumor types, including: breast cancer (Figure 5C), lung cancer (Figure 5C), ovarian carcinoma (Figure 5D) and head and neck and renal carcinomas (Figure 5B).
Table 3: Expression of ISC-466 normal and tumor tissues Contrary to the observation, ISC-466 is involved in colo-rectal carcinomas (Salahshor et al., BMC Cancer, 5:66, 2005), investigations reveal ISC-466 according to the invention as a diagnostic and therapeutic marker. for cancer of the head and neck, breast, ovarian, prosthetic and melanoma.
Example 4: Identification of ISC-518 as target of therapeutic cancer and for diagnosis ISC-518 (SEQ ID NO: 13) codes for a translation product 237 aa (SEQ ID NO: 14). However, there is no data regarding tissue distribution and to date no connection is available for cancer. ISC-518 is a gene / protein predicted hypothetically, bioinformatically. Sequence analyzes revealed that the protein has a transmembrane domain (aa 102-118). The extracellular C term highlights a functional domain that occurs in cell surface glycoproteins. According to the invention, a gene-specific primer pair (SEQ ID NO: 15, 16) for ISC-518 was used in analysis by RT-PCR to amplify cDNA derived from a broad panel of normal and tumor tissues. It was found that the only normal tissue that expresses this gene were the testicles, while none was detected significant expression of ISC-518 in any other normal organ (Figure 6). More surprisingly, when cancer specimens were investigated, high and significant expression levels were found in hepatocellular carcinomas (Figure 7).
Table: Expression of ISC-518 normal and tumor tissues Bioinformatic investigations showed that the protein encoded by ISC-518 represents a cell surface molecule. The previous unknown selective expression of this surface molecule makes it a target for therapeutic purposes and for the development of diagnostic methods for the detection of tumor cells and therapeutic methods for the elimination of tumor cells.
Example 5: Identification of ISC-477 as target of therapeutic cancer and for diagnosis ISC-477 (SEQ ID NO: 17) codes for a translation product 130 aa (SEQ ID NO: 18). ISC-477 is a hypothetical protein. There is no data regarding tissue distribution or a connection to cancer is publicly available. The structural analyzes reveal a hydrophobic region, which could be a transmembrane region or signal peptide. According to the invention, a gene-specific primer pair (SEQ ID NO: 19, 20) for ISC-477 was used in analysis by RT-PCR to amplify the cDNA derived from a broad panel of normal and tumor tissues. It was found that the only normal tissues that express this gene were the placenta and ovary. On the contrary, no significant expression of ISC-477 was detectable in no other normal organ (Figure 8A). More surprisingly, when cancer specimens were investigated, high and significant expression levels were found in lung, ovarian, colon and stomach cancers (Figures 8A-8D). The levels of expression are clearly higher than the expression in normal ovary.
Table 5: Expression of ISC-477 normal and tumor tissues Example 6: Identification of ISC-489 as target of therapeutic cancer and for diagnosis ISC-489 (SEQ ID NO: 21) codes for a translation product 363 aa (SEQ ID NO: 22). The protein is a newly described member of the family of receptors coupled with the G protein. However, there are no data regarding tissue distribution and no connection to cancer is publicly available either. According to the invention, a gene-specific primer pair (SEQ ID NO: 23, 24) for ISC-489, was used in analysis by RT-PCR to amplify the cDNA, derived from a broad panel of normal and tumor tissues. It was found that only the normal tissues expressing this gene were the placenta and esophagus (weak expression). On the contrary, no significant expression of ISC-489 could be detected in any other normal organ (Figure 9A). More surprisingly, when cancer specimens were investigated, high and significant expression levels were found in head and neck and stomach cancers (Figures 9B, 9C). As the member of the G protein-coupled receptor family, ISC-489 is an integral membrane protein with 7 transmembrane domains and various extracellular loops, which can be directed on the cell surface.
Table 6: Expression of ISC-489 in normal and tumor tissues The pronounced expression and the unexpected high incidence of ISC-489 in carcinomas of the head and neck makes this protein according to the invention a very interesting diagnostic and therapeutic marker.
Example 7: Identification of ISC-461 as a target of therapeutic cancer and for diagnosis ISC-461 (SEQ ID NO: 25) codes for a 419 aa protein (SEQ ID NO: 26) with a molecular weight of 47.1 kDa. It belongs to the family of pregnancy-specific glycoproteins. Human pregnancy-specific glycoproteins (PSGs) is a group of molecules that are produced primarily by placental syncytiotrophoblasts during pregnancy and were part of the immunoglobulin superfamily (Beauchemin et al., Exp Cell Res. 252 (2): 243- 9, 1999). Like other PSGs, it has also been reported that ISC-461 will be restricted to the placenta. In accordance with the invention, a gene-specific primer pair (SEQ ID NO: 11, 27) for ISC-461 was used in analysis by RT-PCR to amplify the cDNA derived from a wide panel of normal and tumor tissues. As expected, the placenta was confirmed as the expressed one of this gene, together with the weak expression in testes and ovary (Figures 10A and 10B). However, no significant expression was detected in any other normal organic tissue. More surprisingly, when tissues derived from cancer and cancer cell lines were investigated, high and significant expression levels were found in various tumor types, among which they include breast cancer (Figure 10C), ova carcinoma (Figure 10D) and melanoma (Figures 10B, 10C).
Table 7: Expression of ISC-461 in normal and tumor tissues An additional purpose according to the invention was to identify the splice variants for ISC-461, which can be used both for diagnosis and for therapy. In the investigation of variants of splice, a splice form could be identified (SEQ ID NO: 28) and the protein encoded by it (SEQ ID NO: 29).
Example 8: Identification of ISC-465 as a target of therapeutic cancer and for diagnosis ISC-465 (SEQ ID NO: 30) encodes a 419 aa protein (SEQ ID NO: 31) with a molecular weight of 47.0 kDa. It belongs to the family of pregnancy-specific glycoproteins. Human pregnancy-specific glycoproteins (PSGs) are a group of molecules that are produced mainly in placental syncytiotrophoblasts during pregnancy and are part of the immunoglobulin superfamily (Beauchemin et al., Exp Cell Res. 252 (2): 243 -9, 1999). Like the other PSGs, it has also been reported that ISC-465 will be restricted to the placenta. According to the invention, a gene-specific primer pair (SEQ ID NO: 11, 32) for ISC-465 was used in analysis by RT-PCR to amplify cDNA derived from a broad panel of normal and tumor tissues. As expected, the placenta was confirmed as expressing this gene, together with weak expression in normal ovary (Figure 11A). However, no expression was detected significant in any other normal organic tissue. More surprisingly, when tissues derived from cancer and cancer cell lines were investigated, high and significant expression levels were found in various types of tumor (Figures 11A, 11B), especially breast cancer (Figure 11B).
Table 8: Expression of ISC-461 normal and tumor tissues The selective and high expression of ISC-465 transcripts in tumors, was not previously known and can be used according to the invention for molecular diagnostic methods such as, RT-PCR for the detection of the spread of tumor cells in the serum and bone marrow and for the detection of metastases in other tissues. This molecule can also be used as a specific target for therapeutic approaches.
Example 9: Identification of Mem-030 as target of therapeutic cancer and for diagnosis Mem-030 (SEQ ID NO: 35) codes for a 592 aa protein (SEQ ID NO: 36) with a molecular weight of 67.9 kDa. Mem-030 belongs to the GBP proteins, which are large GTPases that are capable of binding to GTP, PIB, and GMP and catalyze the hydrolysis of GTP to PIB, as well as GMP (Cheng et al., J Biol. Chem. 260 : 15834-9, 1985). GTPases play an important role in cell proliferation, differentiation, signal transduction and intracellular protein transport and can be induced by interferon (Boehm et al., J Immunol. 161 (12): 6715-23, 1998). Also, Mem-030 counteracts the proliferative effect of inflammatory cytokines similar to IFN-g, interleukin 1-b (IL-lb), and factor-a of tumor necrosis (TNF-a) 1 on endothelial cells (Guenzi et al., EMBO J. 20 (20): 5568-77, 2001). According to the invention, a gene-specific primer pair (SEQ ID NO: 37, 38) for Mem-030 was used in analysis by real-time RT-PCR, to amplify the cDNA derived from a broad panel of normal tissues and tumor Mem-030, showed a ubiquitous expression pattern (Figure 12A, Table 9). More surprisingly, when cancer-derived tissues and cancer cell lines were investigated, high and significant levels of overexpression were found in various types of tumor (Figures 12A, 12B), especially carcinomas of the head and neck.
Table 9: Expression of Mem-030 in normal and tumor tissues Tumor type Expression Due to bioinformatic and literature analyzes, a homologous gene of Mem-030 could also be an attractive therapeutic target (SEQ ID NO: 39) and codes for a 586 aa protein (SEQ ID NO: 40) with a molecular weight of 66.6 kDa. Bioinformatic investigations showed that both proteins represent cell surface molecules. The selective over-expression, previously unknown of this surface molecule, makes it a target for therapeutic purposes and for the development of diagnostic methods for the detection of tumor cells and therapeutic methods for the elimination of tumor cells.
Example 10: Identification of Mem-055 as a target of therapeutic cancer and for diagnosis Mem-055 (SEQ ID NO: 41) codes for a 250 aa protein (SEQ ID NO: 42) with a molecular weight of 27.9 kDa. The protein encoded by this gene is a lysosomal thiol reductase that, at low pH, can reduce protein disulfide bonds. The enzyme is constitutively expressed in cells that present antigens and is induced by gamma-interferon in other cell types. This enzyme plays an important role in the processing of antigens restricted to HC class II (Arunachalam et al., Proc Nati Acad Sci 'USA 97 (2): 745-50, 2000). The location of Mem-055 and the protein topology was predicted by the analysis of the putative signal sequences and the transmembrane domains with bioinformatic tools (TMPRED, SOUSI). Mem-055 could have an extracellular C term. According to the invention, a gene-specific primer pair (SEQ ID NO: 43, 44) for Mem-055 was used in real-time RT-PCR analysis to amplify cDNA derived from a broad panel of normal tissues and tumor. Mem-055, showed a ubiquitous expression pattern (Figure 13A, Table 10). More surprisingly, when the expression of Mem-055 was investigated within cancer-derived tissues, high and significant levels of overexpression were found in various types of tumor (Figures 13A, 13B), especially stomach cancers.
Table 10: Expression of Mem-055 in normal and tumor tissues Mem-055 is a white structure for therapeutic and diagnostic purposes, due to putative extracellular domain and unexpected over-expression in different types of carcinoma.
Example 11: Identification of Mem-062 as target of therapeutic and diagnostic cancer Mem-062 (SEQ ID NO: 45) codes for a 271 aa protein (SEQ ID NO: 46) with a molecular weight of 30.7 kDa. Using a computer-based selection method, Mem-062 could be identified previously and described as specifically expressed testes, prostate and placenta (Bera et al., Biochem Biophys Res Commun., 312 (4): 1209-15, 2003). according to the invention, a gene-specific primer pair (SEQ ID NO: 47, 48) for Mem-062, was used in analysis by RT-PCR. Mem-062, surprisingly showed a specific cancer-testes expression pattern (Figure 14A, Table 11). In no other normal organic tissue was any expression detected. More surprisingly, when tissues derived from cancer were investigated, significant levels of Mem-62 expression were found (Figure 14B), especially in ovarian carcinomas.
Table 11: Expression of Mem-062 in normal and tumor tissues The alternative splicing results in an alternative transcription (SEQ ID NO: 49) and its corresponding translation product (SEQ ID NO: 50).
Example 12: Identification of Mem-068 as target of therapeutic cancer and for diagnosis Mem-068 (SEQ ID NO: 61) is a cDNA clone recently identified Using a bioinformatic prediction approach (Genscan) Mem-068 could be described as a gene of multiple exons on chromosome 9 (SEQ ID NO: 62). The deduced protein sequence (SEQ ID NO: 63) has 751 aa and forms a protein with a molecular weight of 82.4 kDa. According to the invention, a gene-specific primer pair for Mem-068 was used in analysis by RT-PCR. Mem-068, surprisingly showed a specific cancer-testes expression pattern (Figure 15A, Table 12). No expression was detected in any other normal organic tissue, except for the placenta (weak expression). More surprisingly, when tissues derived from cancer were investigated, significant levels of expressed Mem-068 were found (Figure 15B), especially in renal cell carcinomas and in stomach cancers.
Table 12: Expression of Mem-068 in normal and tumor tissues Normal tissues Expression Tumor type Expression Brain Carcinoma of the colon + Breast Carcinoma of cells ++ Kidneys Colon Carcinoma of the stomach + Kidney Lung cancer + Liver Breast cancer Lung Ovarian carcinoma Lymph nodes Melanoma Normal tissues Expression Tumor type Expression Ovary - Prosthetic carcinoma - Pancreas - Placenta PBMC - PBMC activated - Prostate - Skeletal muscle - Skin - Stomach - Spleen - Testicles + Uterus - According to the transmembrane prediction program TMpred Mem-068, it could be expressed on the cell surface, which makes it an interesting target for therapeutic or diagnostic purposes.
Example 13: Identification of Mem-071 as target of therapeutic cancer and for diagnosis Mem-071 (SEQ ID NO: 64) is a new cDNA clone, which is encoded in 2 exons or on chromosome 1. According to the invention, a gene-specific primer pair for Mem-071, was used in RT-PCR to amplify the cDNA derived from a broad panel of normal and tumor tissues. It was found that the only normal tissues expressing this gene were the testicles (Figure 16A). In contrast, when they were investigated cancer specimens, expression levels were found high and significant in renal cell carcinomas and stomach cancers (Figure 16B).
Table 13: Expression of Mem-071 in normal and tumor tissues Normal tissues Expression Tumor type Expression Brain - Carcinoma of colon - Breast - Carcinoma of renal cells Colon - Stem carcinoma + Kidney - Lung cancer - Liver - Breast cancer - Lung - Ovarian carcinoma - Lymph nodes - Melanoma - Ovary - Prostatic carcinoma - Pancreas - Placenta PBMC - PBMC activated - Prostate - Skeletal muscle - Skin - Stomach - Spleen - Testicles + Uterus - The unexpected high incidence of Mem-071 in Renal cell carcinomas makes this protein from according to the invention a marker for diagnosis and quite interesting therapeutic.
Example 14: Identification of Mem-072 as target of therapeutic and diagnostic cancer Mem-072 (SEQ ID NO: 65) is a newly identified gene, which is encoded in 3 exons on chromosome 16. According to the invention, a Gene-specific primer pair for Mem-072 was used in RT-PCR analysis to amplify the cDNA derived from a broad panel of normal and tumor tissues. No expression was found within all the normal tissues tested (Figure 17A, Table 14). When tissues derived from cancer and cancer cell lines were investigated, high and significant expression levels were found in lung cancer samples. (Figure 17B).
Table 14: Expression of Mem-072 in normal and tumor tissues Normal tissues Expression Tumor type Expression Brain - Colon carcinoma Breast - Renal cell carcinoma Colon - Kidney carcinoma Kidney - Lung cancer ++ Liver - Breast cancer Lung - Ovarian carcinoma Lymph nodes - Ovarian melanoma - Prostatic carcinoma Pancreas - Placenta PBMC - PBMC activated - Normal tissues Expression Tumor type Expression Prostate - Skeletal muscle - Skin - Stomach - Spleen - Testicles - Uterus - The selective and high expression of Mem-072 in lung tumors was not previously known and can be used according to the invention for molecular diagnostic methods such as, RT-PCR to detect the spread of tumor cells in the serum and bone marrow and to detect metastases in other tissues. This molecule can also be used as a specific target for therapeutic approaches.
Example 15: Identification of Mem-106 as target of therapeutic cancer and for diagnosis Mem-106 (SEQ ID NO: 66) is a newly identified cDNA that is encoded without intron on chromosome 2. According to the invention, a primer pair Gene-specific for Mem-106, was used in analysis by RT-PCR. Mem-106 surprisingly showed a specific cancer-testes expression pattern (Figure 18A, Table fifteen). No expression was detected in any other tissue normal organic More surprisingly, when investigated cancer-derived tissues, they were found Significant levels of Mem-106 expression (Figure 18B), especially in ovarian carcinomas.
Table 15: Expression of Mem-106 in normal and tumor tissues Normal tissues Expression Tumor type Expression Breast Colon carcinoma + Colon Renal cell carcinoma Kidney Carcinoma of the stomach Liver Lung cancer Lung Breast cancer Lymph nodes Ovarian carcinoma ++ Ovarian Melanoma ++ Pancreas Placental carcinoma PBMC PBMC activated Prostate Skeletal muscle Skin Stomach Spleen Testicles Uterus Mem-106 is a white structure for purposes therapeutic and diagnostic, due to unexpected over-expression in different types of carcinomas.
Example 16: Identification of Mem-131 as target of therapeutic and diagnostic cancer Mem-131 (SEQ ID NO: 67) is a cDNA clone recently identified Mem-131 is a gene of 2 exons on chromosome 15.
According to the invention, a gene-specific primer pair for Mem-131 was used in RT-PCR analysis for Amplify the cDNA derived from a broad tissue panel normal and tumor. The analysis by RT-PCR revealed the expression of Mem-131 transcripts only in PBMC normal activated (Table 16, Figure 19A). No significant expression was detected in any other tissue normal organic More surprisingly, when investigated cancer samples, high and significant expression levels were found in several types of tumor, including breast cancer (Figure 19B), lung cancer (Figures 19B + 19C) and ovarian carcinoma (Figure 19C).
Table 16: Expression of Mem-131 in normal and tumor tissues Normal tissues Expression Tumor type Expression Breast Lung Cancer ++ Duodenum Breast cancer ++ Ovarian carcinoma bladder ++ Brain Skin Bone Marrow Colon Table 16: Expression of Mem-131 in normal and tumor tissues Normal tissues Expression Tumor type Expression Liver - Lung - Lymph nodes - Stomach - Spleen - Myocardium - Kidney - Esophagus - Ovarian pancreas PBMC - Activated PBMC - Placenta - Muscle - Testicles - Thymus - The investigations reveal Mem-131 according to the invention as a marker for diagnosis and Therapeutic treatment for lung, breast and ovarian cancers.

Claims (48)

  1. NOVELTY OF THE INVENTION Having described the present invention, it is considered as a novelty and, therefore, the content of the following CLAIMS is claimed as property: 1. A pharmaceutical composition, characterized in that it comprises an agent that: (I) inhibits the expression or activity of a tumor-associated antigen and / or (II) has activity for tumor inhibition, and is selective for cells that express or abnormally express a tumor-associated antigen and / or (III) when administered, selectively increases the number of complexes between an MHC molecule and a tumor-associated antigen or a portion thereof, the tumor-associated antigen has a sequence encoded by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64- 67, a portion or derivative thereof, (b ) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid that degenerates with with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c).
  2. 2. The pharmaceutical composition according to claim 1, characterized in that the low (II) agent causes induction of cell death, reduction in cell growth, damage to the cell membrane or cytokine secretion.
  3. 3. The pharmaceutical composition according to claim 1, characterized in that the low (I) or (II) agent is an antisense nucleic acid that hybridizes selectively with the nucleic acid encoding the tumor-associated antigen.
  4. 4. The pharmaceutical composition according to claim 1, characterized in that the low (I) or (II) agent is an antibody that selectively binds to the tumor-associated antigen.
  5. 5. The pharmaceutical composition according to claim 1, characterized in that the agent comprises one or more components selected from the group consisting of group: (i) the tumor-associated antigen or a portion thereof, (ii) a nucleic acid that codes for the tumor-associated antigen or a portion thereof, (iii) an antibody that binds to the tumor-associated antigen or a portion thereof, (iv) an antisense nucleic acid that hybridizes specifically with a nucleic acid encoding the tumor-associated antigen, (v) a siRNA directed against a nucleic acid encoding the tumor-associated antigen, (vi) a host cell that expresses the tumor-associated antigen or a portion thereof, and (vii) isolated complexes between the tumor-associated antigen or a portion thereof and a MHC molecule.
  6. 6. The pharmaceutical composition according to claim 1, characterized in that the agent comprises two or more agents that in each case selectively inhibit the expression or activity of different tumor-associated antigens, which are in each case selective for cells expressing or expressing abnormally different tumor-associated antigens or that increase the amount of complexes between the MHC molecules and different tumor-associated antigens or portions thereof, with at least one of the tumor-associated antigens having a sequence encoded by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64- 67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid that degenerates with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c).
  7. 7. A pharmaceutical composition, characterized in that it comprises one or more components selected from the group consisting of: (i) a tumor-associated antigen or a portion thereof, (ii) a nucleic acid encoding a tumor-associated antigen or a portion thereof, (iii) an antibody that binds to a tumor-associated antigen or a portion thereof, (iv) an antisense nucleic acid that is hybridizes specifically with a nucleic acid encoding a tumor-associated antigen, (v) a siRNA directed against a nucleic acid encoding a tumor-associated antigen, (vi) a host cell that expresses an antigen tumor-associated or a portion thereof, and (vii) complexes isolated between a tumor-associated antigen or a portion thereof and an MHC molecule, the tumor-associated antigen has a sequence encoded by a nucleic acid that is selected from the group which consists of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid which degenerates with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c).
  8. 8. The pharmaceutical composition according to claim 5 or 7, characterized in that the nucleic acid of (ii) is present in an expression vector.
  9. 9. The pharmaceutical composition according to claim 5 or 7, characterized in that the host cell secretes the tumor-associated antigen or the portion thereof.
  10. 10. The pharmaceutical composition in accordance with claim 5 or 7, characterized in that the host cell further expresses an MHC molecule that binds to the tumor-associated antigen or portion thereof.
  11. 11. The pharmaceutical composition according to claim 10, characterized in that the host cell expresses the MHC molecule and / or the tumor-associated antigen or the portion thereof in a recombinant manner.
  12. 12. The pharmaceutical composition according to claim 10, characterized in that the host cell endogenously expresses the MHC molecule.
  13. 13. The pharmaceutical composition according to claim 5, 7, 10 or 12, characterized in that the host cell is in a cell for presentation of antigens.
  14. 14. The pharmaceutical composition according to claim 4, 5 or 7, characterized in that the antibody is a monoclonal, chimeric or humanized antibody, or is a fragment of an antibody.
  15. 15. The pharmaceutical composition according to claim 4, 5, 7, or 14, characterized in that the antibody is coupled to the therapeutic agent for diagnosis.
  16. 16. The pharmaceutical composition according to claims 1-15, characterized in that it can be used for the treatment or prevention of cancer.
  17. 17. The pharmaceutical composition according to claim 16, characterized in that the cancer is a lung tumor, a breast tumor, a prosthetic tumor, a melanoma, a colon tumor, a gastric tumor, a pancreatic tumor, an ENT tumor, a renal cell carcinoma or a cervical carcinoma, a carcinoma of the colon or a mammary carcinoma.
  18. 18. The pharmaceutical composition according to claims 1-17, characterized in that the tumor-associated antigen comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 2, 6, 10, 14, 18, 22, 26, 29, 31, 36, 40, 42, 46, 50-60, 63, 68, and 69, a portion or derivative thereof.
  19. 19. The pharmaceutical composition according to claims 1, 2, 5-13, and 16-18, characterized in that it is in the form of a vaccine.
  20. 20. The pharmaceutical composition according to claim 19, for therapeutic and / or prophylactic use.
  21. 21. A method of diagnosing or monitoring a disease characterized by the expression or abnormal expression of a tumor-associated antigen, the method comprising detecting or determining the amount: (i) of a nucleic acid encoding the tumor-associated antigen or a portion thereof, and / or (ii) of the tumor-associated antigen or a portion thereof, and / or (iii) of an antibody to the tumor-associated antigen or a portion thereof and / or (iv) of T lymphocytes that are specific for the tumor antigen -associated or for a portion thereof in a biological sample isolated from a patient, with a tumor-associated antigen having a sequence encoded by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence, selected from the group consisting of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, one portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid that degenerates with respect to the nucleic acid of (a) or (b) , and (d) a nucleic acid that is complementary to nucleic acid (a), (b) or (c).
  22. 22. The method according to claim 21, characterized in that the detection or determination of the quantity comprises: (i) contacting the biological sample with a agent that specifically binds to the nucleic acid encoding the tumor-associated antigen or portion thereof, the tumor-associated antigen or portion thereof, the antibody or T lymphocytes, and (ii) detecting the formation or determining the amount of a complex between the agent and the nucleic acid or portion thereof, the tumor-associated antigen or the portion thereof, the antibody or the T lymphocytes. The method according to claim 22, characterized in that the agent which specifically binds to the nucleic acid encoding the tumor-associated antigen or portion thereof is an oligonucleotide or polynucleotide that hybridizes specifically to the nucleic acid or portion thereof. The method according to claim 22, characterized in that the agent that specifically binds to the tumor-associated antigen or the portion thereof is an antibody that binds specifically to the tumor-associated antigen or to the portion thereof. 25. The method according to claim 22, characterized in that the agent that specifically binds to the antibody is a protein or peptide that specifically binds to the antibody. 26. The method according to claim 22, characterized in that the agent that is specifically binds T lymphocytes is a cell that presents the complex between the tumor-associated antigen or the portion thereof and a HC molecule. 27. The method according to any of claims 21 to 26, characterized in that the monitoring of the disease comprises determining the regression, course or appearance of the disease in a sample from a patient who has that disease or is suspected of having the disease. disease. The method according to claim 27, characterized in that it comprises a detection or determination of the quantity in a first sample at a first time point and in a further sample at a second time point and a comparison of the two samples. 29. The method according to any of claims 22-28, characterized in that the agent is marked in a detectable manner. 30. The method according to any of claims 21-29, characterized in that the sample comprises body fluid and / or body tissue. 31. A method for treating or preventing a disease characterized by the expression or abnormal expression of a tumor-associated antigen, the method comprising the administration of a pharmaceutical composition according to Any of claims 1-20, the tumor-associated antigen has a sequence encoded by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid, comprising a nucleic acid sequence, selected from the group consisting of of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof , (b) a nucleic acid hybridizing with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c). 32. A method for treating, preventing, diagnosing or monitoring a disease, characterized by the expression or abnormal expression of a tumor-associated antigen, the method comprising administering an antibody that binds to the tumor-associated antigen or a portion thereof and is coupled to a therapeutic or diagnostic agent, the tumor-associated antigen has a sequence encoded by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence, selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64 -67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c). 33. The method according to claim 24 or 32, characterized in that the antibody is a monoclonal, chimeric or humanized antibody, or is a fragment of an antibody. 34. A method for the treatment of a patient suffering from a disease characterized by the expression or abnormal expression of a tumor-associated antigen, the method comprising: (i) providing a sample containing immunoreactive cells, (ii) contacting the sample with a host cell expressing the tumor-associated antigen or a portion thereof, under conditions that favor the production of cytolytic T lymphocytes or for the release of cytokines against the tumor-associated antigen or the portion thereof, and (iii) introducing cytolytic T lymphocytes or releasing cytokines into the patient in an amount suitable for lysing cells expressing the tumor-associated antigen or a portion thereof, the tumor-associated antigen, has an encoded sequence by a nucleic acid that is selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25 , 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c) ). 35. The method according to claim 34, characterized in that the host cell recombinantly expresses an MHC molecule that binds to the tumor-associated antigen or to a portion thereof. 36. The method according to claim 34, characterized in that the host cell expresses endogenously an MHC molecule that binds to the tumor-associated antigen or to a portion thereof. 37. A method for inhibiting the development of cancer in a patient, the method characterized in that it comprises administering an effective amount of a pharmaceutical composition according to any of the claims. 1-20. 38. The method according to any of claims 21-37, characterized in that the tumor-associated antigen comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31, 36, 40, 42, 46, 50-60, 63, 68, and 69, a portion or derivative thereof. 39. An agent characterized in that it specifically binds to a protein or polypeptide or a portion thereof, the protein or polypeptide is encoded by a nucleic acid selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c). 40. The agent according to claim 39, characterized in that the protein or polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31 , 36, 40, 42, 46, 50-60, 63, 68, and 69, a portion or derivative thereof. 41. The agent according to claim 39 or 40 characterized in that it is an antibody. 42. The agent according to claim 41, characterized in that the antibody is a monoclonal, chimeric or humanized antibody, or is a fragment of an antibody. 43. An antibody characterized in that it binds selectively to a complex of: (i) a protein or polypeptide or a portion thereof and (ii) an MHC molecule to which the protein or polypeptide or portion thereof is bound, with the antibody that does not bind to (i) or (ii) alone and the protein or polypeptide to be encoded by an acid nucleic acid selected from the group consisting of: (a) a nucleic acid comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid that hybridizes with the nucleic acid of (a) under stringent conditions, (c) ) a nucleic acid that degenerates with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c). 44. The antibody according to claim 43, characterized in that the protein or polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 10, 14, 18, 22, 26, 29, 31, 36, 40, 42, 46, 50 -60, 63, 68, and 69, a portion or derivative thereof. 45. The antibody according to claim 43 or 44, characterized in that it is a monoclonal, chimeric or humanized antibody, or is a fragment of an antibody. 46. A conjugate between an agent according to any of claims 39-42, or an antibody according to any of the claims 43-45 and a therapeutic or diagnostic agent. 47. The conjugate according to claim 46, characterized in that the therapeutic or diagnostic agent is a toxin. 48. A kit for detecting the expression or abnormal expression of a tumor-associated antigen, characterized in that the kit comprises the agents to detect or determine the amount of: (i) a nucleic acid encoding the tumor-associated antigen or a portion thereof, and / or (ii) the tumor-associated antigen or a portion thereof, and / or (iii) the antibodies that bind to the tumor-associated antigen or a portion thereof, and / or (iv) of T lymphocytes, which are specific for a complex between the tumor-associated antigen or a portion thereof and an MHC molecule, the tumor-associated antigen has a sequence encoded by a nucleic acid that is selected from the group consisting of: a) a nucleic acid comprising a nucleic acid sequence, selected from the group consisting of SEQ ID NOs: 1, 5, 9, 13, 17, 21, 25, 28, 30, 35, 39, 41, 45, 49, 61, 62, and 64-67, a portion or derivative thereof, (b) a nucleic acid hybridizing with the nucleic acid of (a) under stringent conditions, (c) a nucleic acid degenerating with respect to the nucleic acid of (a) or (b), and (d) a nucleic acid that is complementary to the nucleic acid of (a), (b) or (c).
MX/A/2008/003464A 2005-09-12 2008-03-12 Identification of tumor-associated antigens for diagnosis and therapy MX2008003464A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP05019786 2005-09-12

Publications (1)

Publication Number Publication Date
MX2008003464A true MX2008003464A (en) 2008-10-03

Family

ID=

Similar Documents

Publication Publication Date Title
JP6196338B2 (en) Identification of tumor-associated antigens for diagnosis and treatment
AU2017203332B2 (en) Identification of tumor-associated antigens for diagnosis and therapy
CA2760395A1 (en) Identification of tumor-associated markers for diagnosis and therapy
MX2008003464A (en) Identification of tumor-associated antigens for diagnosis and therapy