MXPA04010091A - Compositions and methods for the diagnosis and treatment of tumor. - Google Patents

Compositions and methods for the diagnosis and treatment of tumor.

Info

Publication number
MXPA04010091A
MXPA04010091A MXPA04010091A MXPA04010091A MXPA04010091A MX PA04010091 A MXPA04010091 A MX PA04010091A MX PA04010091 A MXPA04010091 A MX PA04010091A MX PA04010091 A MXPA04010091 A MX PA04010091A MX PA04010091 A MXPA04010091 A MX PA04010091A
Authority
MX
Mexico
Prior art keywords
antibody
tat
polypeptide
amino acid
seq
Prior art date
Application number
MXPA04010091A
Other languages
Spanish (es)
Inventor
I Wood William
Original Assignee
Genentech Inc
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 Genentech Inc filed Critical Genentech Inc
Publication of MXPA04010091A publication Critical patent/MXPA04010091A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention is directed to compositions of matter useful for the diagnosis and treatment of tumor in mammals and to methods of using those compositions of matter for the same.

Description

COMPOSITIONS AND METHODS FOR THE DIAGNOSIS AND TREATMENT OF TUMORS FIELD OF THE INVENTION The present invention is directed to compositions of the material useful for the diagnosis and treatment of tumors in mammals and methods for using those compositions of matter for them. BACKGROUND OF THE INVENTION Malignant tumors (cancers) are the second leading cause of death in the United States, after heart disease (Boring et al., CA Cancel J. Clin. 43: 7 (1993)). Cancer is characterized by an increase in the number of abnormal or neoplastic cells derived from a normal tissue that proliferates to form a tumor mass, the invasion of adjacent tissues by these tumor neoplastic cells and the generation of malignant cells that eventually spread through from the blood or lymphatic system to the regional lymph nodes and to distant sites through a process called metastasis. In a cancerous state, a cell proliferates under conditions in which normal cells would not grow. Cancer manifests itself in a wide variety of forms, characterized by different degrees of invasiveness and aggressiveness. In attempts to discover cellular-effective targets for cancer therapy, researchers have sought to identify polypeptides that are specifically expressed on the surface of one or more particular types of cancer cells as compared to one or more non-cancerous normal cells. The identification of such tumor-associated cell surface antigen polypeptides has given rise to the ability to specifically target cancer cells for destruction through antibody-based therapies. In this regard, it is noted that antibody-based therapy has proven to be very effective in the treatment of certain cancers. For example, HERCEPTIN® and RITUXAN® (both from Genentech Inc., South San Francisco, California) are antibodies that have been used successfully to treat breast cancer and non-Hodgkin's lymphoma, respectively. More specifically, HERCEPTIN® is a humanized monoclonal antibody derived from recombinant DNA that selectively binds to the extracellular domain of the proto-oncogene of the human epidermal growth factor receptor (HER2). Overexpression of the HER2 protein is observed in 25-30% of primary breast cancers. RITUXAN® is a genetically engineered murine / human monoclonal chimeric antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes. Both antibodies are produced recombinantly in CHO cells.
In other attempts to discover effective cellular targets for cancer therapy, researchers have sought to identify polypeptides that are produced and secreted by a particular type of cancer cells at a level of expression that is higher than that produced and secreted by one or more normal non-cancerous cells. In addition, such polypeptides can be expressed by cells that produce and / or secrete polypeptides that have a potentiating or growth enhancing effect on cancer cells. Such secreted factors are often proteins that provide cancer cells with a growing advantage over normal cells and include such things as, for example, angiogenic factors, cell adhesion factors, growth factors and the like. It would be expected that the identification of antagonists of such secreted polypeptides would serve as effective therapeutic agents for the treatment of such cancers. In addition, the identification of overexpression of such secreted factors would be useful for the diagnosis of particular cancers in mammals. Despite the previously identified advances in cancer therapy in mammals, there is a great need for additional diagnostic and therapeutic agents capable of detecting the presence of tumor in a mammal and to effectively inhibit the growth of neoplastic cells., respectively. Accordingly, it is an object of the present invention to identify cell surface polypeptides that are overexpressed in certain cancer cells as compared to normal cells or in other different cancer cells, or secreted polypeptides that are overexpressed by certain cancer cells compared to normal cells or in other different cancer cells and / or non-cancerous cells that surround them, and use those polypeptides and their encoding nucleic acids, to produce compositions of the useful material in the therapeutic treatment and in the diagnostic detection of cancer in mammals. SUMMARY OF THE INVENTION 1 Modalities In the present specification, the Applicants describe for the first time the identification of several cellular polypeptides (and their encoding nucleic acids or their fragments) that are expressed to a greater degree on the surface or by one or more types of cancer cells compared to the surface or by one or more types of normal non-cancerous cells. Alternatively, such polypeptides are expressed by cells that produce and / or secrete polypeptides that have a potentiating or growth enhancing effect on cancer cells. Such polypeptides are referred to herein as "Tumor-Associated Antigen Target Polypeptides" ("polypeptides") and are expected to serve as effective targets for the therapy and diagnosis of cancer in mammals. embodiment of the present invention, the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding an antigenic target polypeptide associated with the tumor or a fragment thereof (a WTAT polypeptide "). In certain aspects, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80% identity of the nucleic acid sequence, alternatively at least about 81%, 82%, 83%, 84%, 85% , 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity of the nucleic acid sequence , for (a) a DNA molecule encoding a full-length TAT polypeptide having an amino acid sequence as described herein, a TAT polypeptide amino acid sequence lacking the signal peptide as described herein, an extracellular domain of a transmembrane TAT polypeptide, with or without the signal peptide, as described herein or any other specifically defined fragment of an amino acid sequence of the full-length TAT polypeptide as described herein or (b) ) the complement of the DNA molecule of (a).
In other aspects, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80% identity of the nucleic acid sequence, alternatively at least about 81%, 82%, 83%, 84%, 85% , 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity of the nucleic acid sequence , for (a) a DNA molecule comprising the coding sequence of a full length TAT polypeptide cDNA as described herein, the coding sequence of a TAT polypeptide lacking the signal peptide as described in present, the coding sequence of an extracellular domain of a transmembrane TAT polypeptide, with or without the signal peptide, as described herein or the coding sequence of any other specifically defined fragment of the amino acid sequence of the TAT polypeptide of full length as described in the pres or (b) the complement of the DNA molecule of (a). In additional aspects, the invention relates to an isolated nucleic acid molecule comprising a nucleotide sequence having at least about 80% identity of the nucleic acid sequence, alternatively at least about 81%, 82%, 83% , 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity of the nucleic acid sequence for (a) a DNA molecule encoding the same mature polypeptide encoded by the full-length coding region of any of the human protein cDNAs deposited with the ATCC as described in present or (b) the complement of the DNA molecule of (a). In this regard, the term "full length coding sequence" refers to the nucleotide sequence encoding the TAT polypeptide of the cDNA sequence that is inserted into the vector deposited with the ATCC (which is often shown among the start and end codons, inclusive of them, in the accompanying figures). Another aspect of the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a TAT polypeptide that is either a deleted transmembrane domain or an inactivated transmembrane domain, or is complementary to such a coding nucleotide sequence, wherein the transmembrane domain (s) of such polypeptide (s) is (are) described herein. Therefore, the soluble extracellular domains of the TAT polypeptides described herein are contemplated. In other aspects, the present invention is directed to isolated nucleic acid molecules that hybridize to (a) a nucleotide sequence encoding a TAT polypeptide having a full length amino acid sequence as described herein, a sequence of amino acids of TAT polypeptide lacking the signal peptide as described herein, an extracellular domain of a transmembrane TAT polypeptide, with or without the signal peptide, as described herein or any other specifically defined fragment of a sequence of total length TAT polypeptide amino acids as described herein or (b) the complement of the nucleotide sequence of (a). In this regard, one embodiment of the present invention is directed to fragments of a full-length TAT polypeptide coding sequence or its complement, as described herein, which may find its use as, for example, useful hybridization probes. such as, for example, diagnostic probes, antisense oligonucleotide probes or for coding fragments of a full-length TAT polypeptide which optionally can encode a polypeptide comprising a binding site for an anti-TAT polypeptide antibody, an oligopeptide of TAT link or another small organic molecule that binds to a TAT polypeptide. Such nucleic acid fragments are commonly at least about 5 nucleotides in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 , 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75,? 50, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990? 1000 nucleotides in length, wherein in this context the term "approximately" means the reference length of the nucleotide sequence plus or minus 10% of that reference length. It is noted that new fragments of a coding nucleotide sequence of the TAT polypeptide can be determined routinely by aligning the coding nucleotide sequence of the TAT polypeptide with other known nucleotide sequences using any of a variety of well-known sequence alignment programs. and determining which (s) fragment (s) of nucleotide sequence encoding the TAT polypeptide are new. All such new fragments of the nucleotide sequences encoding the TAT polypeptide are contemplated herein. Also contemplated are the TAT polypeptide fragments encoded by these nucleotide molecule fragments, preferably those TAT polypeptide fragments that comprise a binding site for an anti-TAT antibody, a TAT binding oligopeptide or another small organic molecule that binds to a TAT polypeptide. In another embodiment, the invention provides isolated TAT polypeptides encoded by any of the isolated nucleic acid sequences identified hereinbefore. In a certain aspect, the invention relates to an isolated TAT polypeptide, comprising an amino acid sequence having at least about 80% identity of the amino acid sequence, alternatively at least about 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity of the amino acid sequence, for a TAT polypeptide having a full length amino acid sequence as described herein, a TAT polypeptide amino acid sequence that lacks the signal peptide as described herein, an extracellular domain of a Transmembrane TAT polypeptide protein, with or without the signal peptide, as described herein, an amino acid sequence encoded by any of the nucleic acid sequences described herein or any other specifically defined fragment of an amino acid sequence of the TAT polypeptide totality as described herein. In a further aspect, the invention relates to an isolated TAT polypeptide comprising an amino acid sequence having at least about 80% identity of the amino acid sequence, alternatively at least about 81%, 82%, 83%, 84 %, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity of the amino acid sequence, for an amino acid sequence encoded by any of the human protein cDNAs deposited with the ATCC as described herein. In a specific aspect, the invention provides an isolated TAT polypeptide without the N-terminal signal sequence and / or without the start methionine and is encoded by a nucleotide sequence encoding such amino acid sequence as described hereinabove. Also described herein are processes for producing the same wherein those processes comprise culturing a host cell comprising a vector comprising the appropriate coding nucleic acid molecule under conditions suitable for expression of the TAT polypeptide and recovery of the polypeptide. TAT from cell culture. Another aspect of the invention provides an isolated TAT polypeptide that is either deleted from the transmembrane domain or inactivated from the transmembrane domain. Also described herein are processes for producing the same wherein these processes comprise culturing a host cell comprising a vector comprising the appropriate coding nucleic acid molecule under conditions suitable for expression of the TAT polypeptide and recovery of the TAT polypeptide. from cell culture. In other embodiments of the present invention, the invention provides vectors comprising the DNA encoding any of the polypeptides described herein. Host cells comprising any such vector are also provided. By way of example, the host cells can be CHO cells, E. coli cells or yeast cells. A process for producing any of the polypeptides described herein is further provided and comprises culturing the host cells under conditions suitable for expression of the desired polypeptide and recovering the desired polypeptide from the cell culture. In other embodiments, the invention provides isolated chimeric polypeptides that comprise any of the TAT polypeptides described herein fused to a heterologous polypeptide (non-TAT). Examples of such chimeric molecules comprise any of the TAT polypeptides described herein fused to a heterologous polypeptide such as, for example, an epitope tag sequence or an Fe region of an immunoglobulin. In another embodiment, the invention provides an antibody that binds, preferably specifically, to any of the polypeptides described above or later. Optionally, the antibody is a monoclonal antibody, antibody fragment, chimeric antibody, humanized antibody, single chain antibody or an antibody that competitively inhibits the binding of an anti-TAT polypeptide antibody to its respective antigenic epitope. The antibodies of the present invention can optionally be conjugated to a growth inhibitory agent or to a cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like. The antibodies of the present invention can optionally be produced in CHO cells or in bacterial cells and preferably induce death of the cell to which they are linked. For diagnostic purposes, the antibodies of the present invention can be detectably labeled, attached to a solid support or the like. In other embodiments of the present invention, the invention provides vectors comprising the DNA encoding any of the antibodies described herein. The host cell comprising any such vector is also provided. By way of example, the host cells can be CHO cells, E. coli cells or yeast cells. A process for producing any of the antibodies described herein is further provided and comprises culturing host cells under conditions suitable for expression of the desired antibody and recovering the desired antibody from the cell culture. In another embodiment, the invention provides oligopeptides ("TAT-binding oligopeptides") that bind, preferably specifically, to any of the TAT polypeptides described above or later. Optionally, the TAT-binding oligopeptides of the present invention can be conjugated to a growth inhibitory agent or a cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like . The TAT-binding oligopeptides of the present invention can optionally be produced in CHO cells or in bacterial cells and preferably induce death of the cell to which they are linked. For diagnostic purposes, the TAT-binding oligopeptides of the present invention can be detectably labeled, attached to a solid support or the like. In other embodiments of the present invention, the invention provides vectors comprising the DNA encoding any of the TAT linkage oligopeptides described herein. The host cell comprising any such vector is also provided. By way of example, the host cells can be CHO cells, E. coli cells or yeast cells. A process for producing any of the TAT-binding oligopeptides described herein is further provided and comprises culturing the host cells under conditions suitable for the expression of the desired oligopeptide and recovering the desired oligopeptide from the cell culture. In another embodiment, the invention provides small organic molecules ("organic TAT binding molecules") that bind, preferably specifically, to any of the TAT polypeptides described above or later. Optionally, the TAT organic binding molecules of the present invention can be conjugated to a growth inhibitory agent or a cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or Similary. The organic TAT binding molecules of the present invention preferably induce the death of the cell to which they are linked. For diagnostic purposes, the TAT-binding organic molecules of the present invention can be detectably labeled, attached to a solid support or the like. Still in a further embodiment, the invention relates to a composition of matter comprising a TAT polypeptide as described herein, a TAT chimeric polypeptide as described herein, an anti-TAT antibody as described herein, a TAT linkage oligopeptide as described herein or an organic TAT linkage molecule as described herein, in combination with a carrier. Optionally, the vehicle is a pharmaceutically acceptable vehicle. In yet another embodiment, the invention relates to an article of manufacture comprising a container and a composition of matter contained within the container, wherein the composition of matter may comprise a TAT polypeptide as described herein, a TAT chimeric polypeptide. as described herein, a polipanti-TAT antibody as described herein, a TAT-binding oligopeptide as described herein or an organic TAT-binding molecule as described herein. The article optionally may further comprise a fixed label to the container or a packaging insert included with the container, which relates to the use of the composition of matter for the therapeutic treatment or diagnostic detection of a tumor.
Another embodiment of the present invention is directed to the use of a TAT polypeptide as described herein, a chimeric TAT polypeptide as described herein, an anti-TAT polypeptide antibody as described herein, a linkage oligopeptide. TAT as described herein or an organic TAT binding molecule as described herein, for the preparation of a medicament useful in the treatment of a condition responsive to the TAT polypeptide, the TAT chimeric polypeptide, the anti-polypeptide antibody -TAT, to the TAT binding oligopeptide or to the organic TAT binding molecule. 2 Additional Modalities Another embodiment of the present invention is directed to a method of killing a cancer cell that expresses a TAT polypeptide, wherein the method comprises contacting the cancer cell with an antibody, a oligopeptide or a small organic molecule that binds to a TAT polypeptide, which results in the death of the cancer cell. Optionally, the antibody is a monoclonal antibody, a fragment of a body, a chimeric antibody, a humanized antibody or a single chain antibody. The antibodies, TAT-binding oligopeptides and small organic molecules employed in the methods of the present invention can optionally be conjugated to a growth inhibitory agent or cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like. Antibodies and TAT-binding oligopeptides in the methods of the present invention can optionally be produced in CHO cells or in bacterial cells. Another embodiment of the present invention is directed to a method of inhibiting the growth of a cancer cell, wherein the growth of said cancer cell depends at least in part on the effect (s) of increased potency of the TAT polypeptide, in wherein the method comprises contacting the polypeptide with an antibody, an oligopeptide or a small organic molecule that binds to the TAT polypeptide, thereby antagonizing the growth enhancement activity of the TAT polypeptide and, in turn, inhibiting growth of the cancer cell. Preferably the growth of the cancer cell is completely inhibited. Optionally, the antibody is a monoclonal antibody, antibody fragment, chimeric antibody, humanized antibody or a single chain antibody. The TAT-binding oligopeptide antibodies and the organic TAT-binding molecules employed in the methods of the present invention can optionally be conjugated to a growth inhibitory agent or a cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like. The antibodies and TAT-binding oligopeptides used in the methods of the present invention can optionally be produced in CHO cells or bacterial cells. Yet another embodiment of the present invention is directed to a method for therapeutically treating a tumor expressing a TAT polypeptide in a mammal, wherein the method comprises administering to the mammal a therapeutically effective amount of an antibody, an oligopeptide or a small organic molecule. which binds to the TAT polypeptide, thereby resulting in the effective therapeutic treatment of the tumor. Optionally, the antibody is a monoclonal antibody, antibody fragment, chimeric antibody, humanized antibody or single chain antibody. The TAT-binding oligopeptide antibodies and the organic TAT-binding molecules employed in the methods of the present invention can optionally be conjugated to a growth inhibitory agent or a cytotoxic agent such as a toxin, including, for example, a maytansinoid or calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like. The antibodies and oligopeptides used in the methods of the present invention can optionally be produced in CHO cells or in bacterial cells.
Yet another embodiment of the present invention is directed to a method of therapeutically treating a tumor in a mammal, wherein the growth of said tumor depends at least in part on the enhancing effect (s) of the TAT polypeptide, wherein the method comprises administering to the mammal a therapeutically effective amount of an antibody, an oligopeptide or a small organic molecule that binds to the TAT polypeptide, thereby antagonizing the growth of the potency enhancing activity of said TAT polypeptide and resulting in the effective therapeutic treatment of the tumor. Optionally, the antibody is a monoclonal antibody, antibody fragment, chimeric antibody, humanized antibody or a single chain antibody. The antibodies, the TAT-binding oligopeptides and the TAT-binding organic molecules used in the methods of the present invention can optionally be conjugated to a growth inhibitory agent or a cytotoxic agent such as a toxin, including, for example, a maytansinoid or Calicheamicin, an antibiotic, a radioactive isotope, a nucleolytic enzyme or the like. The antibodies and TAT-binding oligopeptides used in the methods of the present invention can optionally be produced in CHO cells or bacterial cells. Yet another embodiment of the present invention is directed to a method of treating or preventing a cell proliferative disorder associated with the expression or altered, preferably increased activity of a TAT polypeptide, the method comprising administering to a subject in need of such treatment, an amount effective of an antagonist of a TAT polypeptide. Preferably, the cell proliferative disorder is cancer and the TAT polypeptide antagonist is an anti-TAT polypeptide antibody, a TAT-binding oligopeptide, an organic TAT-binding molecule or an anti-sense oligonucleotide. The effective treatment or prevention of the cell proliferative disorder may be the result of direct destruction or inhibition of the growth of cells expressing a TAT polypeptide or of antagonizing the cell proliferative activity of a TAT polypeptide. Yet another embodiment of the present invention is directed to a method for determining the presence of a TAT polypeptide in a sample suspected of containing the TAT polypeptide, wherein the method comprises exposing the sample to an oligopeptide antibody or small organic molecule that binds to the TAT polypeptide and determine the binding of the oligopeptide antibody or organic molecule to the TAT polypeptide in the sample, wherein the presence of such a linkage is indicative of the presence of the TAT polypeptide in the sample. Optionally, the sample may contain cells (which may be cancer cells) suspected of expressing the TAT polypeptide. The antibody, the TAT linkage oligopeptide or the TAT organic linker molecule employed in the method may optionally be detectably labeled, attached to a solid support or the like. A further embodiment of the present invention is directed to a method for diagnosing the presence of a tumor in a mammal, wherein the method comprises detecting the level of expression of a gene encoding a TAT polypeptide (a) in a test sample of tissue cells obtained from said mammal and (b) in a control sample of normal non-cancerous cells known from the same tissue origin, wherein a higher level of expression of the TAT polypeptide in the test sample, compared to the sample of control, is indicative of the presence of tumor in the mammal from which the test sample was obtained. Another embodiment of the present invention is directed to a method for diagnosing the presence of a tumor in a mammal, wherein the method comprises (a) contacting a test sample comprising woven cells obtained from the mammal with an oligopeptide antibody. or a small organic molecule that binds to a TAT polypeptide and (b) detect the formation of a complex between the antibody, the oligopeptide or the small organic molecule and the TAT polypeptide in the test sample, wherein the formation of a complex is indicative of the presence of a tumor in the mammal. Optionally, the antibody, the TAT linkage oligopeptide or the organic link molecule TAT employee is detectably labeled, attached to a solid support or the like and / or the test sample of the tissue cells is obtained from an individual suspected of having a cancerous tumor. In still further embodiments, the invention addresses the following set of potential claims for this application: 1. The isolated nucleic acid having at least 80% of the nucleic acid sequence identity for: (a) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or in Figure 2 (SEQ ID NO: 2); (f) the total length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (g) the total length coding sequence of the cDNA deposited under the access ATCC number shown in Table 7; or (h) the complement of (a), (b), (c), (d), (e), (f), or (g). 2. The isolated nucleic acid comprising: (a) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO:), which lacks its associated signal peptide; (c) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) the total length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (g) the total length coding sequence of the cDNA deposited under the access ATCC number shown in Table 7; or (h) the complement of (a), (b), (c), (d), (e), (f), or (g). 3. The isolated nucleic acid that hybridizes to: (a) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) a nucleotide sequence encoding the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) a nucleotide sequence encoding an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) the total length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (g) the total length coding sequence of the cDNA deposited under the access ATCC number shown in Table 7; or (h) the complement of (a), (b), (c), (d), (e), (f), or (g). 4. The nucleic acid of Claim 3, wherein the hybridization occurs under stringent conditions. 5. The nucleic acid of Claim 3, which is at least about 5 nucleotides in length. 6. An expression vector comprising the nucleic acid of Claim 1. 7. The expression vector of Claim 6, wherein said nucleic acid is operably linked to the control sequences recognized by a host cell transformed with the vector. 8. A host cell comprising the expression vector of Claim 7. 9. The host cell of Claim 8 which is a CHO cell, an E. coli cell or a yeast cell. A process for producing a polypeptide comprising culturing the host cell of Claim 8 under conditions suitable for the expression of said polypeptide and recovering said polypeptide from the cell culture. 11. An isolated polypeptide having at least 80% of the amino acid sequence identity for: (a) an amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) an amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 12. An isolated polypeptide comprising: (a) the amino acid sequence shown in the Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO:) (b) the amino acid sequence shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ ID N0: 4), which it lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 13. A chimeric polypeptide comprising the polypeptide of Claim 11 fused to a heterologous polypeptide. . 14. The chimeric polypeptide of Claim 13, wherein said heterologous polypeptide is an epitope tag sequence or an Fe region of an immunoglobulin. 15. An isolated antibody that binds to a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ. ID N0: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide, - (d) a sequence of amino acids from an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 16. The antibody of Claim 15 that binds to a polypeptide comprising: a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 17. The antibody of Claim 15 which is a monoclonal antibody. 18. The antibody of Claim 15 which is an antibody fragment. 19. The antibody of Claim 15 which is a chimeric or humanized antibody. 20. The antibody of Claim 15 which is conjugated to a growth inhibitory agent. 21. The antibody of Claim 15 which is conjugated to a cytotoxic agent. 22. The antibody of Claim 21, wherein the cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. 23. The antibody of Claim 21, wherein the cytotoxic agent is a toxin. 24. The antibody of Claim 23, wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin. 25. The antibody of Claim 23, wherein the toxin is a maytansinoid. 26. The antibody of Claim 15 which is produced in bacteria. 27. The antibody of Claim 15 which is produced in CHO cells. 28. The antibody of Claim 15 that induces the death of the cell to which it binds. 29. The antibody of Claim 15 which is detectably labeled. 30. An isolated nucleic acid comprising a nucleotide sequence encoding the antibody of Claim 15. 31. An expression vector comprising the nucleic acid of Claim 30 operably linked to the control sequences recognized by a host cell transformed with the vector. 32. A host cell comprising the expression vector of Claim 31. 33. The host cell of Claim 32 which is a CHO cell, an E. coli cell or a yeast cell. 34. A process for producing an antibody comprising culturing the host cell of Claim 32 under conditions suitable for the expression of said antibody and recovering said antibody from the cell culture. 35. An isolated oligopeptide that binds to a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ. ID N0: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its signal peptide - - associated; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 36. The oligopeptide of Claim 35 that binds to a polypeptide comprising: a) the amino acid sequence shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ ID NO: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide, - (d) a sequence of amino acids from an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 37. The oligopeptide of Claim 35 which is conjugated to a growth inhibitory agent. . 38. The oligopeptide of Claim 35 which is conjugated to a cytotoxic agent. 39. The oligopeptide of Claim 38, wherein the cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. - - 40. The oligopeptide of Claim 38, wherein the cytotoxic agent is a toxin. 41. The oligopeptide of Claim 40, wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin. 42. The oligopeptide of Claim 40, wherein the toxin is a maytansinoid. 43. The oligopeptide of Claim 35 that induces the death of the cell to which it binds. 44. The oligopeptide of Claim 35 that is detectably labeled. 45. An organic TAT binding molecule that binds to a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ ID N0: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID N0: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ ID N0: 4), with its associated signal peptide; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 46. The organic molecule of Claim 45 that binds to a polypeptide comprising: (a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); (b) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (c) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), with its associated signal peptide; (d) an amino acid sequence of an extracellular domain of the polypeptide shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4), which lacks its associated signal peptide; (e) an amino acid sequence encoded by the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); (f) an amino acid sequence encoded by the full length coding sequence of the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2); or (g) an amino acid sequence encoded by the full-length coding sequence of the cDNA deposited under the accession number ATCC shown in Table 7. 47. The organic molecule of Claim 45 which is conjugated to a growth inhibitory agent. . 48. The organic molecule of Claim 45 which is conjugated to a cytotoxic agent. 49. The organic molecule of Claim 48, wherein the cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. 50. The organic molecule of Claim 48, wherein the cytotoxic agent is a toxin. 51. The organic molecule of Claim 50, wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin. 52. The organic molecule of Claim 50, wherein the toxin is a maytansinoid. 53. The organic molecule of Claim 45 that induces the death of the cell to which it binds. 54. The organic molecule of Claim 45 which is detectably labeled. 55. A composition of matter comprising: (a) the polypeptide of Claim 11; (b) the chimeric polypeptide of the Claim 13; (c) the antibody of Claim 15; (d) the oligopeptide of Claim 35, or (e) the organic TAT binding molecule of the Claim 45 in combination with a vehicle. 56. The composition of matter of Claim 55, wherein said carrier is a pharmaceutically acceptable carrier. 57. An article of manufacture that includes: (a) a container; and (b) the composition of matter of Claim 55 contained within said container. 58. The article of manufacture of Claim 57 further comprising a label attached to said container or a package insert included with said container, which relates to the use of said composition of matter for the therapeutic treatment or diagnostic detection of a cancer. . 59. A method for destroying a cancer cell expressing a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), said method comprising contacting said cancer cell with an antibody, oligopeptide or organic molecule that binds to said polypeptide in said cancer cell, thereby destroying said cancer cell. 60. The method of Claim 59 wherein said antibody is a monoclonal antibody. 61. The method of Claim 59 wherein said antibody is an antibody fragment. 62. The method of Claim 59 wherein said antibody is a chimeric or humanized antibody. 63. The method of Claim 59, wherein said oligopeptide antibody or organic molecule is conjugated to a growth inhibitory agent. 64. The method of Claim 59, wherein said oligopeptide antibody or organic molecule is conjugated to a cytotoxic agent. 65. The method of Claim 64, wherein said cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. 66. The method of Claim 64, wherein the cytotoxic agent is a toxin. 67. The method of Claim 66, wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin. 68. The method of Claim 66, wherein the toxin is a maytansinoid. 69. The method of Claim 59, wherein said antibody is produced in bacteria. 70. The method of Claim 59, wherein said antibody is produced in CHO cells. 71. The method of Claim 59, wherein said cancer cell is subsequently exposed to radiation treatment or a chemotherapeutic agent. 72. The method of Claim 59, wherein said cancer cell is selected from the group consisting of a breast cancer cell, a colorectal cancer cell, a lung cancer cell, an ovarian cancer cell, a cell of central nervous system cancer, a liver cancer cell, a bladder cancer cell, a pancreatic cancer cell, a cervical cancer cell, a melanoma cell and a leukemia cell. 73. The method of Claim 59, wherein said cancer cell overexpresses said polypeptide in comparison to a normal cell of the same tissue origin. associated signal peptide; 74. A method for therapeutically treating a mammal having a tumor comprising cells expressing a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO:); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), said method comprising nistering to said mammal a therapeutically effective amount of an antibody, oligopeptide or organic molecule that binds to said polypeptide, thereby effectively treating said mammal. 75. The method of Claim 74, wherein said antibody is a monoclonal antibody. 76. The method of Claim 74 wherein said antibody is an antibody fragment. 77. The method of Claim 74 wherein said antibody is a chimeric or humanized antibody. 78. The method of Claim 74 wherein said oligopeptide antibody or organic molecule is conjugated to a growth inhibitory agent. 79. The method of Claim 74 wherein said oligopeptide antibody or organic molecule is conjugated to a cytotoxic agent. 80. The method of Claim 79, wherein said cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. 81. The method of Claim 79, wherein the cytotoxic agent is a toxin. 82. The method of Claim 81, wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin. 83. The method of Claim 81, wherein the toxin is a maytansinoid. 84. The method of Claim 74 wherein said antibody is produced in bacteria. 85. The method of Claim 74 wherein said antibody is produced in CHO cells. 86. The method of Claim 74, wherein said tumor is further exposed to radiation treatment or a chemotherapeutic agent. 87. The method of Claim 74, wherein said tumor is a breast tumor, a colorectal tumor, a lung tumor, an ovarian tumor, a tumor of the central nervous system, a tumor of the liver, a tumor of the bladder, a pancreatic tumor or a cervical tumor. 88. A method for determining the presence of a polypeptide in a sample suspected of containing said polypeptide, wherein said polypeptide has at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 ( SEQ ID N0: 3) or Figure 4 (SEQ ID N0: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), said method comprising exposing said parent to an antibody, oligopeptide or organic molecule that binds to said polypeptide and determine the binding of said antibody, oligopeptide or organic molecule to said polypeptide in said sample. 89. The method of Claim 88, wherein said sample comprises a cell suspected of expressing said polypeptide. 90. The method of Claim 89, wherein said cell is a cancer cell. 91. The method of Claim 88, wherein said oligopeptide antibody or organic molecule is detectably labeled. 92. A method for diagnosing the presence of a tumor in a mammal, said method comprising detecting the level of expression of a gene encoding a protein having at least 80% amino acid sequence identity for: (a) the sequence of amino acids shown in Figure 3 (SEQ ID N0: 3) or Figure 4 (SEQ ID NO: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), in a test sample of tissue cells obtained from said mammal and in a control sample from known normal cells of the same tissue origin, wherein a higher level of expression of said polypeptide in the test sample, compared to the control sample, is indicative of the presence of a tumor in the mammal from which the test sample was obtained. 93. The method of Claim 92, wherein the step of detecting the level of expression of a gene encoding said polypeptide comprises employing an oligonucleotide in an in situ hybridization or T-PC analysis. 94. The method of Claim 92, wherein the step of detecting the level of expression of a gene encoding said polypeptide comprises employing an antibody in an immunohistochemical assay. 95. A method for diagnosing the presence of a tumor in a mammal, said method comprising contacting a test sample of tissue cells obtained from said mammal with an oligopeptide antibody or organic molecule that binds to a polypeptide that has at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID NO: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), and detect the formation of a complex between said antibody, oligopeptide or organic molecule and said TAT polypeptide in the test sample, wherein the formation of a complex is indicative of the presence of a tumor in the mammal. 96. The method of Claim 95, wherein said oligopeptide antibody or organic molecule is detectably labeled. 97. The method of Claim 95 wherein said test sample of the tissue cells is obtained from an individual suspected of having a cancerous tumor. 98. A method for treating or preventing a cell proliferative disorder associated with the expression or increased activity of a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in Figure 3 (SEQ. ID N0: 3) or Figure 4 (SEQ ID NO: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), said method comprising administering to a subject who needs such treatment an effective amount of an antagonist of a TAT polypeptide. Preferably, the cell proliferative disorder is cancer and the antagonist of said polypeptide. 99. The method of Claim 98, wherein said antagonist is an anti-TAT-binding oligopeptide TAT polypeptide antibody, TAT binding organic molecule or anti-sense oligonucleotide. Even the additional embodiments of the present invention will be apparent to the skilled artisan upon reading the present specification.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows a nucleotide sequence (SEQ ID NO: 1) of a TAT291 cDNA, wherein SEQ ID NO: 1 is a clone designated herein as "DNA45234-1277". Figure 2 shows a sequence of nucleotides (SEQ ID NO: 2) of a TAT292 cDNA, wherein SEQ ID NO: 2 is a clone designated herein as "DNA37151-1193". Figure 3 shows the amino acid sequence (SEQ ID NO: 3) derived from the coding sequence of SEQ ID NO: l shown in Figure 1. Figure 4 shows the amino acid sequence (SEQ ID NO: 4) derived of the coding sequence of that of SEQ ID NO: 2 shown in Figure 2. DETAILED DESCRIPTION OF THE PREFERRED MODALITIES I. Definitions The terms "TAT polypeptide" and "TAT" as used herein and when immediately followed by a numerical designation, refer to several polypeptides, wherein the complete designation (ie, TAT / number) refers to the sequences of specific polypeptide as described herein. The terms "TAT polypeptide / number" and "TAT / number" wherein the term "number" is given as a current numeric designation as used herein includes naturally occurring polypeptides, polypeptide variants and fragments of - natural sequence polypeptides and polypeptide variants (which are further defined herein). The TAT polypeptides described herein can be isolated from a variety of sources, such as from human tissue types or from other sources or prepared by recombinant or synthetic methods. The term "TAT polypeptide" refers to each individual TAT / number polypeptide described herein. All exposures in this specification that refer to the "TAT polypeptide" refer to each of the polypeptides individually as well as collectively. For example, descriptions of preparation, purification, derivation, formation of antibodies for or against, formation of TAT binding oligopeptides for or against, formation of organic TAT binding molecules for or against, administration, compositions containing, treatment of a disease with, etc., belong individually to each polypeptide of the invention. The term "TAT polypeptide" also includes the variants of the TAT / number polypeptides described herein. A "natural sequence TAT polypeptide" comprises a polypeptide having the same amino acid sequence as the corresponding TAT polypeptide derived from nature. Such naturally occurring TAT polypeptides can be isolated from nature or can be produced by recombinant or synthetic means. The term "Natural sequence TAT polypeptide" specifically includes truncated or secreted forms that occur naturally from the specific TAT polypeptide (eg, an extracellular domain sequence), variant forms that occur naturally (eg, alternatively spliced forms) and allelic variants that occur naturally from the polypeptide. In certain embodiments of the invention, the natural sequence TAT polypeptides described herein are mature or full-length wild-type polypeptides comprising the full-length amino acid sequences shown in the accompanying figures. The start and end codons (if indicated) are shown in bold and underlined in the figures. The nucleic acid residues indicated as "N" in the accompanying figures are any nucleic acid residue. However, although the TAT polypeptides described in the accompanying figures are shown to start with methionine residues designated herein as amino acid position 1 in the figures, it is conceivable and possible that other methionine residues located either upstream or downstream from position 1 of amino acids in the figures as the starting amino acid residue for the TAT polypeptides. The "extracellular domain" or "ECD" of the polypeptide - - AT refers to a form of the TAT polypeptide that is essentially free of the transmembrane and cytoplasmic domains. Commonly, a TAT polypeptide ECD will have less than 1% of such transmembrane and / or cytoplasmic domains and preferably, will have less than 0.5% of such domains. It should be understood that any transmembrane domain identified for the TAT polypeptides of the present invention are identified according to criteria routinely employed in the art to identify that type of hydrophobic domain. The exact limits of a transmembrane domain may vary but more likely by no more than about 5 amino acids in either the domain end as originally identified herein. Therefore optionally an extracellular domain of a TAT polypeptide can contain from about 5 or fewer amino acids on either side of the boundary of the transmembrane domain / extracellular domain as identified in the Examples or the specification and such polypeptides are contemplated by the present invention with or without the associated signal polypeptide and the nucleic acid encoding them. The approximate location of the "signal peptides" of the various TAT polypeptides described herein may be shown in the present specification and / or the accompanying figures. However, it is observed that the limit C- - - The terminal of a signal peptide may vary, but more likely by no more than about 5 amino acids on either side of the C-terminal boundary of the signal peptide as initially identified herein, wherein the C-terminal boundary of the signal peptide it can be identified according to the routine criteria employed in the art to identify that type of amino acid sequence element (eg, Nielsen et al., Prot. Eng. 10: 1-6 (1997) and von Heinje et al. Nucí Acid Res. 14: 4683-4690 (1986)). Furthermore, it should also be recognized that, in some cases, the cleavage of a signal sequence from a secreted polypeptide is not totally uniform, resulting in more than one secreted species. These mature polypeptides are contemplated by the present invention wherein the signal peptide is split into no more than about 5 amino acids on either side of the C-terminal boundary of the signal peptide as identified herein and the polynucleotides that encode it. . The "TAT polypeptide variant" means a TAT polypeptide, preferably an active TAT polypeptide, as defined herein having at least about 80% amino acid sequence identity with a full length natural sequence TAT polypeptide sequence as described herein, a TAT polypeptide sequence that lacks the signal peptide as it is - described herein, an extracellular domain of a TAT polypeptide, with or without the signal peptide, as described herein or any other fragment of a full-length TAT polypeptide sequence as described herein (such as encoded by a nucleic acid representing only a portion of the complete coding sequence for a full-length TAT polypeptide). Such variants of the TAT polypeptide include, for example, the TAT polypeptides wherein one or more amino acid residues are added or deleted, at the N- or C-terminus of the full length natural amino acid sequence. Commonly, a variant of the TAT polypeptide will have at least 80% amino acid sequence identity, alternatively at least about 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity for a full length natural sequence TAT polypeptide sequence as described herein, a TAT polypeptide sequence that lacks the signal peptide as described herein, an extracellular domain of a TAT polypeptide, with or without the signal peptide, as described herein or any other specifically defined fragment of a full-length TAT polypeptide sequence as described herein. Commonly, the TAT variant polypeptides are of at least about 10 amino acids in length, alternatively of at least about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600 amino acids in length or more. Optionally, the TAT variant polypeptides will have no more than one conservative amino acid substitution compared to the natural TAT polypeptide sequence, alternatively no more than 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid substitutions. conservative compared to the natural TAT polypeptide sequence. The "percent (%) identity of the amino acid sequence" with respect to the TAT polypeptide sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the sequence of specific TAT polypeptide, after aligning the sequences and entering the spaces, if necessary, to achieve the maximum percentage of sequence identity and without considering any of the conservative substitutions as part of the sequence identity. Alignment for the purposes of determining the percentage of the amino acid sequence identity can be achieved in various ways which are within the skill in the art, for example, using publicly available computer software such as BLAST software, BLAST- 2, ALIGN or Megalign (DNASTA). Those skilled in the art can determine the appropriate parameters for measuring alignment, including any algorithm necessary to achieve maximum alignment over the full length of the sequences being compared. However, for purposes of the present, amino acid sequence identity% values are generated using the ALIGN-2 sequence comparison computer program, wherein the complete source code for the ALIGN-2 program is provided. in Table 1 below. The ALIGN-2 sequence comparison computer program was authorized by de Genentech, Inc. and the source code shown in Table 1 below was registered with the user documentation in the US Copyright Office, Washington D.C., 20559, where it was registered under the Copyright Registry of E.U. No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or can be collected from the source code provided in Table 1 below. The ALIGN-2 program must be collected for use in a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the - - program ALIGN-2 and do not vary. In situations where ALIGN-2 is used for amino acid sequence comparisons, the% amino acid sequence identity of a given amino acid sequence A for, with or against a given B amino acid sequence (which may alternatively be expressed as a A sequence of amino acids given that it has or comprises a certain% amino acid sequence identity for, with or against a given B amino acid sequence) is calculated as follows: 100 times the X / Y fraction where X is the number of residues of amino acid labeled as identical equivalences by the ALIGN-2 program of sequence alignment in that alignment of the program of A and B and where Y is the total number of amino acid residues in B. It will be appreciated that where the sequence length of amino acids A is not equal to the length of the amino acid sequence B,% amino acid sequence identity from A to B will not be equal to% amino acid sequence identity from B to A. As examples of% amino acid sequence identity calculations using this method, Tables 2 and 3 demonstrate how to calculate the% amino acid sequence identity of the amino acid sequence designated "Comparison Protein" for the amino acid sequence designated "TAT", where "TAT" represents the amino acid sequence of a - - hypothetical TAT polypeptide of interest, the "Comparison Protein" represents the amino acid sequence of a polypeptide against which the TAT polypeptide of interest is compared and "X," Y and "Z each represent different hypothetical amino acid residues. to be specifically stated otherwise, all% of the amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program The "TAT variant polynucleotide" or the "TAT variant nucleic acid sequence" refers to a nucleic acid molecule encoding a TAT polypeptide, preferably an active TAT polypeptide, as defined herein and having at least about 80% sequence identity of nucleic acid with a nucleotide sequence encoding a full length natural sequence TAT polypeptide sequence as disclosed ribe herein, a full length natural sequence TAT polypeptide sequence lacking the signal peptide as described herein, an extracellular domain of a TAT polypeptide, with or without the signal peptide, as described in present or any other fragment of a full length TAT polypeptide sequence as described herein (such as that encoded by an acid - - nucleic which represents only a portion of the complete coding sequence for a full-length TAT polypeptide). Commonly, a TAT variant polynucleotide will have at least about 80% nucleic acid sequence identity, alternatively at least about 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89 %, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% of nucleic acid sequence identity with a nucleic acid sequence encoding a polypeptide sequence TAT of full-length natural sequence as described herein, a full length natural sequence TAT polypeptide sequence lacking the signal peptide as described herein, an extracellular domain of a TAT polypeptide, with or without the signal sequence, as described herein or any other fragment of a full-length TAT polypeptide sequence as described herein. The variants do not cover the sequence of natural nucleotides. Commonly, the TAT variant polypeptides are at least about 5 nucleotides in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 210, - - 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990 or 1000 nucleotides in length, wherein in this context the term "approximately" refers to the length of the nucleotide sequence referred to plus or minus 10% of that referred length. The "percentage (%) of nucleic acid sequence identity" with respect to the TAT coding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical to the nucleotides in the sequence of TAT nucleic acid of interest, after aligning the sequences and introducing the spaces, if necessary, to achieve the maximum percentage of sequence identity. Alignment for the purposes of determining the percentage of nucleic acid sequence identity can be achieved in various ways that are within the skill in the art., for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. However, for purposes of the present,% of the nucleic acid sequence identity - values are generated using the sequence comparison computer ALIGN-2 program, wherein the complete source code for the ALIGN-2 program is provided in Table 1 below. The ALIGN-2 sequence comparison computer program was authorized by Genentech, Inc. and the source code shown in Table 1 below was registered with the user documentation in the US Copyright Office, Washington DC, 20559, where it was registered under the EU Copyright Registry No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or can be collected from the source code provided in Table 1 below. The ALIGN-2 program must be collected for use in a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. In situations where ALIGN-2 is used for nucleic acid sequence comparisons, the% nucleic acid sequence identity of a given nucleic acid C sequence for, with or against a given D nucleic acid sequence (which can be alternatively expressed as a given C nucleic acid sequence having or comprising a certain% nucleic acid sequence identity for, with or against a D sequence of nucleic acid - - given) is calculated as follows: 100 times the W / Z fraction where W is the number of nucleotides marked as identical equivalences by the ALIGN-2 sequence alignment program in that alignment of the C and D program and where Z is the total number of nucleotides in D. It will be appreciated that where the length of the C nucleic acid sequence is not equal to the length of the D nucleic acid sequence, the% nucleic acid sequence identity of C to D does not will be equal to the% nucleic acid sequence identity of D to C. As examples of the% nucleic acid sequence identity calculations, Tables 4 and 5 demonstrate how to calculate the% nucleic acid sequence identity of the nucleic acid sequence designated "Comparison DNA" for the nucleic acid sequence designated "TAT-DNA", wherein "TAT-DNA" represents a hypothetical interest sequence of the nucleic acid encoding TAT, the "Comparison DNA" represe nta the nucleotide sequence of a nucleic acid molecule against which the nucleic acid molecule "TAT-DNA" of interest is compared and "N", WL "and WV" each represent different hypothetical nucleotides. Unless specifically stated otherwise, all% of the nucleic acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program. In other embodiments, the polynucleotides of the TAT variant are nucleic acid molecules that encode a TAT polypeptide and that are capable of hybridizing, preferably under hybridization and stringent washing conditions, to the nucleotide sequences encoding a full-length TAT polypeptide as describes in the present. The TAT variant polypeptides may be those that are encoded by a TAT variant polynucleotide. "Isolated" when used to describe the various TAT polypeptides described herein, refers to the polypeptide that has been identified and separated and / or recovered from a component of its natural environment. The contaminating components of their natural environment are materials that can typically interfere with the diagnosis or therapeutic uses for the polypeptide and may include enzymes, hormones and other proteinaceous or non-proteinaceous solutes. In preferred embodiments, the polypeptide will be purified (1) to a sufficient degree to obtain at least 15 residues of the N-terminal or internal amino acid sequence by use of a rotary cup sequencer or (2) to homogeneity by SDS -PAGE under conditions without reduction or reduction using blue of - - Coomassie or preferably, silver tincture. The isolated polypeptide includes the polypeptide in situ within the recombinant cells since at least one component of the natural environment of the TAT polypeptide will not be present. However, commonly, the isolated polypeptide will be prepared by at least one purification step. A nucleic acid encoding the "isolated" TAT polypeptide or other nucleic acid encoding the polypeptide is a nucleic acid molecule that is identified and separated from at least one contaminating nucleic acid molecule with which it is commonly associated at the source of the nucleic acid encoding the polypeptide. An isolated molecule of nucleic acid encoding the polypeptide is different in the form or placement in which it is found in nature. Therefore, the isolated nucleic acid molecules encoding the polypeptide are distinguished from the nucleic acid molecule encoding the specific polypeptide as it exists in natural cells. However, a nucleic acid molecule encoding the isolated polypeptide includes nucleic acid molecules that encode the polypeptide contained in the cells that commonly express the polypeptide wherein, for example, the nucleic acid molecule is at a different chromosomal location than the of the natural cells. The term "control sequences" refers to the DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. For example, control sequences that are suitable for prokaryotes, include a promoter optionally an operator sequence and a ribosome binding site. Eukaryotic cells are known to use promoters, polyadenylation signals and enhancers. The nucleic acid is "operably linked" when placed in a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory guide is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is placed in order to facilitate translation. Generally, "operably linked" means that the DNA sequences that are linked are contiguous and in the case of a secretory guide, contiguous and in the reading phase. However, breeders do not have to be contiguous. The linkage is carried out by ligation at convenient restriction sites. If such sites do not exist, the adapters or linkers of synthetic oligonucleotides are used according to conventional practice. The "stringency" of the hybridization reactions is easily determined by one of ordinary skill in the art and is generally an empirical calculation that depends on the length of the probe, wash temperature and salt concentration. In general, longer probes require higher temperatures to adequately hybridize, while shorter probes are needed at lower temperatures. Hybridization generally depends on the ability of the denatured DNA to re-hybridize when the complementary filaments are present in an environment below its melting temperature. The higher the degree of desired homology between the probe and the hybridizable sequence, the higher the relative temperature that can be used. As a result, higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures make them lower. For further details and explanation of the stringency of the hybridization reactions, see Ausubel et al. , Current Protocols in Molecular Biology, (Current Protocols in Molecular Biology), Wiley Interscience Publishers, (1995). The "stringent conditions" or "high stringency conditions" as defined herein may be identified by those that: (1) employ low ionic stringency and high temperature for washing, for example, sodium chloride 0.015 M / sodium citrate 0.0015 M / sodium dodecyl sulfate 0.1% at 50 ° C; (2) employ during denaturation a denaturing agent, such as formamide, for example, 50% (v / v) of formamide with 0.1% bovine serum albumin / 0.1% Ficoll / 0.1% polyvinylpyrrolidone / phosphate buffer sodium 50 mM at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 ° C; or (3) hybridization overnight in a solution employing 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 1% sodium pyrophosphate. , 5 x Denhardt's solution, sonicated salmon sperm DNA (50 μ9 / t? 1), 0.1% SDS and 10% dextran sulfate at 42Â ° C, with a 10-minute wash at 42Â ° C in 0.2 x SSC (sodium chloride / sodium citrate) and 50% formamide at 55 ° C followed by a 10 minute high stringency wash consisting of 0.1 x SSC containing EDTA at 55 ° C. The "moderately stringent conditions" can be identified as described by Sambrook et al., Molecular Cloning; ? Laboratory Manual, (Molecular Cloning: a Laboratory Manual), New York: Cold Spring Harbor Press, 1989 and includes the use of washing solution and hybridization conditions. { e.g., temperature, ionic strength and% SDS) less stringent than those described above. A - - example of moderately stringent conditions is incubation overnight at 37 ° C in a solution comprising: 20% formamide, 5 x SSC (150 m NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6) , 5 x Denhardt's solution, 10% dextran sulfate and 20 mg / ml denatured cut salmon sperm DNA, followed by washing the filters in 1 x SSC at approximately 37-50 ° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc., as necessary to adjust the factors such as the length of the probe and the like. The term "labeled epitope" when used herein refers to a chimeric polypeptide comprising a TAT polypeptide or anti-TAT antibody fused to a "marker polypeptide". The marker polypeptide has enough residues to provide an epltope against which an antibody can be made, being still sufficiently short that it does not interfere with the activity of the polypeptide to which it is fused. The marker polypeptide is preferably also clearly unique so that the antibody does not substantially cross-react with other epitopes. Suitable marker polypeptides generally have at least six amino acid residues and usually between about 8 and 50 amino acid residues (preferably, between - about 10 and 20 amino acid residues). "Active" or "activity" for the purposes of the present refers to the form (s) of a TAT polypeptide which retains a biological and / or immunological activity of a native TAT or that occurs naturally, in where the "biological" activity refers to a biological function (either inhibitory or stimulatory) caused by a natural TAT or that occurs naturally different from the ability to induce the production of an antibody against an antigenic epitope possessed by a natural TAT or that occurs naturally and an "immunological" activity refers to the ability to induce the production of an antibody against an antigenic epitope possessed by a natural or naturally occurring TAT. The term "antagonist" is used in the broad sense and includes any molecule that blocks, inhibits or partially or completely neutralizes a biological activity of a natural TAT polypeptide described herein. In a similar form, the term "agonist" is used in the broadest sense and includes any molecule that mimics a biological activity of a natural TAT polypeptide described herein. Suitable agonist or antagonist molecules specifically include antibodies or fragments of agonist or antagonist antibodies, fragments or amino acid sequence variants of TAT polypeptides - natural, antisense oligonucleotide peptides, small organic molecules, etc. Methods for identifying agonists or antagonists of a TAT polypeptide may comprise contacting a TAT polypeptide with a candidate agonist or antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the TAT polypeptide. "Treat" or "treatment" or "relief" refers to both therapeutic treatment and prophylactic or preventive measures, where the object is to prevent or delay (decrease) the targeted pathological condition or disorder. Those in need of treatment include those who are still with the disorder as well as those prone to have the disorder or those in whom the disorder can be prevented. A subject or mammal is "successfully" treated for a cancer expressing the TAT polypeptide if, after receiving a therapeutic amount of an anti-TAT antibody TAT binding oligopeptide or TAT binding organic molecule in accordance with the methods of the present invention , the patient shows the observable and / or measurable reduction in or absence of one or more of the following: reduction in the number of cancer cells or absence of cancer cells; the reduction in tumor size, - the inhibition (i.e., decrease to a certain degree and preferably the - - arrest) of the infiltration of cancer cells towards the peripheral organs including the spread of the cancer in the soft tissue and bone; the inhibition (i.e., decrease to a certain degree and preferably arrest) of the tumor metastasis; the inhibition, to some degree, of tumor growth; and / or relief, to some degree from one or more of the symptoms associated with the specific cancer; morbidity and reduced mortality and improvement in the quality of life. To the extent that the anti-TAT antibody or the TAT-binding oligopeptide can prevent the growth and / or elimination of existing cancer cells, they can be cytostatic and / or cytotoxic. The reduction of these signs or symptoms can also be felt by the patient. The above parameters to assess successful treatment and improvement in the disease are easily measurable by routine procedures familiar to a physician. For cancer therapy, efficacy can be measured, for example, by assessing the time of disease progression (TTP) and / or determining the response rate (RR). Metastasis can be determined by staging and bone scans and tests for the level of calcium and other enzymes to determine diffusion to the bone. CT scans can also be done to look for diffusion to the pelvis and lymph nodes in the area. Chest x-rays and - - measurements of liver enzyme levels by known methods are used to search for metastases to the lungs and liver respectively. Other routine methods to monitor the disease include transrectal ultrasonography (TRUS) and transrectal injection biopsy (TRNB). For bladder cancer, which is a more localized cancer, methods to determine the progress of the disease include urinary cytological evaluation by cystoscopy, monitoring the presence of blood in the urine, visualization of the urothelial tract by sonography or a pyelogram intravenous, computerized tomography (CT) and magnetic resonance imaging (MRI). The presence of distant metastasis can be assessed by CT of the abdomen, chest X-ray or radionuclide image of the skeleton. "Chronic" administration refers to the administration of the agent (s) in a continuous mode as opposed to an acute mode, in order to maintain the initial therapeutic effect (activity) for an extended period. The "intermittent" administration is the treatment that is not done consecutively without interruption, but rather is cyclic in nature. "Mammal" for treatment purposes, alleviating the symptoms of or diagnosing a cancer is - - refers to any animal classified as a mammal, including humans, domestic and farm animals and zoo animals, entertainment or pets, such as dogs, cats, cattle, horses sheep, pigs, goats, rabbits, etc. Animal Preferably, the mammal is human. The administration "in combination with" one or more additional therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order. The "vehicles" as used herein include carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed to these at the dosages and concentrations employed. Frequently the physiologically acceptable carrier is a water buffered solution of pH. Examples of physiologically acceptable carriers include buffers such as phosphate, citrate and other organic acids; antioxidants include ascorbic acid; the low molecular weight polypeptide (less than about 10 residues); proteins, such as serum albumin, gelatin or immunoglobulins, - hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides and other carbohydrates that include glucose, mannose or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; - - counterions that form salts such as sodium; and / or non-ionic surfactants such as TWEEKT®, polyethylene glycol (PEG) and PLURONICS®. By "solid phase" or "solid support" refers to a non-aqueous matrix to which an oligopeptide link TAT antibody or TAT binding organic molecule of the present invention can adhere or join. Examples of solid phases encompassed herein include those partially or wholly formed of crystals. { e.g., controlled porous glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones. In certain embodiments, depending on the context, the solid phase may comprise the cavity of a test plate; in others it is a purification column [e.g., an affinity chromatography column). This term also includes a discontinuous solid phase of separated particles, such as that described in the U.S. Patent. No. 4,275,149. A "liposome" is a small vesicle composed of various types of lipids, phospholipids and / or surfactants that is useful for the delivery of a drug (such as a TAT polypeptide, an antibody thereof or a TAT-binding oligopeptide) to a mammal . The components of the liposome are commonly ordered in a bilayer formation similar to the lipid ordering of the biological membranes. A "small" molecule or organic molecule - - "small" is defined herein to have a molecular weight below about 500 Daltones. An "effective amount" of a polypeptide, TAT binding oligopeptide antibody, TAT binding organic molecule or an agonist or antagonist thereof as disclosed herein is sufficient for carrying out a specifically stated purpose amount. An "effective amount" can be determined empirically and in a routine manner, in relation to the stated purpose. The term "therapeutically effective amount" refers to an amount of an antibody, TAT-binding oligopeptide polypeptide, TAT-binding organic molecule or another drug effective to "treat" a disease or disorder in a subject or mammal. In the case of cancer, the therapeutically effective amount of the drug can reduce the number of cancer cells; reduce the size of the tumor; inhibit (i.e., decrease to a certain degree and preferably arrest) the infiltration of cancer cells into the peripheral organs; inhibit (i.e., decrease to a certain extent and preferably arrest) tumor metastasis; inhibit, to some degree, the growth of the tumor; and / or relief to some degree, from one or more of the symptoms associated with cancer. See, the definition in this "treatment". To the extent that the drug can prevent the growth and / or eliminate existing cancer cells, it can be cytostatic and / or cytotoxic. A "growth inhibitory amount" of an anti-TAT antibody, TAT oligopeptide TAT binding polypeptide or organic TAT binding molecule is an amount capable of inhibiting the growth of a cell, especially tumor, e.g. , cancer cell either in vitro or in vivo. A "growth inhibitory amount" of an anti-TAT antibody, TAT oligopeptide TAT binding polypeptide or organic TAT binding molecule for purposes of inhibiting the growth of the neoplastic cell can be determined empirically and in a routine manner. A "cytotoxic amount" of an anti-TAT antibody, TAT oligopeptide TAT binding polypeptide or organic TAT binding molecule is an amount capable of causing the destruction of a cell, especially tumor, eg, cancer cell either in vitro or in vivo . A "cytotoxic amount" of an anti-TAT antibody, TAT oligopeptide TAT binding polypeptide or TAT binding organic molecule for purposes of inhibiting the growth of the neoplastic cell can be determined empirically and in a routine manner. The term "antibody" is used in the broadest sense and specifically covers, for example, unique anti-TAT monoclonal antibodies (including agonist, antagonist and neutralizing antibodies), anti-TAT antibody compositions with polyepitopic specificity, polyclonal antibodies, single chain anti-TAT antibodies and anti-TAT antibody fragments (see below) while exhibiting the desired biological or immunological activity. The term "immunoglobulin" (Ig) is used interchangeably with antibody herein. An "isolated antibody" is one that has been identified and separated and / or recovered from a component of its natural environment. The contaminating components of its natural environment are materials that can interfere with the diagnosis or therapeutic uses for the antibody and may include enzymes, hormones and other proteinaceous or non-porteinaceous solutes. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method and more preferably more than 99% by weight (2) to a sufficient degree to obtain at least 15 residues of the N-terminal or internal amino acid sequence by using a rotary cup sequencer or (3) up to the homogeneity by SDS-PAGE under reducing or non-reducing conditions using Coomassie blue or preferably, silver tincture. The isolated antibody includes the antibody in itself within the recombinant cells since at least one component of the antibody's natural environment will not be present. However, commonly, the isolated antibody will be prepared by at least one purification step. The basic unit of 4-chain antibody is a heterotetrametric glycoprotex composed of two identical light (L) chains and two identical heavy (H) chains (one IgM antibody consists of 5 of the basic heterotetramer units together with an additional polypeptide called J chain) and therefore contains 10 antigen-binding sites, whereas secreted IgA antibodies can be polymerized to form polyvalent assemblies comprising 2-5 of the basic units of 4 chains together with the J chain). In the case of IgGs, the unit of 4 chains is generally about 150,000 Daltons. Each L chain is linked to an H chain by a covalent disulfide bond, while the two H chains are linked together by one or more disulfide bonds depending on the H chain isotype. Each H and L chain also has disulfide bridges of intra-chain spaced regularly. Each H chain has in the N-terminal, a variable domain (VH) followed by three constant domains (CH) for each of the chains a and? and four CH domains for the μ and e isotypes. Each L chain has in the N-terminal, a variable domain (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the - - heavy chain (CH1). Particular amino acid residues are believed to form an interference between the variable domains of light chain and heavy chain. The pair of one VH and one VL together form a single antigen binding site. For the structure and properties of different classes of antibodies, see e.g., Basic and Clinical Immunology, 8th edition, Daniel P. Stites, Abba I. Terr and Tristram G. Parslo (eds.), Appleton &; Lange, Norwalk, CT, 1994, page 71 and Chapter 6. The L chain of any vertebrate species can be assigned to one or two distinctly distinct types, called kappa and lambda, based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains (CH), immunoglobulins can be assigned to different kinds of isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, which have heavy chains designated a, d, e,? and μ respectively. The classes ? and a are further divided into subclasses on the basis of relatively small differences in sequence and function ¾ / e.g., humans express the following subclasses: IgG1, IgG2, IgG3, IgG, IgA1 and IgA2. The term "variable" refers to the fact that certain segments of the variable domains differ widely in the sequence between the antibodies. The V-domain mediates the binding to antigen and defines the specificity of a particular antibody for its particular antigen. However, the variability is not evenly distributed across the 110 amino acid extension of the variable domains. Instead, V regions consist of relatively invariant extensions called structure regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called "hypervariable regions" that are each 9-12 amino acids long. The variable domains of the heavy and light natural chains each comprise four FRs, greatly adopting a β-sheet configuration, connected by three hypervariable regions, which form connection circuits and in some cases are part of the β-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and with the hypervariable regions of the other chain contributing to the formation of the antigen-binding sites of the antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, (Sequences of Proteins of Immunological Interest), 5 Ed. Public Health Service, National Institutes of Health, Bethesda, D. (1991)). The constant domains do not directly involve the binding to antibody to an antigen, but exhibit several effector functions, such as the participation of the antibody in the - - antibody dependent on cellular cytotoxicity (ADCC). The term "hypervariable region" when used herein refers to the amino acid residues of an antibody that is responsible for the antigen binding. The hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (eg, about approximately residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in VL and around approximately 1-35 (Hl), 50-65 (H2) and 95-102 (H3) in the VH, Kabat et al., Sequences of Proteins of Immunological Interest (Sequences of Proteins of Immunological Interest) , 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) and / or those residues from a hypervariable circuit "(eg, residues 26-32 (Ll), 50-52 (L2) and 91-96 (L3) in the VL and 26-32 (Hl), 53-55 (H2) and 96-101 (H3) in the VH, Chothia and Lesk J. Mol. Biol., 196: 901-917 (1987). The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, ie, the individual antibodies comprising the population are identical except for naturally occurring mutations that may occur in smaller quantities. The monoclonal antibodies are highly specific, being directed against a unique antigenic site. In addition, in contrast to the preparations of the polyclonal antibody that includes different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant in the antigen. In addition to their specificity, monoclonal antibodies are advantageous in that they can be synthesized without being contaminated by other antibodies. The "monoclonal" modifier should not be considered to require the production of the antibody by any particular method. For example, monoclonal antibodies useful in the present invention can be prepared by the hybridoma methodology first described by Kohler et al. , Nature, 256: 495 (1975) or can be made using recombinant DNA methods in bacterial cells, eukaryotic animal or plant (see, e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" can also be isolated from libraries of phage antibodies using the techniques described for example in Clackson et al., Nature, 352: 624-628 (1991) and Marks et al., J. Mol. Biol. , 222: 581-597 (1991). Monoclonal antibodies herein include "chimeric" antibodies in which a portion of the heavy and / or light chain is identical or homologous to the corresponding sequences in antibodies derived from particular species or belonging to a class or subclass of antibody particular, while the remainder of the - (s) chain (s) is identical or homologous to the corresponding sequences in antibodies derived from other species or belonging to another class or subclass of antibody, as well as fragments of such antibodies, provided they exhibit the desired biological activity (see U.S. Patent No. 4,816,567; and Morrison et al., Proc. Nati, Acad. Sci. USA, 81: 6851-6855 (1984)). Chimeric antibodies of interest herein include "primatized" antibodies that comprise variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape, etc.) and human constant region sequences. An "intact" antibody is one that comprises an antigen binding site as well as a CL and at least heavy chain constant domains, CH1, CH2 and CH3. The constant domains can be natural sequence constant domains (e.g., human natural sequence constant domains) or amino acid sequence variants thereof. Preferably, the intact antibody has one or more effector functions. The "antibody fragments" comprise a portion of an intact antibody, preferably the antigen binding or the variable region of the intact antibody. Examples of antibody fragments include Fab, Fab ', F (ab') 2 and Fv fragments; diabodies; linear antibodies (see the - - Patent of E.U. No. 5,641,870, Example 2; Zapata et al., Protein Eng. 8 (10): 1057-10S2 [1995]); single chain antibody molecules; and multispecific antibodies formed from antibody fragments. The papain digestion of the antibodies produces two identical antigen-binding fragments, called "wFab" fragments, and a residual fragment "Fe", a designation that reflects the ability to crystallize easily.The Fab fragment consists of a complete L chain together with the variable region domain of the H chain (VH) and the first constant domain of a heavy chain (CH1) - Each Fab fragment is monovalent with respect to the antigen binding, ie, it has a single antigen-binding site. of the pepsin of an antibody produces a single large F (ab ') 2 fragment corresponding approximately to two disulfide-binding Fab fragments having divalent antigen-binding activity and is still capable of cross-linking to the antigen. They differ from the Fab fragments in that they have a few additional residues in the carboxy terminus of the CH1 domain including one or more cysteines from the art region. antibody iculation. Fab '-SH is the designation herein for Fab' in which the cysteine residue (s) of the constant domains carry a free thiol group. The F (ab ') 2 antibody fragments were originally - - produced as pairs of Fab 'fragments that have articulation cysteines between them. Other chemical couplings of antibody fragments are also known. The Fe fragment comprises the portions of the carboxy terminal of both H chains held together by disulfides. The effector functions of the antibodies are determined by the sequences in the Fe region, whose region is also the part recognized by the Fe (FcR) receptors found in certain cell types. wFv "is the minimum antibody fragment that contains a complete antigen-binding and recognition site.This fragment consists of a dimer of a variable region domain of a heavy and / or a light chain in close non-covalent association. of the functional duality of these two domains emanate six hypervariable circuits (3 circuits each from the H and L chain) that contribute to the amino acid residues for antigen binding and confer the antigen binding specifically to the antibody. , even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind the antigen, albeit at a lower affinity than the entire binding site. simple "also abbreviated as" sFv "or" scFv "are fragments of antibody comprising - - the VH and VL antibody domains connected in a single polypeptide chain. Preferably, the sFv polypeptide further comprises a polypeptide linker between the VH and VL domains which allows the sFv to form the desired structure for antigen binding. For a review of sFv see Pluckthun in The Pharmacology of Monoclonal Antibodies (The Pharmacology of Monoclonal Antibodies) vol. 113, Osenburg and Moore eds. , Springer-Verlag, New York, pp. 269-315 (1994); Borrebaeck 1995, infra. The term "diabodies" refers to the small fragments of antibody prepared by the construction of sFv fragments (see previous paragraph) with short linkers (approximately 5-10 residues) between the VH and VL domains in such a way that the pairs are achieved of the inter-chain but not of the intra-chain of the V domains, resulting in a bivalent fragment, ie, a fragment having two antigen-binding sites. The bispecific diabodies are heterodimers of two "crossover" sFv fragments in which the VH and VL domains of the two antibodies are present in different polypeptide chains. The diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Nati Acad. Sci. USA, 90: 6444-6448 (1993). The "humanized" forms of non-human antibodies (e.g., rodents) are chimeric antibodies that contain the minimum sequence derived from the non-human antibody. For the most part, humanized antibodies are human immunoglobulins (receptor antibody) whose residues from a hypervariable region of the receptor are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or non-human primate that has the specificity, affinity and capacity of the desired antibody. In some cases, the structure region (FR) residues of the human immunoglobulin are replaced by the corresponding non-human residues. In addition, the humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine the performance of the antibody. 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 hypervariable circuits correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a sequence of human immunoglobulin. The humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fe), typically that of a human immunoglobulin. For additional details, see Jones et al., Mature 321: 522-525 (1986); Riechman et al., Nature 332: 323-329 (1988); and Presta, Curr. Op. Struct. Biol. 2: 593-596 (1992). A "species-dependent antibody" eg, a mammalian anti-human IgE antibody is an antibody that has a stronger binding affinity for an antigen from a first mammalian species than it does for a homologue of that antigen from of a second species of mammal. Typically, the species-dependent antibody "specifically bound" to a human antigen (ie, has a binding affinity value (Kd) of not more than about 1 x 10"7 M, preferably not more than about 1 x 10 ~ 8 and more preferably no more than about 1 x 10 ~ 9 M) but has a binding affinity for an antigen homologue from a second non-human mammal species that is at least about 50 times or at least about 500 times or at least about 1000 times, weaker than its binding affinity for the human antigen The antibody dependent on the species can be of any of the various types of antibodies as defined above, but preferably is a humanized or human antibody. "TAG binding oligopeptide" is an oligopeptide that binds, preferably specifically, to a TAT polypeptide as described herein. T can be synthesized chemically - - using known oligopeptide synthesis methodology or can be prepared and purified using recombinant technology. The TAT-binding oligopeptides are usually at least about 5 amino acids in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100 amino acids in length or more, wherein such oligopeptides as described herein are capable of binding, preferably specifically, to a TAT polypeptide. The TAT binding oligopeptides can be identified without undue experimentation using well-known techniques. In this regard, it should be noted that the techniques for classifying oligopeptide libraries for oligopeptides that are capable of specifically binding to a polypeptide target are well known in the art (see, e.g., U.S. Patent Nos. 5,556,762, 5,750,373, 4,708,871, 4,833,092 , 5,223,409, 5,403,484, 5,571,689, 5,663,143; PCT Publications Nos. WO 84/03506 and WO84 / 03564; Geysen et al., Proc. Nati. Acad. Sci. USA, 81: 3998-4002 (1984); Geysen et al., Proc. Nati, Acad. Sci.
- - USA, 82: 178-182 (1985); Geysen et al., In Synthetic Peptides as Antigens (Synthetic Peptides as Antigens), 130-149 (1986); Geysen et al., J. Immunol. eth., 102: 259-274 (1987); Schoofs et al., J. Immunol, 140: 611-616 (1988), Cwirla, S. E. et al., (1990) Proc. Nati Acad. Sci. USA, 87: 6378; Lowman, H.B., et al., (1991) Biochemistry 30: 10832; Clackson, T. et al., (1991) Nature, 352: 624; Marks, J. D. et al., (1991), J. Mol. Biol., 222: 581; Kang, A.S. et al., (1991) Proc. Nati Acad. Sci. USA 88: 8363 and Smith G. P. (1991) Current. Opin. Biotechnol., 2: 668). An "organic TAT binding molecule" is an organic molecule different from an oligopeptide or antibody as defined herein that binds, preferably specifically, to a TAT polypeptide as described herein. Organic TAT binding molecules can be identified and synthesized chemically using known methodology (see e.g., PCT Publication Nos. WO00 / 00823 and WO00 / 39585). Organic TAT binding molecules are usually less than about 2000 Daltons in size, alternatively less than about 1500, 750, 500, 250 or 200 Daltons in size, wherein such organic molecules are capable of binding, preferably specifically, to a TAT polypeptide as described herein can be identified without undue experimentation using - - well-known techniques. In this regard, it should be noted that techniques for classifying libraries of organic molecules for molecules that are capable of binding to a polypeptide target are well known in the art (see, eg, PCT Publications Nos. WO00 / 00823 and WO00 / 39585). An oligopeptide antibody or other organic molecule "linking" an antigen of interest, eg, a tumor-associated polypeptide antigen target, is one that binds to the antigen with sufficient affinity such that the oligopeptide antibody or other organic molecule it is useful as a diagnostic and / or therapeutic agent for targeting a cell or tissue that expresses the antigen and does not significantly cross-react with other proteins. In such embodiments, the degree of binding of the oligopeptide antibody or other organic molecule to an "unfocused" protein will be less than about 10% of the linkage of the oligopeptide antibody or other organic molecule to its particular target protein as determined by the screening analysis. of the fluorescence activated cell (FACS) or by radioimmunoprecipitation (RIA). With respect to the binding of an oligopeptide antibody or other organic molecule to a target molecule, the term "specific binding" or "specifically linked to" or is "specific for" a particular polypeptide or an epitope on a target - - of particular polypeptide refers to the linkage that is measurably different from a non-specific interaction. The specific binding can be measured, for example, by determining the binding of a molecule compared to the binding of a control molecule, which is generally a molecule of similar structure that has no binding activity. For example, the specific binding can be determined by competition with a control molecule that is similar to the target, for example, an excess of unlabeled target. In this case, the specific binding is indicated if the binding of the target tagged to a probe is competitively inhibited by the excess of the unlabeled target. The term "specific binding" or "specifically binds to" or is "specific for" a particular polypeptide or epitope on a particular polypeptide target as used herein may be exhibited, for example, by a molecule having a for the target of at least about 10 ~ 4 M, alternatively at least about 1CT5, alternatively at least about 10 -6 M, alternatively at least about 10 -7 M, alternatively at least about 10 -8, alternatively at least about 10- 9 M, alternatively at least about 10"10 M, alternatively at least about 10" 11, alternatively at least about 10"M or greater.
- - In one embodiment, the term "specific link" refers to the linkage in which a molecule binds to a particular polypeptide or epitope in a particular polypeptide without substantially binding to any other polypeptide or polypeptide epitope. An oligopeptide antibody or other organic molecule that "inhibits the growth of tumor cells expressing a TAT polypeptide" or an oligopeptide antibody or other organic molecule "growth inhibitor" is one that results in the inhibition of measurable cell growth. cancers that express or overexpress the appropriate TAT polypeptide. The TAT polypeptide can be a transmembrane polypeptide expressed on the surface of a cancer cell or it can be a polypeptide that is produced and secreted by a cancer cell. Preferred oligopeptide antibodies or other organic anti-TAT inhibitory molecules inhibit the growth of tumor cells expressing TAT by more than 20%, preferably from about 20% to about 50% and even more preferably, by more than 50% (e.g., from about 50% to about 100%) compared to the appropriate control, the control typically being tumor cells not treated with the oligopeptide antibody or other organic molecule being tested. In one embodiment, the inhibition of growth can be measured at an antibody concentration of about 0.1 to 30 μ / p 1 or about 0.5 nM to 200 nM in a cell culture, wherein the inhibition of growth is determined from 1- 10 days after exposure of the tumor cells to the antibody. Inhibition of the growth of tumor cells in vivo can be determined in various ways as described in the Experimental Examples of the section below. The antibody is growth inhibitory in vivo if administration of the anti-TAT antibody at about 1 g / kg to about 100 mg / kg body weight results in reduction in tumor size or tumor cell proliferation within approximately 5 days to 3 months from the first administration of the antibody, preferably within about 5 to 30 days. An oligopeptide antibody or other organic molecule that "induces apoptosis" is the one that induces programmed cell death as determined by the binding of annexin V, DNA fragmentation, cell decline, dilatation of the endoplasmic reticulum, cellular fragmentation and / or formation of membrane vesicles (called apoptotic bodies). The cell is usually the one that overexpresses a TAT polypeptide. Preferably the cell is a tumor cell, e.g., a prostate, breast, ovarian, stomach, endometrial, lung, kidney, colon, bladder. Various methods are found - - available to evaluate cellular events associated with apoptosis. For example, the translocation of phosphatidyl serine (PS) can be measured by the annexin binding; DNA fragmentation can be assessed through the staggering of DNA; and nuclear / chromatin condensation together with DNA fragmentation can be assessed by any increase in hypodiploid cells. Preferably, the oligopeptide antibody or other organic molecule that includes apoptosis is the one that results in about 2 to 50 times, preferably about 5 to 50 times and more preferably about 10 to 50 times, the induction of the annexin-related linkage. the untreated cell in an annexin binding assay. The "effector functions" of the antibody refer to the biological activities attributable to the Fe region (a Fe region of natural sequence or a Fe region variant of amino acid sequence) of an antibody and varies with the isotype of the antibody. Examples of effector functions of the antibody include: the Clq bond and complement-dependent cytotoxicity; Fe receptor link; antibody-mediated cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor); and the activation of B cells - - "Antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a form of cytotoxicity in which the secreted Ig bound to the Fe (FcRs) receptors present in certain cytotoxic cells (e.g., Natural Eliminator cells) (N), neutrophils and macrophages) allow these cytotoxic effector cells to bind specifically to a target cell that carries the antigen and subsequently eliminate the target cell with the cytotoxins. The antibodies "arm" the cytotoxic cells and are absolutely required for such elimination. The primary cells to mediate the ADCC, NK cells express only FcyRIII, while the monocytes express FcyRI, FcyRII and FcyRIII. The expression FcR in hematopoietic cells is summarized in Table 3 on page 464 of Ravetech and Kinet, Annu. Rev. Immunol. 9: 457-92 (1991). To assess the ADCC activity of a molecule of interest, an ADCC in vitxo assay, such as that described in the E.TJ Patent, can be carried out. No. 5,500,362 or 5,821,337. Effector cells useful for such assays include peripheral blood mononuclear cells (PBMC) and Natural Eliminator (NK) cells. Alternatively or additionally, the ADCC activity of the molecule of interest can be assessed in vivo, e.g., in an animal model such as that described by Clynes et al., (EUA) 95: 652-656 (1998). The "Fe receptor" or "FcR" describes a receiver that - - it binds to the Fe region of an antibody. The preferred FcR is a natural human FcR sequence. In addition, a preferred FcR is one that binds to an IgG antibody (a gamma receptor) and includes receptors of the subclasses FcyRI, FcyRII and FcyRIII, including allelic variants and alternatively spliced forms of these receptors. FcyRII receptors include FcyRIIA (an "activation receptor") and FcyRIIB (an "inhibition receptor"), which have similar amino acid sequences that differ mainly in the cytoplasmic domains thereof. The activation receptor FcyRIIA contains an immunoreceptor activation motive based on tyrosine (ITAM) in its cytoplasmic domain. The inhibition receptor FcyRIIB contains an immunoreceptor motif of inhibition based on tyrosine (ITIM) in its cytoplasmic domain. (See review by M. in Daéron, Annu. Rey, Immunol., 15: 203-234 (1997)). The FcRs were summarized in Ravetch and Kinet, Annu. Rev. Immunol. 9: 457-492 (1991); Capel et al., Immunomethods 4: 25-34 (1994); and de Haas et al., J. Lab. Clin. ed. 126: 330-41 (1995). Other FcRs, including those to be identified in the future, are included by the term "FcR" herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetuses (Guyer et al., J. Immunol. 117: 587 (1976) and Kim et al., Immunol. 24: 249 ( 1994)).
- - "Human effector cells" are leukocytes that express one or more PcRs and carry out effector functions. Preferably, the cells express at least FcyRIII and carry out the effector function of ADCC. Examples of human leukocytes that mediate ADCC include peripheral blood mononuclear cells (PB C), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; PBMCs and NK cells being preferred. Effector cells can be isolated from a natural source, e.g., from the blood. "Complement-dependent cytotoxicity" or "CDC" refers to the lysis of a target cell in the presence of complement. Activation of the classical complement pathway is initiated by the binding of the pirmer component of the complement system (Clq) to the antibodies (of the appropriate subclass) that bind to their cognate antigen. To assess complement activation, a CDC assay can be carried out, e.g., as described in Gazzano-Santoro et al., J. Immunol. Met ods 202: 163 (1996). The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma and leukemia or - - Lymphoid malignancies More particular examples of such cancers include squamous cell cancer (eg, epithelial squamous cell cancer), lung cancer including small cell lung cancer, non-small cell lung cancer, lung adenocarcinoma and squamous cell carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urinary tract cancer, hepatoma, breast cancer, colon cancer, cancer rectal, colorectal cancer, endometrial or uterine carcinoma, carcinoma of the salivary gland, kidney or kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, melanoma, multiple myeloma and cancer B cell lymphoma, brain, as well as head and neck and associated metastasis. The terms "cell proliferative disorder" and "proliferative disorder" refer to disorders that are associated with some degrees of abnormal cell proliferation. In one embodiment, the cell proliferative disorder is cancer. The "tumor" as used herein, refers to all neoplastic and malignant cell growths and proliferations whether malignant or benign and all cells and - - pre-cancerous and cancerous tissues. An oligopeptide antibody or other organic molecule that "induces cell death" is one that causes a viable cell to become non-viable. The cell is that which expresses a TAT polypeptide, preferably a cell that overexpresses a TAT polypeptide as compared to a normal cell of the same type of tissue. The TAT polypeptide can be a transmembrane polypeptide expressed on the surface of a cancer cell or it can be a polypeptide that is produced and secreted by a cancer cell. Preferably, the cell is a cancer cell, e.g. , an ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell. In vitro cell death can be determined in the absence of complement and immune effector cells to distinguish cell death induced by antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). Thus, the assay for cell death can be carried out using heat inactivated serum (i.e., in the absence of complement) and in the absence of immune effector cells. To determine whether the oligopeptide antibody or other organic molecule is capable of inducing cell death, the loss of membrane integrity as assessed by the absorption of propidium iodide (PI), trypan blue - - (see Moore et al., Cytotechnology 17: 1-11 (1995)) or 7AAD can be assessed in relation to untreated cells. Oligopeptide antibodies or other preferred organic molecules that induce cell death are those that induce PI absorption in the PI absorption assay in BT474 cells. A "cell expressing TAT" is a cell that expresses an endogenous or transfected TAT polypeptide either on the cell surface or in a secreted form. A "cancer expressing TAT" is a cancer comprising cells that have a TAT polypeptide present on the cell surface or that produce and secrete a TAT polypeptide. A "cancer expressing TAT" optionally produces sufficient levels of TAT polypeptides on the surface of cells thereof, such that the oligopeptide antibody or other organic anti-TAT molecule can bind thereto and have a therapeutic effect with respect to cancer. . In another embodiment, a "cancer expressing TAT" optionally produces and secretes sufficient levels of TAT polypeptide, such that an oligopeptide antibody or other organic anti-TAT antagonist molecule can be enriched thereto and have a therapeutic effect with respect to cancer. With respect to the foregoing, the antagonist may be an antisense oligonucleotide that reduces, inhibits or prevents the production and secretion of the TAT polypeptide secreted by the cells of - tumor. A cancer that "overexpresses" a TAT polypeptide is one that has significantly higher levels of the TAT polypeptide on the cell surface thereof or produces and secretes compared to a non-cancerous cell of the same type of tissue. Such overexpression can be caused by amplification of the gene or by increased transcription or translation. Overexpression of the TAT polypeptide can be determined in a diagnostic or prognostic assay by evaluating increased levels of the TAT protein present on the surface of a cell or secreted by the cell. { e.g., through an immunohistochemical assay using anti-TAT antibodies prepared against an isolated TAT polypeptide that can be prepared using recombinant DNA technology from an isolated nucleic acid encoding the TAT polypeptide; FACS analysis, etc.). Alternatively or additionally, the levels of nucleic acid encoding the TAT polypeptide or mRNA in the cell can be measured, e.g. , through fluorescent hybridization in si tu using a nucleic acid based on the corresponding probe for a nucleic acid encoding TAT or the complement thereof; (FISH; see W098 / 45479 published October, 1998), Southern blotting, Northern blotting techniques or polymerase chain reaction (PCR), such as quantitative real-time PCR (RT-PCR). You can also study the - - overexpression of the TAT polypeptide by measuring the antigen poured into a biological fluid such as serum, eg, using assays based on the antibody (see also eg, U.S. Patent No. 4,933,294 issued June 12, 1990; WO91 / 05264 published in April 18, 1991; U.S. Patent 5,401,638 issued March 28, 1995; and Sias et al., J. Immunol, Methods 132: 73-80 (1990)). Apart from the above assays, several in vivo assays are available for the expert practitioner. For example, cells within the patient's body can be exposed to an antibody that is optionally labeled with a detectable label, eg, a radioactive isotope and the binding of the antibody to the cells in the patient can be evaluated, eg, by external screening for the radioactivity or by analyzing a biopsy taken from the patient previously exposed to the antibody. As used herein, the term "immunoadhesin" refers to antibody-like molecules that combine the binding specificity of a heterologous protein (an "adhesin") with the effector functions of the immunoglobulin constant domains. Structurally, the immunoadhesins comprise a fusion of an amino acid sequence with the desired binding specificity that is different from the recognition of the antigen and the binding site of an antibody (i.e., is - - "heterologous") and an immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin molecule is typically a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesin can be obtained from any immunoglobulin, such as the subtypes IgG1, IgG2, IgG3 or IgG4, IgA (including IgA1 and IgA2), IgE, IgD or IgM. The word "label" when used herein refers to a detectable compound or composition that is conjugated directly or indirectly to the oligopeptide antibody or other organic molecule in order to generate an oligopeptide antibody or other "labeled" organic molecule. The marker can be detectable by itself. { e.g., radioisotope markers or fluorescent labels) or in the case of an enzymatic marker or can catalyze the chemical alteration of a compound or substrate composition that is detectable. The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents the function of cells and / or causes the destruction of cells. The term is proposed to include radioactive isotopes. { e.g., I131, I125 and 90, Re185, Re188, Sm153, Bi212, P32 and radioactive isotopes of Lu), chemotherapeutic agents, e.g.f methotrexate, adriamycin, vinca alkaloids (vincristine, vinblastine, etoposide) doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents, enzymes and fragments thereof such as nucleolytic enzymes, antibiotics and toxins such as small molecule toxins or enzymatically active toxins of bacterial origin , fungal, plant or animal, including fragments and / or variants thereof and the various anti-tumor or anti-cancer agents described below. Other cytotoxic agents are described below. A tumoricidal agent causes the destruction of the tumor cells. A "growth inhibitory agent" when used herein refers to a compound or composition that inhibits the growth of a cell, especially a cancer cell that expresses TAT either in vitro or in vivo. Thus, the growth inhibitory agent can be one that significantly reduces the percentage of cells expressing TAT in the S phase. Examples of growth inhibitory agents include agents that block the progression of the cell cycle (in a different place than the S phase). ), such as agents that induce retention Gl and retention of phase M. Classical blockers of the M phase include vincapervincas (vincristina and vinblastina), taxanos and inhibitors of topoisomerase II such as doxorubicina, epirubicina, - - daunorubicin, etoposide and bleomycin. These Gl-retaining agents are also poured into the retention of S-phase, for example, DNA alkylation agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil and ara-C.
Additional information can be found in The Molecular Basis of Cancer (The Molecular Bases of Cancer) Mendelsohn and Israel, eds. , Chapter 1, entitled "Cell cycle regulation oncogenes, and antineoplastic drugs" (Regulation of the oncogenic cell cycle and antineoplastic drugs) by Murakami et al., (WB Saunders: Philadelphia, 1995), especially p. 13. The taxanes (paclitaxel and docetaxel) are anti-cancer drugs both derived from the yew tree. Docetaxel (TAXOTERE®, Rhone-Poulenc Rorer), derived from the European yew, is a semi-synthetic analog of paclitaxel (TAXOL®, Bristol-Myers Squibb). Paclitaxel and docetaxel promote the assembly of microtubules from tubulin dimers and stabilize the microtubules by avoiding depolymerization, which results in the inhibition of mitosis in cells. "Doxorubicin" is an anthracycline antibiotic. The complete chemical name of doxorubicin is (8S-cis) -10- [(3-amino-2,3,6-trideoxy-aL-lixo-hexapyranosyl) oxy] -7,8,9,10-tetrahydro-6 , 8, 11-trihydroxy-8- (hydroxyacetyl) -l-methoxy-5, 12-naphtacenedione. The term "cytokine" is a generic term for - - proteins released by a cell population that act on another cell as intercellular mediators. Examples of such cytokines are lymphokines, monokines and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulation hormone (FSH), thyroid stimulating hormone (TSH) and luteinizing hormone (LH); liver growth factor; fibroblast growth factor; prolactin, placental lactogen; tumor necrosis factor-a and -ß; substance that inhibits mullerian; peptide associated with mouse gonadotropin; inhibin; activin; Vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-β; platelet growth factor; transformation growth factors (TGFs) such as TGF-cc and TGF-β; factor I and II of insulin-like growth; erythropoietin (EPO); osteoinductive factors; interferons such as interferon-a, -β and -y; colony stimulation factors (CSRs) such as macrophages-CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12; a tumor necrosis factor such as TNF-α or TNF-β; and other polypeptide factors including LIF and whole ligand (KL). As used herein, the term "cytokine" includes proteins from natural sources or from recombinant cell cultures and biologically active equivalents of naturally occurring cytokines. The term "package insert" is used to refer to instructions that are customarily included in commercial packages of therapeutic products, which contain information about indications, uses, doses, administration, contraindications and / or precautions concerning the use of such therapeutic products.
- - Table 1 C-C increased from 12 to 15 Z is average of EQ B is average of ND compared with unemployment is _M; stop-stop = 0; J (subject) comparison define # _M -8 / * value of a comparison with a stop * / int _day [26] [26] =. { / * ABCDEFGHIJKLMN 0 PQRSTUVWXYZ * / / * A * / 2, 0, -2, 0, 0, -4, 1, -1, -1, 0, -1, -2, -1, 0, JV, 1 0, -2, 1, 1, 0, 0, -6, 0, -3, 0.}. , / * B * / 0, 3, -4, 3, 2, -5, 0, 1, -2, 0, 0, -3, -2, 2, _M, -1, 1, 0, 0, 0, 0, -2, -5, 0, -3, 1.}. , / * C * / -2, -4, 15, -5, -5, -4, -3, -3, -2, 0, -5, -6, -5, -4, _M, -3 , -5, -4, 0, -2, 0, -2, -8, 0, 0, -5} , / * D * / 0, 3, -5, 4, 3, -6, 1, 1, -2, 0, 0, -4, -3, 2, _M, -1, 2, -1, 0 , 0, 0, -2, -7, 0, -4, 2.}. , / * E * / 0.2, -5, 3, 4, -5, 0, 1, -2, 0, 0, -3, -2, 1, _M, -1, 2, -1, 0, 0 , 0, -2, -7, 0, -4, 3.}. , -4, -5, -4, -6, -5, 9, -5, -2, 1, 0, -5, 2, 0, -4, _M, -5, -5, -4, - 3, -3, 0, -1, 0, 0, 7, -5} , / * G * / 1, 0, -3, 1, 0, -5, 5, -2, -3, 0, -2, -4, -3, 0, _M, -1, -1, - 3, 1, 0, 0, -1, -7, 0, -5, 0.}. , / * H * / -1, 1, -3, 1, 1, -2, -2, 6, -2, 0, 0, -2, -2, 2, _M, 0, 3, 2, - 1, -1, 0, -2, -3, 0, 0, 2.}. , 1 * 1 * 1 -1, -2, -2, -2, -2, 1, -3, -2, 5, 0, -2, 2, 2, -2, _M, -2, -2 -2, -1, 0, 0, 4, -5, 0, -1, -2} , * J * I 0, 0, 0.0, 0.0, 0, 0.0,0,0, 0,0,0, _M, 0, 0,0,0, 0,0,0, 0, 0, 0,0} , / * K * / -1, 0, -5, 0, 0, -5, -2, 0, -2, 0, 5, -3, 0, 1, _M, -1, 1, 3, 0 , 0, 0, -2, -3, 0, -4, 0.}. , 1 * 1 * 1 -2, -3, -6, -4, -3, 2, -4, -2, 2, 0, -3, 6, 4, -3, _M, -3, -2 , -3, -3, -l, 0, 2, -2, 0, -1, -2} , / * M * / -1, -2, -5, -3, -2, 0, -3, -2, 2, 0, 0, 4, 6, -2, _M, -2, -1, 0, -2, -1, 0, 2, -4, 0, -2, -1} , / * N * / 0.2, -4, 2, 1, -4, 0, 2, -2, 0, 1, -3, -2, 2, _M, -1, 1, 0, 1, 0, 0, -2, -4, 0, -2, 1.}. , 1 * 0 * 1. { _, _M, _M, _M, _M, _M, _M, _M, _M, _M, _M, _M, _, _M, 0, _M, _M, _M, _M, _M: _M, _M, _M, _M, _M, _M} , I * p * / 1, -1, -3, -1, -1, -5, -1, 0, -2, 0, -1, -3, -2, -l, _M, 6, 0, 0, 1, 0, 0, -1, -6, 0, -5, 0 ,} , / * Q * / 0.1, -5, 2, 2, -5, -1, 3, -2, 0, 1, -2, -1, 1, _M, 0, 4, 1, -1, - 1, 0, -2, -5, 0, -4, 3.}. , / * R * / -2, 0, -4, -1, -1, -4, -3, 2, -2, 0, 3, -3, 0, 0, _M, 0, 1, 6, 0, -1, 0, -2, 2, 0, -4, 0.}. , / * S * / 1, 0, 0, 0, 0, -3, 1, -1, -1, 0, 0, -3, -2, 1, _M, 1, -1, 0, 2, 1, 0, -1, -2, 0, -3, 0.}. 1, 0, -2, 0, 0, -3, 0, -1, 0, 0, 0, -1, -1, 0, _M, 0, -1, -1, 1, 3, 0, 0, -5, 0, -3, 0.}. , / * U * / 0, 0, 0,0, 0,0, 0, 0,0,0,0, 0,0, 0, _M, 0, 0,0,0, 0,0,0, 0, 0, 0,0} , I * Y * / 0, -2, -2, -2, -2, -1, -1, -2, 4, 0, -2, 2, 2, -2, _M, -L -2, -2, -1, 0, 0, 4, -6, 0, -2, -2} , l * W * / -6, -5, -8, -7, -7, 0, -7, -3, -5, 0, -3, -2, -4, -4, _M, -6 , -5, 2, -2, -5, 0, -6.17, 0, 0, -6} , l * X * / 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, _M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0.}. , / * and * / -3, -3, 0, -4, -4, 7, -5, 0, -1, 0, -4, -l, -2, -2, _M, -5, - 4, -4, -3, -3, 0, -2, 0, 0, 10, -4} , / *? * / 0, 1, -5, 2, 3, -5, 0, 2, -2, 0, 0, -2, -1, 1, _M, 0, 3, 0, 0, 0 , 0, -2, -6, 0, -4, 4.}. }; - - / * * / includes # < stdio.h > includes # < ctype h > define # MAXJMP 16 / * max jump in a diag * / define # MAXGAP 24 / * do not continue to penalize spaces larger than this * / define # JMPS 1024 / * max hop in a route * / define # MX 4 / * save if there are at least MX-1 bases since the last jump * / define # DMAT 3 / * value of comparison bases * / define # DMIS 0 / * penalty for decoupled bases * / define # DINSO 8 / * penalty for a space * / define # DINS1 1 / * penalty for base * / define # PINSO 8 / * penalty for a space * / define # PINS1 4 / * penalty for waste * / structure jump { short n [MAXJMP]; / * jump size (neg for dely) * / short without signal x [MAXJMP]; / * without base, from the jump in sec x * / / * limits sec to 2? 16 - 1 * 1}; structure diag { int score; / * score on the last jump * / - - long offset; / * outside the block prev * / short ijmp; / * current jump index * / jump structure jp; / * jump list * /}; path of structure { it spc / * number of guide spaces * / short n [JMPS]; / * sizeofl jump (space) * / int x [JMPS]; / * jump loe (last element before space) * /}; * ofile character; / * output file name * / character * namex [2]; / * names of sec: get secs 0 * / character * pmg; / * prog name for error messages * / character * seqx [2]; / secs: get secs 0 * int dmax; / * diag better: nw 0 * / int dmaxO; / * final diag * / int dna; / * fix if adn: principalO * / int endgaps; / * fix if final penalty spaces * / int gapx, gapy; / * total spaces in secs * / int lenO, lenl; / * sec lens * / int ngapx, ngapy; / * total size of spaces * / int smax; / * max score: nw 0 * / int * xbm; / * bitmap for comparison * / long offset; / * current output in jump file * / struct diag * dx; / * keeps diagonals * / - - estruct path PPP]; / * conserve route for secs * / character * calloc (), * malloc (), * index (), * strcpyO; character getseq (3, g-callocO; / * alignment program Needleman-Wunsch * * use: progs file 1 file2 * where file 1 and file2 are two DNA or two protein sequences. * The sequences can be in case Top or bottom can contain ambiguity * Any line that starts with '>' or '<' is ignored * The maximum length of the file is 65535 (limited by short x with no signal in the jump structure) * A sequence with 1/3 or more of its ACGTU elements is supposed to be DNA * The output is in the file "align.out" * * The program can create a tmp file in / tmp to keep info near the queue of trace * Original version developed under BSD 4.3 in a vax 8650 * / includes # "nw.h" includes * "day.h" static _dbval [26] =. { 1, 14, 2, 13, 0, 0, 4, 11, 0, 0, 12, 0, 3, 15, 0, 0, 0, 5, 6, 8, 8, 7, 9, 0, 10, 0 }; static jpbval [26] =. { 1, 2 I (1 < < (? '-? ·)) | (1 < < CN '-' A ')), 4, 8, 16, 32, 64, - - 128, 256, OxFFFFFFF, 1 < < 10, 1 < < 11, 1 < < 12, 1 < < 13, 1 < < 14, 1 < < 15, 1 < < 16, 1 < < 17, 1 < < 18, 1 < < 19, 1 < < 20, 1 < < 21, 1 < < 22, 1 < < 23, 1 < < 24, 1 < < 25 ¡(1 < < (? '-?')) | (1 «('Q'-W))}; principal (ac, av) principal int ac; character * av []; . { prog = av [0]; yes (ac! = 3). { fprintf (stderr, "use:% s archivol file2 \ n", prog); fprrntf (stderr, "where archivol and file2 are two adn or two sequences of proteinsAn"); fprititf (stderr, "The sequences may be in the upper or lower case '); ^ rintf (stderr," Any line starting with'; 'or' <is ignored \ n "); fprintf (stderr," The output is in the file \ "align.out \" \ n "); output (l);.}. namex [0] = av [l]; namex [l] = av [2]; seqx [0] = getseq (namex [0], & len0); seqxfl] = getseq (namex [l], & lenl); xbm = (dna)? _dbval: _pbval; endgaps = 0; / * 1 to penalize final spaces * / - - ofíle = "aIígn.out"; / * output file * / nwQ; / * fill in the matrix, get the possible jumps * / readjmpsQ; / * get the current jumps * / printQ; / * print states, alignment * / cleanup (O); / * unlinks any tmp file} / * does the alignment, returns to the best score: principalO * adn: values in Fitch and Smith, PNAS, 80, 1382-1386, 1983 * pro: PAM 250 values * When the scores are the same, we prefer to undo any space, prefer * a new space to extend a space in course and prefer a space in seqx * for a space in seq y. * / nwQ. { character * ??, py; / * secs and ptrs * / int * ndely, * dely; / * keeps track dely * / int delx, delx; / * keeps track delx * / int * tmp; / * for row transfer, row 1 * / int mis; / * score for each type * / int insO, insl; / * insertion penalties * / record id; / * diagonal index * / - - registration and; / * jump index * / record * colO, * coll; / * score for current row, last * / record xx and y; / * index in secs * / dx = (strnct diag *) g_calloc ("to get diags", lenO + Ienl + 1, sizeof (struct diag)); ndely (int *) g_calloc ("to getndely", lenl + 1, sizeof (int)); dely = (int *) g_calloc ("to get dely", lenl + 1, sizeof (int)); colO = (int *) g_calloc ("to get colO", lenl + 1, sizeof (int)); coll = (int *) g_calloc ("to get coll", lenl + 1, sizeof (int)); insO = (dna)? DI SO: PENfSO; insl = (dna)? DINS1: PINS1; smax = -10000; yes (final spaces). { for (colO [0] = delyfO] = -insO yy = 1; yy < = lenl; yy + +). { colO [yy] = delyfyy] = colO [yy-l] - insl; ndely [yy] = yy; } colO [0] = 0; / * Waterman Bull Math Biol 84 * /} also for (yy = 1; yy < = lenl; yy + +) delyfyy] = -insO; / * fill the comparison matrix * / for (px seqx [0], xx = 1; xx < = lenO; px + +, xx + +). { / * starts first entry in col * / si (espacial). { if (xx = = 1) coll [0] = delx = - (insO + insl); also coll [0] = delx = col0 [0] - insl ndelx = xx; } Besides . { coll [0] = 0; delx = -insO; ndelx = 0; } ... nw for (py = seqx [l] yy = 1; and < = lenl; py + + yy + +). { mis = col0 [yy-l]; if (dna) my + = (xbm [* px-'A,] & xbm [* py-'A '])? DMAT: DMIS; plus my + = _day [* px-'A '] [* py-'A']; / * updates penalty for the in sec x; * Please again from the envelope in progress * ignore MAXGAP if endgaps are weighted * / if (endgaps 1 1 ndely [yy] <MAXGAP). { yes (colOfyy] - insO > = dely [yy]). { dely [yy] = colO [yy] - (insO + insl); ndely [yy] = 1; } Besides . { dely [yy] - = insl; ndely [yy] + +; } } Besides . { yes (colO [yy] - (insO + insl) > = dely [y]). { dely [yy] = colOfyy] - (insO + insl); ndely [yy] = 1; } also ndely [yy] + +; } / * updates penalty for the in sec and; * Please again from the envelope in progress * / * / si (endgaps 1 1 ndelx <MAXGAP). { yes (coll [yy-l] - insO> = delx). { delx = coll [yy-l] - (insO + insl); ndelx = 1; } Besides . { delx-ins 1; ndelx + +; } } Besides . { yes (coll [yy-l] - (insO + insl) > = delx). { delx = coll [yy-l] - (insO + insl); ndelx = 1; } also ndelx + +; } / * pick up the maximum score; we were favored * my over any del and delx over dely * / ... nw id = XX - yy + lenl - 1; yes (my > = delx & mis > = delyfyy]) coll [yy] = mis; also if (delx > = dely [yy]). { coll [yy] = delx; ij = dx [id] .ijmp; yes (dx [id] .jp.n [0] & (! dna 1 1 (ndelx > = MAXJMP && > dx [id] .jp. x [ij] + MX) [| my > dx [id]. score + DINSO)). { dx [id]. ijmp + +; yes (+ + ij> = MAXJMP). { - - writejmps (id); ij = dx [id] .ijmp = 0; dx [id] .offset = offset; offset + = sizeof (struct jmp) + sizeof (offset); } } dx [id] .jp.n [ij] = ndelx; dx [id] .jp.x [ij] = xx; dx [id] .score = delx; } Besides . { coll [yy] = dely [yy]; ij = dx [id] .ijmp; & (! dna 1 1 (ndely [yy] > = MAXJMP & &xx > dx [id] jp. x [ij] + MX) 1 1 mis > dx [id]. score + DHSTSO)). { dx [id]. ijmp + +; yes (+ + ij> = MAXJMP). { writejmps (id); ij = dx [id] .ijmp = 0; dx [id]. offset = offset; offset + = sizeof (struct jmp) + sizeof (offset); } } dx [id] .jp.n [ij] = -ndely [yy]; dx [id] .jp.x [ij] = xx; - - dx [id] .score = delyfyy]; } yes (xx = = lenO &&yy <; lenl). { / * last col * / if (endgaps) coll [yy] - = insO + insl * (Ienl-yy); yes (coll [yy] > smax). { smax = coll [yy]; dmax = id; } } } yes (endgaps &&xx < lenO) coll [yy-l] - = insO + insl * (IenO-xx); yes (coll [yy-l] > smax). { smax = coll [yy-1]; dmax = id; } tmp = colO; colO = coll; coll = tmp; } (empty) free ((character *) ndely); (empty) free ((character *) dely); (empty) free ((character *) colO) (empty) free ((character *) coll) / * * * printO - only routine visible outside this module * * static: * getmatO - best trace path trace, compare account: printO * pr_align () - print alignment of what is described in order p []: printO * dumpblockO - download a block of lines with numbers, asterisks: pr_align () * numsO - put a line of numbers: dumpblockO * putlíneO - put a line (name, [num], sec, [num]): dumpblockO * starsO - put a line of asterisks: dumpblockO * stripnameO - remove any path and prefix from a sequence name * / include # "nw.h" define # SPC 3 define # P_LI E 256 / * maximum output line * / defme # P SPC 3 / * space between name or num and external _day [26] [26]; int olen; / * establishes line length of exit * / FILE * fx; / * output file * / int lx, ly, first space, last space; / * overlap * / if ((faith = fopen (ofile, "w")) = = 0). { fprintf (stderr, "% s: can not type% s \ n", prog ofile); elirninar (l); } ^ rintf (fk, "<first sequence:% s (length =% d) \ n", namex [0], lenO); fprintf (fx, "< second sequence:% s (length =% d) \ n", namex [l], lenl); olen-60; lx = len0; ly = lenl; first space = last space = 0; yes (dmax < lenl - 1). { / * guiding space in x * / pp [0] .spc = first space = lenl - dmax - 1; ly - = pp [0] .spc; } also if (dmax > lenl - 1). { / * guiding space in y * / pp [l] .spc = first space = dmax - (lenl - 1); lx - = pp [l] .spc; } yes (dmaxO < lenO - 1). { / * dragging space in x * / last space = lenO - dmaxO -1; lx- = last space; also if (dmaxO > lenO - 1). { / * dragging space in y * / last space = dmaxO - (lenO - 1); ly - last space; } getmat (lx, ly, first space, last space); pr_align (); } / * * trace queue the best route, count comparisons * / static getmat (lx, ly, first space, last space) getmat int lx, ly; / * "core" (less final spaces) * / int first space, last space; / * guiding dragging overlay * / . { int nm, iO, il, sizO, sizl; outx caract [32]; double pct; record nO, ni; record character * p0, * pl; / * get total comparisons, score * / iO = il = sizO = sizl = 0; pO = seqx [0] + pp [l]. spc; pl = seqx [l] + pp [0]. spc; ?? = pp [l] .spc + 1; nl = pp [0] .spc + 1; nm = 0; while (* pO && pl). { yes (sizO). { pl + +; nl + +; sizO--; } also if (sizl). { ?? + +; nO + +; sizl-; } Besides . { if (xbm [* pO-'A '] & xbm [* pl-'A']) nm + +; if (nO + + = = pp [0] .x [iO]) sizO = pp [0] .n [iO ++]; if (nl + + = = pp [l] .x [il]) sizl = pp [l] .n [il + +] pO ++; pl ++; } / * homology pct: * if final spaces are penalized, the base is the shortest sec * also eliminates inclinations and takes the shortest kernel * / si (final spaces) lx = (lenO <lenl)? lenO: lenl; also lx = (lx < ly)? lx: ly; pct = 100. * (double) nm / (double) lx; rprintf (fx, "\ n"); "<% d compare% s in an overlap of% d:% .2f percent similarity \ n", nm, (nm = = 1)? "": "is", lx, pct); fprintf (fx, "< spaces in first sequence:% d", gapx); ... getmat si (gapx). { (empty) sprintf (outx, "(% d% s% s)", ngapx, (dna)? "base": "residue", (ngapx = = 1)? "": "s"); f ???? "(??,"% 8"outx); p? 1? (??,", spaces in second sequence:% d ", gapy); if (gapy) { (empty) sprintf (outx, "(% d% s% s)", ngapy, (dna)? "base": "residual", (ngapy = = 1)? "": "s"); fp ??? (G? , "% s" outx);.}. if (dna) fprintf (fx, "\ n < punctuation:% d (comparison =% d, decoupling =% d, space penalty = % d +% d perbase) \ n ", smax, DMAT, DMIS, DINSO, DINS1), plus fprmtf (fx, < punctuation:% d (Dayhoff matrix PAM 250, space penalty =% d +% d per residue ) \ n ", smax, PINSO, PINS1); if (final spaces) ^ rintf (fx, "<final penalized spaces, left end space:% d% s% s, right end space:% d% s% s \ n", first space, (dna)? ":" residue ", (first space = = 1)?" ":" s ", last space, (dna)?" base ":" residue ", (last space = = 1)?" ":" s " ) also fprintf (fx, "<final spaces not penalized \ n");} static nm; / * compare in the kernel - for verification * / static lmax; / * lengths of file names removed * / static ij [2]; / * index jmp for a route * / static nc [2]; / * number in asterisk of current line * / static neither [2]; / * number of current element - to space * / static siz [2] ]; static character * ps [2]; / * ptr for current element * / static character * po [2]; / * ptr for next output character slot * / static character out [2] [P LESIE]; / * output line * / star character [P_LI E]; / * set by asterisks Q * / / * * printed alignment of the described in struct path pp [] * / static pr_alignO pr_align. { int nn; / * character count * / int plus; record i; for (i = 0, lmax = 0; i <2; i + +). { nn = stripname (namex [í]); if (nn> lmax) lmax = nn; nc [i] = 1; ni [i] = 1; siz [i] = ij [i] = 0; ps [i] = seqx [i]; po [i] = out [i]; - - for (n = nm = 0, plus = 1; more;). { ... pr_align for (i = more = 0; i <2; i + +). { / * * Do we have more of this sequence? * / if (! * ps [i]) continues; more + +; yes (pp [i] .spc). { / * guiding space * / * po [i] + + = "; pp [i] .spc ~;.}. Also if (siz [i]) { / * in a space * / * po [i] + + = '-'; siz [i] -;.}. addition { / * we put an element of sec * / * po [i] = * ps [i]; yes (islower (* ps [i] )) * ps [i] = toupper (* ps [i]); po [i] + +; ps [i] ++; / * * are we in the next space for this sec? - - * / si (ni [i] = = pp [i] .x [ij [i]]). { / * * we need to fuse all spaces * in this location * / siz [i] = pp [i] .n [ij [i] + +]; while (ni [i] == pp [i] .x [ij [i]]) siz [i] + = pp [i] .n [ij [i] ++]; } ni [i] + +; } } if (+ + nn = = olen 11! more & amp; nn). { download blockO; for (i = 0; i < 2; i + +) po [i] = out [i]; nn = 0; } } } / * * download a block of lines, including numbers, asterisks: pr_align () * / static - - download block or dumpblock. { record i; for (i = 0; i < 2; i + +) * po [i] - = '\ 0'; ..dumpblock (empty) putc ('\ n', faith); for (i = 0; i <2; i + +). { if (* out [i] & (* out [i]! = "11 * (po [i])! if (i = = 0) nums (i); if (i = = 0 & amp; * out [i]) asterisk O, put line (i), if (i = = 0 & * out [i]) fprintf (faith, asterisk); if (i = = 1) nums (i);.} . } / * * delete number line: download blockO * / static nums (ix) mims int ix; / * index in out [] preserve sec line nline character [P_LINE]; record character record * pn, * px, * py; for (pn = nline, i = 0, i <lmax + P_SPC, i + +, pn + +) * pn = "; for (i = nc [ix], py = out [ix]; * py; py + +, pn + +) { yes (* py = = "|| * py = = '-') * pn ="; in addition { si (i% 10 = = 0 I] (i = = 1 & & nc [ix]! = 1)) { J = (i < 0)? - i: i; for (px = pn; j; j / = 10, px-) * px = j% 10 + '0'; if (i <0) * px = '-';.}. in addition * pn = 'i ++;.}..}. * ?? =' \ 0 '; nc [ix ] = i; for (pn = nline; * pn; pn + +) (empty) putc (* pn, fx); (empty) putcC n !, fx);.}. / * * delete a line (name, [ num], sec, [num]): dumpblockO * / static put line (ix) putline int ix; { ... putline int i; character record px; for (px = namex [ix], i = 0; * px & * px! = ':'; px + +, i + +) (empty) putc (* px, fx); for (; i <lmax + P_SPC; i + +) (empty) putc ('', fx); / * this account from 1: * ni [] is current element (from 1) * nc [] is number in asterisk of current line * / for (x = out [ix]; * px; px + +) (empty) putc (* px &0x7F, faith); (empty) putc ('\ n', faith); } / * * puts a line of asterisks (secs always in out [0] out [l]): dumpblockO * / static asteriskO. { int i; registration feature * p0, * pl, ex, * px; if I return; px = asterisk; for (i = lmax + P_SPC; i; i-) * px ++ = ''; for (p0 = out [0], pl = out [l]; * p0 & pl; p0 ++, pl + +). { yes (isalpha (* pO) & isalpha (* pl)). { yes (xbm [* p0- A *] & xbm [* pl-A ']). { nm ++; } also if (! dna &&_day [* pO-, A,] [* pl-, A,] cx =! '; } also cx = "; * px + + = ex;.}. * px + + = '\ n'; * px = '\ 0';.}. / * * remove path or prefix from pn, return to len: pr_align 0 * / static remove name (pn) stripname character * pn; / * file name (can be path) * / {arch register px, * p py = 0; for (px = pn; * px; px + +) if (* px = = '/') py = px + 1; if (py) (empty) strcpy (pn, py); return (strlen (pn)); / * * clanup 0 - delete any file tmp * getseq 0 - read in sec, set adn, len, maxlen * g_calloc 0 callocO with error verification * readjmps 0 get good jumps, from tmp file if necessary * writejmps 0 write a sorting jumps to a tmp file: * / includes # "nw.h" includes # < sys / archivo.h > character * jname = "/ tmp / homgXXXXXX"; / * tmp file for jumps * / FILE * fj; int cleanup 0; / * delete tmp file * / long lseek; / * * remove any tmp file if we blow * / elirninar (i) cleanup int. { yes (5) (empty) without link (jname); exit (i); } / * - - * read, return ptr to sec, set dna, len, maxlen * skip lines that start with '; ',' < 'or 1 > 'sec in upper or lower case * / character * getseq (file, len) getseq char * file; / * file name * / int * len; / * sec len * / line character [1024], * pseq; feature registration * px, * py; int natgc, tlen; FILE * íp; if ((fp = fopen (file, "r")) = = 0). { fprintf (stderr, "% s: can not read% s \ n", prog, file); exit (l); } tlen = natgc = 0; while (fgets (line, 1024, fp)). { if (* line = = ';' I I * line = = £ < £ 1 1 * lmea = = '>') continuous; for (px = line; * px! = '\?'; px + +) if (isupper (* px) 1 1 islower (* px)) tlen + +; } if ((pseq = malloc ((without signal) (tlen + 6))) = = 0). { fprintf (stderr, "% s: archived malloc to get% d bytes for% s \ n", prog, tlen + 6, file); exit (l); } pseq [0] = pseq [l] = pseq [2] = pseq [3] = '\ 0'; ... getseq py = pseq + 4; * len = tlen; rewind (fp); while (fgets (line, 1024, fp)). { if (* line = = '1 1 * line = =' < '| | * line = =' > ') continuous; for (px = line; * px! - \ n '; px + +). { yes (isupper (* px)) * py + + = * px; Also if (islower (* px)) * ?? + + = toupper (* px); if (index ("ATGCU", * (py-l))) natgc + +; } } * py + + = '\ 0'; * py = < 0 '; (empty) fclose (fp); dna = natgc > (tlen / 3); return (pseq + 4); } char * g_calloc (msg, nx, sz) g_calIoc char * msg; / * program, call routine * / int nx, sz; / * number and size of elements * /. { char * px, * calloc (); if ((px = calloc ((without signal) nx, (without signal) sz)) = = 0). { yes (* msg) { íprintf (stderr, "% s: g-callocO failed% s (n =% d, sz =% d) \ n", prog, msg, nx, sz); exit (l); } } return (px); } / * * get final jumps from dxQ or tmp file, set pp [], reset dmax: mainO * / readjmpsO readjmps - - int fd = -l; int siz, iO, il; record i, j, xx; yes (fj). { (empty) fclose (fj); yes ((fd = openQname or_RDONLY, 0)) < 0). { fprmtf (stderr, "% s: can not openO% s \ n", prog, jname); elirninar (l); } } for (i = iO = il = 0, dmaxO = dmax, xx = lenO;; i + +). { while (1). { for Q = dx [dmax] .ijmp; j > = 0 & & dx [dmax] .jp.x [j] > = xx; j-) ... readjmps yes (j <0 && dxfdmax] .out && fj). { (empty) lseek (fd, dxfdmax] .out, 0); (empty) read (fd, (character *) &dx [dmax] .jp, sizeof (structure jmp)); (empty) read (fd, (character *) &dx [dmax]. outside, sizeof (dx [dmax]. outside)); dx [dmax] .ijmp = MAXJMP-1; } also interruption; } if (i> = JMPS { ¾rintf (stderr, "% s: too many spaces in alignment \ n", prog); delete (l);.}. if (j > = 0) { siz = dx [dmax] .jp.n [j]; xx = dx [dmax] .jp.x [j]; dmax + = siz; yes (siz < 0) { / * space in second sec * / pp [l] .n [il] = -siz; xx + = siz; / * id = xx - y + lenl - 1 * / pp [l] .x [il] = xx - dmax + lenl - 1; gapy + +; ngapy - siz; P when doing final spaces * / siz = (-siz <MAXGAP] | final spacesf)? siz: MAXGAP; il ++;.}. Also if (siz> 0) { / * space in first sec * / pp [0] .n [i0] = siz; pp [0] .x [i0] = xx; gapx + +; ngapx + = siz; ignore MAXGAP when doing final spaces * / siz = (siz <MAXGAP 11 final spaces)? siz: MAXGAP; i0 + +; } } also interruption; } / * invert the order of the jumps * / for (j = 0, i0 ~; j < Í0; j + +, Í0-). { i = pp [0] .n [j]; pp [0] .nD] = pp [0] .n [i0]; pp [0] .n [i0] = i; i = Pp [0] .x [j]; pp [0] .xD] = pp [0] .x [iO]; pp [0] .x [iO] = i; } for (j = 0, il ~; j < il; j + +, il ~). { i = pp [l] .n [j]; pp [l] .n [j] = pp [l] .n [il]; pp [l] .n [il] = i; i = pp [l] .x [j]; pp [l] .xtj] = pp [l] .x [il]; pp [l] .x [il] = i; } if (fd > = 0) (empty) closes (fd); yes (fj). { (empty) without linkname); - - outside = 0; } } / * * write a full structure jmp outside the prev (if any): nw () * / writejmps (ix) writejmps int ix; . { char * mktempO; yes (! fj) { yes (mktempQname) < 0). { fprintf (stderr, "% s: can not mktempO% s \ n", prog, jname); eliminate (l); } if ((fj = fopen (jname, "w")) = = 0). { fprintf (stderr, "% s: can not type% s \ n", prog, jname); exit (l); } } (empty) f rite ((character *) & dx [ix] .jp, sizeof (struct jmp), 1, fj); (empty) fwrite ((char *) &dx [ix] .out, sizeof (dx [ix]. out), 1, fj); Table 2 TAT xxxxxxxxxxxxxxx (Length = 15 amino acids) Comparison Protein XXXXXYYYYYYY (Length = 12 amino acids) % amino acid sequence identity = (the number of amino acid residues identically coupled between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of amino acid residues of the TAT polypeptide) = divided by 15 = 33.3% Table 3 TAT xxxxxxxxxx (Length = 10 arránoácidos) Comparison Protein XXXXXYYYYYYZZYZ (Length = 15 amino acids) % amino acid sequence identity = (the number of amino acid residues identically coupled between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of amino acid residues of the TAT polypeptide) = divided by 10 = 50% - - Table 4 TAT-DNA NNNNNNNNNNNNNN (Length = 14 nucleotides) Comparison DNA NNN NNLLLLLLLLLL (Length = 16 nucleotides) % nucleic acid sequence identity = (the number of nucleotides identically coupled between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the nucleic acid sequence TAT-DNA) = 6 divided by 14 = 42.9% Table 5 TAT-DNA NNNNNNNNNNNN (Length = l 2 nucleotides) Comparison DNA NNNNLLLW (Length = 9 nucleotides) % nucleic acid sequence identity = (the number of nucleotides corresponding identically between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the TAT-nucleic acid DNA sequence) = 4 divided by 12 = 33.3% * II. Compositions and Methods of the Invention A. Anti-TAT Antibodies - - In one embodiment, the present invention provides anti-TAT antibodies that can find their use herein as therapeutic and / or diagnostic agents. Antibodies and enzymes include polyclonal, monoclonal, humanized, bispecific and heteroconjugate antibodies. 1. Polyclonal Antibodies Polyclonal antibodies are preferably cultured in animals by multiple subcutaneous (se) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen (especially when using synthetic peptides) to a protein that is immunogenic in the species to be immunized. For example, the antigen can be conjugated to keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor, using a bifunctional or derivatizing agent, e.g. , maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (via lysine residues), glutaraldehyde, succinic anhydride, S0C12 or R1N = C = NR, wherein R and R1 are different alkyl groups. Animals are immunized against the antigen, immunogenic conjugates or derivatives, by combining, e.g. , 100 μg or 5 μ < 3 of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally in multiple sites. One month later, the animals are boosted with 1/5 to 1/10 of the original amount of the peptide or conjugate in complete Freund's adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later, the animals are bled and the serum is analyzed for antibody titration. The animals are reinforced until the title is established. The conjugates can also be made in a recombinant cell culture as protein fusions. Also, aggregation agents such as alum are suitably used to improve the immune response. 2. Monoclonal Antibodies Monoclonal antibodies can be made using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975) or can be made by recombinant DNA methods (U.S. Patent No. 4), 816, 567). In the hybridoma method, a mouse or other appropriate host animal, such as a hamster, is immunized as described above to produce lymphocytes that produce or are capable of producing antibodies that will bind specifically to the protein used for immunization. Alternatively, the lymphocytes can be immunized in vitro. After immunization, the lymphocytes are isolated and then fused with a myeloma cell line using a suitable fusion agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, ( Goding, Onoclonal Antibodies: Principles and Practice) pp. 59-103 (Academic Press, 1986)). The hybridoma cells thus prepared are seeded and cultured in a suitable culture medium which medium preferably contains one or more substances that inhibit the growth or survival of the original unfused myeloma cells (also referred to as a fusion partner). For example, if the original myeloma cells lack the hypoxanthine guanine phosphoribosyl transferase enzyme (HGPRT or HPRT), the selective culture medium for the hybridomas will typically include hypoxanthine, aminopterin and thymidine (HAT medium), whose substances prevent cell growth. deficient in HGPRT. Preferred fusion partner myeloma cells are those that are efficiently fused, support the stable high-level production of the antibody by means of the cells that produce a selected antibody and are of sense to a selective medium that is selected against the original cells not merged Preferred myeloma cell lines are murine myeloma lines, such as those derived from mouse tumors - - MOPC-21 and MPC-11 available from the Salk Institute Cell Distribution Center, San Diego, California, USA and SP-2 and derivatives e.g., X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, Virginia, USA. Human myeloma and mouse-human heteromyeloma cell lines have also been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133: 3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications, (Techniques of Production and Applications of Monoclonal Antibodies) pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). The culture medium in which the hybridoma cells are grown is analyzed for the production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of the monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as a radioimmunoassay (RIA) or an enzyme-linked immunosorbent assay (ELISA). The binding affinity of the monoclonal antibody can be determined, for example, by the Scatchard analysis described in Munson et al., Anal. Biochem. , 107: 220 (1980). Once the hybridoma cells that produce antibodies of the specificity, affinity and / or desired activity are identified, the clones can be subcloned by limiting the dilution procedures and cultured by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, (Goding, Monoclonal Antibodies : Principles and Practice) page 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM medium or PMI-1640. In addition, the ibridoma cells can be cultured in vivo as ascites tumors in an animal e.g. , by injection i.p. of the cells in mice. The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, the ascites fluid or the serum by conventional methods of antibody purification such as, for example, affinity chromatography (eg, using protein A or protein G-Sepharose). ) or ion exchange chromatography, hydroxylapatite chromatography, gel electrophoresis, dialysis, etc. The DNA encoding the monoclonal antibodies is easily isolated and sequenced using conventional procedures (e.g., using oligonucleotide probes that are capable of specifically binding to genes encoding the heavy and light chains of murine antibodies). Hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA can be placed within expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovarian cells (CHO) or myeloma cells that another way they do not produce antibody protein, to obtain the synthesis of the monoclonal antibodies in the recombinant host cells. Review articles about recombinant expression in DNA bacteria encoding the antibody include Skerra et al., Curr. Opinion in Immunol. , 5: 256-262 (1993) and Plückthun, Immunol. Revs. 130: 151-188 (1992). In a further embodiment, monoclonal antibodies or antibody fragments can be isolated from antibody bacteriophage libraries generated using the techniques described in McCafferty et al., Nature, 348: 552-554 (1990). Clackson et al., Nature, 352: 624-628 (1991) and Marks et al., J. Mol. Biol. , 222: 581-597 (1991) which describe the isolation of murine and human antibodies, respectively, using libraries of bacteriophages. Subsequent publications describe the production of high affinity human antibodies (nM range) by chain redistribution (Marks et al., Bio / Technology, 10: 779-783 (1992)), as well as combinatorial infection and in vivo recombination as strategy to build very large bacteriophage libraries (aterhouse et al., Muc. Acids Res., 21: 2265-2266 (1993)). Thus, these techniques are viable alternatives to the traditional techniques of the monoclonal antibody ibidome for the isolation of monoclonal antibodies. The DNA encoding the antibody can be modified to produce chimeric or fusion antibody polypeptides, for example, by substituting the human heavy chain and light chain constant sequences (CH and CL) for the homologous murine sequences (US Patent No. 4,816,567; and Morrison, et al., Proc. Nati Acad. Sel. USA 81: 6851 (1984)) or by fusing the immunoglobulin coding sequence with all or part of the coding sequence for a non-immunoglobulin polypeptide (heterologous polypeptide). The non-immunoglobulin polypeptide sequences can be substituted for the constant domains of an antibody or substituted for the variable domains of an antigen combining site of an antibody to create a bivalent chimeric antibody comprising an antigen combining site having specificity for an antigen and another antigen combining site that has specificity for a different antigen. 3. Human and Humanized Antibodies The anti-TAT antibodies of the invention may further comprise humanized antibodies or human-antibodies. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab ', F (ab') 2 or other antibody subsequences. of antigen binding) containing the minimal sequence derived from the non-human immunoglobulin. Humanized antibodies include human immunoglobulins (receptor antibody) in which residues from a region of complementarity determination (CD) of the receptor are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit that have the specificity, the affinity and the desired capacity. In some cases, the residues of the Fv structure of the human immunoglobulin are replaced by the corresponding non-human residues. Humanized antibodies may also comprise residues that are not found in the recipient antibody or in the imported CDR 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 FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally will comprise at least a portion of an immunoglobulin constant region (Fe), typically that of a human immunoglobulin [Jones et al., Nature, 321: 522-525 (1986); Riechmann et al., Nature, 332: 323-329 (1988); and Presta, Curr. Op. 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 source that is non-human. 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 collaborators [Jones et al., Nature, 321: 522-525 (1986); Riechman et al., Nature, 332: 323-327 (1988); Verhoeyen et al., Science, 239: 1534-1536 (1988)], substituting the sequences of rodent CDRs or CDRs 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 of 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 residues of analogous sites in rodent antibodies. The choice of the human variable domains, both light and heavy, to be used to prepare the humanized antibodies is very important to reduce the antigenicity and the response ???? (human anti-mouse antibody) when the antibody is proposed for human therapeutic use. According to the so-called "optimal adaptation" method, the variable domain sequence of a rodent antibody is sorted against the entire library of known human variable domain sequences. The sequence of the human V domain that is closest to that of the rodent and the region of human structure (FR) within it accepted for the humanized antibody is identified (Sims et al., J. Immunol, 151: 2296 (1993); Chothia et al., J. Mol. Biol. 196: 901 (1987)). Another method uses a particular region of structure derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same structure can be used for several different humanized antibodies (Cárter et al., Proc Nati, Acad Sci USA, 89: 4285 (1992), Presta et al., J. Immunol. 151: 2623 (1993)). It is also important that the antibodies are humanized with retention of high binding affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, the humanized antibodies are prepared by a process of analysis of the native sequences and various conceptual humanized products using three-dimensional models of the humanized and native sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available that illustrate and display probable three-dimensional conformational structures of selected immunoglobulin candidate sequences. The inspection of these images allows the analysis of the probable role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of the residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this manner, the FR residues can be selected and combined from the receptor and import sequences so that the desired antibody characteristic is achieved, such as the increased affinity for the target antigen (s). In general, hypervariable region residues are directly and more substantially involved in influencing antigen binding. Various forms of a humanized anti-TAT antibody are contemplated. For example, the humanized antibody - - it can be an antibody fragment, such as a Fab, which is optionally conjugated to one or more cytotoxic agents in order to generate an immunoconjugate. Alternatively, the humanized antibody can be an intact antibody, such as an intact IgGl antibody. Human antibodies can be generated as an alternative for humanization. For example, it is now possible to produce transgenic animals (e. < and, mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that homozygous removal of the antibody heavy region binding region (JH) gene in chimeric and germline mutant mice results in complete inhibition of endogenous antibody production. The transfer of the ordering of the germline human immunoglobulin gene to such germline mutant mice will result in the production of human antibodies when testing the antigen. See, e.g., Jakobovits et al., Proc. Nati Acad. Sei USA, 90: 2551 (1993); Jacobovits et al., Nature, 362: 255-258 (1993); Bruggemann et al. Year in Immuno, 7:33 (1993); US Patents Nos. 5,545,806, 5,569,825, 5,591,669 (all from GenPharm); 5,545,807; and O 97/17852. Alternatively, phage imaging technology (McCafferty et al., Nature, 348: 552-553 [1990]) can be used to produce human antibodies and antibody fragments in vitro, from the repertoires of the variable domain immunoglobulin gene. (V) of non-immunized donors. According to this technique, the V domain antibody genes are cloned en bloc within a protein gene either larger or smaller than a filamentous bacteriophage, such as M13 or fd and are shown as functional antibody fragments in the surface of the bacteriophage particle. Because the filamentous particle contains a single-stranded DNA copy of the bacteriophage genome, selections based on the functional properties of the antibody also result in the selection of the gene encoding the antibody that exhibits those properties. Thus, the bacteriophage mimics some of the properties of the B cell. The bacteriophage image can be carried out in a variety of formats, reviewed in, eg, Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology, 3: 564-571 (1993). Several sources of V gene segments can be used for the bacteriophage image. Clackson et al., Nature, 352: 624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice. A repertoire of V genes from non-immunized human donors can be constructed and antibodies can be isolated for a diverse array of antigens (including autoantigens) essentially following the techniques described by Marks et al., J. Mol. Biol. 222: 581-597 (1991) or Griffith et al., EMBO J. 12: 725-734 (1993). See, also, US Patents Nos. 5,565,332 and 5,573, 905. As discussed above, human antibodies can also be generated by B cells activated in vitro (see U.S. Patents 5,567,610 and 5,229,275). 4. Antibody fragments In certain circumstances there are advantages in the use of antibody fragments, instead of whole antibodies. The smaller size of the fragments allows for rapid cleansing and may lead to improved access to solid tumors. Several techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived through the proteolytic digestion of intact antibodies (see, eg, Morimoto et al., Journal of Biochemical and Biophysical Methods, 24: 107-117 (1992); and Brennan et al., Science, 229). : 81 (1985)). However, now these fragments can be produced directly by means of recombinant host cells. All Fab antibody fragments, Fv and ScFv can be expressed in and secreted from E coli, thus allowing the easy production of large quantities of these fragments. Antibody fragments can be isolated from the antibody-treated bacteriophage libraries previously treated. Alternatively, the Fab '-SH fragments can be recovered directly from E. coli and chemically coupled to form F (ab') 2 fragments (Carter et al., Bio / Technology 10: 163-167 (1992)). According to another procedure, the F (ab ') 2 fragments can be isolated directly from the culture of the recombinant host cell. Fab and F (ab ') 2 fragments with an increased in vivo half-life comprising receptor binding epitope recovery residues are described in the U.S. Patent. No. 5,869,046. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of choice is a single chain Fv fragment (scFv). See, W093 / 16185; Patent of E.U. No. 5,571,894; and the U.S. Patent. No. 5,587,458. Fv and sFv are the only species with intact combination sites that are free of constant regions; therefore, they are suitable for the reduced non-specific binding during their in vivo use. The sFv fusion proteins can be constructed to produce the fusion of an effector protein at either the amino or the carboxy terminus of a sFv. See, Antibody Engineering, ed. Borrebaeck, supra. The fragment - - of antibody can also be a "linear antibody", e.g., as described for example in the U.S. Patent. 5,641,870. Such linear antibody fragments may be monospecific or bispecific. 5. Bispecific Antibodies Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Bispecific or exemplary antibodies can bind to two different epitopes of a TAT protein as described herein. Other such antibodies can combine a TAT binding site with a binding site for another protein. Alternatively, an anti-TAT arm may be combined with a branch that binds to an activating molecule on a leukocyte such as a T cell receptor molecule. { e.g., CD3) or Fe receptors for IgG (FcyR), such as FcyRI (CD64), FcyRII (CD32) and FcyRIII (CD16), in order to focus and localize the cellular defense mechanisms for the cell expressing TAT. Bispecific antibodies can also be used to localize cytotoxic agents to cells expressing TAT. These antibodies have a TAT binding branch and a branch that binds the cytotoxic agent. { e.g., saporin, anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as antibodies - - of total length or antibody fragments (e.g., bispecific F (ab ') 2 antibodies). O 96/16613 discloses a bispecific anti-ErbB2 / anti-FcyRIII antibody and the US Patent. No. 5,837,234 discloses a bispecific anti-ErbB2 / anti-FcyRI antibody. A bispecific anti-ErbB2 / Fca antibody is shown in WO98 / 02463. The U.S. Patent No. 5,821,337 shows a bispecific anti-ErbB2 / anti-CD3 antibody. Methods for preparing bispecific antibodies are known in the art. The traditional production of full-length bispecific antibodies is based on the coexpression of two pairs of heavy chain-light chain immunoglobulins, where the two chains have different specificities (Millstein et al., Mature, 305: 537-539 (1983)). ). Due to the random selection of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule, which is commonly done by affinity chromatography steps, is rather complicated and the yield of the product is low. Similar procedures are described in WO 93/08829 and in Traunecker et al., E BO J. 10: 3655-3659 (1991). According to a different procedure, the variable domains of antibody with the desired binding specificities (sites combining antibody-antigen) are fused to constant domain immunoglobulin sequences. Preferably, the fusion is with a heavy chain Ig constant domain, comprising at least part of the articulated regions CH2 and CH3. It is preferred to have present the first heavy chain constant region (CH1) containing the necessary site for the light chain linkage, in at least one of the fusions. The DNAs encoding the heavy chain immunoglobulin fusions and, if desired, the light chain immunoglobulin fusions, are inserted into separate expression vectors and co-transfected into a suitable host cell. This provides greater flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments wherein the unequal ratios of the three polypeptide chains used in the construction provide the optimal yield of the desired bispecific antibody. However, it is possible to insert the coding sequences for two or all three polypeptide chains into a single expression vector when the expression of at least two polypeptide chains in equal proportions results in high yields or when the proportions do not affect significantly the performance of the desired chain combination.
- - In a preferred embodiment of this method, the bispecific antibodies are composed of an immunoglobulin hybrid heavy chain with a first binding specificity in one branch and a hybrid pair of heavy chain-light chain immunoglobulin (which provides a second binding specificity) on the other branch. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, since the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for an easy mode of separation. This procedure is described in WO 94/04690. For further details to generate bispecific antibodies see, for example, Suresh et al., Methods in Enzymology 121: 210 (1986). According to another procedure described in the U.S. Patent. No. 5,731,168, the interface between a pair of antibody molecules can be designed to maximize the percentage of heterodimers that are recovered from the recombinant cell culture. The preferred interface comprises at least a portion of the CH3 domain. In this method, one or more small amino acid side chains of the interphase of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan). The compensatory "cavities" of identical or similar size to - - the large lateral chain (s) are created at the interface of the second antibody molecule by replacing the large amino acid side chains with smaller ones. { e.g. , alanine or threonine). This provides a mechanism to increase the performance of the heterodimer over other undesired end products such as homodimers. Bispecific antibodies include crosslinked or "heteroconjugate" antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies, for example, have been proposed to direct cells of the immune system to unwanted cells (US Patent No. 4,676,980) and for the treatment of HIV infection (WO 91/00360, WO 92/200373 and EP 03089 ). Heteroconjugate antibodies can be made using any convenient method of crosslinking. Suitable crosslinking agents are well known in the art and are described in the U.S. Patent. No. 4,676,980, together with a variety of crosslinking techniques. Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science 229: 81 (1985) describes a method wherein intact antibodies are proteolytically cleaved to generate F (ab ') 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent, arsenite. of sodium, to stabilize neighboring dithioles and prevent intermolecular disulfide formation. The generated Fab 'fragments are then converted into thionitrobenzoate derivatives (TNB). One of the Fab '-T B derivatives is then reconverted to Fab'-thiol by reduction with mercaptoethylamine and mixed with an equimolar amount of the other Fab' -TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immunization of enzymes. Recent progress has facilitated the direct recovery of Fab'-SH fragments from E. coli that can be chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med. 175: 217-225 (1992) describes the production of a fully humanized bispecific antibody F (ab ') 2 molecule. Each Fab 'fragment was secreted separately from E. coli and subjected to direct chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as activating the lytic activity of human cytotoxic lymphocytes against human breast tumor targets. Various techniques for making and isolating bispecific antibody fragments direcfrom the recombinant cell culture have also been described. For example, bispecific antibodies were produced using leucine zippers. Kostelny et al., J. Immunol. 148 (5): 1547-1553 (1992). The leucine zipper peptides of the Fos and Jun proteins were ligated to the Fab 'portions of two different antibodies by fusion of the gene. The antibody homodimers were reduced in the articulated region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be used for the production of antibody homodimers. The iabody technology "described by Hollinger et al., Proc. Nati. Acad. Sci. USA 90: 6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody particles.The fragments comprise a VH connected to a VL By means of a linker that is too short to allow coupling between the two domains in the same chain, according to this, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, by which form two antigen binding sites Another strategy has also been reported for making bispecific antibody fragments by using the Fv - - dimers (sFv) single chain. See, Gruber et al.,. Immunol. , 152: 5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147: 60 (1991). 6. Heteroconjugate Antibodies Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently bound antibodies. Such antibodies, for example, have been proposed to direct cells of the immune system to undesired cells [U.S. Pat. No. 4,676,980] and for the treatment of HIV infection [WO 91/00360; WO 92/200373; EP 03089]. It is contemplated that the antibodies can be prepared in vitro using methods known in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or forming a thioether linkage. Examples of reagents suitable for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those described, for example, in the U.S. Patent. No. 4,676,980. 7. Mutantivalent antibodies A multivalent antibody can be internalized (and / or catabolized) faster than a bivalent antibody - by a cell that expresses an antigen to which the antibodies bind. The antibodies of the present invention can be multivalent antibodies (which are different from those of the IgM class) with three or more antigen binding sites (e. < and, tetravalent antibodies), which can be easily produced by recombinant expression of the nucleic acid encoding the polypeptide chains of the antibody. The multivalent antibody may comprise a dimerization domain and three or more antigen binding sites. The preferred dimerization domain comprises (or consists of) an Fe region or an articulated region. In this scenario, the antibody will comprise an Fe region and three or more amino-terminal antigen binding sites to the Fe region. The preferred multivalent antibody herein comprises (or consists of) three to about eight, but preferably four, antigen binding sites. The multivalent antibody comprises at least one polypeptide chain (and preferably two polypeptide chains) wherein the polypeptide chains (s) comprise two or more variable domains. For example, the polypeptide chain (s) can (n) comprise VD1- (XI) n-vD2- (X2) n -Fc, where VD1 is a first variable domain, VD2 is a second variable domain, Fe is a polypeptide chain of a Fe region, XI and X2 represent an amino acid or polypeptide and n is 0 or 1. For example, the polypeptide chain (s) can (n) comprise: VH-CH1-flexible linker -VH-CHl-chain of Fe region; or VH-CH1-VH-CH1-Fe region chain. The multivalent antibody herein further preferably comprises at least two (and preferably four) light chain variable domain polypeptides. The multivalent antibody herein may comprise, for example, from about two to about eight light chain variable domain polypeptides. The light chain variable domain polypeptides contemplated herein comprise a light chain variable domain and optionally, further comprise a CL domain. 8. Design of the Effector Function It may be desirable to modify the antibody of the invention with respect to effector function, e.g. t / in order to improve the antigen-dependent cell-mediated cytotoxicity (ADCC) and / or the complement-dependent cytotoxicity (CDC) of the antibody. This can be achieved by introducing one or more amino acid substitutions in an Fe region of the antibody. Alternatively or additionally, the cysteine residue (s) can be introduced into the Fe region, thereby allowing formation of the disulfide interchain linkage in this region. The homodimeric antibody thus generated can have an increased internalization capacity and / or increased cell death. mediated by complement and antibody-dependent cellular cytotoxicity (ADCC). See, Carón et al., J. Exp. ed. 176: 1191-1195 (1992) and Shopes, B. J. Immunol. 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolf et al., Cancer Research 53: 2560-2565 (1993). Alternatively, an antibody having dual Fe regions can be designed and whereby it can have improved complement lysis and ADCC capabilities. See, Stevenson et al., Anti-Cancer Drug Design 3: 219-230 (1989). To increase the half-life of the antibody serum, a binding epitope of the recovery receptor must be incorporated within the antibody (especially an antibody fragment) for example as described in the U.S. Patent. 5,739,277. As used herein, the term "recovery receptor binding epitope" refers to an epitope of the Fe region of an IgG molecule (eg, IgG1 # IgG2, IgG3 or IgG4) that is responsible for the increase in life mean in vivo serum of the IgG molecule. 9. Immunoconjugates The invention also relates to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, a growth inhibitory agent, a toxin. { e.g. , a - - enzymatically active toxin of bacterial, fungal, plant or animal origin or its fragments) or a radioactive isotope (i.e., a radioconjugate). The chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that may be used include the diphtheria A chain, active fragments without diphtheria toxin binding, the A chain of exotoxin (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, A chain of modecina, alpha-sarcina, proteins of Aleurites fordii, proteins of diantina, proteins of Phytolaca americana (PAPI, PAPII, PAP-S), inhibitor of momordica charantia, curcina, crotina, inhibitor of sapaonaria officinalis, gelonina, mitogelina, restrictocin, phenomycin, enomycin and the trichothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212Bi, 1311, 131In, 90Y, and 18eRe. The conjugates of the antibody and the cxtotoxic agent are made1 using a variety of protein-binding bifunctional agents such as N-succinimidyl-3- (2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters ( as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as - - glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazoniobenzoyl) -ethylenediamine), diisocyanates (such as tolieno 2,6-diisocyanate) and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). The l-isothiocyanatobenzyl-3-methyldiethylene triaminopentaacetic acid (MX-DTPA) labeled with carbon 14 is a chelating agent for the conjugation of the radionucleotide to the antibody. See, WO94 / 11026. Also contemplated herein are conjugates of an antibody and one or more small molecule toxins, such as a calicheamicin, maytansinoids, a trichothene and CC1065 and the derivatives of these toxins having toxin activity. Maytansin and maytansinoids In a preferred embodiment, an anti-TAT antibody (full-length or fragments) of the invention is conjugated to one or more maytansinoid molecules. Maytansinoids are mitototic inhibitors that act by the polymerization of tubulin. Maytansin was first isolated from the East African bush Maytenus serrata (US Patent No. 3,896,111). Subsequently it was discovered that certain microbes also - - they produce maytansinoids such as maytansinol and maytansinol C-3 esters (U.S. Patent No. 4,151,042). Synthetic maytansinol and its derivatives and analogs are described, for example, in US Patents. Us. 4,137,230; 4,248,870; 4, 256, 746; 4,260,608; 4,265, 814; 4,294, 757; 4,307, 016; 4,308,268 4,308,269; 4,309,428; 4,313,946; 4,315,929, 4,317, 821 4,322,348; 4, 331, 598; 4,361, 650; 4, 364, 866; 4,424,219; 4,450,254; 4,362,663; Y 4,371,533, the descriptions of which are hereby expressly incorporated by reference. Maytansinoid-antibody conjugates In an attempt to improve their therapeutic index, maytansin and maytansinoids have been conjugated to antibodies that bind specifically to tumor cell antigens. Immunoconjugates containing maytansinoids and their therapeutic use are described, for example, in US Patents. Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl, the disclosures of which are hereby expressly incorporated by reference. Liu et al., Proc. Nati Acad. Sci. USA 93: 8618-8623 (1996) described immunoconjugates comprising a maytansinoid designated DM1 linked to the monoclonal antibody C242 directed against human colorectal cancer. The conjugate was found to be highly cytotoxic toward cultured colon cancer cells and showed antitumor activity in a tumor growth analysis in vivo. Chari et al., Cancer Research 52: 127-131 (1992) describes immunoconjugates in which a maytansinoid was conjugated through a disulfide linker to murine antibody A7 that binds an antigen in human colon cancer cell lines or to another antibody murine monoclonal TA.l that binds the oncogene HER-2 / neu. The cytotoxicity of the TA.1-maytansinoid conjugate was tested in vitro in the human breast cancer cell line SK-BR-3, which expresses 3 x 105 HER-2 surface antigens per cell. The drug conjugate achieved a degree of cytotoxicity similar to the free maytansinoid drug, which could be increased by increasing the number of maytansinoid molecules per antibody molecule. The A7-maytansinoid conjugate showed low systemic cytotoxicity in mice. Antibody conjugates of the anti-TAT-maytansinoid polypeptide (immunoconjugates) The anti-TAT-maytansinoid antibody conjugates are prepared by chemically binding an anti-TAT antibody to a maytansinoid molecule without significantly decreasing the biological activity of either the antibody or the antibody molecule. maytansinoid An average of 3-4 conjugated maytansinoid molecules per antibody molecule has shown efficacy in improving the cytotoxicity of the target cells without adversely affecting the function or solubility of the antibody, although a toxin / antibody molecule would even be expected to improve cytotoxicity over the use of the naked antibody. Maytansinoids are well known in the art and can be synthesized by known techniques or isolated from natural sources. Suitable maytansinoids are described, for example, in the U.S. Patent. No. 5,208,020 and in other patent and non-patent publications referred to hereinbefore. Preferred nsinoids are maytansinol and modified maytansinol analogs in the aromatic ring or in other positions of the maytansinol molecule, such as several maytansinol esters. There are many linking groups known in the art for making antibody-maytansinoid conjugates, including, for example, those described in the U.S. Patent. No. 5,208,020 or EP Patent 0 425 235 Bl and Chari et al., Cancer Research 52: 127-131 (1992). The linking groups include disulfide groups, thioether groups, acid-labile groups, photolabile groups, peptidase-labile groups or stearase-labile groups, as described in the above-identified patents, with disulfide and thioether groups being preferred. The antibody and maytansinoid conjugates can be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3- (2-pyridyldithio) propionate (SPDP)., succinimidyl-4- (N-maleimidomethyl) cyclohexane-1-carboxylate, iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde) , bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazonium benzoyl) -ethylenediamine, diisocyanates (such as toluene 2,6-diisocyanate) and bis-active fluorine (such as 1,5-difluoro-2,4-dinitrobenzene.) Particularly preferred coupling agents include N-succinimidyl-3- (2-pyridyldithio) propionate (SPDP) (Carlsson et al., Biochem J. 173: 723-737- [1978]) and N-succinimidyl-4- (2-pyridylthio) pentanoate (SPP) to provide a disulfide bond The linker can be attached to the maytansinoid molecule in various positions, Depending on the type of link, for example, an ester bond can be formed the reaction with a hydroxyl group using conventional coupling techniques. The reaction can occur at the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with a hydroxyl group and the C-20 position having a hydroxyl group. In a preferred embodiment, the bond is formed at the C-3 position of maytansinol or a maytansinol analogue.
- - Calicheamycin Another immunoconjugate of interest comprises an anti-TAT antibody conjugated to one or more calicheamicin molecules. The calicheamicin family of antibiotics is capable of producing double-stranded DNA breaks at sub-picomolar concentrations. For the preparation of conjugates of the calicheamicin family, see Patents of E.U. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, 5,877,296 (all from American Cyanamid Company). Structural calicheamicin analogs that can be used include, but are not limited to, 1, a.21. as1, N-acetyl-yx1, PSAG, and t (Hinman et al., Cancer Research 53: 3336-3342 (1993), Lode et al., Cancer Research 58: 2925-2928 (1998) and US patents previously. mentioned by American Cyanamid). Another anti-tumor drug to which the antibody can be conjugated is QFA which is an antifolate. Both calicheamicin and QFA have intracellular sites of action and do not readily cross the plasma membrane. Consequently, the cellular uptake of these agents through internalization mediated by the antibody greatly improves their cytotoxic effects. Other cytotoxic agents Other antitumor agents that can be conjugated to the anti-TAT antibodies of the invention include BC U, streptozoicin, vincristine and 5-fluorouracil, the family of agents known collectively as LL-E33288 complex described in the US Pat. 5,053,394, 5,770,710, as well as the esperamycins (U.S. patent 5,877,296). The enzymatically active toxins and their fragments that can be used include the diphtheria A chain, non-binding active fragments of the diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, A chain modeccina, alpha-sarcina, proteins of Aleurites fordii, diantine proteins, proteins of Phytolaca americana (PAPI, PAPII and PAP-S), inhibitor of momordica charantia, curcina, crotina, inhibitor of sapaonaria officinalis, gelonin, mitogeline, restrictocin, fenomycin , enomycin and the trichothecenes. See, for example, O 93/21232 published October 28, 1993. The present invention further contemplates an immunoconjugate formed between an antibody and a compound with nucleolytic activity (e.g., a ribonuclease or a DNA endonuclease such as deoxyribonuclease, DNase). For the selective destruction of the tumor, the antibody can comprise a highly radioactive atom. A variety of radioactive isotopes are available for the production of radiocontracted anti-TAT antibodies. Examples include At211, I131, I125 Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu. When the conjugate is used for the diagnosis, it may comprise a radioactive atom for scintigraphic studies, for example tc99ltl or I123 or a rotation mark for the representation of nuclear magnetic resonance (NMR) images (also known as image representation by magnetic resonance, mri), such as again iodine-123 iodine-131, indium-111, fluorine-19, carbon-13, nitrogen-15 oxygen-17, gadolinium, manganese or iron. The radius or other marks can be incorporated into the conjugate in known ways. For example, the peptide can be biosynthesized or synthesized by chemical synthesis of amino acid using suitable amino acid precursors that involve, for example, fluorine-19 instead of hydrogen. Marks such as tc99m or I123, Re186, Re188 and In111 can be linked through a cysteine residue in the peptide. Yttrium-90 can bind through a lysine residue. The IODOGEN method (Fraker et al., (1978) Biochem. Biophys. Res. Commun. 80: 49-57 can be used to incorporate iodine-123. "Monoclonal Antibodies in Immunoscintigraphy" ("Monoclonal Antibodies in Immunoscintigraphy") ( Chatal, CRC Press 1989) describes other methods in detail: The conjugates of the antibody and the cytotoxic agent can be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3- (2-pyridyldithio) -propionate ( SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-1-carboxylate, iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde) , bis-azido compounds (such as bis (p-azidobenzoyl) -hexanediamine), bis-diazonium derivatives (such as bis- (p-diazonium benzoyl) -ethylenediamine, diisocyanates (such as toluene 2,6-diisocyanate) and bis-active fluorine (such as 1,5-difluoro-2,4-dinitrobenzene) For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987). isothiocyanatobenzyl-3-methyldiethylene triaminopentaacetic acid (MX-DTPA) labeled with carbon-14 is an exemplary chelating agent for the conjugation of the radionucleotide to the antibody, see, WO94 / 11026. The linker can be a "binder uncoupling" which facilitates the release of the cytotoxic drug a in the cell. For example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Research 52: 127-131 (1992)) may be used.; Patent of E.U. No. 5,208,020). Alternatively, a fusion protein comprising the anti-TAT antibody and the cytotoxic agent, .g., Can be made by recombinant techniques or peptide synthesis. The length of the DNA may comprise the respective regions coding for the two portions of the conjugate either adjacent to each other or separated by a region encoding a binding peptide that does not destroy the desired properties of the conjugate. In yet another embodiment, the antibody can be conjugated to a "receptor" (such as streptavidin) for use in the pre-tumor target where the antibody receptor conjugate is administered to the patient, followed by removal in the circulation of the conjugate. not linked using a cleaning agent and then the administration of a "ligand". { e.g.r avidin) which is conjugated to a cytotoxic agent (e.g., a radionucleotide). 10. Immunoliposomes The anti-TAT antibodies described herein may also be formulated as immunoliposomes. A "liposome" is a small vesicle composed of several types of lipids, phospholipids and / or surfactant that is useful for the delivery of a drug to a mammal. The liposome components are commonly arranged in a bilayer formation, similar to the lipid arrangement of the biological membranes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al. , Proc. Nati Acad. Scí. USES - - 82: 3688 (1985); Hwang et al., Proc. Nati Acad. Sci. USA 77: 4030 (1980); the Pats. of E.U. Nos. 4,485,045 and 4,544,545; and W097 / 38731 published October 23, 1997. Liposomes with improved circulation time are described in the U.S. Patent. No. 5,013,556. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and phosphatidylethanolamine derived from PEG (PEG-PE). The liposomes are extruded through filters of a defined pore size to produce liposomes with the desired diameter. The Fab 'fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem. 257: 286-288 (1982) through a disulfide exchange reaction. Optionally, a chemotherapeutic agent is contained within the liposome. See, Gabizon et al., J. National Cancer Inst. 81 (19): 1484 (1989). B. TAT Link Oligopeptides The TAT linkage oligopeptides of the present invention are oligopeptides that are linked, preferably specifically to a TAT polypeptide as described herein. The TAT binding oligopeptides can be chemically synthesized using known methodology for the synthesis of oligopeptides or can be prepared and purified using recombinant technology. TAT-binding oligopeptides are commonly at least about 5 amino acids in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1S, 17, 18, 19, 20 , 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45 , 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70 , 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82. 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95 , 96, 97, 98, 99 or 100 amino acids in length or more, wherein such oligopeptides are capable of binding, preferably specifically, to a TAT polypeptide as described herein. TAT binding oligopeptides can be identified without undue experimentation using well known techniques. In this regard, it is noted that techniques for the selection of oligopeptide libraries for oligopeptides that are capable of specifically binding to a polypeptide target are well known in the art (see, eg, U.S. Patent Nos. 5,556,762, 5,750,373, 4,708,871, 4,833,092, 5,223,409, 5,403,484, 5,571,689, 5,663,143; PCT Publications Nos. O 84/03506 and WO 84/03564; Geysen et al., Proc. Nati, Acad. Sci. USA., 81: 3998-4002 (1984); Geysen et al., Proc. Nati, Acad. Sci. USA., 82: 178-182 (1985); Geysen et al., in Synthetic Peptides as Antigens, (Peptides - - Synthetics as Antigens) 130-149 (1986); Geysen et al., J. Immunol. Meth. , 102: 259-274 (1987); Schoofs et al., J. Immunol., 140: 611-616 (1988); Cwirla S.E. et al., (1990) Proc. Nati Acad. Sci. USA., 87: 62TB; Loman H.B. et al., (1991) Biochemistry, 30: 10832; Clackson T. et al., (1991) Nature, 352: 624; Marks J.D. , et al., (1991), J. Mol. Biol. 222: 581; Kang A.S. et al., (1991) Proc. Nati Acad. Sci. USA., 88: 8363 and Smith G.P. (1991) Current Opin. Biotechnol., 2: 668). In this regard, the bacteriophage (phage) image is a well-known technique that allows large oligopeptide libraries to be selected to identify the member (s) of those libraries that are capable of specifically binding to a polypeptide target. Phage imaging is a technique by which variant polypeptides are deployed as fusion proteins for the coat protein on the surface of the bacteriophage particles (Scott, J. and Smith, G.P. (1990) Science 249: 386). The utility of the phage image resides in the fact that large libraries of selectively randomized protein variants (or randomly cloned cDNAs) can be quickly and efficiently sorted for those sequences that bind to a target molecule with high affinity. The image of the peptide libraries (Cwirla S.E. et al. (1990), Proc. Nati. Acad. Sel.
USA, 87: 6378) or protein (Lowman, HB et al., (1991) Biochemistry, 30: 10832; Clackson T. et al., (1991) Nature 352: 624; arks JD et al., (1991) , J. Mol. Biol., 222: 581; anf AS et al., (1991) Proc. Nati, Acad. Sci. USA, 88: 8363) phage have been used to select millions of polypeptides or oligopeptides for some with specific binding properties (Smith GP (1991) Current Opin, Biotechnol., 2: 668). Classification of phage libraries of random mutants requires a strategy for constructing and propagating a large number of variants, a procedure for affinity purification using the target receptor and a means for evaluating the results of link enrichments. US Patents Us. ,223,409, 5,403,484, 5,571,689 and 5,663,143. Although most phage imaging methods have used a filamentous phage, lambdoid phage imaging systems (O 95/34683; US 5,627,024), T4 phage imaging systems (Ren, ZJ et al., (1998)) are also known. Gene 215: 439; Zhu Z. (1997) CAN 33: 534; Jiang J. et al., (1997) can 128: 44380; Ren ZJ. Et al., (1997) CAN 127: 215644; Ren ZJ. ( 1996) Protein Sci. 5: 1833; Efimov, VP et al., (1995) Virus Genes 10: 173) and T7 phage imaging systems (Smith GP and Scott, JK (1993) ethods in Enzymology, 217, 228_257; 5,766,905). Many other improvements have now been developed and - - variants of the basic concept of phage imaging. These improvements increase the ability of the imaging systems to select peptide libraries to bind to selected target molecules and to display functional proteins with the potential to select these proteins for the desired properties. Combinatorial reaction devices have been developed for phage imaging reactions (WO 98/14277) and phage imaging libraries have been used to analyze and control bimolecular interactions (WO 98/20169; WO 98/20159) and the properties of peptides forced coils (WO 98/20036). WO 97/35196 describes a method for isolating an affinity ligand in which a phage imaging library is in contact with a solution in which the ligand will bind to a target molecule and a second solution in which the ligand of Affinity will not bind to the target molecule, to selectively isolate the binding ligands. WO 97/46251 describes a method for the bio-panning of a random phage-image library with a purified affinity antibody and then the isolation of the binding phage, followed by a micro-panning process using micro-plate wells for isolate the high binding affinity phage. The use of Staphylococcus aureus protein A as an affinity tag has also been reported (Li et al., (1998) Mol Biotech., 9: 187). The WO - - 97/47314 describes the use of substrate subtraction libraries to distinguish the specificities of the enzyme using a combinatorial library that can be a phage display library. A method for selecting enzymes suitable for use in detergents using phage imaging is described in O 97/09446. Additional methods for selecting specific binding proteins are described in US Patents. Nos. 5,498,538, 5,432,018 and WO 98/15833. Methods for generating peptide libraries and selecting these libraries are also described in US Patents. Nos. 5,723,286, 5,432,018, 5,580,717, 5,427,908, 5,498,530, 5,770,434, 5,734,018, 5,698,426, 5,763,192 and 5,723,323. C. Organic TAT Linking Molecules Organic TAT linkage molecules are organic molecules different from oligopeptides or antibodies as defined herein that are linked, preferably specifically, to a TAT polypeptide as described herein. Organic TAT binding molecules can be identified and synthesized chemically using known methodology (see, e.g., PCT Publications Nos. WO00 / 00823 and WOOO / 39585). Organic TAT binding molecules are commonly less than about 2000 daltons in size, alternatively less than about 1500, 750, - 8 - 500, 250 or 200 daltones in size, wherein such organic molecules that are capable of binding, preferably specifically, to a TAT polypeptide as described herein can be identified without undue experimentation using well-known techniques. In this regard, it is noted that the techniques for selecting libraries of organic molecules for molecules capable of binding to a polypeptide target are well known in the art (see, PCT Publication Nos. O00 / 00823 and WO00 / 39585). TAT link can be for example, aldehydes, ketones oximes, hydrazones, semicarbazones, carbazides, primary amines, secondary amines, tertiary amines, N-substituted hydrazines, hydrazides, alcohols, ethers, thiols, thioethers, disulfides, carboxylic acids, esters, amides , ureas, carbamates, carbonates, ketals, thioketals, acetals, thioacetals, aryl halides, aryl sulfonates, alkyl halides, alkyl sulfonates, aromatics, heterocyclic compounds, anilines, alkenes, alkynes, diols, aminoalcohols oxazolidines oxazolines, thiazolidines , thiazolines, enamines, sulfonamides, epoxides, aziridines, isocyanates, sulfonyl chlorides, diazo compounds, chlorides, acid or the like. D. Selection of TAT-binding Oligopeptide Anti-TAT Antibodies and Organic TAT Link Molecules with the Desired Properties - - Techniques for generating oligopeptide antibodies and organic molecules that bind to TAT polypeptides have been described above. In addition, oligopeptide antibodies or other organic molecules with certain biological characteristics may be selected, as desired. The growth inhibitory effects of an anti-TAT oligopeptide antibody or other organic molecule of the invention can be assessed by methods known in the art, e.g., using cells that express a TAT polypeptide either endogenously or followed by transfection with the TAT gene. For example, appropriate tumor cell lines and TAT-transfected cells can be treated with an anti-TAT oligopeptide monoclonal antibody, or other organic molecule of the invention at several concentrations for a few days (eg, 2-7 days) and stained with purple crystal or TT or analyzed by some other colorimetric analysis. Another method to measure proliferation would be to compare the 3H thymidine uptake by the treated cells in the presence or absence of an anti-TAT antibody, a TAT-binding oligopeptide or the TAT organic binding molecule of the invention. After treatment the cells are harvested and the amount of radioactivity incorporated within the DNA is quantified in a scintillation counter. Appropriate positive controls include the treatment of a selected cell line with a growth inhibitory antibody known to inhibit the growth of that cell line. Inhibition of the growth of tumor cells in vivo can be determined in various ways known in the art. Preferably, the tumor cell is the one that overexpresses a TAT polypeptide. Preferably, the anti-TAT antibody, the TAT-binding oligopeptide or the TAT-binding organic molecule will inhibit the cell proliferation of a tumor cell that expresses TAT in vi tro or in vivo by approximately 25-100% compared to the non-tumor cell. treated, more preferably, by about 30-100% and even more preferably by about 50-100% or 70-100%, in one embodiment, at an antibody concentration of about 0.5 to 30 μg / ml. Growth inhibition can be measured at an antibody concentration of about 0.5 to 30 μg / ml or about 0.5 to 200 nM in the cell culture, where growth inhibition is determined 1-10 days after exposure of the cells tumor to the antibody. The antibody is growth inhibitory in vivo if administration of the anti-TAT antibody at about 1 g / kg to about 100 mg / kg body weight results in reduction in tumor size or reduction of cell proliferation tumor within about 5 days to 3 months from the first - 91 - administration of the antibody, preferably within approximately 5 to 30 days. To select an anti-TAT antibody, a TAT-binding oligopeptide or an organic TAT binding molecule that induces cell death, the loss of membrane integrity can be assessed as indicated, eg, by the absorption of propidium iodide (PI). ) trypan blue or absorption of 7AAD in relation to the control. An analysis of PI absorption can be carried out in the absence of complement cells and immune effectors. Tumor cells expressing the TAT polypeptide are incubated with a medium alone or with a medium containing the appropriate anti-TAT antibody (e.g., at approximately 10 μg / ml), the TAT-binding oligopeptide or the TAT-binding organic molecule. The cells are incubated for a period of 3 days. After each treatment, the cells are washed and aliquoted into 12 x 75 tubes covered with a 35 mm filter (1 ml per tube, 3 tubes per treatment group) for removal of cell clumps. The tubes then receive PI (10 g / ml). Samples can be analyzed using a FACSCAN® flow cytometer and the CellQuest FACSCONVERT® software (Becton Dickinson). Those TAT-binding oligopeptide anti-TAT antibodies or TAT-binding organic molecules that induce statistically significant levels of cell death as determined by PI-absorption can be selected as TAT-binding oligopeptide TAT antibodies or TAT-binding organic molecules that induce cell death To select oligopeptide antibodies or other organic molecules that bind to an epitope on a TAT polypeptide linked by an antibody of interest, a routine cross-blocking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor, may be performed. Laboratory, Ed Harlow and David Lane (1988). This assay can be used to determine whether an oligopeptide antibody or test organic molecule binds to the same site or epitope as a known anti-TAT antibody. Alternatively or additionally, a representation of the epitope can be carried out by methods known in the art. For example, the antibody sequence can be mutagenized such as by alanine screening to identify contact residues. The mutant antibody is initially tested to bind to the polyclonal antibody to ensure proper folding. In a different method, the peptides corresponding to different regions of a TAT polypeptide can be used in competition assays with the test antibodies or with a test antibody and an antibody with a characterized or known epitope. E. Enzyme-mediated Prodroga Therapy - - Antibody-dependent (ADEPT) The antibodies of the present invention can also be used in ADEPT by conjugating the antibody to a prodrug activation enzyme that converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO81 / 01145) in an active anti-cancer drug. See, for example, O 88/07378 and the U.S. Patent. No. 4,975,278. The enzyme component of the immunoconjugate useful for ADEPT includes any enzyme capable of acting in a prodrug in such a way as to convert it to its more active cytotoxic form. Enzymes that are useful in the method of this invention include, but are not limited to, alkaline phosphatase useful for converting phosphate-containing prodrugs into free drugs; arylsulfatase useful for converting prodrugs containing sulphates into free drugs; cytosine deaminase useful for converting non-toxic 5-fluorocytosine into the anti-cancer drug, 5-fluorouracil; proteases, such as protease serratia, thermolysin subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), which are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, useful for converting prodrugs containing D-amino acid substituents; carboxydrelate cleavage enzymes such as β-galactosidase and neuraminidase useful for converting glycosylated prodrugs - 94 - into free drugs; β-lactamase useful for converting drugs derived with β-lactams into free drugs; and penicillin amidases, such as penicillin V amidase or penicillin G amidase, useful for converting drugs derived in their amine nitrogens with phenoxyacetyl or phenylacetyl groups, respectively, into free drugs. Alternatively, antibodies with enzymatic activity, also known in the art as "abzymes", can be used to convert the prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 328: 457-458 (1987)). Antibody-to-zyme conjugates can be prepared as described herein to deliver the abzyme to a population of tumor cells. The enzymes of this invention can be covalently linked to the anti-TAT antibodies by techniques well known in the art such as the use of the crosslinking eterobifunctional reagents discussed above. Alternatively, fusion proteins comprising at least the antigen binding region of an antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques well known in the art (see , eg, Neuberger et al., Nature 312: 604-608 (1984) F. Total Length TAT Polypeptides - - The present invention also provides newly identified and isolated nucleotide sequences encoding the polypeptides referred to in the present application as TAT polypeptides. In particular, the cDNAs (partial and full length) encoding various TAT polypeptides have been identified and isolated, as described in more detail below in the Examples. As described in the Examples below, several cDNA clones have been deposited with the ATCC. The current nucleotide sequences of those clones can be readily determined by the skilled artisan by sequencing the deposited clone using routine methods in the art. The predicate amino acid sequence can be determined from the nucleotide sequence using routine experience. For the TAT polypeptides and coding nucleic acids described herein, in some cases, the Applicants have identified what is considered to be the best identifiable reading frame with the sequence information available at the moment. G. Variants of Anti-TAT Antibody and Polypeptide TAT In addition to the anti-TAT antibodies and the full-length TAT polypeptides of the native sequence described herein, it is contemplated that variants of the anti-TAT antibody and the TAT polypeptide can be prepared. The anti-TAT antibody and TAT polypeptide variants can be prepared by introducing the appropriate nucleotide changes into the coding DNA and / or by synthesis of the desired antibody or polypeptide. Those skilled in the art will appreciate that amino acid changes can alter the trans-translational processes of the anti-TAT antibody or the TAT polypeptide., such as changing the number or position of the glycosylation sites or altering the anchoring characteristics of the membrane. Variations in the anti-TAT antibodies and the TAT polypeptides described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations published, for example, in the U.S. Patent. No. 5,364,934. The variations may be a substitution, deletion or insertion of one or more codons encoding the antibody or polypeptide that result in a change in the amino acid sequence as compared to the antibody or polypeptide of the native sequence. Optionally, the variation is by substitution of at least one amino acid with any other amino acid in one or more of the anti-TAT antibody or TAT polypeptide domains. The guide for determining which amino acid residue can be inserted, substituted or deleted without adversely affecting the desired activity can be found by comparing the sequence of the anti-TAT antibody or TAT polypeptide with that of known homologous protein molecules and minimizing the number of changes in the amino acid sequence made in regions of high homology. The amino acid substitutions may be the result of the replacement of an amino acid with another amino acid having similar structural and / or chemical properties, such as the replacement of a leucine with a serine, i.e., conservative amino acid replacements. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. The allowed variation can be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and analyzing the resulting variations for the activity exhibited by the full-length or mature native sequence. The fragments of the anti-TAT antibody and the TAT polypeptide are provided herein. Such fragments may be truncated at the N-terminus or C-terminus or may lack internal residues, for example, when compared to a full-length native antibody or protein. Certain fragments lack amino acid residues that are not essential for a desired biological activity of the anti-TAT antibody or the TAT polypeptide. The fragments of the anti-TAT antibody and the TAT polypeptide can be prepared by any of a variety of conventional techniques. The fragments of - - The desired peptide can be synthesized chemically. An alternative method involves generating antibody or polypeptide fragments by enzymatic digestion, eg, treating the protein with a known enzyme to unfold the proteins at defined sites by means of particular amino acid residues or by digesting the DNA with suitable restriction enzymes and isolating the desired fragment. Yet another suitable technique involves isolating and amplifying a DNA fragment encoding a desired antibody or polypeptide fragment by polymerase chain reaction (PCR). The oligonucleotides defining the desired terms of the DNA fragment are used in the 5 'and 3' primers in the PCR. Preferably, the anti-TAT antibody and TAT polypeptide antibody fragments share at least one biological and / or immunological activity with the anti-TAT antibody or the native TAT polypeptide described herein. In particular embodiments, conservative substitutions of interest are shown in Table 6 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes are introduced, termed exemplary substitutions in Table 6 or as further described below with reference to the amino acid classes being introduced and the products selected. Table 6 Residual Substitutions Substitutions Original Examples Preferred val; le; ile val lys; gln; asn lys gln; his; lys; arg gln glu glu be be asn asn asp asp pro; wing wing asn; gln; lys; arg arg 1eu; val; me; ala; phe; norleucine leu norleucine; ile; val; met; to; phe ile arg; gln; asn arg leu; phe; ile leu leu; val; ile; to; tyr leu ala ala thr thr ser ser tyr; phe tyr trp; phe; thr; be phe - - Val (V) ile; leu; met; phe wing; norleucine Substantial modifications are achieved depending on or the immunological identity of the anti-TAT antibody or the TAT polypeptide by selecting substitutions that differ significantly in their effect to maintain (a) the structure of the polypeptide structure in the area of substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site or (c) the volume of the side chain. The naturally occurring residues are divided into groups based on the properties of the common side chain: (1) hydrophobic -. norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic: cys, ser, thr; (3) Acidics: asp, glu; (4) basic: asn, gln, his, lys, arg; (5) residues that influence the orientation of the chain: gly, pro; and (6) aromatics: trp, tyr, phe. Non-conservative substitutions will result in the exchange of a member of one of these classes for another class. Such substituted residues may be introduced at the conservative substitution sites or, more preferably, at the remaining (non-conserved) sites.
- - The variations can be made using methods known in the art such as oligonucleotide-mediated mutagenesis (site-directed), alanine scanning and PCR mutagenesis. Site-directed mutagenesis [Cárter et al. , ucí. Acids Res. 13_: 4331 (1986), Zoller et al., Nucí. Acids Res. 10: 6487 (1987)], cassette mutagenesis [Wells et al., Gene, 3_4: 315 (1985)], restriction selection mutagenesis [Wells et al. , Philos. Trans. R. Soc. London Ser. 317: 15 (198S)] or other known techniques can be carried out on the cloned DNA to produce the variant DNA of the anti-TAT antibody or the TAT polypeptide. Amino acid scan analysis can also be used to identify one or more amino acids along a continuous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine and cysteine. Alanine is typically a preferred amino acid of choice among this group because it removes the side chain beyond beta-carbon and is less likely to alter the conformation of the variant backbone [Cunningham and Wells, Science, 244: 1081- 1085 (1989)]. Alanine is also typically preferred because it is the most common amino acid. In addition, it is frequently found in both occult and exposed positions [Creighton, The Proteins, (W.H.
Freeman & Co., N.Y.); Chothia, J. Mol. Biol. , 150: 1 (1976)]. If the alanine substitution does not produce the adequate amounts of variants, an isoteric amino acid may be used. Any cysteine residue not involved in maintaining the proper conformation of the anti-TAT antibody or TAT polypeptide can also be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking. Conversely, cysteine binding (s) can be added to the anti-TAT antibody or to the TAT polypeptide to improve its stability (particularly when the antibody is an antibody fragment such as an Fv fragment). A particularly preferred type of substitutional variant involves the substitution of one or more hypervariable region residues of a native antibody (e.g., a humanized or human antibody). Generally, the resulting variant (s) selected for further development will have (n) improved biological properties in relation to the native antibody from which it is generated (n). A convenient way to generate such substitutional variants involves affinity maturations using phage imaging. Briefly, several hypervariable region sites (e.g., sites 6-7) are mutated to generate all possible amino acid substitutions at each site. The antibody variants thus generated are deployed in a monovalent manner from filamentous phage particles as fusions for the gene III product of M13 encapsulated within each particle. The phage-displayed variants are then selected for their biological activity (e.g., binding affinity) as described herein. In order to identify candidate hypervariable region sites for modification, alanine scanning mutagenesis can be carried out to identify the hypervariable region residues that significantly contribute to the binding of the antigen. Alternatively or additionally, it may be beneficial to analyze a crystal structure of the antigen-antibody complex to identify contact points between the antibody and the human TAT polypeptide. Such contact residues and neighboring waste are candidates for substitution according to the techniques elaborated herein. Once such variants are generated, the panel of variants is subjected to selection as described herein and antibodies with superior properties can be selected in one or more relevant analyzes for further development. The nucleic acid molecules encoding the amino acid sequence variants of the anti-TAT antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, - - isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis and cassette mutagenesis of a variant prepared with greater anteriority or a non-variant version of the anti-TAT antibody. H. Modifications of Anti-TAT Antibodies and TAT Polypeptides Covalent modifications of anti-TAT antibodies and TAT polypeptides are included within the scope of this invention. One type of covalent modification includes reacting target amino acid residues of an anti-TAT antibody or TAT polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues of the anti-TAT antibody. or TAT polypeptide. Derivatization with bifunctional agents is useful, for example, for the cross-linking of the anti-TAT antibody or TAT polypeptide to a water-insoluble support matrix or surface for use in the method for purifying anti-TAT antibodies and vice versa. Commonly used crosslinking agents include, e.g., 1,1-bis (diazoacetyl) -2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, - 5 - including disuccinimidyl esters such as 3,3'-dithiobis (succinimidylpropionate), bifunctional maleimides such as bis-N-maleimido-1,8-octane and agents such as methyl-3- [(p-azidophenyl) dithio] propioimidate. Other modifications include the deamidation of the glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, the hydroxylation of proline and lysine, the phosphorylation of hydroxyl groups of the seryl or threonyl residues, the methylation of the oc- amino of the side chains of lysine, arginine, and histidine [TE Creighton, Proteins: Structure and Molecular Properties, W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)], the acetylation of the N-terminal amine and the amidation of any C-terminal carboxyl group. Another type of covalent modification of the anti-TAT antibody or TAT polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of the antibody or polypeptide. The "alteration of the native glycosylation pattern" is intended to mean for the purposes herein, to suppress one or more carbohydrate residues found in the anti-TAT antibody or native sequence TAT polypeptide (either by removing the underlying glycosylation site or by suppressing the glycosylation by chemical and / or enzymatic means) and / or by adding one or more glycosylation sites that are not they are present in the anti-TAT antibody or native sequence TAT polypeptide. In addition, the phrase includes qualitative changes in the glycosylation of native proteins, involving a change in the nature and proportions of the various carbohydrate residues present. The glycosylation of antibodies and other polypeptides is typically either N-linked or O-linked. Linked to N refers to the attachment of the carbohydrate residue to the side chain of an asparagine residue. The sequences of tripeptide asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for the enzymatic binding of the carbohydrate residue to the asparagine side chain. Thus, the presence of any of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the binding of one of the sugars of N-acetylgalactosamine, galactose or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine can also be used. The addition of glycosylation sites to the anti-TAT antibody or TAT polypeptide is conveniently achieved by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences. (for glycosylation sites linked to N). The alteration may also be effected by the addition or substitution of one or more serine or threonine residues to the original anti-TAT antibody or TAT polypeptide sequence (for 0-linked glycosylation sites). The amino acid sequence of the anti-TAT antibody or the TAT polypeptide can optionally be altered through changes in the level of DNA, particularly by mutating the DNA encoding the anti-TAT antibody or TAT polypeptide at preselected bases such that codons are generated. which will result in the desired amino acids. Another means of increasing the number of carbohydrate residues in the anti-TAT antibody or TAT polypeptide is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published September 11, 1987 and in Aplin and Wriston, CRC Crit. Rev. Biochem. , pp. 259-306 (1981). The removal of the carbohydrate residues present in the anti-TAT antibody or TAT polypeptide can be achieved chemically or enzymatically or by mutational substitution of codons encoding amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and are described, for example, by Hakimuddin et al., Aren. Biochem.
- - Biophys. , 259: 52 (1987) and by Edge et al., Anal. Biochem. , 118: 131 (1981). Enzymatic cleavage of the carbohydrate residues in the polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., Meth. Enzymol, 138: 350 (1987). Another type of covalent modification of the anti-TAT antibody or TAT polypeptide comprises linking the antibody or polypeptide to one of a variety of non-proteinaceous polymers, eg, polyethylene glycol (PEG), polypropylene glycol or polyoxyalkylenes, in the manner indicated in the Patents from EU Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192; or 4,179,337. The antibody or polypeptide can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin capsules and poly (methylmethacrylate) microcapsules, respectively), in colloidal delivery systems. drugs (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions. Such techniques are described in Remington's Pharmaceutical Sciences, 16th edition oslo, A., Ed., (1980). The anti-TAT antibody or TAT polypeptide of the present invention can also be modified in a manner to form chimeric molecules comprising an antibody. anti-TAT or TAT polypeptide fused to another heterologous polypeptide or amino acid sequence. In one embodiment, such a chimeric molecule comprises a fusion of the anti-TAT antibody or TAT polypeptide with a marker polypeptide that provides an epitope to which an anti-marker antibody can be selectively linked. The epitope tag is generally placed at the amino- or carboxyl- terminus of the anti-TAT antibody or TAT polypeptide. The presence of such epitope-labeled forms of the anti-TAT antibody or TAT polypeptide can be detected using an antibody against the marker polypeptide. Also, the provision of the epitope tag allows the anti-TAT antibody or TAT polypeptide to be easily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag. Various marker polypeptides and other respective antibodies are well known in the art. Examples include poly-histidine (poly-his) or poly-histidine-glycine (poly-his-gly) markers; the FL marker polypeptide and its antibody 12CA5 [Pield et al., Mol. Cell. Biol. , 8 ^: 2159-2165 (1988)]; the c-myc marker and the 8F9, 3C7, 6E10, G4 and 9E10 antibodies for the same [Evan et al., Molecular and Cellular Biology, 5: 3610-3616 (1985); and the glycoprotein D (gD) marker of Herpes Simples virus and its antibody [Paborsky et al. , Protein Engineering, 3_ (6): 547 ~ 553 (1990)]. Other marker polypeptides include the Marked peptide [Hoop et al., BioTechnology, 6 ^: 1204-1210 (1988)]; the epitope KT3 peptide [Martin et al., Science, 255: 192-194 (1992)]; an a-tubulin epitope peptide [Skinner et al., J. Biol. Chem., 266: 15163-15166 (1991)]; and the peptide marker of the T7 gene of protein 10 [Lutz-Freyermuth et al., Proc. Nati Acad. Sei USA, 87: 6393-6397 (1990)]. In an alternative embodiment, the chimeric molecule may comprise a fusion of the anti-TAT antibody or TAT polypeptide with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also referred to as an "immunoadhesin"), such fusion could be for the Fe region of an IgG molecule. Ig fusions preferably include the substitution of a soluble form (deleted or inactivated transmembrane domain) of an anti-TAT antibody or TAT polypeptide in place of at least one variable region within an Ig molecule. In a particularly preferred embodiment, the immunoglobulin fusion includes the joint, CH2 and CH3 or the joint CE, C¾ and the CH3 regions of an IgG1 molecule. For the production of immunoglobulin fusions see also the U.S. Patent. No. 5,428,130 issued June 27, 1995. I. Preparation of Anti-TAT Antibodies and TAT Polypeptides The following description relates mainly to the production of anti-TAT antibodies and TAT polypeptides by culturing cells transformed or transfected with a vector containing an anti-TAT antibody and a TAT polypeptide encoding nucleic acid. Of course, it is contemplated that alternative methods, which are well known in the art, can be employed to prepare anti-TAT antibodies and TAT polypeptides. For example, the appropriate amino acid sequence or portions thereof can be produced by direct synthesis of the peptide using solid phase techniques [see, e.g. , Stewart et al., Solid-Phase Peptide Synthesis, W.H. Freeman Co., San Francisco, CA (1969); Merrifield, J. Am. Chem. Soc. , 5: 2149-2154 (1963)]. In vitro protein synthesis can be carried out using manual techniques or by automation. Automated synthesis can be achieved, for example, by using an Applied Biosystems Peptide Synthesizer (Foster City, CA) using the manufacturer's instructions. Several portions of the anti-TAT antibody or TAT polypeptide can be chemically synthesized separately and combined using chemical or enzymatic methods to produce the desired TAT anti-TAT antibody or polypeptide. 1. Isolation of the Anti-TAT Antibody or TAT polypeptide that encodes DNA - 1 - The anti-TAT antibody or TAT polypeptide encoding DNA can be obtained from a cDNA library prepared from tissue believed to possess the mAR of the anti-TAT antibody or the TAT polypeptide and which expresses it at a detectable level. Accordingly, the human anti-TAT antibody or the TAT polypeptide DNA can conveniently be obtained from a cDNA library prepared from human tissue. The gene encoding the anti-TAT antibody or the TAT polypeptide can also be obtained from a genomic library or by known synthetic methods (e.g., automated nucleic acid synthesis). The libraries can be selected with probes (such as oligonucleotides of at least about 20-80 bases) designed to identify the gene of interest of the protein encoded by the. The selection of the cDNA or genomic library with the selected probe can be carried out using standard procedures, such as those described in Sambrook et al. , Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989). An alternative means for isolating the gene encoding the anti-TAT antibody or the TAT polypeptide is to use PC methodology [Sambrook et al. , supra: Dieffenbach et al. , PCR Primer: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1995)].
- - Techniques for selecting a cDNA library are well known in the art. The oligonucleotide sequences selected as probes must be of sufficient length and sufficiently unambiguous that the false positives are minimized. The oligonucleotide is preferably labeled such that hybridization to the DNA can be detected in the library that is selected. Marking methods are well known in the art, and include the use of radiolabels such as 32 P-labeled ATP, biotinylation or enzyme labeling. Hybridization conditions, including moderate stiffness and high stiffness, are provided in Sambrook et al., Supra. The sequences identified in such library selection methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private databases. Sequence identity (either at the amino acid or nucleotide level) within defined regions of the molecule or through the full length sequence can be determined using methods known in the art and as described herein. Nucleic acid having a protein coding sequence can be obtained by selecting the cDNA or genomic libraries using the deduced amino acid sequence described herein for the first time and, if necessary, using conventional first extension methods as described in Sambrook et al. . , supra, to detect precursors and process mAR intermediates that may not have been reverse-transcribed within the cDNA. 2. Selection and Transformation of Host Cells Host cells are transfected or transformed with expression or cloning vectors described herein for the production of the anti-TAT antibody or TAT polypeptide and cultured in a modified conventional nutrient medium as appropriate to induce promoters, select transformers or amplify the genes encoding the desired sequences. The culture conditions, such as the medium, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation. In general, the principles, protocols and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach, M.Butler, ed. (IRL Press, 1991) and Sambrook et al., Supra. The methods of eukaryotic cell transfection and prokaryotic cell transformation are known to the ordinarily skilled artisan, for example, CaCl2, CaP0, liposome-mediated and electroporation. Depending on the host cell used, the transformation is carried out using standard techniques appropriate for such cells.
The calcium treatment using calcium chloride, as described in Sambrook et al. , supra or electroporation are generally used for prokaryotes. The infection with Agrrojbacterium tumefaciens is used for the transformation of certain plant cells as described by Shaw et al. Gene, 23_: 315 (1983) and WO 89/95859 published on June 29, 1989. For mammalian cells without such cell walls, the calcium phosphate precipitation method of Graham and van der Eb, Virology, 52: 456-457 (1978) may be employed. General aspects of mammalian cell host system transfections have been described in the U.S. Patent. No. 4,399,216. Transformations in yeast are typically carried out according to the method of Van Solingen et al., J. Bact. , 130: 946 (1977) and Hsiao et al., Proc. Nati Acad. Sci. (USA), 76: 3829 (1979). However, other methods can also be used to introduce the DNA into cells such as nuclear microinjection, electroporation, protoplast bacterial fusion with intact cells or polycations, e.g. , polybrene, polyornithine. For various techniques for transforming mammalian cells, see Keown et al., Methods in Enzimology, 185: 527-537 (1990) and Mansour et al., Nature, 336: 348-352 (1988). Suitable host cells for cloning or expressing the DNA in the vectors herein include prokaryote, yeast or larger eukaryotic cells. The - - prokaryotes include but are not limited to eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli. Several species of E. coli are publicly available, such as the species E. coli K12 MM294 (ATCC 31,446); E. coli X1776 (ATCC 31,537); E. coli W3110 (ATCC 27,325) and K5 772 (ATCC 53,635). Other suitable host prokaryotic cells include Enterobacteriaceae such as Escherichia, eg, E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, eg, Salmonella typhimurium, Serratia, eg, Serratia marcescans and Shigella, as well as Bacilli such as B. súbtilis and B. licheniformis. { e.g., B. licheniformis 41P described in DD 266,710 published April 12, 1989), Pseudomonas such as P. aeruginosa and Strepto yces. These examples are illustrative rather than limiting. The W3110 species is a particularly preferred host or native host because it is a common host species for fermentations of recombinant DNA product. Preferably, the host cell secretes minimal amounts of proteolytic enzymes. For example, the W3110 species can be modified to effect a genetic mutation in the genes encoding proteins endogenous to the host, with examples of such hosts including the E. coli W3110 1A2 species, which has the complete tonA genotype; E. coli W3110 9? 4 species, which has the complete tonA ptr3 genotype; - - species E. coli W3110 27C7 (ATCC 55.244), which has the genotype tonA ptr3 phoA E15 complete (argF-lac) 169 degP ompT kanr; species E. coli W3110 37D6, which has the genotype tonA ptr3 phoA E15 complete (argF-lac) 169 degP ompT rbs7 ilvG kanr species E. coli 3110 40B4, which is a 37D6 species with deletion mutation degP not resistant to kanamycin; and the E. coli species having the mutant periplasmic protease described in the U.S. Patent. No. 4,946,783 issued August 7, 1990. Alternatively, in vi tro cloning methods, e.g. , PCR or other nucleic acid polymerase reactions. The full length antibody, the antibody fragments and the antibody fusion proteins can be produced in bacteria, in particular when glycosylation and Fe effector is not required, such as when the therapeutic antibody is conjugated to a cytotoxic agent. { e.g., a toxin) and the immunoconjugate by itself shows effectiveness in the elimination of the tumor cell. The full-length antibodies have a longer half-life in circulation. The production of E. coli is faster and more efficient in cost. For the expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. 5,648,237 (Cárter et al.), U.S. 5,789,199 (Joly et al.) And U.S. 5,840,523 (Simmons et al.) That describe the translation initiation region (TIR) and signal sequences to optimize expression and - 2 - secretion, these patents are incorporated herein by reference. After expression, the antibody is isolated from E.coli cell paste in a soluble fraction and can be purified by, e.g., a protein A or G column depending on the isotype. The final purification can be carried out similar to the process for purifying the antibody expressed e.g., in CHO cells. In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for the vectors encoding the anti-TAT antibody or the TAT polypeptide. Saccharomyces cerevisiae is a minor eukaryotic host microorganism commonly used. Others include, Schizosaccharomyces po be (Beach and Nurse, Nature, 290: 140 [1981]; EP 139, 383 published May 2, 1985); Kluyveromyces hosts (U.S. Patent No. 4,943,529; Fleer et al., Bio / Technology, 9: 968-975 (1991) such as eg, K. lactis (MW98-8C, CBS683, CBS4574; Louvencourt et al., J. Bacteriol., 154 (2): 737-742 [1983]), K.fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K drosophilarum (ATCC 36,906; Van der Berg et al., Bio / Technology, 8: 135 (1990)), K. thermotolerans and K. marxianus, yarrowia (EP 402,226), Pichia pastoris (EP 183,070, Sreekrishna et al., J. Basic Microbiol., 28: 265-278 - -
[1988]; Candida; Trichoderma recia (EP 244,234); Neurospora crassa (Case et al., Proc. Nati, Acad. Sci. USA 76: 5259-5263 [1979]); Schwanniomyces such as Schwanniomyces occidentalis (EP 394,538 published October 31, 1990); and filamentous fungi such as e.g. , Neurospora, Penicillium, Tolypocladium (WO 91/00357 published January 10, 1991) and Aspergilius hosts such as A. nidulans (Ballance et al., Biochem. Biophys., Res. Commmon, 112: 284-289 [1983]; Tilburn et al., Gene, 26: 205-221 [1983], Yelton et al., Proc. Nati, Acad. Sci. USA, 81: 1470-1474 [1984], and A Niger (Kelly and Hynes, EMBO J 4: 475-479 [1985] Methylotropic yeasts are suitable herein and include, but are not limited to, yeast capable of growing in methanol selected from the genus consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharo yces , Torulopsis and Rhodotorula A list of specific species that are exemplary of this class of yeasts can be found in C. Anthony, The Biochemistry of Methylotrophs, 269 (1982) .The host cells suitable for the expression of anti-TAT antibody or of the TAT polypeptide are derived from multicellular organisms Examples of invertebrate cells include the cells of ects such as Drosophila S2 and Spodoptera Sf9, as well as plant cells, such as cell cultures of cotton, corn, soybean, petunia, tomato and tobacco. Numerous baciloviral species and variants and the corresponding permissive insect host cells have been identified from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fly of the f uta) and Bombix mori. . A variety of viral species for transfection are publicly available, eg, the Ll variant of Autographa californica NPV and the Bm-5 species of Bombix mori NPV and such viruses can be used as the virus present according to the present invention, particularly for the transfection of Spodoptera f ugiperda cells. However, the interest has been higher in vertebrate cells and the propagation of vertebrate cells in culture (tissue culture) has become a routine procedure. Examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC C L 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); kidney cells of baby hamster (BHK, ATCC CCL 10); Chinese hamster ovarian cells / -DHFR (CHO, Urlaub et al., Proc. Nati. Acad. Sei. USA 77: 4216 (1980); mouse sertoli cells (TM4, Mather, Biol. Reprod., 23: 243- 251 (1980)), monkey kidney cells (CV1 ATCC CCL 70), African green monkey kidney cells (VERO- - - 76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); renal canine cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 80S5); mouse mammary tumor (MMT 060562, ATCC CCL 51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383: 44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2). The host cells are transformed with the above-described expression or cloning vectors for the production of the anti-TAT antibody or TAT polypeptide and cultured in a modified conventional nutrient medium as appropriate to induce promoters, select transformants or amplify the genes they encode the desired sequences. 3. Selection and use of a Replicable Vector Nucleic acid. { e.g., cDNA or genomic DNA) encoding the anti-TAT antibody or TAT polypeptide can be inserted into a replicable vector for cloning (amplification of the DNA) or for expression. Several vectors are publicly available. The vector, for example, can be in the form of a plasmid, cosmid, viral particle or phage. The appropriate nucleic acid sequence can be inserted into the vector by a variety of methods. In general, the DNA is inserted into the appropriate endonuclease restriction site (s) using techniques known in the art. The components of the vector generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, an augmenting element, a promoter and a transcription termination sequence. The construction of suitable vectors containing one or more of these components employs standard binding techniques that are known to the skilled artisan. TAT can be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which can be a signal sequence or another polypeptide having a specific cleavage site on the N-terminus of the mature protein or polypeptide. In general, the signal sequence may be a component of the vector or it may be part of the anti-TAT antibody or TAT polypeptide that encodes the DNA that is inserted into the vector. The signal sequence may be a prokaryotic signal sequence selected, for example, from the group of alkaline phosphatase, penicillinase, Ipp or heat-stable derivatives of enterotoxin II. For the secretion of the yeast the sequence signal can be e.g., the derivation of yeast invertase, the derivation of the alpha factor (including Saccharomyces and derivations of Kluyveromyces factor ce, the last ones described in the Patent - - of E.U. No. 5,010,182); or derivative of acid phosphatase, the glucoamylase derivative of C. albicans (EP 362,179 published April 4, 1990) or the signal described in WO 90/13646 published November 15, 1990. In the expression of the mammalian cell , mammalian signal sequences can be used for direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory derivations. Both the expression and cloning vectors contain a nucleic acid sequence that allows the vector to be repeated in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast and viruses. The origin of replication from plasmid pBR322 is suitable for most Gram-negative bacteria, the 2m plasmid origin is suitable for yeast and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for the cloning of vectors in mammalian cells. The expression and cloning vectors will typically contain a selection gene, also called a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g. , ampicillin, neomycin, methotrexate or tetracycline, (b) complement auxotropic deficiencies - - or (c) provide critical nutrients not available from the compound medium, e.g., the racemase D-alanine encoding the gene for Bacilli. An example of selectable markers suitable for mammalian cells are those that allow the identification of cells competent to receive the nucleic acid encoding the anti-TAT antibody or the TAT polypeptide, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al., Proc. Nati Acad. Sci. USA 77: 4216 (1980). A suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7 [Stinchcomb et al. , Nature, 282: 39 (1979); Kingsman et al., Gene 7: 141 (1979); Tschemper et al .. Gene 10:57 (1980)]. The trpl gene provides a selection marker for a mutant species of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics, 85:12 (1977)]. Expression and cloning vectors commonly contain a promoter operably linked to the nucleic acid sequence encoding the anti-TAT antibody or TAT polypeptide to direct the direct synthesis of mRNA. Promoters recognized by a variety of potential host cells are well known. Promoters - suitable for use with prokaryotic hosts include the β-lactamase and lactose promoter systems [Chang et al., Ature, 275: 615 (1978); Goeddel et al., Natre, 281: 544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res. , 8: 4057 (1980); EP 36,776] and hybrid promoters such as the tac promoter [deBoer et al., Proc. Nati Acad. Sei USA, 80: 21-25 (1983)]. Promoters for use in bacterial systems will also contain a Shine-Dalgrano (S.D.) sequence operably linked to the anti-TAT antibody or TAT polypeptide encoding the DNA. Examples of suitable promoter sequences for use with yeast hosts include promoters for 3-phosphoglycerate kinase [Hitzeman et al., J. Biol. Chem., 255: 2073 (1980)] or other glycolytic enzymes [Hess et al. al., J. Ad. Enzyme Reg. , 7: 149 (1968); Holland, Biochemistry, 17: 4900 (1978)], such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phospho-fructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, thiosephosphate isomerase, phosphoglucose isomerase and glucokinase. Other yeast promoters, which are inducible promoters that have the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase and enzymes responsible for the use of maltose and galactose. Vectors and promoters suitable for use in the expression of yeast are further described in EP 73,657. A transcription of the anti-TAT antibody or the TAT polypeptide from the vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes or viruses such as polyoma virus, smallpox virus (UK 2,211,504 published on July 5, 1989), adenovirus (such as an Adenovirus 2), bovine papilloma virus, poultry sarcoma virus, cytomegalovirus, a retrovirus, hepatitis B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, eg, the actin promoter or an immunoglobulin promoter and the heat shock promoters, provided that such promoters are compatible with the host cell systems. The transcription of a DNA encoding the anti-TAT antibody or the TAT polypeptide by larger eukaryotes can be increased by inserting an increase sequence into the vector. Augmentators are cis-active elements of DNA, commonly of approximately 10 to 300 bp, - - which act on a promoter to increase its transcription. Many augmentation sequences are now known for mammalian genes (globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, an eukaryotic cell virus enhancer will be used. Examples ide the SV40 enhancer on the back side of the replication origin (bp 100-270), the cytomegalovirus promoter enhancer, the polyoma enhancer on the back side of the replication origin and the adenovirus enhancers. The enhancer can bind within the vector at the 5 'or 3' position to the coding sequence of the anti-TAT antibody or the TAT polypeptide, but is preferably located at a 5 'site from the promoter. Expression vectors used in eukaryotic host cells (yeast, fungus, insect, plant, animal, human or nucleated cells of other multicellular organisms) will also contain the sequences necessary for the termination of transcription and to stabilize the mRNA. Such sequences are commonly available from the 5 'and occasionally 3' untranslated regions of the eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mAR encoding the anti-TAT antibody or the TAT polypeptide.
- - Still other methods, vectors and host cells suitable for their adaptation to the synthesis of an anti-TAT antibody or TAT polypeptide in a vertebrate recombinant cell culture are described in Gething et al., Nature, 293: 620-G25 (1981); Mantei et al., Nature, 281: 40-46 (1979); EP 117,060; and EP 117,058. 4. Culturing the Host Cells The host cells used to produce the anti-TAT antibody and the TAT polypeptide of this invention can be cultured in a variety of media. Commercially available media such as that of Ham FIO (Sigma), Minimal Essential Medium (MEM), (Sigma), PM1.-1640 (Sigma) and Modified Eagle's Medium Dulbecco (DMEM), (Sigma) are suitable for the Culture of the host cells. In addition, any of the means described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102: 255 (1980), Pats. of E.U. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; O 5,122,469; WO 90/03430; WO 87/00195; or Pat of E.U. Re. 30,985 can be used as a culture medium for the host cells. Any of these media can be supplemented as necessary with hormones and / or other growth factors (such as insulin, transferrin or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium and phosphate), buffers (such as HEPES), nucleotides (such as - - adenosine and thymidine), antibiotics (such as the drug GENTAMYCI ®), trace elements (defined as inorganic compounds commonly present in final concentrations in the micromolar range) and glucose or an energy source equivalent. Any other necessary supplements may also be included at the appropriate concentrations that will be known to those skilled in the art. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression and will be apparent to the ordinarily experienced technician. 5. Detection of Gene Amplification / Expression The amplification and / or expression of the gene can be measured in a sample directly, for example, by conventional immunoblotting, immunoblotting to quantitate the transcription of mRNA [Thomas, Proc. Nati Acad. Sei USA, 77: 5201-5205 (1980)], spot staining (DNA analysis) or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies that can recognize specific duplexes, including DNA duplexes, RNA duplexes and hybrid AD-RNA duplexes or DNA-protein duplexes may be employed. The antibodies in turn can be labeled and the analysis can be carried out when the duplexes are linked to a - - surface, so that when duplexes are formed on the surface, the presence of antibody bound to the duplo can be detected. The expression of the gene, alternatively, can be measured by immunological methods, such as immunohistochemical staining of cells or sections of tissue and analysis of cell culture or body fluids, to directly quantify the expression of the gene product. Antibodies useful for immunohistochemical staining and / or analysis of sample fluids can be either monoclonal or polyclonal and can be prepared in any mammal. Conveniently, antibodies against a native sequence TAT polypeptide or against a synthetic peptide can be prepared based on the DNA sequences provided herein or against an exogenous sequence fused to TAT DNA and encoding a specific antibody epitope. 6. Purification of the Anti-TAT Antibody and the TAT Polypeptide The forms of the anti-TAT antibody and the polypeptide TAT can be recovered from the culture medium or lysates of host cells. If it is attached to the membrane, it can be released from the membrane using a suitable detergent solution (e.g., Triton-X 100) or by enzymatic cleavage. The cells employed in the expression of the anti-TAT antibody and the TAT polypeptide can be broken by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption or cell lysate agents. It may be desired to purify the anti-TAT antibody and the TAT polypeptide from recombinant cellular proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation in an ion exchange column; ethanol precipitation; HPLC in reverse phase; chromatography on silica or on a cation exchange resin such as DEAE; chromatofocusing; SDS-PAGE; precipitation of ammonium sulfate; gel filtration using, for example, Sephadex G-75; Protein A Sepharose columns to remove contaminants such as IgG; and metal chelation columns to bind epitope-tagged forms of the anti-TAT antibody and the TAT polypeptide. Various methods of protein purification can be employed and such methods are known in the art and are described for example in Deutscher, Methods in Enzymology, 182 (1990); Scopes Protein Purification: Principles and Practice, Springer-Verlag, New York (1982). The selected purification step (s) will depend, for example, on the nature of the production process used and the anti-TAT antibody or TAT polypeptide produced.
- - When using recogination techniques, the antibody can be produced intraellabularly, in the periplasmic space or secreted directly into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, whether host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio-Technology, 10: 163-167 (1992) describes a method for isolating antibodies that are secreted into the periplasmic space of E. coli. Briefly, the cell paste is melted in the presence of sodium acetate (pH 3.5), EDTA and phenylmethylsulfonyl fluoride (P SF) for about 30 min. The cellular waste can be removed by centrifugation. When the antibody is secreted into the medium, supernatants from such expression systems are generally concentrated first using a commercially available protein filter, for example, an Amicon or illipore ultrafiltration unit. A protease inhibitor such as PMSF can be included in any of the above steps to inhibit proteolysis and antibiotics can be included to prevent the growth of accidental contaminants. The antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, - - dialysis and affinity chromatography, with affinity chromatography being the preferred purification technique. The suitability of protein A as an affinity binding depends on the species and the isotype of any immunoglobulin Fe domain that is present in the antibody. Protein A can be used to purify antibodies that are based on the human?,? 2 or? 4 heavy chains (Lindmark et al., J. Immunol., Meth. 62: 1-13 (1983)). Protein G is recommended for all mouse isotypes and for human? 3 (Guss et al., EMBO J. 5: 1567-1575 (1986)). The matrix to which the affinity ligand is attached is most frequently agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly (styrenedivinyl) benzene allow for faster flow rates or shorter processing times than can be achieved with agarose. When the antibody comprises a CH3 domain, Bakerbond ABX ™ resin (J.T. Baker, Phillipsburg, NJ) is useful for purification. Other techniques for protein purification such as fractionation in an in-exchange column, ethanol precipitation, HPLC in Reverse Phase, silica chromatography, SEPHAROSE ™ heparin chromatography, chromatography on anion or cation exchange resin (such as as a column of polyaspartic acid), chromatofocusing, SDS-PAGE and precipitation of ammonium sulfate are available depending on the antibody to be recovered. Following any preliminary purification step, the mixture comprising the antibody of interest and the contaminants can be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably carried out at low concentrations of salt (e.g., from about 0-0.25M salt). J. Pharmaceutical Formulations Therapeutic formulations of the anti-TAT antibodies, the TAT-binding oligopeptides, the TAT-binding organic molecules and / or the TAT polypeptides used in accordance with the present invention are prepared for storage by mixing the antibody, oligopeptide or organic molecule having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences, 16th edition Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable vehicles, excipients or stabilizers are non-toxic to the receptors in the doses and concentrations employed, and include buffers such as acetate, Tris, phosphate, citrate and other organic acids; - - antioxidants including preservative ascorbic acid and methionine (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcitol; cyclohexanol, 3-pentanol, and m-cresol); low molecular weight polypeptides (less than about 10 residues); proteins, such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers such as polyvinyl pyrrolidone, - amino acids such as glycine, glutamine, asaparagine, histidine, arginine or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose or dextrins; chelating agents such as EDTA; tonicizers such as trehalose and sodium chloride; sugars such as sucrose, mannitol, trehalose or sorbitole; surfactants such as polysorbate; against salt formation ions such as sodium, - metal compounds (e.g., Zn-protein compounds); and / or non-ionic surfactants such as TWEEN®, PLURONICS® or polyethylene glycol (PEG). The antibody preferably comprises the antibody in a concentration of between 5-200 mg / ml, preferably between 10-100 mg / ml. The formulations herein may also contain more than one active compound as necessary for the particular indication to be treated, preferably those with complementary activities that do not adversely affect each other. For example, in addition to an anti-TAT antibody TAT binding oligopeptide or TAT binding organic molecule, it may be desirable to include in the formulation, an additional antibody, eg, a second anti-TAT antibody that binds to a different epitope in the TAT polypeptide or an antibody to some other target such as a growth factor that affects the growth of the particular cancer. Alternatively or additionally, the composition may further comprise a chemotherapeutic agent, cytotoxic agent, cytosine, growth inhibitory agent, anti-hormonal agent and / or cardioprotective agent. Such molecules are suitably present in combination in amounts that are effective for the intended purpose. The active ingredients can also be enclosed in prepared microcapsules, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin microcapsules and poly- (methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (e.g., liposomes , albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions. Such techniques are described in Remington's Pharmaceutical Sciences, 16a. Osol edition, A. Ed. (1980). Release preparations can be prepared - sustained Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films or microcapsules. Examples of sustained release matrices include polyesters, hydrogels (e.g., poly (2-hydroxyethyl-methacrylate) or poly (vinylalcohol)), polylactides (U.S. Pat .: No. 3,773,919), L-glutamic acid copolymers and? ethyl-L-glutamate, non-degradable ethylene vinyl acetate, degradable lactic acid-glycolic acid copolymers such as LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate) and poly-D ~ acid (-) -3-hydroxybutyric. The formulations to be used for in vivo administration must be sterile. This is easily achieved by filtration through sterile filtration membranes. . Diagnosis and Treatment with TAT Link Oligopeptide Anti-TAT Antibodies and TAT Linkage Organic Molecules Several tests are available to determine TAT expression in cancer. In one embodiment, overexpression of the TAT polypeptide can be analyzed by immunohistochemistry (IHC). Sections of tissue immersed in - - Paraffin from a tumor biopsy can be subjected to the IHC analysis and be adjusted to a criterion of intensity of coloration of the TAT protein as follows: Score 0 - no coloration is observed or membrane staining is observed in less than 10% of the tumor cells. Score 1 + - a faint / barely perceptible coloration on the membrane is detected in more than 10% of the tumor cells. The cells are only colored in part of their membrane. Score 2 + - a weak to moderate coloration is observed in the entire membrane in more than 10% of the tumor cells. Score 3 + - moderate to strong coloration is observed in the entire membrane in more than 10% of the tumor cells. Those tumors with a score of 0 or 1+ for the expression of the TAT polypeptide can be characterized as not overexpressing TAT, while those tumors with scores of 2+ or 3+ can be characterized as overexpressing TAT. Alternatively or additionally, FISH analyzes such as INFORM® (sold by Ventana, Arizona) or PATHVISION® (Vysis, Illinois) can be carried out on formalin fixed tumor tissue, immersed in paraffin to determine the degree (if any) ) of TAT overexpression in the tumor. TAT overexpression or amplification can be assessed using in vivo diagnostic analysis, e.g. , administering a molecule (such as an oligopeptide antibody or organic molecule) that binds the molecule to be detected and is labeled with a detectable label. { e.g., a radioactive isotope or a fluorescent tag) and externally scanning the patient for the location of the mark. As described above, the oligopeptide anti-TAt antibodies and organic molecules of the invention have several non-therapeutic applications. The anti-TAT oligopeptide antibodies and organic molecules of the present invention can be useful for the diagnosis and staging of cancers expressing the TAT polypeptide. { e.g. , in radiovisualization). Oligopeptide antibodies and organic molecules are also useful for the purification or immunoprecipitation of the TAT polypeptide from cells, for the detection and quantification of the TAT polypeptide in vitro, eg, in an ELISA or an immunoblot, to destroy and eliminate cells expressing TAT of a population of cells mixed as a step in the purification of other cells. Currently, depending on the cancer stage, cancer treatment involves one or a combination of - - following therapies: surgery to remove cancerous tissue, radiation therapy and chemotherapy. Therapy with anti-TAT oligopeptide antibody or organic molecule may be especially desirable in elderly patients who do not tolerate either the toxicity and side effects of chemotherapy and in metastatic disease where radiation therapy has limited utility. Oligopeptide anti-TAT antibodies and organic molecules that target the tumor of the invention are useful for alleviating TAT-expressing cancers at the initial diagnosis of the disease or during relapse. For therapeutic applications, the anti-TAT oligopeptide antibody or organic molecule can be used alone or in combination therapy with, e.g., hormones, antiangiogenes or radioradiated compounds or with surgery, cryotherapy and / or radiotherapy. The treatment with anti-TAT oligopeptide antibody or organic molecule can be administered in conjunction with other conventional forms of therapy, consequently with either pre- or post-conventional therapy. Chemotherapy drugs such as TAXOTE E® (docetaxel), TAXOL® (palictaxel), estramustine and mitoxantrone are used in the treatment of cancer, particularly in high-risk patients. In the present method of the invention for the treatment or alleviation of cancer, the anti-TAT oligopeptide antibody or organic molecule in - - can be administered to the cancer patient. conjunction with treatment with one or more of the preceding chemotherapeutic agents. In particular, combination therapy with palictaxel and modified derivatives is contemplated (see, e.g., EP0600517). The anti-TAT oligopeptide antibody or organic molecule will be administered with a therapeutically effective dose of the chemotherapeutic agent. In another embodiment, the anti-TAT oligopeptide antibody or organic molecule is administered in conjunction with chemotherapy to improve the activity and efficacy of the chemotherapeutic agent, e.g., paclitaxel. The Physicians' Desk Reference (PDR) describes dosages of these agents that have been used in the treatment of various cancers. The dosage regimen and dosages of these chemotherapeutic drugs mentioned above which are therapeutically effective will depend on the particular cancer to be treated, the extent of the disease and other factors familiar to the physician skilled in the art and can be determined by the physician. In a particular embodiment, the patient is administered a conjugate comprising an anti-TAT oligopeptide antibody or organic molecule conjugated with a cytotoxic agent. Preferably, the immunoconjugate bound to the TAT protein is internalized through the cell, resulting in an increased therapeutic efficacy of the immunoconjugate in the elimination of the cancer cell to which it is linked. In a preferred embodiment, the cytotoxic agent targets or interferes with the nucleic acid in the cancer cell. Examples of such cytotoxic agents are described in the foregoing and include maytansinoids, calicheamicins, ribonucleases and DNA endonucleases. The anti-TAT oligopeptide antibodies, organic molecules or toxin conjugates thereof are administered to a human patient, according to known methods, such as intravenous administration, eg, as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerebroespinal, subcutaneous, intra-articular, intrasynovial, oral intrathecal, topical or inhalation routes. The intravenous or subcutaneous administration of the oligopeptide antibody or organic molecule is preferred. Other therapeutic regimens may be combined with the administration of the anti-TAT oligopeptide antibody or organic molecule. The combined administration includes co-administration using separate formulations or a single pharmaceutical formulation and consecutive administration in any order, wherein preferably there is a period of time while both (or all) active agents simultaneously exert their biological activities. Preferably, such combination therapy results in a synergistic therapeutic effect.
- - It may also be desirable to combine the administration of the oligopeptide anti-TAT antibody or antibodies or organic molecules with the administration of an antibody directed against another tumor antigen associated with the particular cancer. In another embodiment, the therapeutic treatment methods of the present invention involve the combined administration of an oligopeptide anti-TAT antibody (or antibodies) and one or more chemotherapeutic agents or growth inhibitory agents, including co-administration of cocktails. of different chemotherapeutic agents. Chemotherapeutic agents include estramustine phosphate, prednimustine, cisplatin, 5-fluorouracil, melphalan, cyclophosphamide, hydroxyurea and hydroxyureataxanes (such as paclitaxel and doxetaxel) and / or anthracycline antibiotics. The preparation and dosing schedules for such chemotherapeutic agents may be used in accordance with the manufacturer's instructions or as determined empirically by the skilled practitioner. Preparation and dosing programs for such chemotherapy are also described in Chemotherapy Service Ed., .C. Perry, Williams & Williams, Baltimore, MD (1992). The oligopeptide antibody or organic molecule can be combined with an anti-hormonal compound; and. < gr., a - - anti-estrogen compound such as tamoxifen; an anti-progesterone such as onapristone (see, EP 616 812); or an anti-androgen such as flutamide, in known dosages for such molecules. When the cancer to be treated is an independent androgen cancer, the patient may have previously undergone anti-androgen therapy and, after the cancer becomes independent androgen, the anti-TAT oligopeptide antibody or organic molecule may be administered to the patient ( and optionally other agents as described herein). Sometimes, it may be beneficial to also co-administer a cardioprotective (to prevent or reduce myocardial dysfunction associated with the therapy) or one or more cytokines to the patient. In addition to the above therapeutic regimens, the patient may undergo surgical removal of the cancer cells and / or radiation therapy, prior to, concurrently with or subsequent to the oligopeptide antibody or organic molecule therapy. Suitable doses for any of the co-administered agents above are those currently used and may be decreased due to the combined action (synergy) of the agent and the anti-TAT oligopeptide antibody or organic molecule. For the prevention or treatment of the disease, the dose and mode of administration will be selected by the physician according to known criteria. The appropriate dose of the oligopeptide antibody or organic molecule will depend on the type of disease to be treated, as defined above, the severity and course of the disease whether the oligopeptide antibody or organic molecule is administered for preventive or therapeutic purposes, prior therapy , the clinical history of the patient and the response to the oligopeptide antibody or organic molecule and the discretion of the attending physician. The oligopeptide antibody or organic molecule is suitably administered to the patient at one time or over a series of treatments. Preferably, the oligopeptide antibody or organic molecule is administered by intravenous infusion or by subcutaneous injections. Depending on the type and severity of the disease, from about 1 g / kg to about 50 mg / kg of body weight [egf about 0.1-15 mg / kg / dose) of the antibody may be a candidate initial dose for administration to the patient either, for example, by one or more separate administrations or by continuous infusion. A dosage regimen may comprise administration of an initial loading dose of approximately 4 mg / kg, followed by a weekly maintenance dose of approximately 2 mg / kg of the anti-TAT antibody. However, other dosage regimens may be useful.- - A typical daily dose may range from about 1/9 to 100 mg / kg or more, depending on the aforementioned factors. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of the symptoms of the disease occurs. The progress of this therapy can be easily verified by conventional methods and analyzes based on criteria known to the physician or other persons skilled in the art. In addition to the administration of the antibody protein to the patient, the present application contemplates administration of the antibody by genetic therapy. Such administration of nucleic acid encoding the antibody is encompassed by the term "administering an effective therapeutic amount of an antibody". See, for example, WO96 / 07321 published March 14, 1996 concerning the use of gene therapy to generate intracellular antibodies. There are two important attempts to introduce the nucleic acid (optionally contained in a vector) into the patient's cells; in vivo and ex vivo. For in vivo delivery the nucleic acid is injected directly into the patient, commonly at the site where the antibody is required. For the ex vivo treatment, the patient's cells are removed, the nucleic acid is - - introduced into these isolated cells and the modified cells are administered to the patient either directly or, for example, encapsulated within porous membranes that are implanted within the patient (see, e.g., US Patents Nos. 4,892,538 and 5,283,187). There are a variety of techniques available to introduce nucleic acids into viable cells. The techniques vary depending on whether the nucleic acid is transferred into cells grown in vitro or in vivo in the cells of the intended host. Suitable techniques for nucleic acid transfer within mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, calcium phosphate precipitation method, etc. A vector commonly used for the ex vivo delivery of the gene is a retroviral vector. Currently preferred in vivo nucleic acid transfer techniques include transfection with viral vectors (such as adenovirus, Herpes simplex virus or adeno-associated virus) and lipid-based systems (lipids useful for lipid-mediated transfer of the virus). gene are DOTMA, DOPE and DC-Chol, for example). For a review of the currently known genetic marker and gene therapy protocols see Anderson et al., Science 256: 808-813 (1992). See, also WO 93/25673 and references cited therein.
- - The anti-TAT antibodies of the invention can be found in different forms encompassed by the definition of "antibody" herein. Thus, the antibodies include full length or intact antibody, antibody fragments, native sequence antibody or amino acid variants, humanized, chimeric or fusion antibodies, immunoconjugates and their functional fragments. In the fusion antibodies an antibody sequence is fused to a heterologous polypeptide sequence. The antibodies can be modified in the Fe region to provide the desired effector functions. As discussed in greater detail in the sections herein, with the appropriate Fe regions, the naked antibody bound on the cell surface can induce cytotoxicity, eg, through antibody-dependent cellular cytotoxicity (ADCC) or by the complement of supply in the complement-dependent cytotoxicity or some other mechanism. Alternatively, when it is desirable to eliminate or reduce effector function, in order to minimize side effects or therapeutic complications, certain other Fe regions may be used. In one embodiment, the antibody competes to bind or bind substantially to, the same epitope as the antibodies of the invention. Antibodies having the biological characteristics of the present anti-TAT antibodies of the invention are also contemplated, specifically including targeting the tumor in vivo and any inhibition of cell proliferation or cytotoxic characteristics. The methods for producing the above antibodies are described in detail herein. The present anti-TAT oligopeptide antibodies and organic molecules are useful in the treatment of a cancer that expresses TAT or to alleviate one or more symptoms of cancer in a mammal. Such cancer includes prostate cancer, cancer of the urinary tract, lung cancer, breast cancer, colon cancer and ovarian cancer, more specifically, prostatic adenocarcinoma, renal cell carcinomas, colorectal adenocarcinomas, pulmonary adenocarcinomas, squamous cell lung carcinomas and pleural mesothelioma. Cancers encompass metastatic cancers of any of the preceding. The oligopeptide antibody or organic molecule is capable of binding to at least a portion of the cancer cells expressing the TAT polypeptide in the mammal. In a preferred embodiment, the oligopeptide antibody or organic molecule is effective to kill or kill tumor cells expressing TAT or to inhibit the growth of such tumor cells, in vitro or in vivo, by binding to the TAT polypeptide in the cell. Such an antibody includes a naked anti-TAT antibody (non-conjugated to any agent). Naked antibodies that have cytotoxic or cell growth inhibition properties can be further enhanced with a cytotoxic agent to make them even more potent in eliminating the tumor cell. Cytotoxic properties can be conferred to any anti-TAT antibody, e.g. , conjugating the antibody with a cytotoxic agent, to form an immunoconjugate as described herein. The cytotoxic agent or a growth inhibiting agent is preferably a minor molecule. Toxins such as calicheamicin or a maytansinoid and analogs or derivatives thereof are preferred. The invention provides a composition comprising an anti-TAT oligopeptide antibody or organic molecule of the invention and a carrier. For the purpose of cancer treatment, the compositions may be administered to the patient in need of such treatment, wherein the compositions may comprise one or more anti-TAT antibodies present as immunoconjugates or as the naked antibody. In a further embodiment, the compositions may comprise these oligopeptide antibodies or organic molecules in combination with other therapeutic agents such as cytotoxic or growth inhibitory agents, including chemotherapeutic agents. The invention also provides - - formulations comprising an anti-TAT oligopeptide antibody or organic molecule of the invention and a carrier. In one embodiment, the formulation is a therapeutic formulation comprising a pharmaceutically acceptable carrier. Another aspect of the invention is isolated nucleic acids encoding anti-TAT antibodies. The nucleic acids encoding both the H and L chains and especially the hypervariable region residues, the chains encoding the native sequence antibody as well as the variants, modifications and humanized versions of the antibody are encompassed. The invention also provides methods useful for the treatment of a cancer expressing the TAT polypeptide or for alleviating one or more cancer symptoms in a mammal, comprising administering a therapeutically effective amount of an anti-TAT oligopeptide antibody or organic molecule to the mammal. The therapeutic compositions of oligopeptide antibody or organic molecule can be administered in the short term (acute) or chronic or intermittent as directed by the physician. Methods for inhibiting growth and destroying a cell expressing the TAT polypeptide are also provided. The invention also provides equipment and articles of manufacture comprising at least one anti-TAT oligopeptide antibody or organic molecule. The equipment containing anti-TAT oligopeptide antibodies or organic molecules find their use, e.g., for TAT cell elimination analysis, for the purification or immunoprecipitation of the TAT polypeptide of the cells. For example, for the isolation and purification of TAT, the kit may contain an anti-TAT oligopeptide antibody or organic molecule coupled to beads. { e.g., Sepharose pearls). Equipment containing oligopeptide antibodies or organic molecules can be provided for the detection and quantification of TAT in vitro, e.g., in an ELISA or immunoblot. Such an oligopeptide antibody or organic molecule useful for detection can be provided with a label such as a fluorescent radioetube. L. Articles of Manufacture and Equipment Another embodiment of the invention is an article of manufacture containing useful materials for the treatment of cancer expressing anti-TAT. The article of manufacture comprises a container and a packaging label or insert in or associated with the container. Suitable containers include, for example, bottles, vials, syringes, etc. The containers can be formed from a variety of materials such as glass or plastic. The container contains a composition that is effective for treating a cancerous condition and can have a sterile access door (e.g. the container can be an intravenous solution bag or a vial having a pierceable obturator by a hypodermic injection needle) . At least one active agent in the composition is an anti-TAT oligopeptide antibody or organic molecule of the invention. The label or package insert indicates that the composition is used to treat cancer. The label or package insert will further comprise instructions for administering the oligopeptide antibody composition or organic molecule for the cancer patient. Additionally, the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may also include other desirable materials from a commercial and user point of view, including other shock absorbers, diluents, filters, needles and syringes. Also provided are equipment that is useful for various purposes, e.g.f for cell elimination analysis that expresses TAT, for the purification or immunoprecipitation of the TAT polypeptide from the cells. For the isolation and purification of the TAT polypeptide, the kit may contain an anti-TAT oligopeptide antibody or organic molecule coupled to beads. { e.g., Sepharose pearls). Equipment containing the oligopeptide antibodies or organic molecules can be provided for the detection and quantification of the TAT in vitro polypeptide, e.g., in an ELISA or immunoblot. As with the article of manufacture, the equipment comprises a container and a label or packing insert in or associated with the container. The container contains a composition comprising at least one anti-TAT oligopeptide antibody or organic molecule of the invention. Additional containers containing, e.g., diluents and buffers, control antibodies may be included. The packaging insert label can provide a description of the composition as well as instructions for intended in vivo or diagnostic use. M. Uses for Nucleic Acids that Code TAT Polypeptides and TAT Polypeptide The nucleotide sequences (or their complement) that encode TAT polypeptides have several applications in the molecular biology technique, including uses as hybridization probes, in chromosome mapping and gene and in the generation of anti-sense RNA and DNA probes. The nucleic acid encoding TAT will also be useful for the preparation of TAT polypeptides by the recombinant techniques described herein, wherein those TAT polypeptides can find their use, for example, in the preparation of anti-TAT antibodies as described herein.
The full-length TAT gene of the native sequence or its portions can be used as hybridization probes for a cDNA library to isolate the full-length TAT cDNA or to isolate yet other cDNAs (e.g., those encoding TAT variants) that occur naturally or TAT of other species) that have a desired sequence identity for the native TAT sequence described herein. Optionally, the length of the probes will be from about 20 to about 50 bases. Hybridization probes can be derived from at least partially new regions of the native total length nucleotide sequence wherein those regions can be determined without undue experimentation or from genomic sequences including promoters, enhancer elements and TAT introns of native sequence. By way of example, a screening method will comprise isolating the coding region of the TAT gene using the known DNA sequence to synthesize a selected probe of about 40 bases. Hybridization probes can be labeled by a variety of labels, including radionucleotides such as 32P or 35S or enzymatic labels such as alkaline phosphatase coupled to the probe via avidin / biotin coupling systems. Labeled probes having a sequence complementary to that of the TAT gene of the present invention can be used to select human cDNA, genomic DNA or mRNA libraries to determine which members of such libraries the probe hybridizes to. Hybridization techniques are described in greater detail in the examples below. Any of the EST sequences described in the present application can be used similarly as probes, using the methods described herein. Other useful fragments of the TAT-encoding nucleic acids include antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to the TAT (sensitive) mRNA or DNA sequences TAT (anti-sensitive). The antisense or sense oligonucleotides, according to the present invention, comprise a fragment of the TAT DNA coding region. Such a fragment generally comprises at least about 14 nucleotides, preferably from about 14 to 30 nucleotides. The ability to derive an anti-sensitive or sensitive oligonucleotide, based on the cDNA sequence encoding a given protein is described, for example, in Stein and Cohen (Cancer Res., 48: 2659, 1988) and van der Krol et al. al., (BioTechniques 6: 958, 1988). Linking the antisense or sense oligonucleotides to the target nucleic acid sequences results in the formation of duplexes that block the transcription or translation of the target sequence by one of several means, including increased cross-linking of the duplexes, the premature termination of transcription or translation or by other means. Such methods are encompassed by the present invention. Thus, anti-sense oligonucleotides can be used to block the expression of proteins ???, wherein those TAT proteins may play a role in the induction of cancer in mammals. The antisense or sense oligonucleotides further comprise oligonucleotides having modified sugar-phosphodiester (or other sugar linker) structures, such as those described in WO 91/06629 and wherein such sugar linkages are resistant to endogenous nucleases. Such oligonucleotides with resistant sugar bonds are stable in vivo (ie, capable of resisting enzymatic cross-linking) but retain sequence specificity to be capable of binding to the target nucleotide sequences Other examples of sense or anti-sense oligonucleotides include those oligonucleotides that are covalently linked to organic halves, such as those described in WO 90/10048 and to other moieties that increase the affinity of the oligonucleotides for a target nucleic acid sequence, such as poly (L-lysine). Still further, intercalating agents, such as ellipticine and alkylating agents or metal complexes can be attached to sense or anti-sense oligonucleotides to modify the binding specificities of the sensitive or anti-sensitive oligonucleotide for the nucleotide sequence. objective. The antisense or sense oligonucleotides can be introduced into a cell containing the target nucleic acid sequence by any method of gene transfer, including, for example, transfection of CaP04-mediated DNA, electroporation or using gene transfer vectors such like the Epstein-Barr virus. In a preferred method, an anti-sensitive or sensitive oligonucleotide is inserted into a suitable retroviral vector. A cell containing the target nucleic acid sequence is contacted with the retroviral recombinant vector either in vivo or ex vivo. Suitable retroviral vectors include, but are not limited to, those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV) or the double copy vectors designated DCT5A, DCT5B and DCT5C (see WO 90/13641). . Sense or antisense oligonucleotides may also be introduced into a cell containing the target nucleotide sequence by the formation of a conjugate with a ligand-binding molecule, as described in O91 / 04753. Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand-to-ligand molecules does not substantially interfere with the ability of the ligand-binding molecule to bind to its corresponding molecule or receptor or block entry of the sense or antisense oligonucleotide or its conjugated version in the cell. Alternatively, a sense or antisense oligopeptide can be introduced into a cell containing the target nucleic acid sequence by forming an oligonucleotide-lipid complex, as described in WO 90/10448. The sense or antisense oligonucleotide-lipid complex preferably dissociates within the cell by an endogenous lipase. The A or antisense or sense DNA molecules are generally at least about 5 nucleotides in length, alternatively at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, S5, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, - - 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990 or 1000 nucleotides in length, wherein in this context the term "approximately" means the length of the nucleotide sequence referred to plus or minus 10% of that referred length. The probes can also be used in PCR techniques to generate a cluster of sequences for the identification of closely related TAT coding sequences. The nucleotide sequences encoding a TAT can also be used to construct hybridization probes to represent the gene encoding that TAT and for the genetic analysis of individuals with genetic disorders. The nucleotide sequences provided herein may be represented for a chromosome and specific regions of a chromosome using known techniques, such as in situ hybridization, binding analysis against known chromosomal markers and selection by hybridization with the libraries. When the coding sequences for TAT encode a protein that binds to another protein - - (example, where TAT is a receptor), TAT can be used in assays to identify the other proteins or molecules involved in the interaction of the link. By such methods, inhibitors of receptor / ligand link interaction can be identified. The proteins involved in such linkage interactions can also be used to classify the peptide or small molecule inhibitors or agonists of the linkage interaction. Also, the TAT receptor can be used to isolate the correlating ligand (s). Selection tests can be designed to find guiding compounds that mimic the biological activity of a natural TAT or a receptor for TAT. Such screening assays will include docile assays for high throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates. The contemplated small molecules include synthetic organic or inorganic compounds. The assays can be carried out in a variety of formats, including protein-protein binding assays, biochemical selection assays, immunoassays and cell-based assays, which are well characterized in the art. The nucleic acids encoding TAT or its modified forms can also be used to generate either transgenic animals or "knock out" animals which, in turn, - - they are useful in the development and selection of therapeutically useful reagents. A transgenic animal (e.g., a mouse or rat) is an animal that has cells that contain a transgene whose transgene was introduced into the animal or an ancestor of the animal in a prenatal, e.g. an embryonic stage. A transgene is a DNA which is integrated into the genome of a cell from which a transgenic animal develops. In one embodiment, the cDNA encoding TAT can be used to clone genomic DNA encoding TAT according to established techniques and the genomic sequences used to generate transgenic animals that contain cells expressing the DNA encoding TAT. Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in US Patents. Nos. 4,736,866 and 4,870,009. Typically, particular cells can be targeted for incorporation of the TAT transgene with tissue-specific enhancers. Transgenic animals that include a copy of a transgene encoding TAT introduced into the germ line of the animal at an embryonic stage can be used to examine the effect of increased expression of the DNA encoding TAT. Such animals can be used as test animals for the reagents considered to confer protection from, for example, conditions - - pathological conditions associated with its overexpression. According to this facet of the invention, an animal is treated with the reagent and a reduced incidence of the pathological condition, compared to untreated animals carrying the transgene, may indicate a potential therapeutic intervention for the pathological condition. Alternatively, non-human TAT homologs can be used to construct a TAT knock out animal that has a defective or altered gene encoding TAT as a result of homologous recombination between the endogenous TAT-encoding gene and the altered genomic DNA encoding TAT. TAT introduced into an embryonic germ cell of the animal. For example, the cDNA encoding TAT can be used to clone the genomic DNA encoding TAT according to established techniques. A portion of the genomic DNA encoding TAT can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor the integration. Typically, several kilobases of unaltered flanking DNA (both at the 5 'and 3' ends) are included in the vector [see eg, Thomas and Capecchi, Cell, 51: 503 (1987) for a description of homologous recombination vectors. ] The vector is introduced into an embryonic germ cell line (e.g., by electroporation) and the cells in which the introduced DNA is selected are selected. has recombined homologously with endogenous DNA [see e.g., Li et al., Cell, 69: 915 (1992)]. The selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras [see e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach,. { Teratocarcinomas and Embryonic Germ Cells: A Practical Procedure) E. J. Robertson, ed. (IRL Oxford, 1987), pp. 113-152]. A chimeric embryo can then be implanted into a bred animal, a suitable pseudopregnant female and the embryo brought to term to create a "knock out" animal. The offspring that harbor the homologously recombined DNA in their germ cells can be identified by standard techniques and used for created animals in which all the cells of the animals contain the homologously recombined DNA. The knock out animals can be characterized, for example, by their ability to defend against certain pathological conditions and by their development of pathological conditions due to the absence of the TAT polypeptide. The nucleic acid encoding the TAT polypeptides can also be used in gene therapy. In gene therapy applications, genes are introduced into the cells in order to achieve the in vivo synthesis of a therapeutically effective gene product, for example, for the replacement of a defective gene. The "therapy - 2 5 - Genetics "includes both conventional gene therapy where a lasting effect is achieved by a single treatment and in the administration of therapeutic agents of the gene, involving the one-time or repeated administration of a therapeutically effective DNA or mRNA.
Antisense DNAs and RNAs can be used as therapeutic agents to block in vivo expression of certain genes. It has already been shown that short antisense oligonucleotides can be imported into cells where they act as inhibitors, despite their low intracellular concentrations caused by their limited absorption by the cell membrane. (Zamecnik et al., Proc. Nati, Acad. Sci. USA 83: 4143-4146 [1986]). The oligonucleotides can be modified to improve their absorption, e.g., by replacing their negatively charged phosphodiester groups with non-charged groups. There are a variety of techniques available to introduce nucleic acids into viable cells. The techniques vary depending on whether the nucleic acid is transferred into cells grown in vitro or in vivo in the cells of the proposed host. Suitable techniques for the transfer of nucleic acids in mammalian cells in vivo include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, calcium phosphate precipitation method, etc. Currently preferred in vivo gene transfer techniques include transfection with viral vectors (typically retroviral) and transfection mediated by the protein-liposome viral coat (Dzau et al., Trends in Biotechnology 11, 205-210 [1993] ]). In some situations it is desirable to provide the nucleic acid source with an agent that targets the target cells, such as an antibody specific for a cell surface membrane protein or the target cell, a ligand for a receptor in the target cell, etc. Where liposomes are employed, proteins that bind to a cell surface membrane protein associated with endocytosis can be used to direct and / or facilitate absorption, eg, capsid proteins or fragments thereof, tropics for a type of particular cell, antibodies for proteins that undergo internalization in the cycle of proteins that direct intracellular localization and improve the intracellular half-life. The technique of receptor-mediated endocytosis is described, for example, by Wu et al., J. Biol. Chem. 262, 4429-4432 (1987); and Agner et al., Proc. Nati Acad. Sci. USA 87, 3410-3414 (1990). For the review of gene labeling and gene therapy protocols see Anderson et al., Science 256, 808-813 (1992). The nucleic acid molecules encoding the TAT polypeptides or fragments thereof described herein are useful for the identification of the chromosome. In this regard, there is an ongoing need to identify new chromosome markers since relatively few chromosome labeling reagents are currently available, based on the current sequence data. Each TAT nucleic acid molecule of the present invention can be used as a chromosome marker. The TAT polypeptides and nucleic acid molecules of the present invention can also be used diagnostically for histotyping, wherein the TAT polypeptides of the present invention can be differentially expressed in one tissue compared to another, preferably in a diseased tissue compared to a normal tissue of the same type of tissue. The TAT nucleic acid molecules will find use to generate probes for PCR, Northern analysis, Southern analysis and Western analysis. This invention encompasses methods for screening compounds to identify those that mimic the TAT polypeptide (agonists) or prevent the effect of the TAT polypeptide (antagonists). Selection tests for antagonist drug candidates are designed to identify compounds that bind or complex with the TAT polypeptides encoded by the genes identified herein or otherwise interfere with the interaction of - the polypeptides encoded with other cellular proteins, including e.g., inhibit the expression of the TAT polypeptide from the cells. Such screening assays will include available assays for high throughput screening of chemical libraries, marking those particularly suitable for identifying small molecule drug candidates. The assays can be carried out in a variety of formats, including protein-protein binding assays, biochemical selection assays, immunoassays and cell-based assays, which are well characterized in the art. All assays for antagonists are common in that they call for contact with the drug candidate with a TAT polypeptide encoded by a nucleic acid identified herein under conditions and for a sufficient time to allow these two components to interact. In binding assays, the interaction is the binding and the complex formed can be isolated or detected in the reaction mixture. In a particular embodiment, the TAT polypeptide encoded by the gene identified herein or the drug candidate is immobilized on a solid phase, e.g., over a microtiter plate, by covalent or non-covalent linkages. The non-covalent binding is usually carried out by covering the solid surface with a solution of the TAT polypeptide and drying. Alternatively, an immobilized antibody, e.g., a monoclonal antibody, specific for the TAT polypeptide to be immobilized can be used to anchor it to a solid surface. The test is carried out by adding the non-immobilized component, which can be marked by a detectable label, for the immobilized component, e.g., the covered surface containing the anchored component. When the reaction is complete, the unreacted components are removed, e.g., by washing and the complexes anchored on the solid surface are detected. When the originally non-immobilized components carry a detectable mark, the detection of the immobilized mark on the surface indicates that the adjustment occurred. Where the originally non-immobilized components do not carry a label, complexation can be detected, for example, by using a labeled antibody specifically binding the immobilized complex. If the inte candidate compound acts but does not bind to a particular TAT polypeptide encoded by a gene identified herein, its interaction with that polypeptide can be assayed by well-known methods for detecting protein-protein interactions. Such assays include traditional methods, such as, e.g., cross-linking, co-immunoprecipitation and co-purification through gradients or chromatographic columns. In addition, protein-protein interactions can be monitored by using a yeast-based genetic system described by Fields et al. (Fields and Song, Nature (London), 340: 245-246 (1989); Chien et al., Proc. Nati Acad. Sci. USA, 88: 9578-9582 (1991)) as described by Chevray and Nathans, Proc. Nati Acad. Sci. USA, 89: 5789-5793 (1991). Many transcriptional activators, such as yeast GAL4, consist of two physically separate modular domains, one acts as the DNA binding domain, the other functions as the transcription activation domain. The yeast expression system described in the above publications (generally referred to as the "two-hybrid system") takes advantage of this property and employs two hybrid proteins, one in which the target protein is fused to the DNA binding domain of GAL4 and the other in which the candidate activation proteins are fused to the activation domain. The expression of a gene that reports GALl-lacZ low in control of a promoter activated by GAL4 depends on the reconstitution of GAL4 activity through a protein-protein interaction. Colonies containing interacting polypeptides are detected with a chromogenic substrate for β-galactosidase. A complete team (MATCHMAKER ™) to identify protein-protein interactions between two specific proteins using - - The two-hybrid technique is commercially available by Clontech. This system can also be extended to represent the protein domains involved in the specific interactions of the protein as well as to precisely locate the amino acid residues that are crucial in these interactions. Compounds that interfere with the interaction of a gene encoding a TAT polypeptide identified herein and other intra- or extracellular components can be tested as follows: usually a reaction mixture containing the product of the gene and the intra- or extracellular under the conditions and for a time that allows the interaction and union of the two products. To test the ability of a candidate compound to inhibit binding, the reaction is run in the absence and in the presence of the test compound. In addition, a placebo can be added for a third reaction mixture, to serve as a positive control. The binding (complex formation) between the test compound and the intra- or extracellular component present in the mixture is monitored as described here before. The formation of a complex in the control reaction (s) but not in the reaction mixture contains the test compound which indicates that the test compound interferes with the interaction of the test compound and its reaction partner.
For assay of the antagonists, the TAT polypeptide can be added to a cell together with the compound to be selected for a particular activity and the ability of the compound to inhibit the activity of interest in the presence of the TAT polypeptide indicates that the compound is an antagonist for the TAT polypeptide. Alternatively, antagonists can be detected by combining the TAT polypeptide and a potential antagonist with the TAT polypeptide receptors that bind to the membrane or recombinant receptors under conditions appropriate for a competitive inhibition assay. The TAT polypeptide can be labeled, such as by radioactivity, such that the number of TAT polypeptide molecules affixed to the receptor can be used to determine the effectiveness of the potential antagonist. The gene encoding the receptor can be identified by various methods known to those of skill in the art, for example, panning of ligand and FACS classification. Coligan et al., Current Protocols in Immun. 1 (2): Chapter 5 (1991). Preferably, the term "cloning" is used wherein polyadenylated AR is prepared from a cell that responds to the TAT polypeptide and a cDNA library created from this RNA, which is divided into clusters and used to transfect COS cells or other cells that do not respond to the TAT polypeptide. The transfected cells that develop on the glass slides are exposed to the labeled TAT polypeptide. The TAT polypeptide can be labeled by a variety of means including impregnation with iodine or the inclusion of a recognition site for a site-specific protein kinase. After fixation and incubation, the slides are subjected to autoradiographic analysis. Positive clusters are identified and sub-clusters are prepared and re-transfected using an interactive sub-clustering and a re-classification process, eventually producing a single clone encoding the putative receptor. As an alternative method for receptor identification, the labeled TAT polypeptide can be linked by photoaffinity to the cell membrane or extract preparations expressing the receptor molecule. The crosslinked material is reduced by PAGE and exposed to the X-ray film. The labeled complex containing the receptor can be cut, reduced into peptide fragments and subjected to protein micro-sequencing. The amino acid sequence obtained from the micro-sequencing can be used to design a set of degenerate oligonucleotide probes to classify a cDNA library to identify the gene encoding the putative receptor. In another assay for the antagonists, the cells - - of mammal or a membrane preparation expressing the receptor can be incubated with a labeled TAT polypeptide in the presence of the candidate compound. The ability of the compound to improve or block this interaction can then be measured. More specific examples of potential antagonists include an oligonucleotide that binds to immunoglobulin fusions with a TAT polypeptide, and in particular, antibodies that include, without limitation, antibodies and fragments of poly- and monoclonal antibodies, single chain antibodies, anti- idiotypic and chimeric or humanized versions of such antibodies or fragments, as well as antibodies and human antibody fragments. Alternatively, a potential antagonist may be a closely related protein, for example, a mutated form of a TAT polypeptide that recognizes the receptor but imparts no effect, thereby competitively inhibiting the action of the TAT polypeptide. Another potential TAT polypeptide antagonist is an antisense RNA or DNA construct prepared using antisense technology, wherein, eg, an AR molecule or antisense DNA acts to directly block the translation of the mRNA by hybridization to the target ARMn and avoid the translation of the protein. Antisense technology can be used to control the - - expression of the gene through triple helix formation or antisense DNA or RNA, both of these methods being based on the binding of a polynucleotide to DNA or RNA. For example, the 5 'coding portion of the polynucleotide sequence, which encodes the mature TAT polypeptides herein, is used to design an antisense RNA oligonucleotide of from about 10 to 0 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix see Lee et al., Nuci, Acid Res. 5: 3073 (1979), Cooney et al., Science, 241: 456 (1988); Dervan et al., Science, 251: 1360 (1991)), thus avoiding transcription and production of the TAT polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks the translation of the mRNA molecule into the TAT polypeptide (antisense - Okano, Neurochem., 56: 560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression (Oligodeoxynucleotides as Inhibitors Antisense of Gene Expression) (CRC Press: Boca Raton, FL, 1988) The oligonucleotides described above can also be delivered to the cells in such a way that the antisense RNA or DNA can be expressed in vivo to inhibit polypeptide production TAT When antisense DNA is used, the derivative oligodeoxyribonucleotides are preferred from the translation-initiation site, eg, - - between about -10 and +10 positions of the nucleotide sequence of the target gene. Potential antagonists include small molecules that bind to the active site, the receptor binding site or the growth factor or other relevant binding site of the TAT polypeptide, thereby blocking the normal biological activity of the TAT polypeptide. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules, preferably soluble peptides and synthetic organic or inorganic peptidyl compounds. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by specific hybridization of the sequence to the complementary target RNA, followed by endonucleolytic cleavage. Specific ribosome cleavage sites within a potential RNA target can be identified by known techniques. For further details see, e.g., Rossi, Current Biology, 4: 469-471 (1994) and PCT publication No. WO 97/33551 (published September 18)., 1997). Nucleic acid molecules in triple helix formation used to inhibit transcription must be single-stranded and composed of deoxynucleotides. The base composition of these oligonucleotides is designed in such a way as to promote triple helix formation through Hoogsteen base pair rules, which generally require considerable elongations of purines or pyrimidines in a strand of a duplex. For further details see e.g., PCT publication No. WO 97/33551, supra. These small molecules can be identified by any of one or more selection assays described hereinbefore and / or by any other selection technique well known to those skilled in the art. The nucleic acid encoding the isolated TAT polypeptide can be used herein to recombinantly produce the TAT polypeptide using techniques well known in the art as described herein. In turn, the TAT polypeptides produced can be used to generate anti-TAT antibodies using techniques well known in the art and as described herein. Antibodies that specifically bind to the TAT polypeptide identified herein, as well as other molecules identified by the screening assays heretofore described, can be administered for the treatment of various disorders, including cancer, in the form of pharmaceutical compositions. If the TAT polypeptide is intracellular and the whole antibodies are used as inhibitors, the internalization of the antibodies is preferred. Nevertheless, - - The lipofections or liposomes can also be used to deliver the antibody or an antibody fragment into the cells. When the antibody fragments are used, the smaller inhibitory fragment that binds specifically to the binding domain of the target protein is preferred. For example, based on the variable region sequences of an antibody, the peptide molecules can be designed to retain the ability to bind to the sequence of the target protein. Such peptides can be chemically synthesized and / or produced by recombinant DNA technology. See, e.g., Marasco et al., Proc. Nati Acad. Sci. USA, 90: 7889-7893 (1993). The present formulation may also contain more than one active compound as necessary for the particular indication in question, preferably those with complementary activities that do not adversely affect each other. Alternatively or in addition, the composition may comprise an agent that enhances its function, such as for example, a cytotoxic agent, cytokine, chemotherapeutic agent or growth inhibitory agent. Such molecules are suitably present in combination in amounts that are effective for the purpose proposed. The following examples are offered for purposes of illustration only and are not intended to limit the scope of the present invention in any way. All patents and literature references cited in the present specification are incorporated herein by reference in their entirety. EXAMPLES Commercially available reagents referred to in the examples were used in accordance with the manufacturers' instructions unless otherwise indicated. The source of these cells identified in the following examples and through the specification, using the ATCC access numbers is the American Type Culture Collection, Manassas, VA. EXAMPLE 1; Tissue Expression Profile Using GeneExpress® The patented database containing the gene expression information (GeneExpress®, Gene Logic Inc., Gaithersburg, MD) was analyzed in an attempt to identify polypeptides (and their nucleic acids). coding) whose expression is significantly upregulated in a particular tumor tissue (s) of interest in comparison to other tumor (s) and / or normal tissues. Specifically, the analysis of the GeneExpress® database was conducted using either software available through Gene Logic Inc., Gaithersburg, MD, to be used with the database - - GeneExpress® or with the patented software written and developed in Genentech, Inc., to be used with the GeneExpress® database. The classification of the positive ranges in the analysis are based on several criteria including, for example, tissue specificity, tumor specificity and level of expression in normal essential tissues and / or normal proliferation. The following is a list of molecules whose tissue expression profile as determined from an analysis of the GeneExpress® database evidences high tissue expression and significant up-regulation of expression in a tumor or specific tumors compared to other tumor (s) and / or normal tissues and optionally relatively low expression in normal and / or normal proliferating tissues. As such, the molecules listed below are excellent polypeptide targets for the diagnosis and therapy of cancer in mammals.
Molecule Overregulation of the Compared to: Expression in: DNA45234-1277 (TAT291) breast tumor normal breast tissue DNA45234-1277 (TAT291) ovarian tumor normal ovarian tissue DNA45234-1277 (TAT291) Uterine uterine tissue DNA37151-1193 (TAT292) Breast tumor normal breast tissue DNA37151-1193 (TAT292) Tubal tumor of normal fallopian tube tissue - Fallopian DNA37151-1193 (TAT292) Normal ovarian tissue ovarian tumor DNA37151-1193 (TAT292) Uterine Tumor Uterine Normal Tissue EXAMPLE 2: Microassay Analysis to Detect Over-regulation of TAT Polypeptides in Cancerous Tumors Nucleic acid microassays often contain thousands of gene sequences, which are useful for differentially identifying genes expressed in diseased tissues compared to their normal counterparts. Using nucleic acid microassays, tests and samples of AR m control tests and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The cDNA tests are then hybridized to an immobilized nucleic acid array on a solid support. The arrangement is configured in such a way that the sequence and position of each member of the arrangement is known. For example, a selection of genes known to be expressed in certain disease states can be arranged on a solid support. Hybridization of a labeled probe with a particular disposed member indicates that the sample from which it is derived expresses that gene. If the hybridization signal of a probe from a test sample (diseased tissue) is greater than the hybridization signal of a probe from a control sample (normal tissue), the gene is identified or - - genes over-expressed in diseased tissue. The implication of this result is that an overexpressed protein in a diseased tissue is useful not only as a diagnostic marker for the presence of the disease condition, but also as a therapeutic target for the treatment of the disease condition. The methodology of hybridization of nucleic acids and micro-assay technology is well known in the art. In one example, the specific preparation of nucleic acids for hybridization and probes, slides and hybridization conditions are all detailed in PCT Patent Application Series No. PCT / US01 / 10482, filed on March 30, 2001 and which is incorporated in the presentation by reference. In the present example, cancerous tumors derived from various human tissues were studied for upregulated expression of the gene in relation to cancerous tumors of different types of tissue and / or non-cancerous human tissues in an attempt to identify those polypeptides that are found Overexpressed in particular cancerous tumor (s). In certain experiments, human tissue from cancerous tumor and non-cancerous tumor human tissue of the same type of tissue (often from the same patient) was obtained and analyzed for the expression of TAT polypeptides. Additionally, cancerous human tissue was obtained from any of a variety of different human tumors and compared with a "universal" epithelial control sample that was prepared by pooling non-cancerous human tissues of epithelial origin, including hepatic, renal and pulmonary. The mRNA isolated from the stored tissues represents a mixture of the gene products expressed from these different tissues. The micro-assay hybridization experiments using the stored control samples generated a linear scheme in a 2-color analysis. The curve of the line generated in a 2-color analysis was then used to normalize the proportions (test detection: control) within each experiment. The normalized ratios of several experiments were then compared and used to identify gene expression pools. In this way, the "universal control" sample stored not only allowed effective relative determinations of gene expression in a simple comparison of 2 samples, but also comparisons of multiple samples through several experiments. In the present experiments, the nucleic acid probes derived from the nucleic acid sequences encoding the TAT polypeptide described herein were used in the creation of the micro assay and the RNA from various tumor tissues was used for hybridization thereto.
- - Below are shown the results of these experiments, demonstrating that several of the TAT polypeptides of the present invention are significantly overexpressed in various human tumor tissues compared to other human tumor tissues and / or non-cancerous human tissue (s) (s). ). As described above, these data demonstrate that the TAT polypeptides of the present invention are useful not only as diagnostic markers of the presence of one or more cancerous tumors, but also that they serve as therapeutic targets for the treatment of those tumors. Molecule Overregulation of the E comparison to: Expressed in: DNA45234-1277 (TAT291) breast tumor Epithelial control of normal breast tissue EXAMPLE 3: Verification of the Differential Expression of TAT polypeptides by GEPIS TAT polypeptides that may have been identified as tumor antigen as described in one or more of the above Examples were analyzed and verified as follows. We investigated an ADM database of expressed sequence mark (EST) (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) and interesting EST sequences were identified by GEPIS. The profiling of gene expression in silicon (GEPIS) is a tool of - - bioinformatics developed in Genentech, Inc. that characterizes the genes of interest for new therapeutic targets of cancer. GEPIS takes advantage of large amounts of information from the EST sequence and from the library to determine the expression profiles of the gene. GEPIS is able to determine the expression profile of a gene based on its proportional correlation with the number of its occurrences in the EST databases, and it works by integrating the LIFESEQ® EST relational database and the Genentech own information of a rigorously and statistically significant way. In this example, GEPIS is used to cross-validate and validate the new tumor antigens, although GEPIS can be configured to perform both very specific analyzes and wide-ranging tasks. For initial screening, GEPIS is used to identify the EST sequences of the LIFESE Q® database that correlate with expression in a particular tissue or tissues of interest (often a tumor tissue of interest). The EST sequences identified in this initial selection (or the consensus sequences obtained from the alignment of multiple related and overlapping EST sequences obtained from the initial selection) were then subjected to a proposed selection to identify the presence of at least one transmembrane domain in the encoded protein. Finally, GEPIS was used to generate a complete profile of tissue expression for the various sequences of interest. Using this type of selection bioinformatics, several TAT polypeptides (and their nucleic acid coding molecules) were identified as significantly overexpressed in a particular type of cancer or certain cancers compared to other cancers and / or non-cancerous normal tissues. The classification of GEPIS hits is based on several criteria including, for example, tissue specificity, tumor specificity and the level of expression in normal and / or normal tissues of proliferation. The following is a list of molecules whose tissue expression profile determined by GEPIS demonstrates high tissue expression and significant upregulation of expression in a tumor or specific tumors compared to other tumor (s) and / or normal tissues and optionally relatively low expression in normal and / or normal proliferation tissues. As such, the molecules listed below are excellent polypeptide targets for the diagnosis and therapy of cancer in mammals. Molecule Upregulation of the Compared to: Expression in: DNA45234-1277 (TAT291) breast tumor normal breast tissue DNA45234-1277 (TAT291) ovarian tumor normal ovarian tissue DNA45234-1277 (TAT291) uterine tumor normal uterine tissue - - DNA37151-1193 (TAT292) breast tumor normal breast tissue DNA37151-1193 (TAT292) fallopian tube tissue tumor of normal fallopian tube DNA37151-1193 (TAT292) ovarian tumor normal ovarian tissue DNA37151-1193 (TAT292) uterine tumor normal uterine tissue EXAMPLE 4: The use of TAT as a hybridization probe The following method describes the use of a nucleotide sequence encoding TAT as a hybridization probe for ie, the diagnosis of the presence of a tumor in a mammal. The DNA comprising the full-length coding sequence or the mature TAT as described herein can also be used as a probe for the selection of homologous DNAs (such as those encoding the naturally occurring TAT variants) in the human tissue cDNA libraries or the human tissue genomic libraries. Hybridization and washing of the filters containing either library DNAs are carried out under the following highly stringent conditions. Hybridization of the radiolabelled TAT-derived probe for the filters is carried out in a 50% formamide solution, 5 x SSC, 0.1% SDS, 0.1% sodium pyrophosphate, 50 mM sodium phosphate, pH 6.8, 2 x Denhardt's solution and 10% dextran sulfate at 42 ° C for 20 hours. The washing of the filters was carried out in an aqueous solution of 0.1 x SSC and 1% SDS at 42 ° C. The DNAs that have a desired sequence identity with the DNA that encodes ??? of full-length native sequence can then be identified using standard techniques known in the art. EXAMPLE 5: Expression of TAT in E. coli This example illustrates the preparation of a non-glycosylated form of TAT by recombinant expression in E. coli. The DNA sequence encoding TAT is initially amplified using selected PC primers. The primers must contain restriction enzyme sites corresponding to the restriction enzyme sites in the selected expression vector. A variety of expression vectors can be employed. An example of a suitable vector is pBR322 (derived from E. coli, see Bolivar et al., Gene, 2_: 95 (1977)) which contains genes for resistance to ampicillin and tetracycline. The vector is digested with the restriction enzyme and dephosphorylated. The amplified PCR sequences are then ligated to the vector. The vector will preferably include sequences coding for an antibiotic resistance gene, a trp promoter, a polyhis conductor (including the first six STII codons, the polyhis sequence and the cleavage site of the eteroquinasa), the TAT coding region, the lambda transcripcxonal terminator and an argU gene. The ligation mixture is then used to transform a selected E. coli strain using the methods described by Sambrook et al., Supra. Transformants are identified by their ability to grow on LB plates and antibiotic-resistant colonies are then selected. Plasmid DNA can be isolated and confirmed by restriction analysis and DNA sequencing. The selected clones can grow overnight in a liquid culture medium such as LBed supplemented with antibiotics. The overnight culture can be used subsequently to inoculate a larger scale culture. The cells then grow to a desired optical density during which the expression promoter is initiated. After culturing the cells for several more hours, the cells can be harvested by centrifugation. The cell pellet obtained by centrifugation can be solubilized using various agents known in the art and the solubilized TAT protein can then be purified using a metal chelating column under conditions that allow the tight junction of the protein. TAT can be expressed in E. coli in a poly-His labeled form, using the following procedure. The DNA encoding TAT is initially amplified using selected PCR primers. The primers will contain restriction enzyme sites corresponding to the restriction enzyme sites in the selected expression vector and other useful sequences provided for efficient and reliable initiation of translation, rapid purification on a metal chelation column and removal of the proteolytic with enterokinase. The poly-His labeled sequences amplified by PCR are then ligated to an expression vector, which is used to transform an E. coli host on the basis of strain 52 (W3110 fuhA (tonA) Ion galE rpoHts (htpRts) clpP (laclq The transformants grow first in LB containing 50 mg / ml carbenicillin at 30 ° C with shaking until an OD600 of 3-5 is reached.The cultures are then diluted 50-100 fold in the CRAP medium (prepared at mix 3.57g of (NH4) 2S0, 0.71 g of sodium citrate-2H20, 1.07 g of KCl, 5.36 g of Difco yeast extract, 5.36 g of SF Sheffield hicase in 500 ml of water, as well as 110 mM of MPOS, pH 7.3, 0.55% (w / v) glucose and 7 mM MgSO4) and grows for approximately 20-30 hours at 30 ° C with shaking.The samples are removed to verify expression by SDS-PAGE analysis and culture Volume is centrifuged to pellet the cells.The pellets of cells are frozen until purification and - - fold. The paste of E. coli from 0.5 to 1 1 of fermentations (pellets of 6-10 g) is resuspended in 10 volumes (/ v) in 7 M of guanidine, 20 m of Tris, buffer of pH 8. Sodium sulfite Solid and sodium tetraionate are added to make the final concentrations of 0.1 M and 0.02 M respectively and the solution is stirred overnight at 4 ° C. This step results in a denatured protein with all the cysteine residues blocked by sulfitolization. The solution is centrifuged at 40,000 rpm in a Beckman Ultracentrxfuga for 30 minutes. The supernatant was diluted with 3-5 volumes of metal chelate column buffer (6 M guanidine, 20 mM Tris, pH 7.4) and filtered through 0.22 micron filters to clarify. The clarified extract was loaded onto a 5 ml Quiagen Mi-NTA metal chelate column equilibrated with the metal chelate column buffer. The column was washed with additional buffer containing 50 mM imidazole (Calbiochem, class Utrol), pH 7.4. The protexin was extracted with buffer containing 250 mM of imidazole. The fractions containing the desired protease are pooled and stored at 4 ° C. The concentration of the protexin is estimated by its absorbance at 280 nm using the destruction coefficient calculated on the basis of its amino acid sequence.
The proteins are refolded by diluting the sample slowly in fresh prepared reflux buffer consisting of: 20 mM Tris, pH 8.6, 0.3 M NaCl, 2.5 M urea, 5 mM cysteine, 20 mM glycine and 1 mM of EDTA. The fold volumes are chosen such that the final concentration of the protein is between 50 to 100 micrograms / ml. The refolding solution is gently stirred at 4 ° C for 12-36 hours. The refolding reaction is cooled by the addition of TFA to a final concentration of 0.4% (pH of about 3). Before further purification of the protein, the solution is filtered through a 0.22 micron filter and the acetonitrile is added to the final concentration of 2-10%. The refolded protein is chromatographed on a Poros Rl / H reversed phase column using a movable shock absorber of 0.1% TFA with elution with an acetonitrile gradient of 10 to 80%. The aliquots of the fractions with an A280 absorbance are analyzed on SDS polyacrylamide gels and fractions containing the homogeneous refolded protein are pooled. Generally, adequately refolded species of most proteins are eluted at lower concentrations of acetonitrile since those species are the most compact with their hydrophobic interiors protected from interaction with the reverse phase resin. Aggregate species elute - - usually at higher acetonitrile concentrations. In addition to solving the unfolded forms of the proteins from the desired form, the reverse phase step also removes endotoxin from the samples. The fractions containing the desired refolded TAT polypeptide are pooled and the acetonitrile is removed using a gentle stream of nitrogen directed to the solution. The proteins are formulated in 20 mM Hepes, pH 6.8 with 0.14 M sodium chloride and 4% mannitol by dialysis or by gel filtration using GF Superfine resins (Pharmacia) equilibrated in the buffer of the formulation and filtered in a manner sterile Certain of the TAT polypeptides described herein have been expressed and successfully purified using this technique (s). EXAMPLE 6: Expression of TAT in mammalian cells This example illustrates the preparation of a potentially glycosylated form of TAT by recombinant expression in mammalian cells. The vector, pRK5 (see EP 307,247 published in March of 1989) is used as the expression vector. Optionally, the TAT DNA is ligated into pRK5 with selected restriction enzymes to allow insertion of the TAT DNA using ligation methods such as described by Sambrook et al., supra. The resulting vector is called - - pRK5 - TAT. In one embodiment, the selected host cells may be 293 cells (ATCC CCL 1573) that grow to confluent in the tissue culture plates in the medium such as DMEM supplemented with fetal calf serum and optionally nutrient components and / or antibiotics. Approximately 10 g of pRK5-TAT DNA are mixed with about 1 g of DNA encoding the RNA VA gene [Thimmapaya et al., Cell, 31: 543 (1982)] and dissolved in 500 μ? of 1 mM Tris-HCl, 0.1 mM EDTA, 0.227 M CaCl2. To this mixture is added dropwise, 500 μ? 50 mM HEPES (pH 7.35), 280 mM NaCl, 1.5 mM NaP04 and a precipitate is left to form for 10 minutes at 25 ° C. The precipitate is suspended and added to the 293 cells and allowed to stand for about four hours at 37 ° C. The culture medium is left to extract and 2 ml of 20% glycerol are added for 30 seconds. The 293 cells are then washed with a serum-free medium, the fresh medium is added and the cells are incubated for approximately 5 days. Approximately 24 hours after the transfections, the culture medium is removed and replaced with a culture medium (alone) or culture medium containing 200 μ ?? / ??? of 35S-cysteine and 200 of 35S-methionine. After 12 hours of incubation, the conditioned medium is collected, concentrated in a centrifugal filter and loaded in 15% SDS gel. The processed gel can be dried and exposed to the film for a selected period of time to reveal the presence of the TAT polypeptide. Cultures containing transfected cells may undergo additional incubation (in serum-free medium) and the medium is tested in the selected bioassays. In an alternative technique, TAT can be introduced temporarily into the 293 cells using the dextran sulfate method described by Somparyrac et al., Proc. Nati Acad. Sci. , 12: 7575 (1981). The 293 cells grow to the maximum density in a rotating flask and 700 μg of pRK5-TAT DNA are added. The cells are first concentrated from the spinner flask by centrifugation and washed with PBS. The DNA dextran precipitate is incubated in the cell pellet for four hours. The cells are treated with 20% glycerol for 90 seconds, washed with the tissue culture medium and reintroduced into the spinner flask containing the tissue culture medium, 5 μg / ml bovine insulin and 0.1 μg / ml. of bovine transferrin. After about four days, the conditioned medium is centrifuged and filtered to remove the cells and debris. The sample containing expressed TAT can then be concentrated and purified by any selected method, such as dialysis and / or column chromatography. In another embodiment, TAT can be expressed in CHO cells. PRK5-TAT can be transfected into CHO cells using known reagents such as CaP04 or DEAE-dextran. As described above, the cell cultures can be incubated and the medium replaced with the culture medium (alone) or the medium containing a radiolabel such as 35S-methionine. After determining the presence of the TAT polypeptide, the culture medium can be replaced with the serum free medium. Preferably, the cultures are incubated for approximately 6 days and the conditioned medium is then harvested. The medium containing TAT expressed can then be concentrated and purified by any selected method. The labeled TAT of Epitope can also be expressed in the CHO host cells. The TAT can be subcloned out of the pRK5 vector. The subclone insert can be subjected to PCR to fuse into the structure with a selected epitope tag such as a poly-his tag in a Baculovirus expression vector. The labeled poly-his TAT insert can then be subcloned into a driven SV40 vector containing a selection marker such as DHFR for the selection of stable clones. Finally, CHO cells can be transfected (as described above) with the driven SV40 vector. The marking can be carried out, as described above, to verify the expression. The culture medium containing the expressed poly-His TAT can then be concentrated and purified by a selected method, such as by affinity chromatography Ni2 + -kelate. TAT can also be expressed in CHO and / or COS cells by a transient expression method or in CHO cells by another stable expression method. Stable expression in CHO cells is carried out using the following procedure. The proteins are expressed as an IgG construct (immunoadhesin), in which the coding sequences for the soluble forms. { e.g., extracellular domains) of the respective proteins are fused to an IgG1 constant region sequence containing the CH2 joint and the CH2 domains and / or is a poly-His labeled form. After PCR amplification, the respective DNAs were subcloned into a CHO expression vector using standard techniques as described by Ausubel et al. , Current Protocols of Molecular Biology, Unit 3.16, John Wiley and Sons (1997). CHO expression vectors are constructed to have compatible restriction sites 5 'and 3' of the DNA of interest to allow convenient movement of cDNAs. The vector that uses expression in CHO cells as described by Lucas et al., Nucí. Acids Res. 24: 9 (1774-1779 (1996) and uses of the promoter / enhancer to direct the expression of ADMc of interest and reductase dihydrofolate (DHFR) .The expression of DHFR allows selection for stable maintenance of the plasmid following the Transfection: Twelve micrograms of the desired plasmid DNA was introduced into approximately 10 million GHO cells using commercially available transfection reagents Superfect® (Quiagen), Dosper® or Fugene® (Boehringer Mannheim) .The cells grew as described by Lucas et al. ., supra, Approximately 3 x 107 cells were frozen in an ampoule for growth and additional production as described below.The vials containing the plasmid DNA were dissolved by placing them in a water bath and mixing them by vortex stirrer. The contents were pipetted into a centrifuge tube containing 10 ml of medium and centrifuged at 1000 rpm for 5 minutes. the cells were resuspended in 10 ml of the selective medium (0.2 μt? of PS20 filtered with 5% of 0.2 μp? of diafiltered fetal bovine serum). The cells were then aliquoted in a 100 ml centrifuge containing 90 ml of the selective medium. After 1-2 days, the cells were transferred in a 250 ml centrifuge filled with 150 ml of the selective growth medium and incubated at 37 ° C. After another 2-3 days, centrifuges of 250 ml, 500 ml and 2000 ml were seeded with 3 x 10 5 cells / ml. The cell medium was exchanged with fresh medium by centrifugation and resuspension in the production medium. Although any suitable CHO medium can be employed, a production medium can currently be used as described in the U.S. Patent. No. 5,122,469, issued June 16, 1992. A 3 1 production centrifuge is seeded at 1.2 x 10 6 cells / ml. On day 0, the pH of the cell number is determined, for example. On day 1 start with the centrifuge that is sampled and sprayed with filtered air. On day 2 the centrifuge is sampled, the temperature changes to 33 ° C and 30 ml of 500 g / 1 of glucose and 0.6 1 of 10% of antifoam are taken (eg 35% of polydimethylsiloxane emulsion, Dow Corning 365 Medical Grade) Emulsion). Throughout the entire production, the pH was adjusted as necessary to keep it at around 7.2 After 10 days or until the viability dropped below 70%, the cell culture was collected by centrifugation and filtered through a 0.22 μp filter ?. The filter was stored either at 4 ° C or immediately loaded into the columns for purification. For the labeled poly-His constructs, the proteins were purified using a Ni-NTA column (Quiagen). Before purification, imidazole was added to the medium conditioned at a concentration of 5 ttiM. The conditioned medium was pumped to another Ni-NTA column of 6 ml equilibrated in 20 mM Hepes, pH 7.4, buffer containing 0.3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml / min. at 4 ° C. After loading, the column was washed with additional equilibration buffer and the protein was eluted with equilibrium buffer containing 0.25 M imidazole. The highly purified protein was desalted in a storage buffer containing 10 mM Hepes, 0.14 M NaCl and 4% mannitol, pH 6.8, with a G25 Superfine column (Pharmacia) of 25 ml and stored at -80 ° C. . The immunoadhesin constructs (containing Fe) were purified from the conditioned medium as follows. The conditioned medium was pumped into a 5 ml Protein A (Pharmacia) column which had been equilibrated with 20 mM phosphate buffer, pH 6.8. After loading, the column was extensively washed with equilibration buffer before elution with 100 mM citric acid, pH 3.5. The eluted protein was immediately neutralized by collecting fractions of 1 ml in tubes containing 275 μ? of 1 M Tris buffer, pH 9. The highly purified protein was subsequently desalted in a store buffer as described above for labeled poly-His proteins. Homogeneity was assessed by SDS polyacrylamide gels and by N-terminal amino acid sequencing by Edman degradation.
- - Certain of the TAT polypeptides described herein have been expressed and successfully purified using this technique (s). EXAMPLE 7: Expression of TAT in Yeast The following method describes the recombinant expression of TAT in the yeast. First, the expression vectors of the yeast are constructed for the production or intracellular secretion of TAT from the ADH2 / GAPDH promoter. The TAT encoding DNA and the promoter are inserted into suitable restriction enzyme sites in the selected plasmid to direct the intracellular expression of TAT. For secretion, DNA encoding TAT can be cloned into the selected plasmid, along with DNA encoding the ADH2 / GAPDH promoter, a natural TAT signal peptide or other mammalian signal peptide or for example, a yeast alpha factor or signal / conductive secretory invertase and linker sequences (if needed) for the expression of TAT. Yeast cells, such as yeast strain AB110, can then be transformed with the expression plasmids described above and cultured in the selected fermentation medium. The transformed yeast supernatants can be analyzed by precipitation with 10% trichloroacetic acid and separation by SDS-PAGE, followed by dyeing of the gels with Coomassie Blue dye. The recotnbinant TAT can be subsequently isolated and purified by removing the yeast cells from the fermentation medium by centrifugation and then concentrating the medium using selected cartridge filters. The concentrate containing TAT can be further purified using selected column chromatography resins. Certain of the TAT polypeptides described herein have been expressed and successfully purified using this technique (s). EXAMPLE 8 Expression of TAT in Insect Cells Infected with Baculovirus The following method describes the recombinant expression of TAT in insect cells infected with Baculovirus. The sequence coding for TAT is fused upstream of an epitope tag contained within a baculovirus expression vector. Such epitope tags include poly-his tags and immunoglobulin tags (such as IgG Fe regions). A variety of plasmids can be employed, including plasmids derived from commercially available plasmids such as pVL1393 (Novagen). Briefly, the sequence encoding TAT or the desired portion of the TAT coding sequence tal- - as the sequence encoding an extracellular domain of a transmembrane protein or the sequence encoding the mature protein if the protein is extracellular is amplified by PCR with primers complementary to the 5 'and 3' regions. The 5 'primer can incorporate flanking the restriction enzyme sites (selected). The product is then digested with those selected restriction enzymes and subcloned into the expression vector. Recombinant baculovirus is generated by co-transfection of the above plasmid and BaculoGold ™ virus DNA (Phngen) in Spodoptera frugiperda cells. ("Sf9") (ATCC CRL 1711) using lipofectin (commercially available by GIBCO-BRL). After 4-5 days of incubation at 28 ° C, the released viruses were harvested and used for further amplifications. Viral infection and protein expression is carried out as described by O'Reilley et al. , Baculovirus expression vectors:? Laboratory Manual (baculovirus expression vectors: A Laboratory Manual) oxford: Oxford University Press (1994). The expressed labeled poly-his TATs can then be purified, for example, by Ni2 + affinity chromatography-chelate as follows. The extracts were prepared from recombinant Sf9 cells infected with virus as described by Rupert et al. , Nature, 362: 175-179 (1993). Briefly, the Sf9 cells were washed, resuspended in sonication buffer (25 ml of Hepes, pH 7.9; 12.5 mM MgCl2; 0.1 mM EDTA; 10% glycerol; 0.1% of NP-40; 0.4 M of KC1) and sonicated twice for 20 seconds on ice. The sonicates were clarified by centrifugation and the supernatant was diluted 50-fold in charge buffer (50 mM phosphate, 300 mM NaCl, 10% glycerol, pH 7.8) and filtered through a 0.45 μp filter. A Ni2 + -NTA agarose column (commercially available from Qiagen) was prepared with a bed volume of 5 ml, washed with 25 ml of water and equilibrated with 25 ml of charge buffer. The filtered cell extract was loaded onto the column at 0.5 ml per minute. The column was washed for a baselin A28o with charge buffer, at which point the collection of the fraction began. The column was then washed with a secondary wash buffer (50 mM phosphate, 300 mM NaCl, 10% glycerol, pH 6.0), which elutes non-specific binding protein. After reaching the A2SOI baseline again, the column was developed with a gradient of Imidazole from 0 to 500 mM in the secondary wash buffer. One ml fractions were collected and analyzed by SDS-PAGE and silver stain or Western blot with Ni2 + -NTA conjugate for alkaline phosphatase (Qiagen). Fractions containing eluted labeled His-TAT are pooled and dialyzed - - against the load absorber. Alternatively, the purification of the TAT of labeled IgG (or labeled Fe) can be carried out using known chromatography techniques, including for example, Protein A or column chromatography of the G protein. Certain of the TAT polypeptides described herein have been expressed and purified successfully using this technique (s). EXAMPLE 9: Preparation of Antibodies That Bind to TAT This example illustrates the preparation of monoclonal antibodies that can specifically bind to TAT. Techniques for producing monoclonal antibodies are known in the art and are described, for example, in Goding, supra. Immunogens that can be employed include purified TAT, fusion proteins containing TAT and cells expressing recombinant TAT on the cell surface. The selection of the immunogen can be made by the person skilled in the art without undue experimentation. Mice such as Balb / c were immunized with the TAT immunogen emulsified in the complete Freund's adjuvant and injected subcutaneously or intraperitoneally in an amount from 1-100 micrograms. Alternatively, the immunogen was emulsified in the MPL-TDM adjuvant (Rbi Immunochemical Research, Hamilton, MT) and injected into the plants of the hind paws of the animal. The immunized mice were then supercharged 10 to 12 days later with additional immunogen emulsified in the selected adjuvant. Serum samples can be obtained periodically from the mice by retro-orbital bleeding to be tested in ELISA assays to detect anti-AT antibodies. After a suitable antibody titer has been detected, animals "positive" for the antibodies can be injected with a final intravenous injection of TAT. Three to four days later, the mice are sacrificed and the spleen cells are harvested. Spleen cells are then fused (using 35% polyethylene glycol) to a murine myeloma cell line such as P3X63AgU.l, available from ATCC, No. CRL 1597. The fusions generate hybridoma cells which can then be plated on plates of tissue culture of 96 cavities containing the HAT medium (hypoxanthine, aminopterin and thymidine) to inhibit the proliferation of unfused cells, myeloma hybrids and spleen cell hybrids. Hybridoma cells were classified in an ELISA for reactivity against TAT. The determination of "positive" in the selection of hybridoma cells of the desired monoclonal antibodies against TAT is found by the person skilled in the art.
- - Hybridoma positive cells can be injected intraperitoneally into syngeneic Balb / c mice to produce ascites containing the anti-TAT monoclonal antibodies. Alternatively, the hybridoma cells can grow in tissue culture bottles or cylindrical bottles. The purification of the monoclonal antibodies produced in the ascites can be carried out using ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography can be employed based on the binding of the antibody to protein A or protein G. Antibodies directed against certain of the TAT polypeptides described herein have been successfully produced using this technique (s). ). EXAMPLE 10: Purification of TAT Polypeptides Using Specific Antibodies Natural or recombinant TAT polypeptides can be purified by a variety of standard techniques in the purification of protein. For example, the pro-TAT polypeptide, the complete TAT polypeptide or the pre-TAT polypeptide is purified by immunoaffinity chromatography using antibodies specific for the TAT polypeptide of interest. In general, an immunoaffinity column is constructed by covalently coupling the anti-TAT polypeptide antibody to a chromatographically activated resin. Polyclonal immunoglobulins are prepared from immune sera either by precipitation with ammonium sulfate or by purification on immobilized Protein A (Pharmacia L B Biotechnology, Piscataway, N.J.). Similarly, monoclonal antibodies are prepared from fluids of mouse ascites by precipitation or chromatography with ammonium sulfate on immobilized Protein A. A partially purified immunoglobulin is covalently linked to a chromatographic resin such as the SEpAROSE ™ CnBr-activated (Pharmacia LKB Biotechnology). The antibody is coupled to the resin, the resin is blocked and the bypass resin is washed according to the manufacturers' instructions. Such an immunoaffinity column is used in the purification of the TAT polypeptide by preparing a fraction from the cells containing the TAT polypeptide in a soluble form. This preparation is derived by the solubilization of the whole cell or of a subcellular fraction obtained via differential centrifugation by the addition of the detergent or by other methods known in the art. Alternatively, the soluble TAT polypeptide containing a signal sequence can be scavenged in a useful amount in which the cells grow. A preparation containing the soluble TAT polypeptide is passed over the immunoaffinity column and the column is washed under conditions that allow preferential absorbency of the TAT polypeptide (e.g., high ionic strength buffers in the presence of a detergent). Then, the column is eluted under conditions that break the binding of the TAT antibody / polypeptide. { e.g., a low pH buffer such as about pH 2-3 or a high concentration of a chaotrope such as urea or thiocyanate ion) and the TAT polypeptide is collected. Material Deposit The following materials have been deposited with the American Type Culture Collection, 10801 University Blvd-, anassas, VA 20110-2209, USA (ATCC): Table 7 Material ATCC Dept. No. Date of Deposit DNA45234-1277 209654 March 5, 1998 DNA37151-1193 209393 October 17, 1997 These deposits were prepared under the provisions of the Budapest Treaty in the International Recognition of the Deposit of Micro-organisms for the Purpose of Patent Procedure and the Regulations, (International Recognition of Deposit of Microorganisms for the Purpose of the Procedure and the Regulation of Patents) hereinafter (Budapest Treaty). This ensures the maintenance of a viable crop of the deposit for 30 years from the date of deposit. The deposits will be available by ATCC under the terms of the Budapest Treaty and subject to an agreement between Genentech, Inc. and ATCC, which ensure the permanent and unrestricted availability of the crop progeny for the public until the patent is published. from EU relevant or even to the public exposure of any US patent application. or foreign, whatever is presented first and ensure the availability of the progeny to someone determined by the US Patent and Trademark Commissioner. to be entitled to the foregoing in accordance with 35 USC § 122 and the rules of the Commissioner in accordance with the same (including 37 CFR § 1.14 with particular reference to 886 OG 638). The assignee of the present application has agreed that if a culture of the materials on deposit must die or be lost or destroyed when monitored under suitable conditions, the materials will be immediately replaced upon notification with another of the same. The availability of the deposited material is not considered as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws. The above written specification is considered sufficient to enable an expert in the art to practice the invention. The present invention sh not be limited in scope by the deposited construction since the deposited mode is proposed as a simple illustration of certain aspects of the invention and any construction that is functionally equivalent is within the scope of this invention. The deposit of the material herein does not constitute an admission that the written description contained herein is inadequate to allow the practice of any aspect of the invention, including the best mode thereof, nor is it considered as limiting the scope of the claims the specific illustrations that are represented. Indeed, various modifications of the invention in addition to those shown as described herein, will be apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims.

Claims (15)

  1. CLAIMS 1. A method for killing a cancer cell that expresses a polypeptide having at least 80% amino acid sequence identity for: (a) the amino acid sequence shown in the Figure 3 (SEQ ID NO: 3) or Figure 4 (SEQ ID ??: 4); or (b) an amino acid sequence encoded by a nucleotide sequence comprising the nucleotide sequence shown in Figure 1 (SEQ ID NO: 1) or Figure 2 (SEQ ID NO: 2), said method comprising contacting said cancer cell with an antibody, oligopeptide or organic molecule that binds said polypeptide in said cancer cell, thereby destroying said cancer cell.
  2. 2. The method of claim 1, wherein said antibody is a monoclonal antibody.
  3. 3. The method of Claim 1 wherein said antibody is an antibody fragment.
  4. 4. The method of Claim 1 wherein said antibody is a chimeric or humanized antibody.
  5. 5. The method of Claim 1 wherein said oligopeptide antibody or organic molecule is conjugated to a growth inhibitory agent.
  6. 6. The method of Claim 1 wherein said oligopeptide antibody or organic molecule is conjugated to a cytotoxic agent.
  7. The method of Claim 6 wherein said cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
  8. 8. The method of Claim 6 wherein the cytotoxic agent is a toxin.
  9. The method of Claim 8 wherein the toxin is selected from the group consisting of maytansinoid and calicheamicin.
  10. 10. The method of Claim 8 wherein the toxin is a maytansinoid.
  11. 11. The method of Claim 1 wherein said antibody is produced in a bacterium.
  12. 12. The method of Claim 1 wherein said antibody is produced in CHO cells.
  13. The method of Claim 1 wherein said cancer cell is further exposed to radiation treatment or a chemotherapeutic agent.
  14. The method of claim 1, wherein said cancer cell is selected from the group consisting of a breast cancer cell, an ovarian cancer cell, and a uterine cancer cell.
  15. 15. The method of Claim 1, wherein said cancer cell overexpresses said polypeptide as compared to a normal cell of the same tissue origin.
MXPA04010091A 2002-04-17 2002-04-17 Compositions and methods for the diagnosis and treatment of tumor. MXPA04010091A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2002/012206 WO2003088898A2 (en) 2002-04-17 2002-04-17 Compositions and methods for the diagnosis and treatment of tumor

Publications (1)

Publication Number Publication Date
MXPA04010091A true MXPA04010091A (en) 2004-12-13

Family

ID=29247990

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA04010091A MXPA04010091A (en) 2002-04-17 2002-04-17 Compositions and methods for the diagnosis and treatment of tumor.

Country Status (7)

Country Link
EP (1) EP1494709A4 (en)
JP (1) JP2005523311A (en)
KR (1) KR20040111525A (en)
AU (1) AU2002254651A1 (en)
CA (1) CA2481085A1 (en)
MX (1) MXPA04010091A (en)
WO (1) WO2003088898A2 (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030049676A1 (en) * 1997-09-17 2003-03-13 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6291663B1 (en) * 1999-03-03 2001-09-18 Board Of Trustees Of The University Of Arkansas TADG-12: a novel transmembrane serine protease overexpressed in a ovarian carcinoma
EP1252300B1 (en) * 2000-02-03 2011-01-19 Dendreon Corporation Nucleic acid molecules encoding transmembrane serine proteases, the encoded proteins and methods based thereon
AU6802801A (en) * 2000-03-01 2001-09-24 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001083781A2 (en) * 2000-04-28 2001-11-08 Millennium Pharmaceuticals, Inc. 14094, a novel human trypsin family member and uses thereof

Also Published As

Publication number Publication date
KR20040111525A (en) 2004-12-31
WO2003088898A2 (en) 2003-10-30
CA2481085A1 (en) 2003-10-30
JP2005523311A (en) 2005-08-04
AU2002254651A1 (en) 2003-11-03
EP1494709A4 (en) 2006-06-28
WO2003088898A3 (en) 2004-09-16
EP1494709A2 (en) 2005-01-12

Similar Documents

Publication Publication Date Title
AU2003249014B2 (en) Compositions and methods for the diagnosis and treatment of tumor
US7514538B2 (en) Compositions and methods for the diagnosis and treatment of tumor
US20130210116A1 (en) Composition and methods for the diagnosis and treatment of tumor
MXPA05001933A (en) Compositions and methods for the diagnosis and treatment of tumor.
MXPA04010092A (en) Compositions and methods for the diagnosis and treatment of tumor.
AU2001270118A1 (en) Compositions and methods for the diagnosis and treatment of tumor
MXPA04012243A (en) Compositions and methods for the diagnosis and treatment of tumor.
US6914130B2 (en) Compositions and methods for the diagnosis and treatment of tumor
EP1311662A2 (en) Compositions and methods for the diagnosis and treatment of tumor
AU2001286785B2 (en) Compositions and methods for the diagnosis and treatment of tumor
US8097700B2 (en) TAT294 polypeptides
AU2001286785A1 (en) Compositions and methods for the diagnosis and treatment of tumor
AU2006230703A1 (en) Compositions and methods for the diagnosis and treatment of tumor
US20020161199A1 (en) Compositions and methods for the diagnosis and treatment of tumor
AU2001284906A1 (en) Compositions and methods for the diagnosis and treatment of tumor
US20070031901A1 (en) Compositions and methods for the diagnosis and treatment of tumor
MXPA04010091A (en) Compositions and methods for the diagnosis and treatment of tumor.
MXPA04005945A (en) Compositions and methods for the diagnosis and treatment of tumor.
WO2003090600A2 (en) Compositions and methods for the diagnosis and treatment of tumor
ZA200408159B (en) Compositions and methods for the diagnosis and treatment of tumor

Legal Events

Date Code Title Description
FG Grant or registration