MXPA06014344A - Antibodies for selective apoptosis of cells - Google Patents

Antibodies for selective apoptosis of cells

Info

Publication number
MXPA06014344A
MXPA06014344A MXPA/A/2006/014344A MXPA06014344A MXPA06014344A MX PA06014344 A MXPA06014344 A MX PA06014344A MX PA06014344 A MXPA06014344 A MX PA06014344A MX PA06014344 A MXPA06014344 A MX PA06014344A
Authority
MX
Mexico
Prior art keywords
cells
antibody
pathogens
mhc
recombinant
Prior art date
Application number
MXPA/A/2006/014344A
Other languages
Spanish (es)
Inventor
Reiter Yoram
Klechevsky Eynav
Denkberg Galit
Original Assignee
Denkberg Galit
Klechevsky Eynav
Reiter Yoram
Technion Research & Development Foundation Ltd
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 Denkberg Galit, Klechevsky Eynav, Reiter Yoram, Technion Research & Development Foundation Ltd filed Critical Denkberg Galit
Publication of MXPA06014344A publication Critical patent/MXPA06014344A/en

Links

Abstract

A recombinant isolated antibody and a pharmaceutical composition containing same, capable of specifically recognizing an MHC-peptide complex with an affinity in a nanomolar range and of inducing apoptosis in cancer or pathogen infected cells is provided. Also provided are method for treating and diagnosing cancer or pathogen infection in a subject using the recombinant isolated antibody of the present invention.

Description

ANTIBODIES FOR SELECTIVE FIELD CELL APOPTOSIS AND BACKGROUND OF THE INVENTION The present invention relates to isolated recombinant antibodies that specifically recognize complexes of MHC-peptide and are capable of inducing apoptosis in a restricted manner of peptide-specific MHC, and more particularly , to methods for treating and diagnosing cancer or pathogen infections using such TCR-like, recombinant and isolated antibodies. The ligation of several surface receptors is known to induce apoptosis. In activated B cells, Fas ligation induces apoptosis by activating a number of specific caspases. In addition, the cross-linking of B cell receptors initiates a different apoptotic pathway at certain stages of B cell development. The Major Histocompatibility Complex of class I (MHC-I) plays a central role in the human immune system by acting as a structure for the presentation of peptide to CD81 T cells (Boon T. van der B.P., 1996). MHC-1 molecules were recently recognized as important signal transducing molecules involved in the regulation and fine tuning of immune responses. For example, while cross-linking the aC domain of MHC-I induced apoptosis in the latent B cells, the anti-MHC-I (Ab) antibody, BAL-1, induced apoptosis in the activated B cells. While the apoptotic pathway induced by the MHC-I ligation of T cells is distinctly different from that of Fas, the apoptotic pathway induced by the cross-linking of MHC-I in B cells is not completely understood, one of the events that follows the cross-linking of MHC-I is the mobilization of intracellular calcium. Thus, crosslinking the MHC-I molecules using an anti-β2 microglobulin antibody (Ab) resulted in protein tyrosine phosphorylation of protein tyrosine kinases p53 / 561in and p72sik, leading to an increase in intracellular Calcium (Nagy ZA, et al., 2002, Nagy ZA, et al., 2003; Longo, DL, 2002. Vidovic D, Toral JI; 1998; Pedersen AE, et al., 1999; Ruh ald M, et al., 1999; Mori M, et al., 1999; Genestier L, et al., 1997; Genestier L, et al., 1997; Skov S, et al., 1997). Significant progress in understanding the mechanisms that lead to the cellular immune response against tumor cells was achieved by characterizing the MHC-associated tumor-associated antigens and the arrival of the appropriate methodology developed to monitor immune recognition [Altman et al., 1996; Boon, 1996; Lee et al., 1999; Rosenberg, 2001; Renkvist et al., 2001]. Cytotoxic T lymphocytes recognize antigens as short peptides linked to MHC-I molecules [Klein and Sato, 2000]. Most of the presented peptides of MHC-I are derived from the degradation of intracellular proteins by the proteasome [Niedermann, 2002]. The segmented peptides are transported in the lumen of the endoplasmic reticulum (ER) by a transporter associated with a processing molecule (TAP), possibly protected by the heat shock proteins accompanying the complete degradation before entering the ER. The trimmed peptides bind to the cleft of the assembled MHC-I molecule in the ER and the complex is then transported to the cell surface [Yewdell, 2001; Yewdell and Bennink, 2001]. The selection of peptides and their presentation on the surface of the cell depends on several factors such as the presence of endogenous or exogenous proteins in intracellular compartments, the appropriate degradation of such proteins in intracellular compartments, the ability of degraded peptides to bind the cleavage of the particular HLA molecule, and the successful transport of such molecules to the surface of the cell. Several studies have shown that the inability of the patient's immune system to induce an effective immune response against the tumor is often due to the deficient presentation of the antigen [Restifo et al., 1993; Seliger et al., 2000]. To study the presentation of the MHC-peptide on the surface of macrophages, dendritic cells, tumor cells or virus-infected cells, recombinant soluble T cell receptors (TCRs) were produced in E. coli However, such TCRs exhibited unstable and inherently low affinity for ligands [Wulfing and Pluckthun, 1994]. Thus, it was recognized that antibodies that would recognize MHC-peptide complexes associated with tumor with the same specificity as the TCR would be valuable reagents for studying the presentation of the antigen by the tumor cells, to visualize the MHC-peptide complexes on the cells and eventually to monitor the expression of specific complexes during immunotherapy. In addition, they can serve as good candidates for targeting reagents - in the construction of recombinant immunotoxins, fusions with cytokine molecules or for bi-specific antibody therapy. Antibodies that specifically recognize the peptide-MHC class I complexes were prepared in murine systems [Murphy et al., 1989; Aharoni et al., 1991; Andersen et al., 1996; Reiter et al., 1997; Porgador et al., 1997; Day and collaborators, 1997; Dadaglio et al., 1997; Zhong et al., 1997a; Zhong et al., 1997b; Krogsgaard et al., 2000; Polakova et al., 2000]. However, antibodies with such specificity similar to the intense T cell receptor have proven difficult to produce using conventional hybridoma techniques or immunization strategies even in combination with the selection in vi tro of phage display libraries. The direct selection of such antibodies from libraries of phage antibodies, which are not very large, was only recently demonstrated [Chames et al., 2000; Proc. Nati Acad. Sci USA. 97 (14): 7969-74]. Using this route, a Fab derived from the phage display (ie, associated with phage) was isolated which recognizes the MAGE-Al melanoma antigen in the complex with the human MHC-A1 MHC molecule. However, such an antibody (G8) exhibited a low affinity towards the MHC complex in the 250 nM range (Chames P, et al., 2002, J. Immunol., 169: 1110-8). To overcome the low affinity limitation, Chames P, et al., (2002, J. I munol.169: 1110-8) grafted the T cells expressing the G8 antibody designed in vitro and prepared cell-specific antibodies. However, such antibodies are impractical to treat cancer or infection with pathogens since they require gene therapy manipulations on a patient-specific basis. The present inventors have previously isolated the TCR-like antibodies from a murine scFv phage display library constructed of transgenic HLA-A2 mice that were immunized with the stable, single chain HLA-A2-β2- β molecule formed in complex with a peptide derived from the melanoma differentiation Ag gplOO (Denkberg G, et al, The Journal of Immunology, 2003; 171 (5): 2197-207). In contrast to prior art attempts, this scFv, secreted, soluble recombinant antibody (Gl) exhibited a high affinity (within the nanomolar range, eg, 5 nM) to the HLA-A2-G9-209 complex, in a manner MHC-restricted, peptide-specific, similar to the specificity of TCRs (Denkberg G, 2003, Supra). While the present invention is reduced to practice, the present inventors have discovered that isolated recombinant antibodies that specifically recognize MHC-peptide complexes can be used as potent apoptosis-inducing agents and thus treat cancer or infection by pathogens. BRIEF DESCRIPTION OF THE INVENTION According to one aspect of the present invention there is provided a method for inducing apoptosis in cancer cells or cells infected by pathogens, the method comprising contacting or expressing in the cells a recombinant isolated antibody capable of specifically binding an MHC-peptide complex specifically expressed on the cells, thereby inducing apoptosis in cancer cells or cells infected by pathogens. According to another aspect of the present invention there is provided a method for treating cancer or infection with pathogens comprising administering to, or expressing in the cells of a subject in need thereof a therapeutically effective amount of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, the isolated recombinant antibody is capable of inducing apoptosis of cancer cells or cells infected by pathogens, cancer cells or cells infected with pathogens that specifically express the MHC-peptide complex, treating in this way cancer or infection by pathogens in the subject. According to yet another aspect of the present invention there is provided the use of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, such as a pharmaceutical substance, the isolated recombinant antibody is capable of inducing apoptosis of cancer cells or cells infected by pathogens that specifically express the MHC-peptide complex. According to yet another aspect of the present invention there is provided the use of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, the recombinant isolated antibody is capable of inducing apoptosis of cancer cells or cells infected by pathogens that specifically express the MHC-peptide complex, for the manufacture of a drug identified for the treatment of cancer or infection by pathogens. According to yet another aspect of the present invention there is provided a pharmaceutical composition for treating cancer or infection by pathogens comprising as an active ingredient a recombinant isolated antibody capable of specifically binding an MHC-peptide complex in cancer cells or cells infected by Pathogens that specifically express the MHC-peptide complex, the isolated recombinant antibody is capable of inducing apoptosis in cancer cells or cells infected by pathogens and a pharmaceutically acceptable carrier. According to a further aspect of the present invention there is provided a method for diagnosing cancer or infection by pathogens in a subject, comprising: (a) contacting a biological sample of the subject with a recombinant isolated antibody capable of specifically binding a complex of MHC-peptide under conditions suitable for the formation of the immunocomplex; the isolated recombinant antibody is capable of inducing apoptosis in cancer cells or cells infected by target pathogens, and (b) detecting the formation of the immune complex, thereby diagnosing cancer or infection by pathogens in the subject. According to still a further aspect of the present invention there is provided a method for diagnosing cancer or infection by pathogens in a subject, comprising: (a) contacting a biological sample of the subject with a recombinant isolated antibody capable of binding specifically an MHC-peptide complex under conditions suitable for immunocomplex formation, the isolated recombinant antibody is capable of inducing apoptosis in cancer cells or cells infected by objective pathogens; and (b) detecting a level of apoptosis in the cells of the biological sample thereby diagnosing the cancer or infection by pathogens in the subject. According to yet a further aspect of the present invention there is provided a device for diagnosing cancer or infection by pathogens in a subject, the equipment comprises packaging materials and at least one agent identified by the packaging materials as they are suitable for a immunocomplex formation between a recombinant isolated antibody capable of specifically binding an MHC-peptide complex and the MHC-peptide complex, the isolated recombinant antibody is capable of inducing apoptosis in cancer cells or in cells infected by pathogens.
According to a further aspect of the present invention there is provided a recombinant antibody capable of inducing apoptosis in cancer cells or cells infected by pathogens. According to still another aspect of the present invention there is provided a pharmaceutical composition comprising a recombinant antibody capable of inducing apoptosis in cancer cells or cells infected by pathogens, and an acceptable pharmaceutical carrier. According to yet a further aspect of the present invention a 9H recombinant scFv is provided as set forth in SEQ ID NO: 1. According to yet another aspect of the present invention, a recombinant CLA12 scFv is provided as set forth in SEQ ID NO: 17. According to yet a further aspect of the present invention there is provided a recombinant antibody comprising at least one CDR selected from the group consisting of SEQ ID NOs: 5-16 and 19-24. According to additional features in preferred embodiments of the invention described below, the method further comprises detecting apoptosis in cancer cells or cells infected by pathogens after contact or expression.
According to still further features in preferred embodiments of the invention, the detection of apoptosis is effected by a method selected from a group consisting of the FACS analysis of propidium iodide, FACS Annexin V analysis, Ethidium homodimer-1 spotting, live / dead assay viability / cytotoxicity and Tunnel assay. According to still further features in preferred embodiments of the invention, the recombinant isolated antibody is selected from the group consisting of a Fab fragment and a ScFv. According to still further features in preferred embodiments of the invention, the recombinant antibody is at least bivalent. According to still further features in preferred embodiments of the invention, the ScFv is 9H (SEQ ID NO: 1), Gl (SEQ ID NO: 2) or CLA12 (SEQ ID NO: 17). According to still further features in preferred embodiments of the invention, the peptide is derived from a polypeptide selected from a group consisting of MUC1, gplOO, HTERT, TAX and MARTI. According to still further features in preferred embodiments of the invention, the recombinant antibody comprises at least one CDR selected from the group consisting of SEQ ID NOs: 5-16 and 19-24.
According to still further features in preferred embodiments of the invention, the recombinant antibody. It is an IgG subtype. According to still further features in preferred embodiments of the invention, the cancer is melanoma or breast cancer. According to still further features in preferred embodiments of the invention, the kit further comprises at least one agent identified by the packaging materials as is suitable for detecting a level of apoptosis in cancer cells or cells infected by pathogens. The present invention successfully addresses the deficiencies of currently known configurations by providing a method for inducing apoptosis in cancer and cells infected by pathogens. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein may be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the patent specification, including definitions, will control it. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting. BRIEF DESCRIPTION OF THE DRAWINGS The invention is described herein, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is emphasized that the alles shown are by way of example and for illustrative discussion purposes of the preferred embodiments of the present invention only, and are presented in order to provide what is believed. which is the most useful and easily understood description of the principles and conceptual aspects of the invention. In this regard, no attempt is made to show the structural details of the invention in greater detail than is necessary for a fundamental understanding of the invention, the description taken with the drawings which makes evident to those skilled in the art how the various forms of the invention can be included in practice. In the drawings: Figs. la-f represent the binding of TCR-like antibodies to antigen presenting cells. Figures la-c - RMAS-HHD cells were loaded with the HLA-A2 restricted peptides derived from gplOO of melanoma differentiation antigen G9-209 (Figure la), G9-280 (Figure Ib) and G9-154 (Figure le), or with control HLA-A2 peptides (Figures la-c). Peptide loaded cells were incubated with specific soluble purified ScFv antibodies: ScFv 1A7 (specific to G9-209; Figure, la), ScFv 2F1 (specific to G9-280) and Fab G2D12 (specific to G9-154). Antibody binding was detected using a FITC-labeled anti-human Fab fragment. Figures ld-f -Detection of the HLA-A2 / MUC1-D6 complexes (MUC = mucin) on the surface of cells using a M3al Fab tetramer. JY cells (Figure Id), breast carcinoma cells MDA-MB-231 (Figure 1), or mature HLA-A2 + dendritic cells (DC, Figure 1f) were pulsed with the restricted peptide of MUC1-D6 HLA- A2 derived from mucin. Cells pulsed with peptide were then incubated with the M3A1 tetramer labeled with pseudomonas exotoxins (PE), specific HLA-A2 / MUC1-D6 or monomer antibodies. The binding of the Fab monomer was detected with the anti-human Fab labeled with PE. Control = uncharged cells were labeled with the M3A1 tetramer. Figs. 2a-i represent the detection of specific HLA-A2 / peptide complexes by TCR-like antibodies after naturally occurring active intracellular processing. The APCs of JY HLA-A2 + (Figure 2a) or APD HLA-A2- (Figure 2b) were transfected with the control vector pCDNA or with the pCDNA containing the Tax gene derived from intact full-length HTLV-1 (pCTAX ). Processing of this viral transcription factor gene was shown to result in the generation of the restricted peptide epitope of HLA-A2 Ta n-19. Twelve to twenty-four hours after transfection, the cells were stained by flow cytometry using the T3F2 specific Fab of HLA-A2 / Taxu_? 9 or a Fab similar to G2D12 TCR specific for the epitope gplOO of melanoma differentiation antigen. , G9-154. The efficiency of transduction of the Tax gene in JY and APD cells was monitored by transfection of the pCDNA vector carrying the GFP gene (not shown). Figure 2c - The cells HLA-A2 + RSCD4 and HLA-A2-HUT102, which are lines of human TCD4 + cells infected with HTLV-1, were stained with the Fab tetramer labeled with PE T3F2 and the control G2D12 as shown in FIG. indicates Figures 2d-i- Immunohistochemical staining of HLA-A2 / Tax complexes after active intracellular processing. JY HLA-a2 + or HLA-A2-APD APCs were transfected with a pCDNA expression vector containing the Tax gene (Figures 2d, e, g, h) or with the expression vector alone (Figures 2f, i). ). Twelve to twenty-four hours after transfection, the cells were absorbed onto the slides coated with poly-L-lysine and stained with the specific HLA-A2 / Tax Fab T3F2 (Figures 2d, e, f, h, i) or with Fab G2D12 similar to TCR specific for the epitope of HLA-A2 / G9-154 melanoma gplOO (Figure 2g). Figure 2e is an enlargement (x 60) of the image presented in Figure 2d (x 40). Figs. 3a-h represent the binding of the scfv gl to JY APCs pulsed with peptide (figures 3a-c), melanoma FM3D cells (3d-f figures) or kb3-l cells (figures 3g-h), as determined by the flow cytometry. The cells were pulsed for 3 hours at 37 ° C with peptides G9-280, G9-209, G9-209M or restricted from HLA-A2 Tax as indicated. Black = uncharged cells. Figures 3a-c - Cells were loaded with the peptides G9-209M and G9-280 and incubated with the soluble purified scFv Gl antibody (Figure 3c), the anti-HLA antibody W6 / 32 (Figure 3a), or the antibody anti-HLA-A2 BB7.2 (Figure 3b). The bound antibodies were detected using an anti-Myc antibody labeled with FITC. Note that while both antibodies W6 / 32 and BB7.2 detected the complexes of HLA peptides on the cell surface (Figures 3a and b), the Gl antibody specifically detected the HLA-G9-209M complex but not the HLA-G9 complex. 280 (Figure 3c). Figures 3d-f- The melanoma FM3D cells were pulsed with the peptides derived from gplOO G9-209M (Figure 3e), G9-209 (Figure 3f) or the control G9-280 (Figures 3d, e and f) and subsequently stained with anti-HLA-A2 BB7.2 (Figure 3d) or Gl scFv antibodies (Figures 3e, f). Note that while the BB7.2 antibody detected the cells that were pulsed with the control peptide G9-280 (Figure 3d), the Gl antibody specifically detected the cells that were pulsed with the G9-209 (Figure 3f) or the G9-209M (Figure 3e). Also note that the fraction of the Gl-positive cells loaded with the mutant G9-209M peptide was significantly higher than that of the cells loaded with the G9-209M peptide. Figures 3g-h - Negative KB3-1 cells of HLA-A2 were pulsed with peptides derived from gplOO G9-209 or G9-280 and stained with antibodies BB7.2 (Figure 3g) or Gl scFv (Figure 3h ). Note the absence of stained cells in cells lacking endogenous HLA-A2 molecules. Figs. 4a-g are immunohistochemical analyzes representing the staining of melanoma cells with the Gl scFv antibody. Melanoma cells expressing gplOO, HLA-A2-positive (FM-3-29, M-ww, M-PAT, M-141, FM3-D; Figures 4a-d and f) or negative (G-43; Figures 4e) or the control breast carcinoma cells (MDA-MB-231; Figures 4g) were stained with the Gl scFv antibody. Cells (5 x 10 6) were incubated for one hour on ice with 20-30 μg of Gl scFv in the RPMI culture medium containing 10% fetal calf serum (FCS) followed by a 45 minute incubation on ice with an HRP-anti-Myc antibody. The cell suspension was applied on the slide pre-coated with 0.1% Poly-L-Lysine (Sigma) as described (Harlow and Lane, 1988). The cells were then incubated for 1 hour at room temperature, washed 3 times with phosphate buffer saline (PBS), incubated for 1 hour with a solution of DAB + (Dako), after which the excess staining reagent was washed with PBS. The nuclei of the cells were stained with hematoxylin (Sigma). FM3-D, FM-3-29, M-ww, M-PAT, M-141 and G-43 are melanoma cell lines derived from patients with melanoma; MDA-MB-231 is a cell line derived from a patient with breast carcinoma. Figures 5a-d are FACS analyzes that represent the cytotoxicity of Gl scFv to APCs pulsed with peptides. The RMAS-HHD (not shown) and the JY APCs were loaded with the G9-209 or the Tax control peptides. Peptide loaded cells were subsequently incubated with 25-50 μg of Gl scFv (Figures 5c and d) or remained untreated (Figures 5a and b). The cells were stained with anti-Annexin V (a marker for early apoptosis) and Propidium iodide (Pl) and subjected to flow cytometry. Note the significant increase in the cells labeled with Annexin V in the presence of the Gl scFv antibody in the cells loaded with the G9-209 peptide (Figure 5c) as compared to the cells loaded with the control Tax peptide (Figure 5d). Also note that in the absence of the Gl scFv antibody, no difference was observed in the Annexin C labeling between the cells loaded with G9-209 (Figure 5a) and the cells loaded with the control Tax peptide. Fig. 6 is a graph representing the cytotoxicity assays with. the antibody of Gl scFv on the melanoma cells. Several melanoma cell lines [HLA-A2 + / gpl00 cells (526, 501, 624), HLA-A2- / gpl00 + cells (G-43) and HLA-A2 + cells / gpl00- (1938)] were incubated with the concentrations Increase of Gl scFv antibody and inhibition of protein synthesis was determined by measuring uptake of 3H-Leucine in cellular proteins. The IC50 was determined as the concentration of Gl scFv required to inhibit protein synthesis by 50%. The IC5o was found to be 60 μg / ml for the melanoma cell lines 526, 624 and 501A. These results demonstrate that TCR-like antibodies can induce apoptosis in melanoma target cells. Figures 7a-c are graphs representing the cytotoxic activity of Gl scFv-PE38 on APCs loaded with peptide. TCR-like antibody Gl was fused to a truncated version of pseudomonas exotoxin (PE) a, PE38. The RMAS-HHD cells (Figure 7a) or JY (Figure 7b) were loaded with peptides restricted from HLA-A2 G9-209M and G9-280V peptides derived from the peptide gplOO, T865 (derived from the catalytic subunit of telomerase), the peptide TAX (derived from the Tax protein HTLV-1) or B3 (Fv) -PE38 (a non-specific control immunotoxin targeting the Y Lewis antigen). Cells loaded with peptide were incubated with the increasing concentrations of Gl scFv-PE38. Protein synthesis was determined by the incorporation of 3H-Leucine into cellular proteins. Note, the specific toxicity observed in cells loaded with the peptide G9-209M in the presence of the specific HLA-G9-209 complex. Figure 7c-excess (0.15-0.25 mg / ml) of the indicated scHLA-A2 peptide complex is added to the wells before the addition of Gl scFv-PE38 (25-50 ng / ml). In the control, the MHC-peptide complex. Note the complete inhibition of protein synthesis in cells expressing the scHLA-A2-G9-209 complex. Figs. 8a-e are immunoblotted (Figures 8a-c) and graphs (Figures 8d-e) representing the internalization of TCR-like antibodies (Figures 8a-c) and the killing of melanoma cells (Figures 8d-e). Figures 8a-c - TCR-like antibody 2F1 scFv targeting the epitope gplOO 280 was fused to a truncated version of pseudomonas exotoxin (PE) A, PE38 and was marked with FITC. Fluorescence was monitored by confocal microscopy at time 0 (Figures 8a), 15 minutes (Figures 8b), in 360 minutes (Figures 8c) at 37 ° C. The termination was observed after 15 minutes and after 360 minutes the significant blooming was observed in the cytosol indicative of efficient internalization. Figures 8d-e are graphs depicting the effect of 2F1 directed to gplOO (Figures 8d) or scAv CLA12 directed to an HLA-A2 peptide complex derived from MARTI (Figures 8e) on protein synthesis (followed by measuring the incorporation of 3H-Leucine into cellular proteins) of melanoma cell lines derived from several patients. Inhibition of protein synthesis was calculated as a percentage of cells that do not undergo antibody treatment. Note that while all melanoma cells HLA-A2 + / gpl00 + (526, 501A, 624.38) were sensitive to 2F1 cytotoxicity, melanoma cell lines HLA-A2- / gpl00 + (G-43) or HLA-A2 + / gpl00- (1938) were resistant to antibody treatment (Figure 8d ). For comparison, note the effect of the CLA12 antibody (SEQ IN NO: 17) on the inhibition of cell synthesis of 624-38 and 526 cells but not of G-43 (Figure 8e). FIGs. 9a-J are FASC analyzes that represent cell death of melanoma cells induced by the scFv fragment of TCR-like antibody (free of naked toxin). Figures 9a-h - cells that are peptide HLA-A2 + / antigen + (Figures 9e-h), peptide HLA-A2 / antigen + (Figures 9c-d) or peptide HLA-A2 + / antigen- (Figures 9a-b) are treated with the naked TCR-like antibody fragments 2F1 (Figures 9a and c), 9H (Figures 9b and g), and (Figure 9f) directed to peptides derived with gplOO, the naked TCR-like antibody with CLA 12 (Figures 9d and e). ) are directed against the peptide derived from MARTI or with the control antibody 1A7 (which is specific against HLA-A2-G9-209M but does not recognize the HLA-A2-G9-209 complex). The measurement of cell death was made by spotting the flow cytometry of propidium iodide (Pl) and the percentages of Pl positive cells are indicated. Figures 9i-j - Melanoma cells (624) that are gpl00 + / HLA-A2 + were incubated with antibodies Gl (which is specific against the complex of HLA-A2-G9-209) (Figure 9j) or 1A7 of control (Figure 9i). The measurement of cell death by flow cytometry of propidium iodide (Pl) and Annexin V and percentages are shown for positive cells. FIGs. lOa-d are flow cytometric analyzes that represent the transformation of the ScFv fragments of TCR-like antibody into the complete IgG molecule and the induction of cell death in melanoma cells. The TCR-like antibody 9H targeting the HLA-A2 peptide (G9-209 gplOO) was transformed into a complete IgG molecule and its reactivity was tested on peptide-pulsed JY cells (Figures 10A-d) Four transfected clones are shown different: 1-40 (Figure 10a), 1-35 (Figure 10b), 1-12 (Figure 10c) and 1-65 (Figure 10O) Red -G9-209, green - EBV, blue - Mucol, purple - Tax, black - CMV Controls are several restricted peptides of HLA-A2 (Tax, EBV, Mucl, CMV) DESCRIPTION OF THE PREFERRED MODALITIES The present invention is a method for inducing apoptosis in cells expressing a complex of MHC peptide in a peptide-specific MHC restricted fashion Specifically, the present invention can be used to diagnose and treat cancer or pathogen infection using bare, toxin-free and highly potent antibodies capable of specifically binding the MHC complexes. peptide with an affinity in e The nanomolar interval. The principles and operation of the method for treating and diagnosing cancer or infection by pathogens according to the present invention can be better understood with reference to the accompanying drawings and descriptions. Before explaining at least one embodiment of the invention in detail, it will be understood that the invention is not limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other modalities or of being practiced or carried out in various ways. Also, it will be understood that the phraseology and terminology employed herein is for the purpose of the invention and should not be considered as limiting. The major histocompatibility complex of class I (MHC-I) plays a central role in the human immune system, by acting as a structure for the presentation of peptides to T-CD81 cells (Boon T. van der B.P., 1996). The peptides presented by the MHC complex (HLA-A-β2 microglobulin) are products of degradation of several proteins expressed endogenously, exogenous proteins (for example, that are derived from pathogens such as viruses or bacteria) and / or cancer-related proteins (for example the melanoma differentiation protein gplOO) which are obtained via the proteasome. The MHC-peptide complex is then recognized as an antigen by cytotoxic T lymphocytes [Klein and Sato, 1000]. To study the presentation of MHC-peptide on the surface of macrophages, dendritic cells, tumor cells or virus-infected cells, soluble T cell receptors (TCRs) were prepared in E. coli However, such TCRs were unstable and exhibited low affinity for their ligands [ulfing and Pluckthun., 1994]. Several studies attempted to prepare the TCR-like antibody in murine systems [Murphy et al., 1989; Aaron and collaborators, 1991; Andersen et al., 1996; Reiter et al., 1997: Porgador et al., 1997; Day and collaborators, 1997; Dadaglio et al., 1997; Zhong et al., 1997a; Zhong et al., 1997b: Krogsgaard et al., 2000; Polakova et al., 2000]. However, antibodies with a specificity intensity similar to the T-cell receptor have proved difficult to produce the use of conventional hybridoma techniques or immunization strategies even in combination with the selection in vi tro of the phage display libraries. The direct selection of such antibodies from very large non-immune phage antibody libraries was only recently demonstrated [Chames et al., 2000; Supra]. The use of this route, a phage clone (G8) which recognizes the MAGE-A1 melanoma antigen in the complex was isolated with the human HLA-Al molecule MHC. However, the affinity of the G8 phage towards the MHC-peptide complex was relatively low (250 nM). To overcome the low affinity limitation, Chames p., Et al., (2002; J. Immunol., 169: 1119-8) grafted the T cells with the G8 antibody designed in vitro and prepared the cell-specific antibodies. However, such antibodies are impractical to treat cancer or infection by pathogens since they require manipulations of gene therapy on a patient-specific basis. The present inventors have previously isolated the Gl TCR-like antibody from a murine scFv phage display library constructed of transgenic HLA-A2 mice which were immunized with the stable chain alone HLA-A2-β2 molecule complexed with the melanoma specific peptide G9-209 (Denkberg G., et al, The Journal of Immunology, 2003; 171 (5): 2197-207). In contrast to the prior art attempts, this recombinant scFv (Gl) antibody, which was isolated and soluble (ie, is not a phage clone), exhibited a high affinity (within the nanomolar range, for example, 5 nM) to the HLA-A2-G9-209 complex, in a restricted MHC, peptide-specific manner, similar to the specificity of the TCRs (Denkberg G., 2003, Supra). In sharp contrast to the prior art, the present inventors have discovered that isolated TCR-like (i.e., cell-free) recombinant antibodies, which specifically bind MHC-peptide complexes with affinity in the nanomolar range (e.g. , 5 nM), are capable of inducing apoptosis in cells expressing such complexes. In addition, the present inventors have discovered novel recombinant and isolated scFv antibodies [clones 9H (SEQ ID NO: 1) and CLA 12 (SEQ ID NO: 17)] of a large human synthetic chain single Fv antibody library Azriel-Rosenfeld R, Valensi M, Benhar I. A human synthetic combinatorial library of single chain antibodies that are arranged based on the intermixing of CDRs formed in vivo in the regions of general structure. J. Mol Biol. 2004 Jan 2; 335 (1); 177-92. "These antibodies are capable of specifically binding the complex HLA-A2-G9-209 (scFv 9H) or the complex HLA-A2-MART1 (scFv CLA12) with an affinity of 0.5 nM (for scFv 9H) or nM (for scFv CLA12) Thus, as shown in Figures 3a-h, 5a-d, 6 and 9a-j and the section of Examples below, the previously isolated Gl antibody (Demlberg et al., 2003. Supra), antibodies 9H (SEQ ID NO: 1) and CLA12 (SEQ ID NO: 17) can be used as potent, toxin-free, nude agents for the selective induction of apoptosis of cells expressing MHC-peptide complexes specific (cancer cells) in a peptide-specific HLA-restricted manner Thus, according to one aspect of the present invention, a method for inducing apoptosis in cancer cells or cells infected by pathogens is provided. effect by contacting or expressing in the cells a recombinant isolated antibody capable of and specifically binding an MHC-peptide complex specifically expressed on the cells, thereby inducing apoptosis in cancer cells or cells infected by pathogens.
As used herein the term "apoptosis" refers to the programmed cell death by which the cell executes a "cell suicide" program. Apoptosis plays an important role in a number of physiological events including embryogenesis, regulation of the immune system and homeostasis. Thus, apoptosis could be in response to diverse signals such as limb and neural development, neurodegenerative diseases, radiotherapy and chemotherapy. Apoptotic processes are usually characterized by the uncoupling of mitochondrial oxidation, decreased levels of dinucleotide adenine nicotinamide phosphate [NAD (P) H], cytochrome c release, caspase activation, DNA fragmentation and phosphatidylserine externalization (a membrane phospholipid normally restricted to the inner leaflet of the lipid bilayer) to the outer leaflet of the plasma membrane. As used herein the phrase "MHC-peptide complex" refers to a protein complex formed between any molecule (or complex) of MHC and a peptide. The term MHC includes several classes of MHC complexes (e.g., class I, II, III and the like of MHC). According to a preferred embodiment of the present invention, the term MHC refers to class I of MHC. MHC class I molecules are expressed on the surfaces of most cells and are recognized by CD8 positive cytotoxic CT cells. Class I molecules of MHC are heterodimers composed of subunits of a and β. The a subunit includes three structural domains (al, a2, a3) and is encoded by the MHC gene cluster (for example, the HLA-Al or HLA-A2 genes). The structural domains al and a2 associate to form the cavity that links the peptide and the distal region of the membrane of the molecule. The third domain structures of the a-a3 subunit - are non-covalently associated with the β2 microglobulin of the β subunit; ß2m), forming the proximal region of the membrane of the molecule. The ß2m is not encoded by the MHC gene cluster. The peptide that binds to the MHC molecule can be any peptide obtained by the proteasomal degradation of a protein. Such a protein can be from a pathogenic protein, which is derived from the pathogen [eg, bacteria, viruses, fungi and the like], just as a protein is usually not expressed in normal cells, but rather is expressed only in cancer cells. or precancerous (e.g., tumor promoting proteins, tumor differentiation proteins and the like). Non-limiting examples of MHC-peptide complexes include HLA-A2-G9-209, HLA-A2-G9-280 and HLA-A2-MART1. Additional examples are provided in the Examples section which follows. Preferably, the peptide of the MCH-peptide complex is derived from a protein such as MUC1 (see Cohen et al., 2002), gplOO (Access of GenBank No. AAB31176), HTERT (Access of GenBank No. AAD30037), TAX (see Cohen et al., 2003), and MARTI (Access of GenBank No. Q16655). According to the method of the present invention, the isolated recombinant antibody is capable of specifically binding an MHC-peptide complex expressed specifically in the cells. For example, as shown in Figures 3a. -h, cells pulsed with an antigenic peptide were specifically labeled with the Gl antibody only when expressing the specific HLA-A2 allele. In addition, as shown in Figures 4a-g, the TCR-like antibody Gl specifically recognized natural non-pulsed melanoma cells expressing the G9-209 peptide derived from gplOO. The term "recombinant antibody" as used in this invention includes intact molecules as well as functional fragments thereof, such as Fab, F (ab ') 2, Fv or single domain molecules such as VH and VL to an epitope of a antigen at least partially generated by recombinant DNA technology. These functional antibody fragments are defined as follows: (1) Fab, the fragment which contains a monovalent antigen binding fragment of an antibody molecule, can be produced by digestion of all the antibody with the enzyme of papain for produce an intact light chain and a portion of a heavy chain; (2) Fab ', the fragment of an antibody molecule that can be obtained by treating the whole antibody with pepsin, followed by reduction, to produce an intact light chain and a portion of the heavy chain; two Fab 'fragments are obtained per antibody molecule; (3) (Fab ') 2, the fragment of the antibody that can be obtained by treating the whole antibody with the pepsin enzyme if the subsequent reduction, F (ab') 2 is a dimer of two Fab 'fragments held together by two disulfide bonds; (4) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; (5) single chain antibody ("SCA"), a genetically engineered molecule containing the variable region of the light chain and the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule (scFv); and (6) domain antibodies are only composed of a VH or VL domain which exhibits sufficient affinity for the antigen. The term "antibody" as used herein is not only inclusive of antibodies generated by immunization and recombinant phage display techniques, but also includes any polypeptide that is generated to include at least one region of complementary determination (CDR) ) which is capable of binding specifically to the MHC-peptide complex of the present invention (see, for example, Azriel-Rosenfeld R. et al., 2004; J Mol Biol. 33581): 177-92). Thus, the antibody of the present invention can be expressed (as hereinafter additionally described) of a polynucleotide sequence that includes a coding sequence for at least one antibody CDR. To increase the avidity of the antibody towards the MHC-peptide complex, the antibodies of the present invention are preferably at least bivalent. Such antibodies can be cloned into the subtype (which is bivalent), IgM (which is penta-valent), or selected from such subtypes using methods known in the art. Alternatively, the antibodies can be chemically conjugated such as by using a cross-linker or a sequence tag (for example BirA, see Example 1 of the Examples section which follows). Methods for producing polyclonal and monoclonal antibodies as well as fragments thereof are well known in the art (See for example, Harlow and Lane, Antibodies: A 'Laboratory Manual, Cold Spring Harbor Laboratory, New Cork, 1988, incorporated herein by reference). Antibody fragments according to the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli or mammalian cells (e.g., culture of Chinese hamster ovary cells or other protein expression systems) of DNA that encodes a fragment. Antibody fragments can be obtained by digestion of pepsin or papain from whole antibodies by conventional methods. For example, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F (ab ') 2. This fragment can be further segmented using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from the cleavage of the disulfide bonds, to produce the Fab '3.5S monovalent fragments. Alternatively, an enzymatic cleavage using pepsin produces the monovalent Fab 'fragments and a Fe fragment directly. These methods are described, for example, by Goldenberg, U.S. Patent Nos. 4,036,945 and 4,331,647, and the references contained therein, the patents which are hereby incorporated by reference in their entirety. See also Porter, R.R.
[Biochem. J. 73: 119-126 (1959)]. Other methods to segment antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, in addition to fragment cleavage, or other enzymatic, chemical or genetic techniques can also be used, while fragments are linked to the antigen that is recognized by the intact antibody. The Fv fragments comprise an association of VH and VL chains. This association may be non-covalent, as described in Invar. And collaborators [Proc ,. Nat'l Acad. Sci. USA 69: 2659-62 (19720) Alternatively, variable chains can be linked via an intermolecular or crosslink disulfide bond to chemicals such as glutaraldehyde Preferably, the Fv fragments comprise VH and VL chains connected by a linker. These proteins that bind chain-chain-only antigens (scFv) are prepared by constructing a structural gene comprising DNA sequences encoding the VH and VL domains connected by an oligonucleotide.The structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli Recombinant host cells synthesize a polypeptide chain alone with a linker peptide bridging the two V domains. Methods for producing scFvs are described, for example, by hitlow and Filpula, Methods 2: 97-105 (1991); Bird et al., Science 242: 423-426 (1988); Pack et al., Bio / Technology 11:12 71-77 (1993); and U.S. Patent No. 4,946,778, which are hereby incorporated by reference in their entirety. Another form of an antibody fragment is a peptide coding for a region of complementary determination alone (CDR). CDR peptides ("minimum recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region of the RNA of the cells that produce antibodies. See, for example, Larrick and Fry [Methods, 2: 106-10 (1991)]. For example, such a peptide can be at least one peptide from the group of peptides set forth by SEQ ID NOs: 5-10 (which are derived from 9H scFv), SEQ ID NOs: 11-16 (which are derived from Gl scfv ) or SEQ ID NOs: 19-24 (which are derived from CLA12 scfv). Humanized non-human antibody forms (eg, murine) are chimeric immunoglobulin molecules, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab ', F (ab') 2 or other sequences that bind antigens of antibodies) containing minimal sequences derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which the residues of a region of complementary determination (CDR) of the container are replaced by the residues of a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit that have the specificity, affinity and desired capacity. In some cases, the residues of the Fv structure of the human immunoglobulin are replaced by the corresponding non-human residues. The humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the CDR involved or structure sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the regions of Structure (FR) are those of a human immunoglobulin condensation sequence. The optically humanized antibody will also comprise at least a portion of an immunoglobulin constant region (Fe), typically that of a human immunoglobulin [Jones et al., Nature, 3212: 522-525 (1986): Riecmann et al., Nature, 332 : 323-329 (1988); and borrow, Curr. O Struct. Biol., 2: 593-596 (1992)]. Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced therein from a non-human source. These non-human amino acid residues are often referred to as import residues, which are typically taken from a variable import domain. Humanization can be carried out essentially following the method of inter and co-workers [Jones et al., Nutre, 321: 522-525 (1986); Riechmann et al., Nature 332: 323-327 (1988); Verhoeyen et al., Science, 239: 1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such humanized antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been replaced by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are replaced by analogous site residues in rodent antibodies. Human antibodies can also be produced using various techniques known in the art, including phage display libraries [Hoogenboom and Winter, J. Mol. Biol., 227; 381 (1991; Marks et al., J. Mol. Biol., 222: 581 (1991).] The techniques of Colé et al., And Boerner et al. Are also available for the preparation of human monoclonal antibodies ( Colé et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p.77 (1985) and Boerner et al., J. Immunol., 147 (1): 86-95 (1991); Chames P., et al. 2000, Proc Nati Acad Sci USA 97 (14): 7969-74, Azriel-Rosenfeld R, et al., 2004, J. Mol. Biol. 33581): 177-p2; Denkberg G., et al., 2001, J Immunol., 167: 270-276; Denkberg J. Immunol, Benhar, I., and Reiter, Y., 2002, Phage display of dingle-chain antibody (scfv) constructs Current Protocols in Innunology 48: 104 pp 59 -87; Brekke OH, Loset GA, New Technologies in therapeutic antibody development, Curr Opin Pharmacol 2003, 3 (5): 544-50, Benhar I, Biotechnological applications of phage and cell display, Biotechnol Adv. 2001, 19 (1 ): 1-33] Similarly, the an Human antibodies can be made by introducing human immunoglobulin sites into the transgenic animals, for example, mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. In stimulation, the production of human antibodies is observed, which resembles exactly what is seen in humans in all aspects, including gene rearrangement, assembly and antibody repertoire. This process is described, for example, in U.S. Patent Nos. 5,545,807, 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in the following Marks et al. Publications, Bio / Technology 10,: 779-783 (1992); Lonberg et al., Nature 368: 856-859 (1994); Morrison, Nature 368 812-13 (1994); Fishwild et al., Nature Biotechnology 14, 845-51 (1996); Neuberger, Nature Biotechnology 14: 826 (1996); and Lonberg and Huszar, Intern. Rev. Immunol. 13, 65-93 (1995). According to a preferred embodiment, the antibody of the present invention is isolated from a non-immune phage library essentially as described in Cohen et al., 2002; Lev et al., 2002; Denkberg et al., 2002a, Cohen et al. 2003, which are fully incorporated herein by reference. According to the preferred embodiments of the present invention, the recombinant isolated antibody is a recombinant polypeptide comprising at least one DCR sequence as set forth in SEQ ID NOs: 5-10, 11-16 and / or 19-24. Examples include, but are not limited to, the 9H scFv fragment (SIQ ID NO: 1, Gl (SEQ ID NO: 2) or CLA12 (SEQ ID NO: 17)] and / or any IgG clone that includes at least a CDR or scFv sequence Such recombinant polypeptide can be expressed from a polynucleotide coding such as at least one CDR or scFv of a DNA construct as further described hereinbelow. IgG is produced (using recombinant DNA techniques known in the art and as described further hereinafter) 1, such IgG antibody can be further digested to produce a Fab fragment which is shorter and thus more soluble than the IgG antibody. Complete IgG antibody In addition, as shown in Figures 7a-c, 8d-ey is described in the Examples section which follows, when the coding sequences of the scFvs Gl, 2F1, or C1A12 were fused to encode the sequence of the exot pseudomonas oxin (PE38) an even more highly specific, more potent inhibition of protein synthesis was observed. Thus, using such immunotoxins, 50% inhibition of protein synthesis in melanoma cells was achieved in the presence of only 10 ng / ml of the Gi scFv PE immunotoxins (Figure 7a), 15 ng / ml of the 2F1 scFv PE38 (Figure 8d) and 10-100 ng / ml of Clal2 scFv PE38 (Figure 8e). On the other hand, as shown further in Figures 7a, 8d and e, an almost complete inhibition of protein synthesis was achieved in the presence of 100 ng / ml of the specific immunotoxins. These results suggest the use of antibodies fused to the toxins for the treatment of cancer cells or cells infected by pathogens.
Thus, according to a preferred embodiment of the present invention, the recombinant isolated antibody of the present invention is fused or conjugated to a cytotoxic agent to thereby form a specific immunotoxin. Such toxin agents may be, for example, Pseudomonas exoxins PE35, PE38, PE40, pseudomonas aeroginosa exotoxin A (ETA '), diphtheria toxin (DT390), and the like. Additionally or alternatively, the recombinant isolated antibody of the present invention can be fused to or conjugated to a chemotherapeutic agent (e.g., Mechlorethamine, Florouracil, Dacarbazine, Docetaxel, Carmustine, Vindesine and the like), the antibiotic agent or the antiviral agent. It will be appreciated that such immunotoxins and / or chemotherapy agents can be generated using recombinant DNA techniques (e.g., by ligating the coding sequence of the agent molecule to the recombinant antibody coding sequence, usually downstream of the sequence of encoding the recombinant antibody) or by covalently conjugating the toxin or chemotherapeutic agents to the polypeptide sequence of recombinant isolated antibody (e.g., to the polypeptide set forth by SEQ ID NO: 1, 2, or 17) by methods known in the art . For example by using a variety of agents that couple the bifunctional protein such as N-succinimidyl-3- (2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL) active esters ( such as disuccinimidilsuberate), aldehydes (such as glutaraldehyde), bisazido compounds (such as bis- (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazoniobenzoyl) -ethylenediamine), diisocyanates ( as toliene 2, 6-diisocyanate), and bis-active fluorine compounds (such as 1, 5-difluoro-2,4-dinitrobenzene). For example, a 5-FU peptide conjugate can be generated. as described by Semko (1996) Peptides Abs. 24th Symp. Eur. Pept. Soc P26. A ricin-peptide conjugate can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). L-isothiocyanatobenzyl-3-methyldiethylene-triaminopentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation or radionucleotide to the peptide (see WO94 / 11026). Cancer cells in which apoptosis is induced can be derived from any kind of tumor, including solid tumors (for example, breast cancer, colon cancer, lung cancer, melanoma) and non-solid tumors (for example, leukemia and lymphoma). Preferably, the cancer cells of the present invention are breast cancer cells or melanoma cells. In addition, such cells can be derived by mutagenesis or transfection of normal cells with an oncogene using methods known in the art. Cells infected by pathogens in which apoptosis is induced can be any of the eukaryotic cells, preferably mammals, more preferably human cells, which are infected with the pathogen, i.e. any known viruses, bacteria and / or fungi in the techniques Such cells can be, for example, hematopoietic cells, lymphoid cells, dendritic cells, macrophages, skin cells, neurons, fibroblasts, endothelial cells, smooth muscle cells, and epithelial cells. The virus can be any virus that is capable of infecting the cells of the present invention. For example, human immunodeficiency virus (HIV), hepatitis A, B and C virus, herpes virus, cytomegalovirus (CMV) and Rous sarcoma virus (RSV). The bacterium may be bacteria that is intracellular such as Listeria moncitogenes, Salmonella tifimurio, and Micobaterium tuberculosis (Soloski MJ, et al., Proc Soc Exp Biol. Med. 2000, 224: 231-9). According to the method of the present invention, the recombinant isolated antibody is contacted or expressed in cancer cells or cells infected by pathogens of the present invention. Methods for administering antibodies to cells are known in the art and include, for example, the addition of the antibody from the cell environment such as blood, plasma, regulators, tissue culture medium and the like. For example, as shown in Figures 8a-c, the antibody 2F1 which is directed to the epitope gplOO 280 was added to the medium The culture of the cells was found inside the cells within 360 minutes. Alternatively, the recombinant isolated antibody of the present invention can be expressed in cells (e.g., mammalian cells, bacterial cells, plant cells, yeast cells) as part of a nucleic acid construct that encodes polynucleotides by example, the Fab fragment, the scFV, the full igG antibody or any of the CDRs of the recombinant isolated antibody of the present invention. To express the recombinant isolated antibody of the present invention in mammalian cells, a polynucleotide sequence encoding at least one CDR sequence as set forth by SEQ ID NOs: 5-16 and 19-324 is preferably linked in a construct of nucleic acid suitable for expression of mammalian cells. Such a polynucleotide can be, for example, the polynucleotide set forth by SEQ IN DO: 3 (for 9H scFv), SEQ ID NO: (for Gl scFv), SEQ ID NO_18 (for CLA12 scFv). The nucleic acid construct includes a promoter sequence for directing the transcription of the polynucleotide sequence in the cell in a constitutive or inducible manner. Suitable constitutive promoters for use with the present invention are promoter sequences that are active under most environmental conditions and most cell types such as cytomegalovirus (CMV) and Rous sarcoma virus (RSV). Suitable inducible promoters for use with the present invention include, for example, the tetracycline-inducible promoter. (Zabla M, et al., Cancer Res. 2004, 64 (8): 2799-804). The nucleic acid construct (also referred to herein as an "expression vector") of the present invention includes additional sequences that render this vector suitable for replication and integration into prokaryotes, eukaryotes, or preferably both (e.g. shuttle). In addition, typical cloning vectors may also contain an initiation and translation sequence, the transcription and translation terminator and a polyadenylation signal. By way of example, such constructs will typically include a 5 'LTR, a tRNA binding site, a packaging signal, an origin of DNA synthesis in the second strand, and a 3' LTR or a portion thereof. The nucleic acid construct of the present invention typically includes a signal sequence for the secretion of the peptide from a host cell in which it is placed. Preferably the signal sequence for this purpose is a mammalian signal sequence or the signal sequence of the polypeptide variants of the present invention. Eukaryotic promoters typically contain two types of recognition sequences, the TATA box and the upstream promoter elements. The TATA box, located in the base pairs 25-30 upstream of the transcription initiation site, is thought to be involved in directing the RNA polymerase which is the RNA synthesis. The other upstream promoter elements terminate the rate at which transcription is initiated. Preferably, the promoter used for the nucleic acid construct of the present invention is active in the population of transformed specific cells. Examples of cell-type and / or tissue-specific promoters that include promoters such as albumin that is liver specific [Pinkert et al., (1987) Genes Dev. 1: 268-277], lymphoid-specific promoters [Cameme] and collaborators, (1988) Adv. Innunol 43: 235-257]; in particular promoters of T cell receptors [Winoto et al., (1989) EMBO J. 8: 729-733] and immunoglobulins; [Banerji et al., (1983) Cell 33729-740], neuron-specific promoters such as neurofilament promoter [byrne et al.c. Nati Acad. Scí. USA 86: 5473-5477], pancreas-specific promoters [Edlunch et al., (1985) Science 230: 912-916] or mammary gland-specific promoters such as whey promoters (U.S. Patent No. 4,873,316 and Publication of European Application No. 264,166). The enhancer elements can stimulate up to 1,000-fold transcription of homologous or heterologous bound promoters. The enhancers are active when placed downstream or upstream of the transcription initiation site. Many enhancer elements derived from viruses have a broad host range and are active in a variety of tissues. For example, the SV40 early gene enhancer is suitable for many cell types. Other enhancer / promoter combinations that are suitable for the present invention include those derived with polyoma virus, human or murine cytomegalovirus (CMV), long-term repeat of various retroviruses such as murine leukemia virus, murine sarcoma virus or Rous and HIV. See, Enhancers and Eukaryotic Expression, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. 1983, which is incorporated herein by reference. In the construction of the expression vector, the promoter is preferably positioned approximately at the same distance from the initiation site of the heterologous transcription and that this is from the transcription initiation site in its lateral placement. As is known in the art, however, some variation in this distance can be accommodated without the loss of the promoter function. The polyadenylation sequences can also be added to the expression vector in order to increase the efficiency of the mRNA translation of the recombinant isolated antibody. Two distinct sequence elements are referred for precise and efficient polyadenylation: the GU or U rich sequences located downstream of the polyadenylation site and a highly conserved sequence of 6 nucleotides, AAUAAA, located upstream of nucleotides 11-30. The termination and polyadenylation signals that are suitable for the present invention include those derived from S40. In addition to the elements already described, the expression vector of the present invention may typically contain other specialized elements proposed to increase the level of expression of the cloned nucleic acids or to facilitate identification of the cells carrying the recombinant DNA. For example, a number of animal viruses contain DNA sequences that promote extra chromosomal replication of the viral genome in offensive cell types. The plasmids carrying these viral replicons are replicated episomally while 'appropriate factors are provided by the genes either carried on the plasmid or with the genome of the host cell. The vector may or may not include a eukaryotic replicant. If a eukaryotic replicator is present, then the vector is amplifiable in eukaryotic cells that use the appropriate selectable marker. If the vector does not comprise a eukaryotic replicant, no episomal amplification is possible. In contrast, the recombinant DNA is integrated into the genome of the designed cell, where the promoter directs the expression of the desired nucleic acid. The expression vector of the present invention can additionally include additional polynucleotide sequences that allow for example, the translation of several proteins from a single mRNA such as an internal ribosome entry site (IRES) and the sequences for the denomic integration of the polypeptide promoter-chimeric. Examples for mammalian expression vectors include, but are not limited to, pcDNA3, pcDNA3.1 (+/-), pGL3, pZeoSV2 (+/-), pSecTAG_2_, pDisplay, pEF / myc / cyto, pCMV / myc / cyto, pCR3.1, pSinRepd, DH26S, DHBB, pNMTl, pNMT41, pNMTdl, which are available from Invitrogen, pCI which is available from Promega, pMbac, pPbac, pBK-RSV and pBK-CMV which are available from Strategene, pTRES which is available from Clontech and its derivatives. Expression vectors containing regulatory elements of eukaryotic viruses such as retroviruses can also be used. SV40 vectors include pSVT7 and pMT2. Vectors derived from bovine papilloma virus include pBV-IMTHA, and vectors derived from Epstein Bar virus include pHEBO, and p205. Other exemplary vectors include pMSF, pAV009 / A +, pMTO10 / A +, pMAMneo-5, baculovirus pDSVE, and any other vector allowing the extrusion of the proteins under the direction of the SV-40 early promoter.1, the SV-40 late promoter, etalothionein promoter, murine mammary tumor virus promoter, Rous sarcoma promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells. As described in the above, viruses are highly specialized infectious agents that have developed, in many cases, to bypass host defense mechanisms. Typically, viruses infect and spread in specific cell types. The objective specificity of the viral vectors utilizes their natural specificity for the specifically objective predetermined cell types and thus introduces a recombinant gene into the infected cell. As well as the type of vector used by the present invention will depend on the type of transformed cell. The ability to select suitable vectors according to the type of transformed cell is well within the capabilities of the ordinary skilled person and as such no general description of the selection consideration is provided herein. Bone marrow cells can be targeted using type I of the human T cell leukemia virus (HTVL-I) and kidney cells can be targeted using the heterologous promoter present in the nucleopolyhedrovirus calculogenic baculovirus (AcMNPV) as described in Liang CY et al., 2004 (Aren Virol. 149: 51-60). Recombinant viral vectors are useful for the in vivo expression of the isolated antibody of the present invention since they offer advantages such as lateral infection and objective specificity. Lateral infection is inherent in the life cycle of, for example, retrovirus is the process by which an infected cell alone produces many progeny virions that sprout and infect neighboring cells. The result is that a large area becomes infected quickly, most of which was not initially infected with the original viral particles. It is in contrast to the vertical type of infection in which infectious agents are spread only through the offspring of the daughter. Viral vectors can also be produced that are capable of laterally propagating. This feature can be useful if the desired purpose is to introduce a specified gel into only a localized number of target cells. Several methods can be used to introduce the expression vector of the present invention into the cells. Such methods are generally described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Springs Harbor Laboratory, New York (1989, 1992), in Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, Md. 1989), Chang et al., Somatic Gene Therapy, CRC Press, Ann Arbor, Mich. (1995), Vega et al., Gene Targeting, CRC Press, Ann Arbor Mich. (1995), Vector: A Survey of Molecular Cloning Vectors and Their Uses, Butterworths, Boston Mass. (1988) and Gilboa et al. [Biotechniques 4 (6): 504-512, 1986] and include, for example, stable transfection or transient, lipofection, electroporation and infection with recombinant viral vectors. In addition, see U.S. Patent Nos. 5,464,764 and 5, 487,992 for positive-negative selection methods. The introduction of nucleic acids by viral infection offers several advantages over other methods such as lipofection and electroporation, since the efficiency of the highest transfection can be obtained due to the infectious nature of the viruses.
Currently preferred in vivo nucleic acid transfer techniques (gene therapy in vivo) include transfection with viral or non-viral constructs, such as adenoviruses, lentiviruses, Herpes simplex viruses, or adeno-associated viruses (AAV) and systems based on lipid Lipids useful for the lipid-mediated transfer of the gene are, for example, DOTMA, DOPE, and DC-Chol [Tonkinson et al., Cancer Investigation, 14 (1): 54-65 (1996)]. The most preferred constructs for use in gene therapy are viruses, much more preferably adenovirus, AAV, lentivirus, or retrovirus. A viral construct such as a retroviral construct includes at least one transcriptional promoter / enhancer or site definition element (s), or other elements that control the expression of the gene by other means such as alternating splicing, nuclear RNA export. , or post-translational modification of the messenger. Such vector constructs also include a packaging signal, long terminal repeats (LTRs) or portions thereof, and binding sites of the negative strand primer appropriate to the virus used, unless it is already present in the viral construct. In addition, such a construct typically includes a signal sequence for the secretion of the peptide from a host cell in which it is placed. Preferably the signal sequence for this purpose is a mammalian signal sequence or the signal sequence of the polypeptide variants of the present invention. Optionally, the construct may also include a signal that directs the polyadenylation, as well as one or more restriction sites and an exemplary translation termination sequence, such constructs will typically include 5 'LTR, an RNA binding site, a signal of packaging, an origin of DNA synthesis of the second strand, and a 3 'LTR or a portion thereof. Other vectors can be used that are not viral, such as cationic lipids, polylysine and dendrimers. Unlike the one that contains the elements necessary for the transcription and translation of the inserted coding sequence, the expression construct of the present invention may also include sequences designed to increase the stability, production, purification, yield or toxicity of the expressed peptide. For example, the expression of a fusion protein or segmentable fusion protein comprising the Met variant of the present invention and a heterologous protein can be designed. Such a fusion protein can be designed so that the fusion protein can be easily isolated by affinity chromatography; for example, by immobilization on a specific column for the heterologous protein. Where a cleavage site is designed between the Met portion and the heterologous protein, the Met portion can be released from the chromatographic column by treatment with an appropriate enzyme or agent that cleaves the cleavage site [eg, see Booth et al., (1988) Immunol. Lett. 19: 65-70; and Gardella et al., (1990) J. Biol. Chem. 265: 15854-15859]. It will be appreciated that a variety of prokaryotic or eukaryotic cells can be used as host expression systems to express the isolated recombinant antibody (the polypeptide) of the present invention. These include, but are not limited to, microorganisms, such as bacteria transformed with a recombinant bacteriophage DNA, plasmid DNA expression vector or cosmid DNA containing the coding sequence, the yeast transformed with the recombinant yeast expression vectors containing the coding sequence; plant cell systems infected with recombinant virus expression vectors (eg, cauliflower mosaic virus, CaMV, tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors, such as Ti plasmid, which contains the coding sequence. Mammalian expression systems can also be used to express the peptides of the present invention. Examples of bacterial constructs include the pET series of E. coli expression vectors [Studier et al. (1990) Methods in Enzimol. 185: 60-89). In yeast, a number of vectors containing the constitutive or inducible promoters can be used, as disclosed in U.S. Patent No. 5,932,447. Alternatively, the vectors can be used which promote the integration of the foreign DNA sequence into the chromosome of the yeast. In cases where the plant expression vectors are used, the expression of the coding sequence can be directed by a number of promoters. For example, viral promoters such as the 35S RNA and 19S RNA promoters of CaMV [Brisson et al. (1984) Nature 310: 511-514] or the TMV layer protein promoter [Takamatsu et al. (1987) EMBO J. 6: 307-311] can be used. Alternatively, plant promoters such as the small subunit of RUBISCO [Coruzzi et al., (1984) EMBO J. 3: 1671-1680 and Brogli et al., (1984) Science 224: 838-843] or the promoters can be used. of heat shock, for example, soy hsp 17.5-E or hpsp 17.3-B [Gurley et al., (1986) Mol. Cell. Biol. 6: 559-565]. These constructs can be introduced into the plant cells using the Ti plasmid, the Ri plasmid, the viral plant vectors, the direct DNA transformation,. microinjection, electroporation and other techniques well known to the skilled person. See, for example, Weissbach &; Weissbach, 1988, Methods by Plant Molecular Biology, Academia Press, NY, Section VIII, pp 421-463. Other expression systems such as insect and mammalian host cell systems are well known in the art and can be used by the present invention. The recovery of the recombinant polypeptide is effected after an appropriate time in the culture. The phrase "recovery of the recombinant polypeptide" refers to the collection of the complete fermentation medium containing the polypeptide and does not need to involve additional steps of separation or purification. Not in opposition to the above, the polypeptides of the present invention can be purified using a variety of standard protein purification techniques, such as, but not limited to, affinity chromatography, ion exchange chromatography, filtration, electrophoresis, interaction chromatography. hydrophobic, gel filtration chromatography, reverse phase chromatography, chromatofocusing and differential solubilization. According to preferred embodiments of the present invention, the method according to this aspect of the present invention further comprises a step to detect apoptosis in cancer cells or viral infected cells after administration or expression of recombinant isolated antibody in the present invention. Apoptosis induced by the recombinant isolated antibody of the present invention can be detected using various methods known in the art. Staining of homodimer-1 of ethidium - Apoptosis can be detected by dyes that bind specifically to cells with compromised membranes, that is, dead cells. Briefly, non-fixed cells such as suspension cells, tissue culture, tissue sections and the like are stained with the Etidio homodimer-1 fluorescent dye (excitation, 495 nm, emission, 635 nm). In this assay, living cells have a green fluorescent cytoplasm but no EthD-1 signal, whereas dead cells lack green fluorescence and are stained with EthD-1. Tixpnel assay (Roche, Basel, Switzerland) - marks DNA breaks that are characteristic of cells that undergo apoptosis with fluorescein (excitation 450-500 nm, emission 515-565 nm). Two-Color Fluorescence Assay / Live / Dead Cytotoxicity Assay (Molecular Prohes, Inc., -3224, Eugene, OR, USA) - This assay measures the activity of intracellular esterase with a permeable cell substrate (Calcein-AM) which converts living cells to a fluorescent derivative (polyanion calcein, excitation, 495 nm, emission, 515 nm) which is then retained by the intact plasma membrane of living cells. FACS analysis - using molecules capable of specifically binding cells that undergo apoptosis, such as propidium iodide and Annexin V (see the description of the drawings and the Examples section below). Annexin V is a human protein characterized by the high calcium-mediated binding affinity for phosphatidylserine that subjects externalization to the outer side of the plasma membrane during early apoptosis. DNA fragmentation by gel electrophoresis - Briefly, DNA is extracted from non-fixed cells and subjected to gel electrophoresis (eg, 1.5-2% agarose gel) and the degree of DNA fragmentation is evaluated using any DNA stain such as Ethidium bromide, Syber's Green and the like. As further shown in Figure 6 and described in Example 3 of the Examples section that follows, the naked Gl scFv antibody was able to inhibit protein synthesis of melanoma cells 526, 624 and 501A (and thus , "extermination" of melanoma cells) is a restricted form of MHC, specific to the peptide. In addition, the inhibition of protein synthesis was dose dependent with an IC5 (ie, the concentration of the antibody required to achieve 50% inhibition of protein synthesis) of 60 μg / ml for all melanoma cells HLA-A2 + / gpl00 +. These results strongly suggest the use of the TCR-like recombinant antibodies isolated from the present invention in the treatment of cancer or infection by pathogens. Thus, according to another aspect of the present invention, there is provided a method for treating cancer or infection by pathogens. The method is effected by contacting, or expressing in the cells of a subject in need thereof in a therapeutically effective amount of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, the isolated recombinant antibody which is capable of to induce apoptosis of cancer cells or pathogen-infected cells, cancer cells or pathogen-infected cells specifically express the MHC-peptide complex, thereby treating cancer or infection by pathogens in the subject. The term "treat" refers to inhibiting or interrupting the development of a disease, disorder or condition (i.e., for example, cancer or infection by pathogens) and / or causing the reduction, remission or regression of a disease, disorder or condition. . Those of skill in the art will understand that various methodologies and assays can be used to estimate the development of a disease, disorder or condition, and similarly, various methodologies and assays can be used to estimate the reduction, remission or regression of a disease, disorder. or condition. As used herein, the term "subject" (or "individual" that is interchangeably used herein) includes mammals, preferably humans of any age who suffer from the disease, disorder or condition. Preferably, this term includes individuals who are at risk for developing the disease, disorder or condition. For example, individuals who express low levels of a cancer differentiation protein in a pre-malignant state, or individuals who are infected with a pathogen that is still in a latent phase. As used herein the phrase "cells of a subject" includes any of the cells that are derived from the subject and are taken from the subject (i.e., ex vivo gene therapy) or cells that are part of the subject (i.e. in vivo gene as described hereinabove). It will be appreciated that for ex vivo gene therapy, cells that are derived from the subject (either the subject in need of treatment or a r subject) are cultured in the presence of the appropriate culture medium and transfected ex vivo with a vector of expression containing the polynucleotide designed to express and secrete the isolated recombinant body of the present invention to the target cells (e.g., cancer cells or viral infected cells of the subject), essentially as described hereinabove. Such cells can be, for example, kidney cells, bone marrow cells, keratinocyte cells and lymphocyte cells. The administration of the antibody expressing cells of the present invention can be carried out using any suitable route such as intravenous, intraperitoneal, intra kidney, intra gastrointestinal, subcutaneous, transcutaneous, intramuscular, intracutaneous, intrathecal, epidural and rectal. According to presently preferred embodiments, the cells expressing the antibody of the present invention are introduced to the individual using intravenous, intra-kidney, intra-gastrointestinal and / or intraperitoneal administrations. The cells expressing antibody of the present invention can be derived from either autologous sources such as bone marrow cells alone or from allogenic sources such as bone marrow or other cells derived from non-autologous sources. Since non-autologous cells are likely to induce an immune reaction when the body is administered several procedures have been developed to reduce the likelihood of rejection of non-autologous cells. These include either suppression of the recipient immune system or encapsulation of non-autologous cells or tissues in the immunoisolation, of semipermeable membranes before transplantation. Encapsulation techniques are generally classified as either microencapsulation, involving small spherical vehicles and macroencapsulation, which involves the larger flat sheets and hollow fiber membranes (Uludag, H. et al., Technology of mammalian cel encapsulation.) Adv Drug Deliv Rev. 2000; 42: 29-64). Methods for preparing the microcapsules are known in the art and include for example those disclosed by Lu MZ, et al., Cell encapsulation with alginate and 'alpha-phenoxycinnamilidene-acetylated poly (allylamine). Biotechnology Bioeng. 2000, 70: 479-83, Chang TM and Prakash S. Procedures for microencapsulation of enzymes, cells and genetically engineered microorganisms. Mol Biotechnol. 2001, 17: 249-60, and Lu MZ, et al., A novel cell encapsulation method using photosensitive poly (alilamine alfa-cyanocinnamylideneacetate). J Microencapsul. 2000, 17: 245-51. For example, the microcapsules are prepared by complexing the modified collagen with a terpolymer shell of 2-hydroxyethylmethylacrylate (HEMA), methacrylic acid (MAA) and methyl methacrylate (MMA), resulting in a capsule thickness of 2-5 μm. . Such microcapsules can be further encapsulated with additional 2-5 μm Pert-polymer shells in order to impart a negatively charged smooth surface and to minimize the absorption of plasma protein (Chia, S.M et al., Multi-layered microcapsules for cell encapsulation Biomaterials. 2002 23: 849-56). Other microcapsules are based on alginate, a marine polysaccharide (Sambanis, A. Encapsulated islets in diabetes treatment Diabetes Thechnol, Ther 2003, 5: 665-8) or its derivatives. For example, the microcapsules can be prepared by the complex formation of the polyelectrolyte between the sodium alginate of polyanions and the sodium cellulose sulfate with poly (methylene-co-guanidine) hydrochloride polycation in the presence of calcium chloride. It will be appreciated that the encapsulation of the cell is improved when the smaller capsules are used. Thus, quality control, mechanical stability, diffusion property, and in vitro activities of the encapsulated cells are improved when the capsule size is reduced from 1 mm to 400 μm (Canaple L. et al., Improving cell encapsulation through size control J Biomater Sci Plym Ed. 2002; 13: 783-96). On the other hand, nanoporous biocapsules with well controlled pore size as small as 7 nm, adjusted surface chemistries and precise microarchitectures were found to successfully immunoisolate microenvironments for cells (Willians D. Small is beautiful: microparticle and nanoparticle technology in medical devices, Med Device Technol 1999, 10: 6-9, Desai, TA Microfabrication technology for pancreatic cell encapsulation, Expert Opin Biol Ther 2002, 2: 633-46). The recombinant isolated antibody of the present invention, the nucleic acid construct encoding the same or the cells expressing same may be administered to an organism per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients. As used herein a "pharmaceutical composition" refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.The purpose of a pharmaceutical composition is to facilitate the administration of A compound to an organism In the present term the term "active ingredient" refers to the recombinant isolated antibody of the present invention or the polynucleotide or the cells expressing the same accountant for the biological effect. "physiologically acceptable" and "pharmaceutically acceptable" which can be used interchangeably refer to a carrier or a diluent that does not cause significant irritation to an organism and does not negate the biological activity and properties of the compound administered.An adjuvant is included under these phrases. In the present the term "excipient" is refers to an inert substance added to a pharmaceutical composition to further facilitate the administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for the formulation of drug administration can be found in "Remington's Pharmaceutical Sceinces," Makc Publishing Co. , Easton, PA, latest edition, which is incorporated herein by reference. Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramodular injections as well as intrathecal, direct intraventricular, intracardiac, intravenous, intraperitoneal, intranasal, or intraocular injections. . Alternatively, the pharmaceutical composition can be administered in a local rather than systemic manner, for example, by the injection route of the pharmaceutical composition directly into a tissue region of a patient. The pharmaceutical compositions of the present invention can be manufactured by processes well known in the art, for example, by means of conventional polishing, and solution, granulation, dragee manufacture, levigation, emulsification, encapsulation, atrophy or lyophilization processes. The pharmaceutical compositions for use according to the present invention can thus be formulated in the conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate the processing of the active ingredients in the preparations which can be used pharmaceutically . The appropriate formulation is dependent on the chosen route of administration. For injection, the active ingredients of the pharmaceutical composition can be formulated in aqueous solutions, preferably in physiologically compatible regulatory solutions such as Hank's solution, Ringer's solution, or physiological salt buffer solution.
For transmucosal administration, the appropriate penetrants to the barrier to be penetrated are used in the formulation. Such penetrants are generally known in the art. For oral administration, the pharmaceutical composition can be easily formulated by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers allow the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, watered pastes, suspensions and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, then adding the appropriate auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, which include lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth gum, methyl cellulose, hydroxypropylmethyl cellulose, sodium carbomethyl cellulose; and / or physiologically acceptable polymers such as polyvinylpyrrolidone. { PVP). If desired, disintegrating agents can be added, such as crosslinked polyvinylpyrrolidone, agar or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions can be used which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. The dye materials or pigments can be added to the coatings of tablets or dragees for identification or to characterize different combinations of active compound doses. Pharmaceutical compositions that can be used orally include push-fit capsules made of gelatin as well as sealed, soft capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Push-fit capsules may contain the active ingredients in the mixture with fillers such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, the stabilizers can be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. For administration by nasal inhalation, the active ingredients for use according to the present invention are conveniently supplied in the form of an aerosol spray presentation of a pressurized pack or a nebulizer with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane or carbon dioxide. In the case of a pressurized aerosol, the dosing unit can be determined or provided with a valve to supply a measured quantity. Capsules and cartridges of, for example, gelatin for use in a dispenser can be formulated containing a mixture of powder of the compound and a suitable powder base such as lactose or starch. The pharmaceutical composition described herein may be formulated for parenteral administration, for example, by bolus injection or continuous infusion. Formulations for injection may be present in the unit dosage form, for example, in ampules or in multiple dose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulating agents such as suspending, stabilizing and / or dispersing agents. Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in the water-soluble form. Additionally, suspensions of the active ingredients can be prepared as appropriate oily or water-based injection suspensions. Suitable lipophilic solvents or vehicles include fatty acids such as sesame oil, or synthetic fatty acid esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain stabilizers or suitable agents that increase the solubility of the active ingredients to allow the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution for a suitable vehicle, eg, sterile, pyrogen-free water-based solution, before use. The pharmaceutical composition _ the present invention can also be formulated in rectal compositions such as suppositories or retention enemas, using, for example, conventional suppository bases such as cocoa butter or other glycerides. Pharmaceutical compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients (the isolated recombinant antibody of the present invention or the polynucleotide or cells expressing same) effective to prevent, mitigate or ameliorate symptoms of a disorder (e.g., cancer or viral infection) or prolong the survival of the subject being treated. The determination of a therapeutically effective amount is well within the ability of those skilled in the art, especially in the illustration of the detailed description provided herein. For any preparation used in the methods of the invention, the therapeutically effective amount of times can be estimated initially from in vitro cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more precisely determine useful doses in humans.
The toxicity and therapeutic efficacy of the active described active ingredients described herein can be determined by standard pharmaceutical procedures in vi tro, in cell cultures or experimental animals. The data obtained from these in vitro assays and culture of cells and animal cultures can be used in formulating a dosage range for use in humans. The dosage may vary depending on the dosage form used and the route of administration used. The exact formulation, route of administration and dosage can be chosen by the physician or individual in view of the condition of the patient. (See, for example, Fingí, et al, 1975, in "The Pharmacological Basis of Therapeutics," C. 1, p.1). For example, as shown in Figure 6 and the Examples section that follows, the Gl scFV antibody caused a 50% inhibition of protein synthesis in melanoma cells at a concentration of 60 μg / ml, and a 80% inhibition at a concentration of 120μg / ml. In addition, as shown in Table 2 of the Examples section below, while the 2F1 ScFv antibody exhibited an IC50 of 130 μg / ml on protein synthesis in melanoma cells, the full-length IgG clone which derived from such Fab fragment exhibited an IC50 significantly lower than 4 μg / ml.
In addition, as shown in Figures la-c, 8d-ey is described in the Examples section that follows, when immunotoxins were used inhibition of melanoma cells was achieved with IC50 concentrations of 10 ng / ml of Fl scFv Pe38 (Figure 7a), 15 ng / ml of the 2F1 scFv PE38 (Figure 8d) and 10-100 ng / ml of the CLA12 scFv PE38 (Figure 8e). The amount and range of dosage can be adjusted individually to provide the cancer cells or viral cells with the active ingredient that are sufficient to induce apoptosis (minimum effective concentration, MEC). The MEC will vary for each preparation, but it can be estimated from the in vitro data. The dosages necessary to achieve the MEC will depend on the individual characteristics and administration routes. Detection assays can be used to determine plasma concentrations. Depending on the severity and sensitivity of the condition being treated, the dosage may be from one or a plurality of administrations, with the course of treatment lasting from several days to several weeks or until the cure is effected or the decrease in state of illness is achieved. The amount of a composition that is administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc. The compositions of the present invention may, if desired, be present in a package or dispensing device, such as an FDA approved equipment, may contain one or more unit dosage forms containing the active ingredient. The package may, for example, comprise a thin sheet of metal or plastic, such as a vesicular packing. The packaging or dispensing device may be accompanied by instructions for administration. The package or dispenser can also be accommodated by a notice associated with the container in a predescribed form - by a government agency that regulates the manufacture, use or sale of pharmaceutical materials, which the notice is reflective of approval by the agency of the form of the compositions for human or veterinary administration. Such notice, for example, may be labeling approved by the United States Food and Drug Administration for the prescription of drugs or drugs. of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier can also be prepared, placed in an appropriate container, and labeled for treatment of a stated condition, as is further detailed in the foregoing. It will be appreciated that the method of the present invention can also be used to treat various other pathologies that are associated with produced levels of apoptosis. These include, for example, psoriasis, (Víctor FC and Gottlieb AB, 2002, J. Drugs Dermatol.1: 264-75), ichthyosis (Melino G, et al., 2000, Methods Enzi ol. 322: 433-72), common warts, keratoacanthoma (Tsuji T, 1997, J. Cutan, Pathol 24: 409-15), seborrheic keratosis (Satchell AC, et al, 2004, Br. J. Dermatol 151: 42-9), seborrhea, carcinomas of scaly cell (SCC; Seta C, et al., 2000. J. Oral Pathol. Med. 29: 271-8) basal cell carcinoma (BCC, Li C, et al., 2004, Oncogene, 2004, 23: 1608-17) nonmelanoma skin cancer (NMSC) and multiple human tumors. While further reducing the present invention in practice, the present inventors have discovered that the isolated recombinant antibody of the present invention can be used to diagnose cancer or infection by pathogens. As shown in Figure 4a-f and described in Example 2 of the examples section below, the Gl scFv antibody was capable of specifically immunoblotting FM-3-29 expressing HLA-A2-positive / gplOO, M -ww, M-PAT, M-141 and the natural melanoma cells FM3-D, but not the natural melanoma cells G-43 that express the HLA-A2 negative / gplOO. Thus, according to a further aspect of the present invention there is provided a method for diagnosing cancer or viral infection in a subject. The method is effected by (a) containing a biological sample of the subject with a recombinant isolated antibody capable of specifically binding an MHC-peptide complex under conditions suitable for immunocomplex formation; the recombinant isolated antibody that is capable of inducing apoptosis in the objective cancer cells or viral infected cells, and (b) detecting the formation of the immunocomplex, thus diagnosing the cancer or viral infection in the subject. As used - herein the phrase "diagnose" refers to the amplification of a disease (cancer or infection by pathogens) or a symptom, which determines a severity of the disease, which monitors the progression of disease, predicts a result of illness and / or recovery prospects. As used herein, "biological sample" refers to a sample of tissue or fluid isolated from a subject, including but not limited to, for example, blood cells, bone marrow cells and specifically macrophages, fluid lymphoid, various tumors, neuronal cells, dendritic cells, organs and also samples of cell culture constituents in vivo. It should be noted that a "biological sample of the subject" may also optionally comprise a sample that has not been physically removed from the subject. According to one of the preferred embodiments of the present invention, the cancer which is diagnosed using the method according to this aspect of the present invention is a solid tumor (e.g., breast cancer, melanoma and the like). Preferably, the biological sample obtained from the subject is a tissue specimen such as a tissue biopsy which is preferably cut into sections (e.g., paraffin sections or cryosections). The diagnosis of cancer or infection by pathogens according to the present invention can be made by contacting the biological sample of the subject with the isolated recombinant antibody of the present invention under conditions suitable for the formation of the immune complex. As used herein the term "immunocomplex" refers to a complex formed between an antibody (e.g., the isolated recombinant antibody of the present invention as described herein above) and its specific antigen (e.g., a complex of MH-specific peptide such as the HLA-A2-G9-209 complex, the HLA-A2-G9-280 complex, the HLA-A2-MART1-35 complex). The immunocomplex of the present invention can be formed in a variety of temperatures, salt concentration, and pH values that can vary depending on the antibody used and the cells that present the antigen and those skilled in the art are capable of adjusting the appropriate conditions for the formation of each immunocomplex. For example, as described in Example 2 in the description of Figures 4a-g, for the formation of the immunocomplex F1-HLA-A2-G9-209, melanoma cells (e.g., cells FM-3-29, M-ww, M-PAT, M-141) were incubated for 1 hour on ice with 20-30 μg Gl scFv in RPMI culture medium containing 10% fetal calf serum (FCS). According to this method of the present invention, the detection of the immunocomplex formation is indicative of a diagnosis of disease, cancer or viral infection. Various methods can be used to detect the immunocomplex of the present invention and those of skill in the art are able to determine which method is suitable for each immunocomplex and / or the type of cells used for diagnosis. For example, the immunocomplex can be detected by immunohistochemistry or conventional immunofluorescence, FACS, ELISA, Western blot and RIA analysis, or by a method based on molecular weight. . Immunohistochemistry or Immunofluorescence Analysis - This method involves the detection of an antigen (e.g., the MHC-peptide complex) in situ in fixed cells by antigen-specific antibodies (i.e., the isolated recombinant antibody of the present invention). Antigen-specific antibodies can bind to enzymes or bind to fluorophores. The detection is by means of microscopy and subjective or automatic evaluation. If enzyme-linked antibodies are used, a colorimetric reaction may be required. It will be appreciated that immunohistochemistry is frequently followed by the counter staining of cell nuclei using for example Hematoxylin or Giemsa stain. Classification of fluorescent activated cell (FACS) - This method involves the arrest of an antigen in situ in cells by antigen-specific antibodies. The antigen-specific antibodies bind to the fluorophores. The detection is by means of a cell sorting machine which is the wavelength of the light emitted from each cell as it passes through a luminous ace. This method can use two or more antibodies simultaneously. Enzyme Linked Immunosorbent Assay (ELISA) -This method involves fixing a sample (e.g., fixed cells or a proteinaceous solution) containing an antigen (e.g., the MHC-peptide complex) to a surface such as a cavity of a plate of icrotítulo. An antigen-specific antibody coupled to an enzyme is applied and allowed to bind to the antigen. The presence of the antibody is then detected and quantified by a calorimetric reaction employing the enzyme coupled to the antibody. The enzymes commonly used in this method include horseradish peroxidase and alkaline phosphatase. If it is well calibrated and is within the linear range of response, the amount of substrate present in the sample is to provide the amount of color produced. A substrate standard is clearly used to improve quantitative accuracy. Western blotting - This method involves the separation of a substrate from another protein by means of an acrylamide gel followed by the transfer of the substrate to a membrane (eg, nylon or PVDF). The presence of the substrate is then detected by the substrate-specific antibodies, which in turn are detected by the reagents that bind to the antibody. Reagents that bind antibodies can be, for example, protein A, or other antibodies such as those in the ECL kit (Amersham Biosciences Ine, Piscataway, NJ, USA). Reagents that bind antibodies can be radiolabeled or linked to enzymes as described hereinabove. The detection can be by autoradiography, colorimetric reaction or quinolinescence. This method allows both the quantification of a quantity of the substrate, and the determination of its identity by a relative position in the membrane which is indicative of a migration distance in the acrylamide gel during electrophoresis. It will be appreciated that in the case of the MHC-peptide complex, non-denaturing gel electrophoresis is preferably employed. Radio-immunoassay (RIA): In one version, this method involves the precipitation of the desired antigen (ie, the MHC-peptide complex) with a specific antibody and the protein that binds radiolabelled antibodies (eg, protein A labeled with I 125 ) immobilized on a precipitable carrier such as agarose beads. The number of beads in the pellet pellet is to provide the amount of antigen. In an alternate version of the RIA, a labeled antigen and a protein that binds unlabeled antibodies are used. A sample containing an unknown quantity of antigens is added in varying amounts. The decrease in the precipitated counts of the labeled antigen is to provide the amount of the antigen in the added sample. Method based on molecular weight - It will be appreciated that the immunocomplex formed between the MHC-peptide complex exhibits a higher molecular weight than its components, i.e., the isolated recombinant antibody or the MHC-peptide complex. Thus, methods capable of detecting such a change in molecular weight can also be employed. For example, the immune complex can be detected by a gel retardation assay. Briefly, a non-denaturing acrylamide gel is loaded with the samples containing the recombinant isolated antibody and the MHC-peptide complex before and after the immunocomplex formation. A change in the size (molecular weight) of the protein product as compared to its components is indicative of the presence of an immunocomplex. Such a change to a higher molecular weight can be observed using a non-specific protein stain such as silver staining or commassie blue staining. Alternatively, the MHC or recombinant isolated antibody can be labeled (eg, with a radioactive label) before gel electrophoresis. Additionally or alternatively, cells expressing the MHC complex can be radioactively labeled prior to protein extraction. Preferably, the method for detecting immunocomplex formation is immunohistochemistry (including immunofluorescence) which is performed on tissue sections (e.g., paraffin sections or cryosections) of a solid tumor. It will be appreciated that diagnosis of cancer or infection by pathogens can also be effected upon detection of apoptosis (as described hereinabove) in the cells after contact of the cells with the isolated recombinant antibody of the present invention. Therefore, according to yet a further aspect of the present invention there is provided a method for diagnosing cancer or pathogen infection in a subject. The method is effected by (a) putting a biological sample of the subject with a recombinant isolated antibody capable of specifically binding an MHC-peptide complex under conditions suitable for immunocomplex formation; the isolated recombinant antibody that is capable of inducing apoptosis in objective cancer cells or cells infected by pathogens; and (b) detecting a level of apoptosis in the cells of the biological sample thereby diagnosing the cancer or infection by pathogens in the subject. The agents described hereinabove for the detection of immunocomplex formation can be included in a diagnostic equipment / article of manufacture preferably along with appropriate instructions for use and labels indicating FDA approval for use in the diagnosis and / or estimation of a cancer security from a viral infection.
Such a kit may include, for example, at least one container that includes at least one of the diagnostic agents described above (eg, Gl, 9H or CLA12 antibody) and one imaging reagent packaged in another container (eg, enzymes, secondary antibodies, regulatory solutions, chromogenic substrate, fluorogenic material). The team can also include appropriate and conservative regulatory solutions to improve the shelf life of the equipment. According to preferred embodiments of the present invention, the diagnosis of cancer or viral infection can also be made by detecting the level of apoptosis in the cells after the contact of the recombinant isolated antibody with the cells. Such methods are described hereinabove. As used herein, the term "approximately" refers to ± 10%. Objects, advantages and additional novel features of the present invention will become apparent to one ordinarily skilled in the art in the summary of the following examples, which are not intended to be limiting. Additionally, each of the various embodiments and aspects of the present invention as described hereinabove and as claimed in the claims section immediately find experimental support in the following examples. EXAMPLES Reference is now made to the following examples, which in conjunction with the foregoing descriptions, illustrate the invention in a non-limiting manner. Generally, the nomenclature used herein and the laboratory procedures used in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are fully explained in the literature. See, for example, "Molecular Cloning: A Laboratory Manual" Sambrook et al. (1989); "Current Protocois in Molecular Biology" Volumes 1-111 Ausubel, R. M., Ed. (1994); Ausubel et al., "Current Protocois in Molecular Biology," John Wleyley and Sons, Baltimore, Maryland (1989); Perbal, "A Practical Guide to Molecular Cloning," John Wiley & Sons, New York (1988); Watson et al. "Recombinant DNA", Scientific American Books, New York; Birren et al. (Eds.) "Genome Analysis: A Laboratory Manual Series", Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801.53 1; 5,192,659 and 5,272,057; "Cell Biology: A Laboratory Handbook", Volumes 1-111 Cells ,. J. E., Ed. (1994); "Culture of Animal Cells - A Manual of Basic Technique" by Freshney, Wiley-Liss, N. Y. (1994), Third Edition; "Current Protocols in Irnmunology" Volumes 1-111 Coligan J. E., Ed. (1994); Stites et al. (Eds.), "Basic and Clinical Immunology" (8th Edition), Appleton & Lange, Norwallc, CT (1994); Misheli and Shiigi (Eds.), "Selected Methods in Cellular Immunology," W. H. Freeman and Co. , New York (1980); immunoassays are extensively described in the patent and the scientific literature, see, for example, U.S. Patent Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; "Oligonucleotide Synthesis" Gait, M. J., Ed. (1984); "Nucleic Acid Hybridization "Hames, B.D., and Higgins S.J., Eds. (1985); "Transcription and Translation" Hames, B.D., and Higgins S.J., Eds. (1984); "Animal Cell Culture" Fresbney, R. 1., Ed. (1986); "Inimobilized Cells and Enzymes" IRL Press, (1986); • "A Practice Guide to Molecular Cloning" Perbal, B., (1984) and "Methods in Enzymology" Vol. 1-317, Academic Press; "Protocois PCR: A Guide to Methods and Applications", Academic Press, San Diego, CA (1990); Marshak et al. "Strategies for Protein Purification and Characterization - A Laboratory Course Manual "CSHL Press (nineteen ninety six); "The function of MHC molecules" Iii "Funda p Immunology, "Paul WE (Ed.), 1999, Forth edition, Lippincott-Raven publisher, all of which are incorporated by reference as fully set forth herein, and other general references are provided throughout this document. present are believed to be well known in the art and are provided for the convenience of the reader All the information contained herein is incorporated herein by reference EXAMPLE 1 PREPARATION OF TCR SIMILAR ANTIBODIES Isolation and Characterization of TCR-like antibodies - The model system used in the present study was the HLA-A2 human superhaplotype molecule, which is the most frequent MHC allele in the Caucasian population (approximately 40%) .The recombinant MHC-peptide complexes that exhibit the epitopes of the T cell peptide of the associated tumor derived peptides and viral antigens, generated by using a MHC single-chain construct ( scMHC) [Denkberg et al., 2000; Denkberg et al., 2001]. Lev et al., 2002, used recombinant designed single chain MHC-peptide complexes to isolate antibodies with TCR-like specificity than a large non-immune library repertoire of 'phage antibodies. The complexes of HLA-A2 / peptide-classified targets included a variety of epitopes derived from tumor-associated antigens such as the catalytic subunit of telomerase (hTERT) widely expressed in many tumor cells, the melanoma differentiation antigen gplOO [Denkberg et al. collaborators, 2002a], the mucin associated with the epithelial cell (MUCI) related to breast carcinomas [Cohén et al, 2002] or viral epitopes derived from the human T cell lymphotropic virus type I (HTLV-1) of the transcription factor (TAX) [Cohén et al., 2003], epitope of influenza matrix protein or the epitope derived from Epstein-Bar virus (EBV). After two and three cycles of selection on peptide-MHC complexes, an unexpected and surprisingly high frequency (6-80%) of antibodies recognizing human HLA-A2-MCH molecules formed in complex with several peptides was observed. On the other hand, a significantly high proportion of these antibodies (20-80%) exhibited T cells similar to the properties binding the specific, MHC-restricted peptide. Unlike previous reports in murine systems where only a rare antibody clone could only be isolated from the hybridoma or a phage display library, here a panel of different antibodies for each MHC-classified peptide complex was isolated [Cohen and collaborators, 2002; Lev et al., 2002; Denkberg et al., 2002a; Cohen et al., 2003]. These antibodies, with the restricted specificity of MHC-specific antigen of T cells bound to their corresponding MHC-peptide complex with a high affinity in the nanomolar range. In addition, these antibodies were selective to the MHC complex only when the specific peptide was displayed. Accordingly, these antibodies exhibit binding and kinetic properties of antibodies but copy the fine specificity of the T cells so that antibodies similar to TCR were classified. To demonstrate the specific binding of the isolated antibody fragments (scFv) to the MHC-peptide complex in the native form, as expressed on the surface of the cell, several strategies were employed. These experiments showed that the specific scFv bound only to the cells that exhibit the peptide "appropriate specific (Denkberg, G., et al., 2002a; Lev, A., and collaborators, 2002: Cohen CJ. and collaborators, 2002; Lev, A., and collaborators, 2002, Human Recombinant Antibodies with MCH-restricted T Cell Receptor-Like Spedificity: Recognition of distinct T cell epitopes derived from a variety of tumor associated antigens. Tumor Microenvironment: Progretíon, Therapy and Prevention Monduzzi editore press Pp 163-167; Cohen, CJ. , et al., 2003, J. Immunol 170: 4349-4361; Denkberg, G. et al., 2003, J. hn unol. 171: 2197-2207; Cohen C.J., et al., 2003, J. Mol. Pick up 16: 324-332; Yamano, Y., et al., 2004, J. Exp. Med. 199: 1367-1377; Held, G., and collaborators, 2004, Eur. J. Immunol. 34: 2919-2929). For example, the ScFvs 1A7, 2F1 and G2D12 reacted only with their RMA-S-HHD cells loaded with the epitope G9-209, G9-280 and G9-154 derived from respective gplOO and not with the cells loaded with the control peptides ( Figures la-c). Several other APCs were used to demonstrate peptide specific binding of these Fabs, such as HLA-A2 + JY cells from B-lymphoblast transformed with TAP + EBV or mature dendritic cells (DCs). In these cells, the peptide loading is facilitated by the exchange of endogenously derived peptides with HLA-A2 restricted peptides supplied externally by incubating the cells with the desired peptides. TCR-like Fab fragments reacted with JY cells or mature DCs incubated with the specific peptide whereby they were selected but not with the control cells exhibiting other control-restricted HLA-A2 peptides as shown in Figure Ib . The unexpectedly high frequency of these antibodies and the ability to isolate several different antibodies directed at either the complex even more surprisingly in view of previous reports, in which the use of immunized or natural phage libraries resulted in only one clone. of antibody alone [Andersen et al., 1996; Porgador et al., 1997; .Dadaglio et al., 1997; Zhong et al., 1997b; Krogsgaard et al., 2000; Polakova et al., 2000; Cohen et al., 2003]. Study of antigen presentation using TCR-like antibodies - TCR-like antibodies are tools to study antigen presentation in transfected models as well as in tumor cells. For example, to examine the ability of TCR-like Fab antibodies to detect HLA-A2 / Tax complexes produced under physiological antigen processing conditions, JY cells that display the HLA-2-positive B cell antigen transformed with EBV were transfected with the HTLV-1 Tax gene. Twenty-four hours after transfection, the previous control of significant spotting could be clearly observed using TCR-like antibodies only with the JY HLA-A2-positive cells transfected with the Tax gene but not with the APD-negative HLA-A2 cells transfected with the Tax gene (Figures 2d-i). The restricted pattern of MCH, peptide-specific reactivity by Fabs anti TAX / HLA-A2 was not due to differences in the efficiency of transfection or HLA expression of JY and APD cells because the percentage of Transfected cells were similar as determined in the control experiments using a GFP construct. These results indicate that TCR-like Fab antibodies are capable of detecting the MCH-specific peptide complex of the endogenous and endogenous intracellular processing that occurs naturally. Since the density of a particular peptide-HLA complex on the tumor cells is expected to be low compared to the pulsed transfected peptide APCs, the activity of several scFvs was increased by scoring the scFv tetramers, with the fluorescent probes directly marked. To form the scFv tetramers, each scFv fragment was designed to include a BirA sequence tag for site-specific biotinylation at the C-terminal site of the CL or CH1 domain, and after biotinylation, each of the streptavidin molecule was capable of to link four fragments of scFv. Another advantage of using fluorescent tetramers is that only one staining step alone is required, whereas unlabeled monomeric scFVs require a fluorescently labeled secondary antibody. Using these highly sensitive scFv tetramers, it was possible to specifically detect the occurrence of desired peptide / MHC complexes on the surface of breast carcinoma cells. A tetramerized scFv directed towards a mucin epitope and a complex with the HLA-A2 molecule was able to detect the MUC1-D6 complexes on the surface of HLA-A2 +, MCF7 and MDA-MB 231 cells, which express MUC1 but not on HLA-A2 + melanoma FM3D cells, MUC1 negative on MUC1 negative HLA-A2 and G-43 cells either. The M3A1 tetramer exhibited a low background staining with the SK-BR3 negative cells of HLA-A2, positive for MUC1. Such molecules also provide new means for quantitating specific peptide / MHC complexes as they occur on the surface of APCs and the tumor cell. This procedure represents the first attempt to quantify, using a direct means, the number of TCR ligands associated with tumor (peptide-MHC complexes) on the surfaces of tumor cells. The fluorescence intensity of the stained cells was measured and compared with that of the calibration beads with the known numbers of fluorophores (Cohen, CJ., Et al., 2002, Cancer Res. 62: 5835-5844). This method provides an easy and sensitive means to quantify cells stained with fluorophore with a flow cytometer. The TCR-like Fab of antimucine was used to quantify initially the number of MUC1-D6 / HLA-A2 complex that are exhibited on peptide-pulsed cells as well as the naturally unpulsed tumor cell: the pulse of the JY APCs with the peptide resulted in the ability to detect as much as 1.2 x 10 5 D6HLA-A2 complex per cell. The latter result is in complete agreement with the recent quantification of the murine H-2kb bound to the ovalbumin peptide SIINFEKL after infection of the vaccinia virus of the cells in vitro using an antibody similar to murine-specific TCR [Porgador et al. , 1997]. The number of complexes derived from D6-MUC1 on the surface of tumor cells expressing MUC1 was estimated to reach only several hundred per cell (Table 1, hereinafter). Table 1 Quantification of the number of D6-HLA-A2 complexes on the surfaces of tumor cells Cells Average number of sites / cell JY pulsed (1 mM peptide) 120,959 ± 16,934 pulsed MCF7 (1 M peptide) 5,539 ± 1,828 MDA MB 231 (1 M peptide) 10,022 ± 2,004 MCF7 284 ± 170 MDA MB 231 173 ± 112 Background 26 ± 11 Table 1: the fluorescence intensity of the stained cells in each experiment was compared to the fluorescence intensities of the calibration beads with the known numbers of the phycoertrine (PE) molecules on account (PE beads QuantiBRITE, Becton-Dickinson) and the number of sites per experiment was determined. The number of non-specific sites was determined by the intensity of staining of cells that do not express the HLA-A2 peptide. The number of specific sites for each experiment was then calculated for each experiment. The average number of sites was calculated from 10 independent experiments / determinations. The deviation in the number of sites depends on the sensitivity of the detection and the physiological state of the cells in each individual determination. The background number of the sites was determined as described, using SK-BR3 cells (HLA-A2- / MUC1 +), FM3D (HLA-A2 + / MUC1-), and G-43 (HLA-A2- / MUC1) as controls. The JY cells are B cells. The pulse with the peptide serves as a means to quantify the potential total HLA-A2 molecules on the surface of the cells. In a viral model, it was possible to detect HLA-A2 / Tax complexes on the HTLV-1 infected cells. HTLV-1 is involved in many pathologies including lympho / adult T cell leukemia (ATLL) and neurological disorders (HAM / TSP). A significant spotting with the anti-Tax / HLA-A2 Fab was observed on the RSCD4 (HLA-A2 +) cells infected with HTLV-1 but not on the HUT 102 (HLA-A2), which indicates that the TCR-like Fab is capable of detecting the specific HLA-A2 / Tax complex on the surface of cells infected with viruses. The staining pattern revealed two sub-populations with a high and moderate low reactivity with the TCR-like Fab, which may indicate the variability of the specific Tax epitope expression within the sub-populations of the cells infected with RSCD4. 1-. Similar variability was observed in spotting experiments with an anti-Tax protein antibody. This analysis revealed that the RSCD4 cells infected with the virus, which could also be divided into two sub-populations with high and moderate reactivity, exhibit on their surface 3 x 104 HLA-A2 / Tax complexes and several hundred sites respectively. The results clearly demonstrate the power of such TCR-like antibodies to provide useful and easily obtained direct quantitative data on the expression of specific peptide-MHC complexes on each cell in a population. In addition, these results demonstrate the high level expression of the specific MHC class I molecules that exhibit determinants derived from the proteins produced in the transfection with an intact whole gene or directly on the virus infected cells. These results are surprising in view of the fact that the number of MHC-peptide complexes on the surface of the tumor cell is low and were measured to be in the range of a few hundred complexes per cell (Table 1). EXAMPLE 2 SIMILAR ANTIBODIES TO TCR ARE CAPABLE OF RECOGNIZING SPECIFIC MHC-PEPTIDE COMPLEXES ON MELANOMA CELLS To test the biological activity of a TCR-like antibody on tumor cells, the Gl antibody, which recognizes HLA-A2 in a complex with the peptide derived from pglOO 209, was used in vitro , as follows. The procedures used in the experiments reported in Examples 2-5 (for example, apoptosis, cell death, FACS, flow cytometry and antibody binding) are included in Denkberg, G., et al., 2002, Proc. Nati Acad. Sci. USA. 99: 9421-9426; Lev, A., et al., 2002, Cancer Res, 62: 3184-3194; Denkberg, G., et al., 2003, J. Immunol. 171: 2197-2207; all of which are fully incorporated herein by reference in their entirety. The melanoma tumor cells were used to determine the reactivity of the recombinant Fab or scFv antibodies with the HLA-A2 / peptide complexes expressed on the surface of the cell. Approximately 106 cells were washed two, times with serum-free RPMI and incubated for 60-90 minutes at 40 ° C with recombinant TCR-like antibodies (10-100 μg / ml) in 100 μl. After three washes the cells were incubated with the FITC-labeled anti-human Fab (Jakson Immunoresearch, West Grove, PA, USA). After a final wash, the cells were resuspended in cold PBS as ice. Adherent tumor cells were harvested by trypsinization and resuspended in cold RPMI. All subsequent washes and incubations were performed in cold PBS such as ice. Cell analyzes were performed using a FACStar flow cytometer (Becton Dickinson and Co, San José, CA, USA) and the results were analyzed with the EinMDI program (Trotter J., http // facs. Scripps. Edu /) . Stable HLA-A2 complexes are formed on peptide loaded cells - To demonstrate the ability of Gl antibody to specifically bind an HLA-G9-209 complex, cells expressing HLA-G9-209 or HLA-G9 complexes -280, uncharged cells were incubated with the antibody W6 / 32 (an antibody directed against HLA), BB7.2 (an antibody directed against HLA-A2) or Gl. As shown in Figures 3a-c, the expression and stabilization of HLA molecules on the surface of the cell is dependent on an MHC-peptide complex. In addition, the Gl antibody was found to interact specifically with the cell presenting the MHC-G9-209 complex. To demonstrate that the Gl scFv can bind the MHC-specific peptide complex exhibited on the surface on the tumor cells, the melanoma FM3D cells HLA-A2 + were used. The cells were pulsed with the peptide G9-209 derived from specific gplOO or the control peptide (Figure 3d). Similar to JY APCs, the pulse of the melanoma cells allows the display of the exogenously supplied peptide facilitated by peptide exchange. Using this strategy, a mixture of the exogenously and endogenously derived peptides presented on the HLA-A2 that are exhibited on the surface of the cell was obtained. As shown in Figure 33, the Gl scFv antibody is capable of spotting and detecting HLA-A2 / G9-209 complexes on FM3D cells pulsed with G9-209 M but not on FM3D cells pulsed with the G9- peptide. 280 derived from gplOO. The Gl scFv antibody was also able to stain the FM3D cells pulsed with the native unmodified G9-209 peptide (Figure 3f), her, the control negative HLA-A2 KB3-1 cells (Figure 3g) pulsed with the G9 peptides -209M or G9-209 were not recognized by the Gl scFv antibody (Figure 3h). Taken together, these results demonstrate that the Gl scFv antibody can recognize the MHC-specific peptide complex on the surface of peptide pulsed and non-pulsed tumor melanoma cells. EXAMPLE 3 IDENTIFICATION OF RECOMBINANT ANTIBODIES 9H AND CLA21 A library of large human synthetic single chain Fv antibody was classified for antibodies capable of binding the MHC-peptide complexes. In this library the complementary determination regions formed in vivo (CDRs) were combinatorially intermixed in the structures of the human variable region derived from I germline [Azriel-Rosenfeld R, Valensí M, Benhar IA human synthetic combinatorial library of arrayable single chain antibodies on shuffling in vivo formed CDRs into general fra ework regions. J Mol Biol. 2004 Jan 2; 335 (1): 177-92]. The classification was carried out essentially as described elsewhere (Denkberg, G., and collaborators, 2002, Proc. Nati, Acad. Sci. USA 99: 9421-9426) using the following antigens: the HLA-A2- molecule complexes. ß2m of single chain with the peptide G9-209 derived from the melanoma gplOO protein and the single chain HLA-A2-ß2m molecule formed in complex with the peptide 26-35 derived from the MARTI melanoma protein. Two clones of scFv antibodies were isolated as soluble antibodies: -i 1. The 9H scFv antibody (sequence encoding the scFv is set forth by SEQ ID NO: 3; sequence of the scFv polypeptide is set forth by SEQ ID N0: 1 and 9H CDRs are provided in SEQ ID Nos: 5-10) which is a human antibody specific to the HLA-A2-G9-209 complex with a binding affinity of 0.56 nM (not shown). 2. The CLA12 scFv antibody (sequence encoding the scFv is set forth by SEQ ID NO: 18, sequence of the scFv polypeptide is set forth by SEQ ID NO: 17 and CDRs CLA12 is given in SEQ ID Nos: 19-24) which is a human antibody specific to the peptide HLA-A2-MART26-35 derived from the MARTI melanoma protein. EXAMPLE 4 TCR SIMILAR ANTIBODIES ARE CAPABLE OF DETECTING MHC COMPLEXES FROM IN-SITU TUMOR DERIVATIVES Another major potential use for TCR-like antibodies is direct visualization of the cells that carry ligand within the unaltered tissues. * -They use immunohistological methods. As a first step to estimate this potential, the potential of Gl scFv to directly visualize cells that carry ligand (complexes of HLA-A2 / G9-209) was tested on melanoma cells. Melanoma cells were derived from patients with melanoma and were cordially provided with National Cancer Institute, NIH. In situ detection of peptide-MHC complexes HLA-A2 / G9-209 on melanoma cells - To further characterize the ability of the "Gl antibody to recognize the G9-209 epitope derived from gplOO on the surface of the cell , the non-pulsed melanoma cells were used in an immunohistochemical analysis (Figures 4a-g), the HLA-A2-positive and negative melanoma cells expressing the melanoma gplOO were subjected to immunohistochemical staining with the Gl scFv antibody. followed by a secondary staining step with the anti-Myc antibody labeled with HRP As shown in Figures 4a-g, these experiments showed a strong positive staining of the HLA-A2-positive melanoma lines, which express gplOO FM- 3-29 (Figure 4a), M-ww (Figure 4b), M-PAR (Figure 4c) M-141 (Figure 4d) and FM3-D (Figure 4f) but not G-43 melanoma cells of HLA-A2-negative expressing gplOO (Figure 4e) either of the breast carcinoma cells MDA-MB-2312 of gplOO negative, HLA-A2-positive (Figure 4g). Gl scFv did not stain the additional melanoma 6 non-tested cell lines (data not shown). These data show the ability of the TCR-like antibody Gl scFv to detect in situ-specific peptide-MHC complexes on cells. and potentially the tissue sections after the active intracellular processing that occurs naturally. This is the first demonstration of the in situ detection of a tumor-derived T cell epitope using recombinant scFv antibodies similar to TCR. EXAMPLE 5 TCR-LIKE ANTIBODIES CAN INDUCE THE DEATH OF THE CELL IN A SPECIFIC PEPTIDE-MHC COMPLEX-RESTRICTED To determine the ability of TCR-like antibodies to induce cell death and eliminate the cells that present ' antigens, APCs loaded with peptides were subjected to the treatment with the antibody Gl scFv and the staining of propidium iodide (Pl) or annexin V was used. Pl stains the adherent apoptotic cells and Annexin V detects the exposure of the membrane to phosphatidyl serine (PS), indicative of early apoptosis events in damaged cells. Results Gl scFv can induce cell death of melanoma cells loaded with HLA-A2 peptides. JY cells were loaded with epitopes derived from gplOO G9-209M and G9-280V as well as with other peptides restricted from HLA-A2 control. As mentioned previously, FACS analysis with the anti-HLA-A2 antibody revealed a similar expression pattern of the HLA-A2 molecules with G9-209M, G9280V and other cells loaded with control peptides (Figures 3a-h). As shown in Figures 5a-d, staining of Annexin V revealed that Gl scFv can induce death of cells loaded with the specific G9-209 peptide (Figure 5c) but not of cells loaded with a control peptide. (Figure 5d). The non-cytotoxic activity was observed on RMA-HHD cells that were loaded with the G9-280V epitope derived from gplOO or with other control-restricted HLA-A2 peptides or cells that were not loaded with the peptide (not shown). In JY cells transformed with EBV, which express normal TAP, the display of the exogenously supplied peptide is facilitated by the exchange of peptides (ie, the peptide from the outside is replacing that peptide that is entering from the inside. for the first time, that TCR-like antibodies can induce cell death in a restricted form of peptide-specific MHC.GlscFv can induce cell death in untreated melanoma cells - to further investigate the death of induced cell ("the effect of I to extermination") the influence of Gl scFv was "tested on established melanoma cell lines of human patients.
Melanoma lines that are positive gplOO and HLA-A2 (526, 624. and 501A), positive gplOO but HLA-A2 negative (G43)., - and negative gplOO and positive HLA_A2 (1938) were subjected to the Gl antibody treatment and the effect of the antibody on the synthesis of the protein was measured. As shown in Figure 6, the Gl scFv antibody induced cell death, in a dose-dependent manner, of all melanoma cells positive for tantota HLA-A2 protein as the tumor-specific protein gplOO. part, as shown further in Figure 6, protein synthesis of melanoma cell lines 1938 or G43 that do not express the gplOO antigen or the correct MHC allele, respectively, were not affected by the antibody. for the first time, that an antibody similar to TCR, such as Gl can induce efficient cell death of the target cells.These results can pave the way for a new family of antibody-based molecules that can be used as agents Therapeutics for eliminating in an antigen (peptide) and the restricted populations of MHC from diseased cells such as tumor cells or cells infected with viruses which express disease-specific MHC-peptide complexes. Since the T cell epitopes derived from melanoma differentiation antigen alone most characterized TAAS and many immunotherapy strategies were developed using these objectives such as adoptive transfer strategies and vaccination with peptides and dendritic cells (DCs), the present inventors have generated a panel of antibodies similar to the T cell receptor (TCRL) directed to melanoma differentiation antigens such as gplOO, MARTI and tyrosinase and other MHC-associated peptide complexes with tumor as well as viral targets. (Denkberg., Et al., 2002, Proc. Nati, Acad. Sci. USA, 99: 9421-9426, Denkberg, G., et al., 2033, J. Immunol., 171: 2197-2207; Lev, A., et al, 2002, Cancer Res. 62: 3184-3194; Cohen, CJ., et al., 2002, Cancer Res. 62: 5835-5844; Cohen, Cj. et al., 2003, J. Im unol 170: 4349- 4361). These TCRL antibodies were able to analyze the pulsed peptide APCs (Figure 1-3) as well as the lines of o "melanoma cells derived from patients with melanoma that are HLA-A2 positive (Figures 4a-d). Thus, TCRL antibodies were found capable of binding the MCH-authentic peptide complex as expressed on the surface of the cell and this reactivity was correlated and dependent on the expression of HLA-A2 and antigen Fused antibodies of TCRL-PE exhibit anti-tumor activity - TCRL antibodies were genetically fused to a truncated form of the Pseudomonas exotoxin (PE38) in which the cell-binding domain of the toxin is delayed and the translocation and ribosylation domains of ADP are fused to the TCLR antibody. The effect of such fused TCRL molecules on the synthesis of the protein was tested on cells loaded with various peptides. As shown in Figures la-c, a specific protein inhibition activity was achieved when the cells were loaded with the specific gplOO-derived peptide and thus exhibited the HLA-peptide complex (eg, G9-209). Figures 8a-d depict the internalization of TCRL in melanoma cells and the TCRL-specific 2F1 cytotoxicity effect (e.g., antibody 2F1) over melanoma cells HLA-a2 + / gpl00 + (e.g., 526, 501A, 624.38) ) but not on melanoma cell lines HLA ~ A2 + / gpl00 + (G-43) or HLA-A2 + / gpl00- 81938). In addition, as shown further in Figure 8d, such an indication of cell death was achieved in the presence of relatively low TCRL concentrations, with an efficient IC 50 of 15-30 ng / ml. These results suggest remarkably the potential use of TCRL antibodies to targeted drugs or toxins specifically in tumor cells. Bare TCRL antibodies exhibit potent inhibition of protein synthesis in a specific manner of HLA-A2 and antigen - There are strong indications in the literature that antibodies to class I and MHC class II have biological effects on the target cells in particular its ability to induce the signal transduction events that will lead to the apodosis of the target cell (Nagy ZA, and collaborators, 2002; Nagy ZA and Money NA., 2003; Longo, D.L., 2002; Vidovic D and Toral JI. 1998; Pedersen AE. and collaborators, 1999; Ruhwald M, et al., 1999; Mori M, et al., 1999; Genestier L. et al., 1997; Genestier L, 1 et al., 1997; Skov S. et al., 1997). These studies were performed with the anti-class I and class II antibodies that are specific for MHC but are not specific for alleles and peptides such as the TCRL antibodies of the present invention. To test the ability of the TCRL antibodies of the present invention to exert biological activities towards target cells such as naked antibodies, melanoma cell lines derived from patients were incubated with the TCRL antibodies of gplOO and MARTI specific. Bare TCRL antibodies induced apoptosis in melanoma cells in a specific manner of HLA-peptide - Melanoma cells were subjected to antibody fragments similar to TCRL bare 2F1, 9H and Gl directed to apoptosis of gplOO or MARTI (CLA12) and the cell was measured using a flow cytometric staining of Propidium iodide (Pl). Figures 9a-j represent the results of these experiments. While in the cells that were HLA-A2 + / antigen + the fraction of the positive Pl cells was in the range of 44-58% (Figures 9e-g), in the cells HLA-A2 (+) gplOO (-) MART ( -) or HLA-A2 (-) pglOO (+) MART (+), the fraction of the positive Pl cells was in the range of 7-14% (Figures 9a-d). To test the specificity of naked TCRL antibodies to the HLA-peptide complex, 624 melanoma cells that are gpl00 + / HLa-A2 + were treated with TCRL Gl and control 1A7 antibodies (specific to HLA-A2 in a complex with the peptide G9-209M mutant but not in a complex with the natural G9-209 peptide) and the cells were subjected to the flow cytometric analysis of Pl Annexin V. As shown in Figures 9i-k, while in the cells treated with the Gl TCRL the fractions of the Pl and Annexin V positive cells were "" ~ 20% and 53%, respectively, in the cells treated with the control 1A7 TCRL antibody, the fractions of Pl and Andesina V-positive cells ~ 7a% and 12%, respectively. Cloning of TCRL antibodies - It is not known from the literature whether the signaling pathways involved through the binding of antibodies to class I require dimerization. The present inventors have hypothesized that the high concentrations of TCRL scFv specific for melanoma are due to the fact that in high concentrations there is the formation of spontaneous dimers. Thus, the TCRL scFv fragment was transformed into a complete IgG molecule by cloning the Fd and CL Fab domains into an IgG1 backbone and a complete TCRL IgG molecule was formed in the HEK293 cells and the TCRL IgG from the supernatants was purified. of culture. The various clones were isolated essentially as described in Thomas Jostocka, et al., Rapad generation of functional human IgG antibodies deriv from Fab-on-phage display libraries, Journal of Immunological Methods, Volume 289, Still 1-2, June 2004, Pages 65-80). The cloned TCRL IgG exhibits increased specificity towards the HLA-peptide complex - the TCR-like antibody 9H, which targets the gplOO peptide HLA-A2 (G9-209, was transformed into a complete IgG molecule and its reactivity was tested on JY cells pulsed with peptide As shown in Figures lOa-d, the reactivity and specificity of the TCRL IgG molecules to the APCs pulsed with the melanoma peptide derived from specific gplOO was maintained and even improved due to the increase in their Similarly, an IgG clone was prepared from 2F1 scFv, which targets the complex of HLA-A2-G9-280 Table 2, hereinafter, presents a comparison between the activity of TCRL IgG molecules Naked and scFv molecules on the melanoma cell lines As shown in Table 2, the activity of cell death induction was maintained in the clonal TCRLs. On the other hand, the concentration of the TCRL IgG molecule required to achieve a significant level of 50% of the cell death was significantly reduced compared to the scFv antibody. Table 2 Induction of cell death in tumor cells by TCR-like antibodies: The influence of avidity of the monovalent antibody against the bivalent Table 2: Concentration of the antibody 2F1 scFv (monovalent) and 2F1 complete IgG (bivalent) required to achieve cell death specific to melanoma cells with a TCR-like antibody that targets the melanoma epitope gpl00-G9-280 . These results demonstrate the potential use of TCRL antibodies to serve as objective portions for melanoma cells as well as considering their potential enlargements as molecules that can induce peptide-specific apodosis, restricted from specific MHC in target cells and can be considered as a new entity that can be developed pre-clinically as a therapeutic immunotherapeutic modality for melanoma and other types of tumor. It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is proposed to cover all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents and patent applications mentioned in this specification are hereby incorporated in their entirety by reference in the specification, to the same degree as if each individual publication, patent or patent application were specifically and individually indicated to be incorporated in the present by reference. In addition, the citation or identification of any reference in this application will not be considered as an admission that such a reference is available as a prior art for the present invention. REFERENCES (Additional references are cited in the text) 1. Aharoni R., Teitelbaum D., Arnon R., Puri J. 1991. Immunomodulation of experimental allergic encephalomyelitis by antibodies to the antigen-the complex. Nature, 351: 147-150. 2. Alt an J.D., Moss P.A.H., Goulder P.J.R., Barouch D.H., McHeyzer Willianis M.G., Beli 'J.I., McMichael A.J., Davis M.M. 1996. Phenotypic analysis of antigen-specific T lymphocytes [erratum appears in Science 1998 Jun 19; 280 (5371): 1821]. Science, 274: 94-96. 3. Andersen P.S., Stiyhn A., Hansen B.E., Fugger L., Engberg J., Buus S. 1996. A recombinant antibody with the antigen-specific, major histocompatibility complex-restricted speciflcity of T cells. Proc. Nati Acad. Sci. U. S. A, 93: 1820-1824. 4. Boon T.van der B.P. 1996. Human tumor antigens recognized by T lymphocytes. J. Exp. Mcd. , 183: 725-729. 5. Cloutier SM, Couty S., Terskikh A., Marguerat L., Crivelli Y., Pugnieres M., Mani JC, Leisinger HJ, Mach JP, Deperthes D. 2000. Streptabody, a high avidity molecule made by tetramerization of in live bictinylated, phage display-selected scFv fragments on streptavidin Mol. Immunol., 37: 1067-1077. 6. Cohen CJ, Hoffmann N., Farago M., Hoogenboom HR, Eisenbach L., Reiter Y. 2002. Direct detection and quantitation of a distinct T-cell epitope derived from turaor-specific epithelial cell mucin using human recombinant antibodies endowed with The antigen-specific, major histoco patibility complex-restricted specificity of T cells. Cancer Res., 62: 5835-5844. 7. Cohen CJ, Sarig O., Yamano Y., Tomaru U., Jacobson S., Reiter Y. 2003. Direct Phenotypic Analysis of human MHC Class 1 Antigen Presentation: Visualization, Quantitation In Situ Detection of Human Viral Epitopes Using Peptide- Specific, MBC-restricted Human Recombinant Antibodies. J. Immunol. 170: 4349-4361, 2003. 8. Dadaglio G., Nelson C.A., Deck M.B., Petzold S.J., Unanue E.R. 1997. Characterization and quantitation of peptide-MHC complexes produced from hen egg lysozyme using a monoclonal antibody. Immunity , 6: 727-738. 9. Day P.M., Yewdell J.W., Pergador A., Ger ain R.N., Bennink J.R. 1997. Direct delivery of exogenous MHC class 1 molecule-binding oligopeptides to the endoplasmic reticulum of viable cells. Proc. Nati Acad. Sci. U. S. A, 94: 8064-8069.
. Denkberg G., Cohen C.J., Sega! D., Kirkin A.F., Reiter Y. 2000. Recombinant human single-chain MHC-peptide complexes made from E. coli by in vitro refolding: funetional single-chain MHC-peptide complexes and tetramers with tumor associated antigens. Eur. J Immunol., 30: 3522-3532. 11. Denkberg G., Cohen C.J., Reiter Y. 2001. Critical role for CD8 in bindi? G of MHC tetramers to TCR: CD8 antibody block speciflc binding of human tumor-specific MHC-peptide tetramers to TCR. J Im unol. , 167: 270-276. 12. Denkberg G., Cohen C.J., Lev A., Chames P., Hoogenboom H.R., Reiter Y. 2002a. Direct visualization of distinct T cell epitopes derived from a tumor-associated melanoma antigen by using human recombinant antibodies with MHC- resfricted T cell receptor-like specificity. Proc. Nati Acad. Sci. U. S. A, 99: 9421-9426. 13. Denkberg G., Klechevsky E., Reiter Y. 2002b. Modification of a tumor-derived peptide at an HLA-A2 anchor residue can alter the conformation of the MHC-peptide complex: probing with TCR-like recombinant antibodies. J. Immunol., 169: 4399-4407. 14. Krogsgaard M., Wucherpfennig KW, Canella B., Hansen BE, Svejgaard A., Pyrdol J., Ditzel H., Raine C, Engberg J., Fugger L. 2000. Visualization of yelin basic protein (MBP) T cell epitopes in multiple sclerosis lesions using a monoclonal antibody specific for the human histocompatibility leukocyte antigen (HLA) -DR2-MBP 85-99 complex. J Exp. Med., 191: 1395-1412. 15. Lev A., Denkberg G., Cohen CJ, Tzukerman M., Skorecki KL, Chames P., Hoogenboom HR, Reiter Y. 2002. Isolation and characterization of human recombinant antibodies endowed with the antigen-specific, major histocompatibility complex- restricted specificity of T cells directed to the wide 1 and expressed tumor T-cell epitopes of the telomerase catalytic subunit. Cancer Res., 62: 3184-3194. 16. Polakova K., Plaksin D., Chung D.H., Belyakov I.M., Berzofsky J.A., Margulies D.H. 2000. Antibodies directed against the MHC-I molecule H-2D (d) complexed with an antigenie peptide: similarities to a T cell receptor with the same specificity [In Process Citation]. J Immunol., 165: 5703-5712. 17. Porgador A., Yewdell J.W., Deng Y., Bennink J.R., Germain R.N. 1997. Localization, quantitation, and in situ detection of speciflc peptide- MHC class 1 complexes using a monoclonal antibody. Jinmunity , 6: 7 -15-726. 18. Reiter Y., Di Cario A., Fugger L., Engberg J., Pastan 1. 1997. Peptide specific killing of antigen-presenting cells by a recombinant antibody-toxin fusion protein targeted to major histocompatibility complex / peptide class 1 complexes with T cell receptor-like specificity. Proc. Nati Acad. Sci. U. S. A, 94: 463 1-4636. 19. Rognan D., Stryhn A., Fugger L., Lyngbaek S., Engberg J., Andersen P.S., Buus S. 2000. Modeling the interactions of a peptide-major histocompatibility class 1 ligand with its receptors. 1. Recognition by two alpha beta T cell receptors. J. Co put. Aided Mol. Des., 14: 53-69. 20. Stryhn A., Andersen PS, Pedersen LO, Svejgaard A., Blm A., Thorpe CJ, Fugger L., Buus S., Engberg J. 1996. Shared faith specificity between T cell receptors and an antibody recognizing a peptide. major histocompatibility class 1 complex. Proc. Nati Acad. Sci. U. S. A, 93: 10338-10342. 21. Wulfing C, Pluckthun A. 1994. Correctly folded T-cell receptor fragments in the periplasm of Escherichia coli. Influence of folding catalysts. J. Mo! . Biol., 242: 65 5-669. 22. Zhong G., Reis e Sousa, Germain R.N. 1997a. Antigen-unspecific B cells and lymphoid dendritic cells both show extensive surface expression of processed antigen-major histocompatibiity complex class II complexes after soluble protein exposure iii vivo or in vitro. J. Exp. Med., 186: 673-682. 23. Zhong G., Reis e Sousa, Germain R.N. 1997b. Production, specificity, and functionality of monoclonal antibodies to specific peptide-major histocompatibiity complex class II complexes formed by processing of exogenous protein. Proc. Nati Acad. Sci. U. S. A, 94: 13856-13861. 24. Yamano Y, Cohen CJ, Takenouchi N, Yao K, Tomaru U, Li HC, Reiter Y, Jacobson S. Increased Expression of Human T Ly phocyte Virus Type 1 (HTLV-I) Taxll-19 Peptide-Human Histocompatibility Leukocyte Antigen A * 201 Complexes on CD4 + CD25 + T Cells Detected by Peptide-specific, Major Histocompatibility Complex-restricted Antibodies in Patients with HTLV-I associated Neurologic Disease. J Exp Med. 199: 1367-1377, 2004. 25. Zehn, D., Cohen, CJ., Reiter, and Waiden, P.
Extended presentation of specific MHC-peptide complexes by mature dendritic celia compared to other types of antigen-presenting cells. Eur. J. Inimunol.34: 155 1-1560, 2004. 26. Nagy ZA, Hubner B, Lobning C, Rauchenberger R, Reiffert S, Thomassen-Wolf E, Zahn S, Leyer S, Schier EM, Zahradnik A, Brunner C, Lobenwein K, Rattel B, Stanglmaier M, Hailek M, Wing M, Anderson S, Dunn M, Kretzschmar T, Tesar M. 2002. Fully human, HLA ~ DR-specific monoclonal hypoxia induces progranimed death of malignant lymphoid cells. Nat Mcd. 8: 801-7. 27. Nagy ZA, Mooney NA. 2003. A novel, alternative pathway of apoptosis triggered through class II major histocompatibiity complex molecules. J Mol. Med. 81: 757-65 28. Longo, D.L., 2002. DR 's orders: human antibody kilis tumors by direct signaling. Nature Mcd. 8: 781-3. 29. Vidovic D, Toral JI. 1998. Selective apoptosis of neoplastic cells by the HLA-DR-specific monoclonal antibody. Cancer Lett. 128: 127-35. 30. Pedersen AB, Bregenholt 'S, Johansen B, Skov S, Claesson MH. 1999.MHC-I-induced apoptosis in human B-lymphoma cells is dependent on protein tyrosine and serine / threonine kinases. E p Celi Res. 25 1: 128-34. 31. Ruhwald M, Pedersen AB, Claesson MH. 1999. MHC class 1 cross taik with CD2 and CD28 induces specific intracellular signaling and leads to growth retardation and apoptosis via p56 (Ick) -dependent mechanism. Exp Clin Immunogenet.; 16: 199-211 32. I died M, Terui Y, Ikeda M, Tomizuka H, Uwai M, Kasahara T, Kubota N, Itoh T, Mishima Y, Douzono-Tanaka M, Ya ada M, Shimamura S, Kikuchí J, Furukawa Y, Jshizaka Y, Ikeda K, Hand H, Ozawa K, Hatake K. 1999. Beta (2) -microglobulin identi fi ed as an apoptosis-inducing factor and its characterization. Blood. 94: 2744-53. 33. Genestier L, Pailiot R, Bonnefoy-Berard N, Meffre G, Flacher M, Fevre D, Liu YJ, Le Bouteiller P, Waldmann H, Engelhard VH, Banchereau J, Revillard IP. 1997. Fas-independent apoptosis of activated T cells induced by antibodies to the HLA class 1 alphal domain. Blood. 90: 3629-39. 34. Genestier L, Meffre G, Garrone P, Pm JJ, Liu YJ, Banchereau 3, Revillard IP. 1997. Antibodies to HLA class 1 alphal domain trigger apoptosis of CD40-activated human B lymphocytes. Blood. 90: 726-35. 35. Skov S, Klausen P, Claesson MH. 1997. Ligation of major histocompatability complex (MHC) class 1 molecules on human T cells induces cell death through PI-3 kinase-induced c-Jun NH2-terminal kinase activity: a novel apoptotic pathway distinct from Fas-induced apoptosis. J Cell Biol. 139: 1523-31.

Claims (30)

  1. CLAIMS 1. A method for inducing apoptosis in cancer cells or cells infected by pathogens, the method characterized in that it comprises contacting or expressing in the cells a recombinant isolated antibody capable of specifically binding a complex of .MHC-peptide specifically expressed on the cells, in this way induce apoptosis in cancer cells or cells infected by pathogens.
  2. 2. A method for treating cancer or infection by pathogens, characterized in that it comprises administering to, or expressing in the cells of a subject in need thereof a therapeutically effective amount of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, the isolated recombinant antibody that is capable of inducing apoptosis of cancer cells or cells infected by pathogens, cancer cells or 'cells infected by pathogens that specifically express the MHC-peptide complex, in order to thereby treat cancer or infection by pathogens in the subject.
  3. 3. Use of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, characterized in that it is as a pharmaceutical substance, the isolated recombinant antibody that is capable of inducing apoptosis of cancer cells or cells infected by pathogens that express specifically the MHC-peptide complex.
  4. 4. Use of a recombinant isolated antibody capable of specifically binding an MHC-peptide complex, the isolated recombinant antibody that is capable of inducing apoptosis of cancer cells or cells infected by pathogens that specifically express the MHC-peptide complex, characterized because it is for the manufacture of a drug identified for the treatment of cancer or infection by pathogens.
  5. 5. A pharmaceutical composition for treating cancer or infection by pathogens, characterized in that it comprises as an active ingredient a recombinant isolated antibody capable of specifically binding an MHC-peptide complex in cancer cells or cells infected by pathogens that specifically express the MHC complex peptide, the recombinant isolated antibody that is capable of inducing apoptosis in cancer cells or cells infected by pathogens and a "pharmaceutically acceptable carrier"
  6. 6. The method according to claim 2, characterized in that it also comprises detecting apoptosis in cancer cells or cells infected by pathogens after administration or expression.
  7. 7. The method according to claim 6, characterized in that the detection of apoptosis is carried out by a method selected from the group consisting of FACS analysis of propidium iodide, FACS analysis of Annexin V, staining of Ethidium homodimer-1, viability / live / dead cytotoxicity and Tunnel test.
  8. 8. The method, use or pharmaceutical composition according to any of claims 1, 2, 3, 4 and 5, characterized in that the isolated recombinant antibody is selected from the group consisting of a Fab fragment and a ScFv.
  9. 9. The method, use or pharmaceutical composition according to claim 8, characterized in that the recombinant antibody is at least bivalent.
  10. 10. The method, use or pharmaceutical composition according to claim 9, characterized in that the ScFv is 9H (SEQ ID N0: 1), Gl (SEQ ID NO: 2) or CLA12 (SEQ ID NO: 17).
  11. 11. The method, use or pharmaceutical composition according to any of claims 1, 2, 3, 4 and 5, characterized in that the peptide is derived from a polypeptide selected from the group consisting of MUC1, gplOO, HTERT, TAX, MARTI .
  12. The method, use or pharmaceutical composition according to claim 8, characterized in that the recombinant antibody comprises at least one CDR selected from the group consisting of SEQ ID Nos: 5-16 and 19-24.
  13. 13. The method, use or pharmaceutical composition according to any of claims 1, 2, 3, 4 and 5, characterized in that the recombinant antibody is a subtype of IgG.
  14. 14. The method, use or pharmaceutical composition according to any of claims 1, 2, 3, 4 and 5, characterized in that the cancer is melanoma or breast cancer.
  15. 15. A method for diagnosing cancer or infection by pathogens in a subject, characterized in that it comprises: (a) contacting a biological sample of the subject with a recombinant isolated antibody capable of specifically binding an MHC-peptide complex under conditions suitable for immune complex formation; the isolated recombinant antibody that is capable of inducing apoptosis in objective cancer cells or cells infected by pathogens, and (b) detecting the formation of the immune complex, so as to diagnose cancer or infection by pathogens in the subject.
  16. 16. A method for diagnosing cancer or infection by pathogens in a subject, characterized in that it comprises: (a) contacting a biological sample of the subject with a recombinant isolated antibody capable of specifically binding an MHC-peptide complex under conditions suitable for the formation of the immunocomplex, the isolated recombinant antibody that is capable of inducing apoptosis in target cancer cells _ or cells infected by pathogens, and (b) detecting a level of apoptosis in the cells of the biological sample in order to diagnose cancer or infection by pathogens in the subject.
  17. 17. A device for diagnosing cancer or infection by pathogens in a subject, the device characterized in that it comprises packaging materials and at least one agent identified by the packaging materials as being suitable for an immunocomplex formation between a recombinant isolated antibody capable of specifically binding an MHC-peptide complex and the MHC-peptide complex, the isolated recombinant antibody that is capable of inducing apoptosis in cancer cells or cells infected by pathogens.
  18. The equipment according to claim 17, characterized in that it also comprises at least one agent identified by the packaging materials as being suitable for detecting a level of apoptosis in cancer cells or cells infected by pathogens.
  19. 19. The method or equipment according to any of claims 15, 16 and 17, characterized in that the isolated recombinant antibody is a fragment of Fab or scFv.
  20. 20. The method or equipment according to claim 19, characterized in that the ScFv is 9H (SEQ ID NO: l), Gl (SEQ ID NO: 2) or CLA12 (SEQ ID NO: 17).
  21. The method or equipment according to any of claims 15, 16 and 17, characterized in that the antibody is a subtype of IgG.
  22. 22. The method or equipment according to any of claims 14, 15 and 16, characterized in that the cancer is melanoma or breast cancer.
  23. 23. A recombinant antibody, characterized in that it is capable of inducing apoptosis in cancer cells or cells infected by pathogens.
  24. 24. A pharmaceutical composition, characterized in that it comprises a recombinant antibody capable of inducing apoptosis in cancer cells or cells infected by pathogens and an acceptable pharmaceutical carrier.
  25. 25. The recombinant antibody or pharmaceutical composition according to any of claims 23 and 24, characterized in that the recombinant antibody is selected from the group consisting of a Fab fragment and a ScFv fragment.
  26. 26. The recombinant antibody or pharmaceutical composition according to claim 25, characterized in that the ScFv is 9H as set forth by SEQ ID NO: 1, Gl as set forth by SEQ ID NO: 2 or CLA12 as set forth by the SEQ ID NO: 17.
  27. 27. The recombinant antibody or pharmaceutical composition according to any of claims 23 and 24, characterized in that the recombinant antibody comprises at least one CDR selected from the group consisting of SEQ ID NOs: 5-16 and 19-24.
  28. 28. A 9H recombinant scFv, characterized in that it is as set forth in SEQ ID NO: l.
  29. 29. A recombinant scFv CLA12, characterized in that it is as set forth in SEQ ID NO: 17.
  30. 30. A recombinant antibody, characterized in that it comprises at least one CDR selected from the group consisting of SEQ ID NOs: 5-16 and 19-24.
MXPA/A/2006/014344A 2004-06-09 2006-12-07 Antibodies for selective apoptosis of cells MXPA06014344A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/577,920 2004-06-09

Publications (1)

Publication Number Publication Date
MXPA06014344A true MXPA06014344A (en) 2008-09-26

Family

ID=

Similar Documents

Publication Publication Date Title
CA2570458C (en) Antibodies for selective apoptosis of cells
US7632923B2 (en) Compositions capable of specifically binding particular human antigen presenting molecule/pathogen-derived antigen complexes and uses thereof
Cohen et al. Direct phenotypic analysis of human MHC class I antigen presentation: visualization, quantitation, and in situ detection of human viral epitopes using peptide-specific, MHC-restricted human recombinant antibodies
Denkberg et al. Selective targeting of melanoma and APCs using a recombinant antibody with TCR-like specificity directed toward a melanoma differentiation antigen
Cohen et al. Recombinant antibodies with MHC‐restricted, peptide‐specific, T‐cell receptor‐like specificity: new tools to study antigen presentation and TCR–peptide–MHC interactions
US20180071398A1 (en) Polypeptide that binds aberrant cells and induces apoptosis
US6992176B2 (en) Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
US20100158927A1 (en) Antibodies, methods and kits for diagnosing and treating melanoma
US8097704B2 (en) Antibody specifically binding to DR5 and composition for preventing or treating cancers comprising the same
CA2876133C (en) Human bispecific egfrviii antibody engaging molecules
JP6786722B2 (en) New Antibodies Against Programmed Cell Death Protein (PD-1) and Their Uses
JP2013535963A (en) An isolated high affinity entity with T cell receptor-like specificity for a natural complex of MHC class II and a diabetes-related autoantigenic peptide
JP2022523710A (en) CD44-specific antibody
JP2022514693A (en) MUC18-specific antibody
JP2023153779A (en) Anti-hK2 chimeric antigen receptor (CAR)
JP2022514786A (en) MUC18-specific antibody
MXPA06014344A (en) Antibodies for selective apoptosis of cells
US20240034790A1 (en) Antibody specific for cd47 and uses thereof
US20170198046A1 (en) Compositions capable of specifically binding particular human antigen presenting molecule/ antigen complexes and uses thereof
JP2024517985A (en) Anti-CD300c monoclonal antibody and its biomarker for preventing or treating cancer
WO2023044466A2 (en) Herv-k antibody, cell, vaccine, and drug therapeutics