![]() methods and compositions to modulate pd1
专利摘要:
FINGER ZINC PROTEIN THAT CONNECTS TO TARGET SITE IN PROGRAMMED-DEATH GENE-1 (PD1), POLYNUCLEOTIDE THAT UNDERSTANDS IT AND USE THE SAME IN TREATING DISEASES AND DISORDERS RELATED TO THE PD1 GENE, AS WELL AS A MODE FOR MODEING EXPRESSION GENE CELL.The present invention relates to the zinc finger protein that binds to a target site in a programmed death gene - 1 (PD1) and to a polynucleotide that comprises it. Furthermore, the present invention relates to a method for modulating expression of a PD1 gene in a cell, comprising introducing said polynucleotide into the cell, under conditions such that the expression of the PD1 gene is modulated and the use of said polynucleotide for the preparation of a drug to treat an individual having a disease or disorder with the characteristic of aberrant expression or regulation of a PD1 gene. 公开号:BR112013012124A2 申请号:R112013012124-6 申请日:2011-11-17 公开日:2020-09-01 发明作者:Philip D. Gregory;Michael C. Holmes;Matthew C. Mendel;Xiangdong Meng;David Paschon;Andreas Reik;Fyodor Urnov 申请人:Sangamo Biosciences, Inc.; IPC主号:
专利说明:
Invention Patent Descriptive Report for "PROTEIN FINGER OF ZINC THAT CONNECTS TO A TARGET SITE IN A PROGRAMMED-DEATH GENE-1 (PD1), A POLINUCLEOTIDE THAT UNDERSTANDS IT AND USE IT IN THE TREATMENT OF DISEASES AND DISORDERS RELATED TO THE PD1 GENE, AS WELL AS A MODE FOR MODULATING EXPRESSION OF GENE PD1 IN CELL ". DECLARATION OF RIGHTS TO INVENTIONS MADE UNDER RESEARCH SPONSORED BY THE GOVERNMENT Not applicable TECHNICAL FIELD The present invention relates to fields of genome engineering and nuclease identification. BACKGROUND Nucleases, including zinc finger nucleases and homing endonucleases such as SceI, which are designed to specifically bind to target sites have been shown to be useful in genome engineering. For example, zinc finger nucleases (ZFNs) are proteins comprising projected site-specific zinc fingers fused to a nuclease domain. Said ZFNs have been used successfully for genome modification in a variety of different species. See, for example, North American publications 20030232410; 20050208489; 20050026157; 20050064474; 20060188987; 20060063231; and international publication WO 07/014275, the disclosures of which are incorporated in their entirety for all purposes. These ZFNs can be used to create a double-strand (DSB) break in a target nucleotide sequence, which increases homologous recombination in an targeted location more than 1000 times. In addition, inaccurate repair of a site-specific DSB by joining the non-homologous end (NHEJ) may still result in a gene break. The creation of said DSBs results in the deletion of large regions arbitrarily. The programmed death receptor (PD1, also known as PDCD1) has been shown to be involved in regulating the balance between active- Below is sheet 1a / 57 1a / 57 tion of T cell and T cell tolerance in response to chronic antigens. During infection with HIV1, expression of PD1 was shown to be increased in CD4 + T cells. Upward regulation of PD1 is believed to be somehow linked to T cell exhaustion (defined as a progressive loss of key effector functions) when T cell dysfunction is observed. Following is sheet 2/57 Patent Specification Report for "METHODS AND COMPOSITIONS FOR MODULAR PDl". - 'DECLARATION OF RIGHTS TO INVENTIONS MADE UNDER RESEARCH SPONSORED BY THE GOVERNMENT 5 Not applicable TECHNICAL FIELD The present invention relates to fields of genome engineering and nuclease identification. BACKGROUND - 10 Nucleases, including zinc finger nucleases and homing endonucleases like Scel, which are designed to specifically bind to target sites have been shown to be useful in genome engineering. For example, zinc finger nucleases (ZFNS) are proteins comprising projected site-specific zinc fingers fused to a nuclease domain. 15 Said ZFNS have been used successfully for genome modification in a variety of different species. See, for example, North American publications 20030232410: 20050208489: 20050026157: 20050064474; 20060188987; 20060063231: and international publication WO 07/014275, the disclosures of which are incorporated in their entirety for all 20 purposes. These ZFNS can be used to create a double-strand break (DSB) in a target nucleotide sequence, which increases homologous recombination in an targeted location more than 1000 times. In addition, inaccurate repair of a site-specific DSB by joining the non-homologous tip (NHEJ) can still result in gene breakdown. The creation of said 25 DSBS results in the deletion of large regions arbitrarily. The programmed death receptor (PDl, also known as PDCDI) has been shown to be involved in regulating the balance between T cell activation and T cell tolerance in response to chronic antigens. During HlV1 infection, PDl expression was shown to be increased in CD4 + T cells. Upward regulation of PDl is believed to be somehow linked to T cell exhaustion (defined as a progressive loss of key effector functions) when T cell dysfunction is observed. · Widespread in the presence of chronic antigen exposure as is the case in HlV infection, upward regulation of PDl may still be associated with increased apoptosis in the same sets of cells during infection + viral (see, Petrovas et al., (2009) J / mmuno / .183 (2): 1120-32). PDl 5 can also play a role in tumor-specific escape from immune surveillance. PDl has been shown to be highly expressed in tumor-specific cytotoxic T lymphocytes (CTLs) in both chronic myeloid leukemia (CML) and acute myeloid leukemia (AML). PDl is further regulated upward in T lymphocytes that infiltrate melanoma (TILS) (see, Dotti (2009) - 10 B / ood 114 (8): 1457-58). Tumors expressly demonstrated the PD1 ligand (PDL) which, when combined with upward regulation of PDl in CTLs, may be a contributing factor in the loss of T cell functionality and the inability of CTLs to mediate an effective anti-tumor response. Researchers have shown that in mice 15 chronically infected with lymphocytic chorionomeningitis virus (LCMV), administration of anti-PD1 antibodies blocked the PDI-PDL interaction and was able to restore some T cell functionality (cytokine proliferation and secretion) , and led to a reduction in viral load (Barber et al. (2006) Nature 439 (9): 682-687). Deregulation of PDl may also play a role in autoimmune disease. PDl SNPs (in particular PD 1.3) were still associated with an increased risk for systemic lupus erythematosus (SLE). It has been shown that SLE patients have a higher frequency of PD 1.3 PDl allele, and that these patients showed reduced expression of PDI in their activated CD4 + T cells (see, Bertsias et al., (2009) Arthritis Rheum. 25 60 (1 ): 207-18). Thus, there remains a need for modulators targeting PDl, for example, nucleases targeting PDl or transcription factors that can be used in research and therapeutic applications. SUMMARY 30 The present invention relates to the development of PDl-targeted nucleases, for example, engineered meganucleases and zinc finger nuclease (ZFNS). The present invention demonstrates active zinc finger proteins specific for a rodent PD1 and fusion proteins, including zinc finger protein transcription factors (ZFP-TFS) or zinc finger nucleases (ZFNS), comprising these finger proteins PDl-specific zinc 5 - The proteins comprising PDl-specific zinc finger proteins of the invention can be used for research and therapeutic purposes, including for the treatment of any disease or disorder in which PDl is abnormally expressed, or where the pathway of PDl is abnormally used due to overexpression of a PDl linker. For example, finger nuclease - 10 zinc targeted to PDl sites on T cells can be used to block PDl-dependent immune suppression in both chronic diseases and infectious diseases. Alternatively, a defective PD1 site can be remedied using ZFN-dependent targeted insertion of wild-type sequences, or a 15 zinc finger protein transcription factor (ZFP TF) can be used to modulate (for example, suprarular or infrared) the expression of defective PDl. In addition, a ZFP TF targeted to the PD1 site can be used to modulate a wild type PD1 gene. In another aspect of the invention, the fusion proteins comprise zinc finger nucleases (ZFNS) that are specific to the human PD1 gene. In certain embodiments, the zinc finger domains of nuclease fusion proteins comprise non-naturally occurring recognition helices shown in Table 1 and / or that bind to the active sites shown in Table 2- In yet another aspect, they are provided here ZFP-TFs capable of modulating the expression of a PD1 gene. In certain embodiments, the ZFP-TFS zinc finger domains comprise the non-naturally occurring recognition propellers shown in Tables 1 or 5 and / or bind to the target sites shown in Tables 2 or 6. In another aspect, they are provided here are methods and compositions 30 for the regulation of the PD1 gene. In certain embodiments, the methods comprise introducing a fusion protein comprising the zinc finger protein that is designed to bind to a target site in a PD1 gene (or "polynucleotide (which encodes a fusion protein) in cells of a patient with a disease or disorder in which the disease or disorder is characterized "caused by abnormal expression of PDl and / or undesirable use of the PDl pathway, caused by overexpression of PDl ligands. The methods can be used in the treatment and / or prevention of chronic infections such as HlV and HCV. Similarly, the methods and compositions can be used in the treatment and / or prevention of cancer and malignancy. Non-limiting examples of cancers that can be treated and / or prevented include lung cancer, pancreatic cancer, liver cancer, bone cancer, breast cancer, colorectal cancer, leukemia, ovarian cancer, lymphoma, cancer of brain and the like. The methods and compositions described here can be used as a standard isolated treatment, or can be used in combination with other antiviral or anticancer therapies. These methods and compositions 15 can be provided with antiviral or anticancer therapies in a sequential manner, or they can be administered concomitantly. The methods and compositions provided here can also be used to modify the expression of PDl in a patient afflicted with an autoimmune disease, or can be used to treat said patient by integrating into a wild type PDl 20 allele, or a PDl allele with altered characteristics if this patient contains a defective or undesirable allele. Cell lines can be constructed to specifically alter the PD1 gene sequence to create therapeutic compounds that can alter the regulation or functionality of a PDl gene. 25 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a graph showing the breakdown of the PDl gene in human PBMCS using PDl-specific zinc finger nucleases, as determined by the Cel-l-based SURVEYORtm nucleoase assay that measures the percentage of inserted mutations by the non-homologous end junction 30 (NHEJ) which is induced when the indicated ZFNs are introduced into these cells. The cells were treated with ZFN pairs that each combined ZFN 12942 with a different variant of ZFN that bound to the opposite strand of DNA and could form a functional nuclease in binding to the target site with 12942. The SBS number for the second ZFN of the pair is indicated> "below each graph. The leftmost bar of each pair shows NHEJ percentages of three days after nucleofection for cells incubated at 37 ° C. 5 The second bar on the left in each indicated pair shows NHEJ percentages ten days after nucleofection for cells incubated at 37 ° C. The second bar on the right of each indicated pair shows NHEJ three days after nucleofection for cells incubated at 30 ° C and the rightmost bar of each indicated pair shows NHEJ percentages ten days after nucleofection for cells incubated at 30 ° C. Ç. Figure 2 represents the results of an analysis to determine the sequence of PDl site in CD8 + T cells after treatment with the specific pair for PDl ZFN 12942 and 12947 that directs exon 1. The inserts are described in large bold letters . Deletions are denoted with a (-). As can be seen from the figure, several insertions and deletions were observed close to the ZFN cut site as a result of DSB repair via NHEJ. Figure 3 represents the results after transfection of splenocytes derived from transgenic TCR / Ragl - / - mice with murine PD1-specific ZFNs 20. The cells were stimulated with anti-CO3 antibodies, and then stained for PD1. The plots show the percentage of CD1 + CD8 + cells positive for PD1 in each group and the median fluorescence in PD1 for each group, and the data are represented in the Table format below. As can be seen in the figure, the expression of PDl reduces 25 in cells that received the specific ZFNS for PDI, even in the presence of CD3 stimulation. Figure 4 demonstrates that the reduction in PDl expression was evident at later time points. The cells were also collected at 72 hours after stimulation with CD3, and stained for PDl. Histograms- 30 but higher show c) percentage of CD1 + CD8 + cells positive for PDl in each group and median fluorescence PDl for each group- Lower Plots show the frequency of cells expressing PDI / CFSE. This figure demonstrates that PD1 expression is further reduced in cells treated with PD1-specific ZFNs still 72 hours after "CD3" stimulation. Figure 5 represents the results for the PD1-specific ZFN pairs tested on CD4 + T cells and analyzed using the Cel-1 assay for genome editing activity. In these cells, up to 44% editing was observed with some pairs. Figure 6 represents the purification of PDl (-) cells after treatment with PDF-specific ZFN pairs in CD4 + T cells. The 10 percent edit or NHEJ was measured by the Cel-1 assay as described above, and up to 44% edition was observed with some of the PDL-specific ZFN Pairs. After treatment, the cells were stimulated with a first exposure to anti-CD3 / CD8 beads to induce the transgenic ZFN and then re-stimulated and subjected to a purification procedure, 15 by FACS or by affinity chromatography. The cells were collected and analyzed by PD1 edition by the Cel-l assay (described above), (i) after the first stimulation, (ii) after the second stimulation but before any purification, (iii) after cell classification for CD25 "(an activation marker), PD1 (-), or (iv) after affinity chromatography. As shown, 20 using the cell classification technique, up to 56% of the recovered cells demonstrated to be modified. PD1 (-) cells purified by affinity chromatography showed an overall PDl modification of up to 42% as assessed by Cel-1 analysis. Figure 7 is a graph describing the results for PDl-specific ZFNS 25 tested on T cells. In this experiment, mR-NAS encoding PDl-specific ZFNS were translated into CD8 + T and the percentage PDl modification was analyzed by the Cel I assay. The amount of modification observed was related to the amount of mR-NA used, with smaller amounts of resulting input mRNA lower percentage of target modification. These results demonstrate that PD1-specific ZFNS described here are able to modify the PDl site in cell lines and in primary T cells. m DETAILED DESCRIPTION Compositions and methods for tri-acting systems in high yield in vivo to identify functional nucleases are described here. In particular, assays use a reporter system to monitor a nuclease's ability to induce a break in double-stranded in its active site. In addition, assays can be used to determine the effect of nucleasis on cell growth (toxicity). Designed nuciease technology is based on the engineering of naturally occurring DNA-binding proteins. , engineering · 10 of endonucleases homing with adapted DNA binding specificities has been described Chames et al. (2005) Nuc / eic Acids Res 33 (20): e178; Arnold et al. (2006) J. Mol Biol. 355: 443-458, In addition, ZFP engineering has been described, see, for example, US patents 6,534,261; 6,607,882; 6,824,978; 6,979,539; 6,933,113; 7,163,824; and 15 7,013,219. In addition, ZFPs have been linked to the nuclease domains to create ZFNs - a functional entity that is able to recognize its intended target gene through the projected DNA binding domain (ZFP) and the nuclease causes the gene to be cut close to the site ZFP Connection. 20 See, for example, Kim et al. (1996) Proc Nat'l Acad Sci USA 93 (3): 1156-1160. More recently, ZFNS have been used for genome modification in a variety of organisms. See, for example, US Patent Publications 20030232410; 20050208489; 20050026157; 20050064474; 20060188987: 20060063231: and International publication WO 07/014275. 25 Although the rules that allow engineering of ZFPs to bind to specific DNA sequences are well characterized and precisely identify specific ZFPS, these same ZFPS may not bind with equal affinity and / or specificity when incorporated into a ZFN. For example, it is possible that a chromosomal substrate may affect the precise dimerization of nuclease domains in living cells, consequently decreasing the potential for cleavage, and that the chromatin architecture needed by a given genomic site will differentially affect the "ZFNS ability to bind and cleave its intended target sequence. In addition, it is difficult if not impossible for in vitro assays to imitate the parameters, "research features that a designed DNA binding domain is subjected to. ¥ do when presented with a cell genome in chromatinized form. 5 As a result, it is essential to test numerous variants in the relevant organism, or cell line, to identify a ZFN presenting the ideal characteristics for gene modification. In addition, since each in vivo system has its own peculiarities, it is necessary to develop detection tests to determine - 10 ZFN action. Thus, unlike the in vivo screening methods previously described for homing endonucleases with different binding specificity from naturally occurring endonuclease homing, the methods described here provide a fast and efficient way of ranking nucleoses known to bind to a particular target site by predicting their in vivo functionality as well as the toxicity of a nuclease to the host cell. Thus, the methods and compositions described here provide highly fast and efficient methods for identifying nucleases that are biologically active in vivo. In addition to precisely predicting nuclease functionality in vivo, the assays described here can also be used to determine nuclease toxicity, thus allowing identification of safer and functionally active active proteins- General The practice of the methods, as well as preparation and use of the compositions disclosed herein employ, unless otherwise indicated, conventional techniques in molecular biology, biochemistry, chromatin structure and analysis, computational chemistry, cell culture, recombinant DNA and related fields are within the skill of the art. . These techniques are fully explained in the literature. See, for example, Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL, Second edition, Cold 30 Spring Harbor Laboratory Press, 1989 and Third edition, 2001; Ausubel et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John VViley & Sons, New York, 1987 and periodic updates; the METHODS IN ENZYMO- series LOGY, Academic Press, San Diego; Wolffe, CHROMATIN STRUCTURE AND FUNCTION, Third edition, Academic Press, San Diego, 1998; ME- 'THODS IN ENZYMOLOGY, Vol. 304, "Chromatin" (P.M. Wassarman and A. P. Wolffe, eds.), Academic Press, San Diego, 1999: and METHODS IN MOLE-. 5 CULAR BIOLOGY, VoL 119, "Chromatin Protocols" (P.B. Becker, ed.) Human Press, Totowa, 1999. Definitions The terms "nucleic acid," "polynucleotide," and "oligonucleotide" are used interchangeably and refer to a deoxy - 10 ribonucleotide or ribonucleotide, in linear or circular conformation, and in the form of single or double tape. For the purposes of the present invention, these terms are not to be construed as limiting the length of a polymer. The terms may include known analogs of natural nucleotides, as well as nucleotides that are modified at the base, fractions of sugar and / or phosphate (for example, phosphorothioate structures). In general, an analogue of a particular nucleotide has the same base pairing specificity; that is, an analogue of A will pair on the basis of T. The terms "polypeptide," "peptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues. The term still applies to amino acid polymers in which one or more amino acids are chemical analogues or modified derivatives of corresponding naturally occurring amino acids. "Binding" refers to a specific sequence, non-covalent interaction between macromolecules (for example, between a protein and a nucleic acid). Not all components of a linkage interaction need to be sequence specific (for example, counted with phosphate residues in a DNA structure), as long as the interaction as a whole is sequence specific. These interactions are generally characterized by a dissociation constant (Ki) of 10 "'M"' or less. "Affinity" refers to 30 if the form of binding: increased binding affinity being correlated with a lower Ki. A "binding protein" is a protein that is capable of binding not covalently to the other molecule. A binding protein can bind to, for example, a DNA molecule (a protein binding to P 'DNA), an RNA molecule (an RNA binding protein) and / or a protein molecule (a protein binding protein). In the case of a protein-binding protein, it can bind itself (to form homodimers, homotrimers, etc.) and / or it can bind to one or more molecules of a different protein or proteins. A binding protein can have more than one type of binding activity. For example, zinc finger proteins have DNA binding, RNA binding and protein binding activity. A "zinc finger DNA binding protein" (or binding domain) is a protein, or a domain within a larger protein, which binds DNA in a sequence-specific way through one or more zinc fingers, which are regions of amino acid sequences within the binding domain whose structure is stabilized by coordination of a zinc ion The term zinc finger DNA binding protein is generally shortened as zinc finger protein or ZFP. Zinc finger binding domains can be "designed" to bind to a predetermined nucleotide sequence. Non-limiting examples 20 for zinc finger protein engineering are designed and selected A designed zinc finger protein is a protein that does not occur in nature whose design / composition results mainly from rational criteria. They include the application of substantial rules and computerized algorithms for processing 25 information in a database that stores information on existing designs of ZFP projects and connection data. See, for example, US Patents 6,140,081; 6,453,242; and 6,534-261; see also VVO 98 / 530q8: VVO 98/53059; WO 98/53060; WO 02/016536 and WO 03/016496. A "selected" zinc finger protein is a protein not found in nature whose production results mainly from an empirical process such as phage display, interaction trap or hybrid selection. See, for example, US 5,789,538; US 5,925,523: US 6,007,988; US íKm - 6,013,453; US 6,200,759; WO 95/19431; WO 96/06166; WO 98/53057; WO 98/54311: WO 00/27878; WO 01/60970 WO 01/88197 and WO 02/099084. 0 "Cleavage" refers to the square of the covalent structure of a% DNA-Cleavage molecule can be initiated by a variety of methods, including, but not limited to, enzymatic or chemical hydrolysis of a phosphodiester bond. Both single-strand cleavage and double-strand cleavage are possible, and double-strand cleavage may occur as a result of two distinct single-strand cleavage events. DNA cleavage can result in the production of blunt ends or staggered ends. - 10 In certain embodiments, fusion polypeptides are used for double stranded DNA cleavage. A "cleavage half-domain" is a sequence of polypeptides that, together with a second polypeptide (identical or different) forms a complex containing cleavage activity (preferably double-stranded cleavage activity). The terms "first and second half-domains of civilization;" "+ and - cleavage half domains" and "right and left cleavage half domains" are used interchangeably to refer to the pairs of cleavage half domains that dimerize. A "projected cleavage half-domain" is a 20-cleavage half-domain that has been modified to form mandatory heterodimers with another half-cleavage domain (for example, another projected cleavage half-domain). See also US patent applications 10 / 912,932 and 11 / 304,981 and provisional application US 60 / 808-486 (filed on May 25, 2006), incorporated here by reference in their entirety. The term "sequence" refers to a nucleotide sequence of any length, which can be DNA or RNA; it can be linear, circular or branched and it can be single or double tape. The term "donor sequence" refers to a nucleotide sequence that is inserted into a genome. A donor sequence can be of any length, for example, between 2 and 10,000 nucleotides in length (or any integer between them or above), preferably between about 100 and 1,000 nucleotides in length (or any integer between them), plus prefer- preferably between 200 and 500 nucleotides in length. "Chromatin" is the nucleoprotein structure comprising the cell genome. Cell 'chromatin' comprises nucleic acid, mainly DNA, and protein, including chromosomal proteins histones and non-histones. Most of the chromati-. 5 in the eukaryotic cell it exists in the form of nucleosomes, in which a nucleosome nucleus comprises approximately 150 base pairs of DNA associated with an octamer comprising two each of histones H2A, H2B, H3 and H4; and DNA ligand (of variable length depending on the organism) extends between nucleosome nuclei. A H1 - 10 H1 molecule is usually associated with the DNA ligand. For the purposes of the present invention, the term "chromatin" is intended to include all types of cell nucleoproteins, both prokaryotic and eukaryotic. Cell chromatin includes both chromosomal and episomal chromatin. A "chromosome," is a complex chromatin comprising all or part of a cell's genome. The genome of a cell is generally characterized by its karyotype, which is the collection of all the chromosomes that comprise the cell's genome. The genome of a cell can comprise one or more chromosomes. An "episome" is a replicating nucleic acid, complex of 20 nucleoproteins or another structure comprising a nucleic acid that is not part of a cell's chromosome karyotype. Examples of episodes include plasmids and certain viral genomes. A "target site" or "target sequence" is a nucleic acid sequence that defines a portion of a nucleic acid to which a binding molecule will bind, provided that sufficient conditions exist for binding. For example, the 5'-GAATTC sequence -3 'is a target site for the restriction endonucleate Eco R1. A "chronic infectious disease" is a disease caused by an infectious agent in which the infection has persisted. Such a disease may include hepatitis (A, B, or C), herpes virus (eg, VZV, HSV-l, HSV-6, HSV-ll, CMV, and EBV), and HIV / AIDS. Non-viral examples may include chronic fungal diseases such as pergylosis, Candidiasis, Coccidioidomycosis, and diseases diseases associated with Cryptococcus and Histoplasmosis. Non-limiting examples of bacterial infection agents can be Chlamydia pneumoniae, - Listeria monocytogenes, and Mycobacterium tuberculosis. The term "cancer" refers to any disease in which there is an unpressed proliferation of. 5 cells, whether within an organ or body tissue. Thus, the term includes any type of cancer or malignancy, including, but not limited to, ovarian cancer, leukemia, lung cancer, colorectal / colon cancer, CNS cancer, melanoma, renal cell carcinoma, plasmacytoma / myeloma, prostate cancer, breast cancer, and the like. As used herein, the term "moral tissue" refers to abnormal growth of malignant-type cells or tissues, unless specifically stated otherwise and does not include a benign tissue type. The term "inhibit or inhibit" as used here means to reduce growth / replication. The term "autoimmune disease" refers to any disease or disorder in which the subject assembles a destructive immune response against its own tissues. Autoimmune disorders can affect almost any organ system in the subject (eg, hurnan), including, but not limited to, diseases of the nerves, gastrointestinal, and endocrine systems, as well as skin and other connective tissues, eyes, blood, and blood vessels. Examples of autoimmune diseases include, but are not limited to, Hashimoto's thyroiditis, systemic lupus erythematosus, Sjogren's syndrome, Graves' disease, Scleroderma, rheumatoid arthritis, multiple sclerosis, Myasthenia gravis and Diabetes. An "exogenous" molecule is a molecule that is not normally present in a cell, but can be introduced into a cell by one or more genetic, biochemical or other methods, "Normal presence in the cell" is determined with respect to particular development stage and environmental conditions of the cell. Thus, for example, a molecule that is present only during embryonic development is an exogenous molecule with respect to an adult muscle cell. Similarly, a heat shock induced molecule is an exogenous molecule with respect to a cell treated with heat shock. An exogenous molecule can comprise, for example, a working version of "a malfunctioning endogenous molecule or a malfunctioning version of a normally functioning endogenous molecule. An exogenous molecule can be, among other things, a small . small molecule, as it is generated by a combinatorial chemical process, or a macromolecule as a protein, nucleic acid, carbohydrate, lipid, glycoprotein, lipoprotein, polysaccharide, any modified derivative of the above molecules, or any complex comprising one or more of the above molecules . Nucleic acids include DNA and RNA, can be single or double stranded; they can be linear, branched or circular; and can be - 10 of any length. Nucleic acids include those capable of forming doubles, as well as triple forming nucleic acids. See, for example, US patents 5,176,996 and 5,422,251. Proteins include, but are not limited to, DNA binding proteins, transcription factors, chromatin remodeling factors, methylated DNA binding proteins, polymerases, methylases, demethylases, acetylases, deacetylases, kinases, phosphatases, integrases, recombinases, ligases, topoisomerases, girases and helicases. An exogenous molecule can be the same type of molecule as an endogenous molecule, for example, an exogenous protein or nucleic acid. For example, an exogenous nucleic acid may comprise an infectious viral genome, a plasmid or episome introduced into a cell, or a chromosome that is not normally present in the cell. Methods for introducing exogenous molecules into cells are known to those skilled in the art and include, among others, lipid-mediated transfer (ie, liposomes, including neutral and cationic liplets), electroporation, direct injection, fusion of cell, particle bombardment, calcium phosphate co-precipitation, DEAE-dextran-mediated transfer and vector-mediated transfer. In contrast, an "endogenous" molecule is one that is normally present in a particular cell at a stage of development under particular environmental conditions. For example, an endogenous nucleic acid may comprise a chromosome, the genome of a mitochondria, chloroplast or other organelle, or a naturally occurring episomal nucleic acid. Additional endogenous molecules can include proteins, for example, transcription factors and enzymes. 0 A "fusion" molecule is a molecule in which two or more , subunit molecules are linked, preferably covalently. 5 Subunit molecules can be the same chemical type of molecule, or they can be different chemical types of molecules. Examples of the first type of fusion molecule include, but are not limited to, fusion proteins (for example, a fusion between a ZFP DNA binding domain and a cleavage domain) and fusion nucleic acids (for example, a nucleic acid (encoding the fusion protein described above). Examples of the second type of fusion molecule include, but are not limited to, a fusion between a triple-formed nucleic acid and a polypeptide, and a fusion between a minor ligand groove and an acid. Expression of a fusion protein in a cell can result from the release of the fusion protein to the cell or by release of a polynucleotide that encodes the fusion protein to a cell, in which the polynucleotide is transcribed, and the transcript is translated, to generate the fusion protein. Trans-splicing, polypeptide cleavage and polypeptide binding may also be involved in the expression of a protein in a cell. Methods for delivering polynucleotide and polypeptide to cells are presented elsewhere in this invention. A "gene," for the purposes of the present invention, includes a region of DNA that encodes a gene product (see, infra), as well as all regions of DNA that regulate the production of the gene product, whether or not - 25 regulatory sequences are adjacent to the coding and / or transcribed sequences. Thus, a gene includes, but is not necessarily limited to, promoter sequences, terminators, regulatory translation sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, isolators, boundary elements, origins 30 replication, matrix binding sites and Iocal control regions. "Gene expression" refers to the conversion of information, contained in a gene, into a gene product. A gene product can be the product of direct transcription of a gene, for example, mRNA, tRNA, rRNA, antisense RNA, ribozyme, structure RNA or other type of RNA or a protein produced by translation of an mRNA. The gene products are still they include RNAs that are modified by processes such as capping, poly-denylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, ADP-ribosylation, myristylation, and glycosylation. "Modulation" of gene expression refers to a change in the level of expression of a gene. Expression modulation can include, among 10 others, gene activation and gene suppression. Modulation can also be complete, among others, in which the expression of the gene is totally inactivated or is activated in levels of wild type or beyond; or it can be partial, in which the expression of the gene is partially reduced, or partially activated to some fractions of wild type levels. "Eukaryotic" cells include, among others, fungal cells (such as yeast), plant cells, animal cells, mammalian cells and human cells (for example, T cells). The terms "operative link" and "operatively linked" (or "operatively linked") are used interchangeably with reference to a juxtaposition of two or more components (such as sequence elements), in which components are arranged so that both components function normally and allow the possibility that at least one of the components can mediate a function that is exercised in at least one of the other components. By way of illustration, a regulatory transcription sequence, like a promoter, is operatively linked to a coding sequence if the regulatory transcription sequence controls the transcription level of the coding sequence in response to the presence or absence of one or more regulatory transcription factors. A regulatory transcription sequence is generally operationally linked in cis with a coding sequence, but need not be directly adjacent to it. For example, an enhancer is a regulatory transcription sequence that is operationally linked to a sequence. coding frequency, although not contiguous. With regard to fusion polypeptides, the term "operably linked" can refer to the fact that each of the components performs the "same function in linking to another component as it would have if it were not linked. For example, with respect to a 5-fusion polypeptide that a ZFP DNA Binding domain is fused to a cleavage domain, the ZFP DNA binding domain and cleavage domain are operatively linked if, at fusion polypeptide, the portion of the ZFP DNA binding domain is capable of binding its target site and / or its binding site, while the cleavage domain is capable of cleaving DNA in the vicinity of the target site, similarly, with respect to to a fusion polypeptide in which a ZFP DNA binding domain is fused to an activation or repression domain, the ZFP DNA binding domain and activation or repression domain are in operative connection if, in the fusion polypeptide , the portion of the ZFP DNA binding domain is able to bind to its target site and / or its binding site, while the activation domain is able to suppress gene expression or the repression domain is capable of infrarregular gene expression. A "functional fragment" of the protein, polypeptide or nucleic acid is a protein, polypeptide or nucleic acid whose sequence is not identical to the full-length protein, polypeptide or nucleic acid, yet retains the same function as the protein of total length, polypeptide or nucleic acid. A functional fragment can have more, less or the same number of residues as the corresponding native molecule, and / or it can contain one or more nucleic acid or nucleotide substitutions. Methods for determining the function of a nucleic acid (for example, coding function, ability to hybridize to another nucleic acid) are well known in the art. Similarly, methods for determining protein function are well known. For example, the DNA-binding function of a polypeptide can be determined, for example, by filter binding, electrophoretic mobility change, or immunoprecipitation assays. DNA cleavage can be assessed by gel electrophoresis. See Ausubel et al., Supra. The ability of the protein to interact with another protein can be determined, for example, by co-immunoprecipitation, two-hybrid assays or complementation, both genetic and biochemical. See, for example, Fields et al. (1989) Nature 340: 245-246; US patent 5,585,245 and PCT WO 98/44350. A "vector" is capable of transferring gene sequences to target cells- Typically, "vector construct," "expression vector," and "gene transfer vector," means that any nucleic acid construct capable of targeting the expression of a gene of interest and that can transfer gene sequences to target cells. Thus, the term includes cloning, and expression vehicles, as well as vectors of integration. A "reporter gene" or "reporter sequence" refers to any sequence that produces a protein product that is easily measured, preferably although not necessarily in a routine assay. Suitable reporter genes include, but are not limited to, sequences encoding proteins that mediate antibiotic resistance (for example, ampicillin resistance, neomycin resistance, G418 resistance, pure mycine resistance), sequences encoding colored proteins or fluorescent or luminescent (for example, green fluorescent protein, improved green fluorescent protein, red fluorescent protein, Iuciferase), and proteins that mediate improved cell growth and / or gene amplification (for example, dihydrofolate reductase). Epitope markers include, for example, one or more copies of FLAG, His, myc, Tap, HA or any detectable amino acid sequence. "Expression markers" include sequences that encode reporters that can be operatively linked to a desired gene sequence to monitor expression of the gene of interest. Overview Zinc finger protein transcription factors (ZFP-TFS) are described here and / or nucleases (e.g., ZFNs) targeting the PDl gene as well as compositions comprising and methods of using these ZFP-TFs and / or nucleases to treat disease or disorders in which PDl is abnormally or undesirably expressed, or in which pathway PDl is abnormally or undesirably used due to overexpression of a PDl ligand, including, for example, treatment of chronic infectious diseases, cancers, and / or autoimmune diseases. For the treatment of a subject with a disease or disorder that is alleviated by the modulation of PD1 expression, the ZFP-TFS and nucleases described here can be introduced into the cells in vivo or ex vivo (for example, primary cells isolated from a patient afflicted with said disease). After treating ZFP-TF and / or ZFN, cells can be reintroduced into the patient for use as a medication in the treatment of a chronic infectious disease or cancer. Similarly, stem cells can be used that have been treated with 10 PDF-specific ZFNS and / or ZFP-TFs. These cells can be infused into a distressed patient for treatment of said medical condition. . The compositions and methods described herein, therefore, allow modulation of a PD1 gene. PDl expression can be inactivated, silenced, up-regulated or down-regulated using ZFP TFS or 15 PDF-specific ZFNs, depending on the need. PDl expression can be downregulated with ZFP-TFS or one or more ZFNS specific for PDl, for example, in patients afflicted with chronic infectious diseases or cancers, and can be upward regulated in patients, for example, patients with autoimmune disease. The methods and compositions of the invention further provide therapeutics comprising a polypeptide encoding ZFP TFS and / or PDl-specific nucleases, in which the polynucleotide is administered directly to the patient. In addition, the invention provides methods and compositions in which the polynucleotide encoding ZFP TFs and / or nucleases can be incorporated into a vector as a viral delivery vehicle for systemic administration as a therapeutic for an afflicted patient. DNA binding domains Compositions comprising a DNA binding domain that specifically binds to an active site in a 30 PD1 gene are described here. Any DNA binding domain can be used in the compositions and methods described herein, including but not limited to a zinc finger DNA binding domain or a DNA binding domain of a meganuclease- . In certain embodiments, the DNA-binding domain comprises a zinc finger protein. Preferably, the finger protein of zinc is non-naturally occurring in that which is designed to bind to a target site of choice. See, for example, Beerli et al- (2002) Nature Biotech-. 5 nol. 20: 135-141: Pabo et al. (2001) Ann. Rev. Bioche / n. 70: 313-340; lsalan et al. (2001) Nature Biotechnol. 19: 656-660; Segal et al- (2001) Curr. Opin. Biotechnol. 12: 632-637; Choo et al. (2000) Curr. Opin. Struct. Biol. 10: 411- 416: US patents 6,453,242; 6,534,261; 6,599,692; 6,503,717; 6,689,558; 7,030,215; 6,794,136; 7,067,317; 7,262,054; 7,070,934; 7,361,635; 7,253,273; 10 and US patent publication 2005/0064474; 2007/0218528; 2005/0267061, all of which are incorporated by reference in their entirety. A designed zinc finger binding domain may have a new binding specificity, compared to a naturally occurring zinc finger protein. Engineering methods include, but are not limited to, rational design 15 and various types of selection. The rational design includes, for example, using databases comprising triplet nucleotide sequences (Olj quadruplets) and individual zinc finger amino acid sequences, in that each triplet or quadruplet sequence is associated with one or more zinc finger amino acid sequences that bind to the triplet or quadruplet particle sequence. See, for example, US patents of co-ownership 6,453,242 and 6,534-261, incorporated herein by reference in their entirety. Exemplary selection methods, including phage display and two hybrid systems, are described in U.S. Patents 25 5,789,538; 5,925-523; 6,007,988; 6,013,453; 6,410,248; 6,140,466; 6,200,759; and 6,242-568; as well as WO 98/37186; WO 98/53057; WO 00/27878; WO 01/88197 and GB 2,338,237. In addition, the improvement in the binding specificity for zinc finger binding domains has been described, for example, in WO 02/077227 co-ownership. 30 In addition, as revealed in these and other references, zinc finger domains and / or multi-finger zinc finger proteins can be linked together using any linker sequence, including, for example, ligands of 5 or more amino acids in length. See also US patent applications 6,479,626; 6,903-185; and 7,153,949 for sequence. exemplary linkers 6 or more amino acids in length. The proteins described here can include any combination of appropriate ligands between individual zinc fingers of the protein. In addition, the improvement in binding specificity for zinc finger binding domains has been described, for example, in co-ownership WO 02/077227. The selection of target sites; ZFPs and methods for designing and building fusion proteins (and polynucleotides that encode them) 10 are known to those skilled in the art and described in detail in US patents 6,140.0815: 789,538; 6,453,242; 6,534,261: 5,925,523; 6.007.988: "6,013,453; 6,200,759; WO 95/19431; WO 96/06166; WO 98/53057; WO 98/54311; WO 00/27878: WO 01/60970 WO 01/88197; WO 02 / 099084; WO 98/53058; WO 98/53059: WO 98/53060; WO 02/016536 and WO 03/016496.15 In addition, as disclosed in these and other references, zinc finger domains and / or finger proteins multi-finger zinc can be linked together using any appropriate linker sequence, including, for example, ligands of 5 or more amino acids in length. See also US patent applications 6,479,626; 6,903,185; and 7,153,949 for exemplary linker sequences 6 or more amino acids in length. The proteins described here can include any combination of appropriate ligands between individual zinc fingers of the protein. Alternatively, the DNA-binding domain can be derived from a nuclease. For example, the recognition sequences of 25 homing endonucleases and meganucleases such as l-Scel, l-Ceul, Pl-Psp !, Pl-Sce, l-ScelV, l-Csml, | -Panl, l-Scell, l-Ppol , l-Scell !, l-Crel, 1-Tevl, 1-Tevll and | - Tevlll are known. See, also U.S. patent No. 5,420,032; U.S. patent No. 6,833,252; Beifort et al. (1997) Nuc / eic Acids Res. 25: 3379-3388: Dujon et al. (1989) Gene 82: 115-118; Perler et al. (1994) Nucleic Acids Res. 22, 30 1125-1127; Jasin (1996) Trends Genet. 12: 224-228; Gimble et al. (1996) j. Mol. Biol. 263: 163-180; Argast et al. (1998) J. Mol. Biol. 280: 345-353 and the New England Biolabs catalog. In addition, the link specificity "to the DNA of homing endonucleases and meganucleases can be designed to bind unnatural target sites. See, for example, Chevalier et al. (2002) "" Mo / ec-Cell 10: 895-905; Epinat et al. (2003) Nuc / eic Acids Res. 31: 2952- 2962; Ashworth et al. (2006) Nature 441: 656-659; Paques et al. (2007) Curent 5 Gene Therapy 7: 49-66; US patent publication 20070117128. In certain embodiments, the DNA binding domain is a designed zinc finger protein that binds (in a sequence-specific way) to a target site in a PD1 gene and modulates PDl expression. ZFPs can selectively bind to a mutant PD1 allele or a wild-type PD1 sequence. Targeted PD1 sites typically include at least one zinc finger, but may include a zinc finger pIurality (eg, 2, 3, 4, 5, 6 or more fingers). Generally, ZFPs include at least three fingers. Certain ZFPs include four, five or six fingers. ZFPS that include three fingers typically has a target site that includes 9 or 10 15 nucleotides; ZFPS that include four fingers typically recognize a target site that includes 12 to 14 nucleotides; while ZFPS containing six fingers can recognize target sites that include 18 to 21 nucleotides. The ZFPS can also be fusion proteins that include one or more regulatory domains, whereas these regulatory domains can be activation of transcription or repression domains. In some embodiments, the DNA binding domain is a projected domain of a TAL effector derived from the plant pathogen Xanthonas (see, Boch et al, (2009) Science 29 Oct 2009 (10-1126 / science- 117881) and Moscow and Bogdanove, (2009). Science 29 Oct 2009 (10.1126 / sci 25 ence.1178817). See also US patent application 13 / 068,735. Fusion proteins Fusion proteins comprising DNA-binding proteins (for example , ZFPS) as described here and a heterologous (functional) regulatory domain (or functional fragment thereof) are still provided 30 Common domains include, for example, transcription factor domains (activators, repressors, co-activators, co -repressors), silencers, oncogenes (for example, members of the myc family, jun, fos, myb, max, mad, rel, ets, bcl, myb, mos etc.); DNA repair enzymes and their associated and modifying factors; DNA rearrangement enzymes and their associated factors. data and modifiers; chromatin-associated proteins and their modifiers (for example kinases, acetylases and deacetylases); and DNA modifying enzymes (eg, methyltransferases, topoisomerases, helicases, ligases, kinases, phosphatases, polymerases, endonucleases) and their associated and modifying factors. US patent application publication 20050064474; 20060188987 and 2007/0218528 for details regarding fusions of DNA binding domains and nuclease cleavage domains, incorporated 10 by reference in their entirety here. Appropriate domains for obtaining activation include the HSV VP16 activation domain (see, for example, Hagmann et al., J. Virol. 71, 5952-5962 (1997)) nuclear hormone receptors (see, for example, Torchia et al., Curr. Opin. Ce / l. Biol. 10: 373-383 (1998)); the p65 subunit of factor nu- 15 clear cover B (Bitko & Barik, J. Virol. 72: 5610-5618 (1998) and Doyle & Hunt, Neuroreport 8: 2937-2942 (1997)); Liu et al., Cancer Gene Ther. 5: 3-28 (1998)), or artificial chimeric functional domains such as VP64 (Beerli et al., (1998) Proc. Natl. Acad. Sci. USA 95: 14623-33), and degron (Molinari et al., (1999) EMBO J. 18, 6439-6447). Exemplary additional activation domains, Oct 1, OCt-2A, Spl, AP-2, and CTFI (Seipel et al., EMBO J. 11, 4961- 4968 (1992) as well as p300, CBP, PCAF, SRCl PvALF, AtHD2A and ERF-2. See, for example, Robyr et al. (2000) Mol. Endocrinol. 14: 329-347; Colling-wood et al. (1999) J. Mol. Endocn'nol. 23 255-275; Leo et al. (2000) Gene 245: 1-11; Manteuffel-Cymborowska (1999) Acta Biochim. Pol. 46: 77-89; Mc- 25 Kenna et al. (1999) J. Steroid Biochem. Mol. Biol. 69: 3-12; Malik et al. (2000) Trends Biochem. Sci. 25 277-283; and Lemon et al. (1999) Curr. Opin. Genet. Dev. 9: 499-504. Additional exemplary activation domains include, but are not limited to, OsGAl, HALF-I, Cl, APl, ARF-5, -6, -7, and -8, CPRFI, CPRF4, MYC-RP / GP, and TRABI. See, for example, Ogawa et al. (2000) Gene 245: 21-29; 30 Okanami et al. (1996) Genes Cells 1: 87-99; Goff et al. (1991) Genes Dev. 5: 298-309; Cho et al. (1999) P / ant Mol. Biol. 40: 419-429; Ulmason et al. (1999) Proc. Natl. Acad. Sci. USA 96: 5844-5849; Sprenger-Haussels et al. (2000) PIant J. 22: 1-8; Gong et al. (1999) P / ant Mol. Biol. 41: 33-44; and Hobo et al. (1999) Proc. Natl. Acad. Sci. USA 96: 15,348-15,353. B . It will be clear to those skilled in the art that in the formation of a fusion protein (or a nucleic acid encoding it) between a DNA binding domain and a functional domain, an activation domain or a molecule that interacts with an activation domain is appropriate as a functional domain. Essentially any molecule capable of recruiting an activation complex and activation activity (such as histone acetylation) to the target gene is useful as an activation domain 10 of a fusion protein. Isolating domains, localization domains, and chromatin remodeling proteins as domains containing ISW1 and / or methyl-binding protein domains for use as functional domains in fusion molecules are described, for example, in patent applications. North American co-ownership2002 / 0115215 and 2003/0082552 and in 15 WO 02/44376 of co-ownership. Exemplary repression domains include, but are not limited to, KRAB NB, KOX, TGF-beta-inducible (TIEG), v-erbA, SID, MBD2, MBD3, members of the DNMT family (eg, DNMTI, DNMT3A, DNMT3B ), Rb, and MeCP2. See, for example, Bird et al. (1999) Ce // 99: 451-20 454; Tyler et al. (1999) Ce // 99: 443-446; Knoepfler et al. (1999) Ce // 99: 447-450; and Robertson et al. (2000) Nature Genet. 25: 338-342. Additional exemplary domains of repression include, among others, ROM2 and AtHD2A, See, for example, Chem et al. (1996) Plant Ce // 8: 305-321; and Wu et al. (2000) P / antj. 22: 19-27. 25 Fusion molecules are constructed by biochemical cloning and conjugation methods that are well known to those skilled in the art. Fusion molecules comprise a DNA binding domain and a functional domain (for example, a transcriptional or repressive activation domain). Fusion molecules still optionally comprise nuclear localization signals (such as that of SV40 antigen T medium) and epitope markers (such as, for example, FLAG and hemagglutinin). Fusion proteins (and nucleic acids that encode them) are designed so that the translation reading region is preserved between the components of the fusion. . Fusions between a polypeptide component of a functional domain (or a functional fragment thereof) on the one hand, and a non-protein DNA binding domain (eg, antibiotic, interleaver, minor groove ligand, nucleic acid) by on the other hand, they are constructed by the biochemical conjugation methods known to those skilled in the art. See, for example, the Pierce Chemical Company catalog (Rockford, IL). Methods and compositions for preparing fusions between a minor groove linker and a polypeptide have been described. Mapp et al- (2000) Proc. Natl. Acad. Sci. USA 97: 3930-3935. In certain embodiments, the target site linked by the zinc finger protein is present in an accessible region of cellular chromatin. Accessible regions can be determined as described, for example, in the international co-ownership publication WO 01/83732. If the target site is not present in an accessible region of cell chromatin, one or more accessible regions can be generated as described in WO 01/83793 of co-ownership. In additional embodiments, the DNA binding domain of a fusion molecule is capable of binding cellular chromatin regardless of whether its target site is an accessible region or not. For example, said DNA binding domains are capable of binding the DNA ligand and / or nucleosomal DNA. Examples of this type of "pioneer" DNA-binding domain are found in a given steroid receptor and in heparocyte nuclear factor 3 (HNF3). Cordingley et al. (1987) Cell 48: 261-270; Pina et al. (1990) 25 Ce // 60: 719-731; and Cirillo etal. (1998) EMBO J. 17: 244-254. The fusion molecule can be formulated with a pharmaceutically acceptable carrier, as is known to those skilled in the art. See, for example, Remington's Pharmaceutical Sciences, 17th ed., 1985; and WO 00/42219 co-ownership. 30 The component / functional domain of a fusion molecule can be selected from any of a variety of different components capable of influencing the transcription of a gene once the fusion molecule binds to a target sequence through its DNA-binding domain- Thus, the functional component can include, among others, several domains. transcription factor, such as activators, repressors, co-activators, m co-repressors, and silencers. 5 Additional exemplary functional domains are described, for example, in the US patent co-ownership 6,534,261 and US patent application publication 2002/0160940. Functional domains that are regulated by small exogenous Olj ligand molecules can still be selected. For example, RheoSwitch® technology can be used in which a functional domain only assumes this active conformation in the presence of an external Rheo- 'Chem' ligand (see, for example, US 20090136465). Thus, the ZFP can be operated linked to the adjustable functional domain in which the resulting ZFP-TF activity is controlled by the external linker. In certain embodiments, the fusion protein comprises a DNA binding domain and a cleavage domain (nuclease). As such, gene modification can be achieved using a nuclease, for example, a designed nuclease. Nuclease engineering technology is based on the engineering of naturally occurring DNA-binding proteins. The methods and compositions described here 20 are widely applicable and can involve any nuclease of interest. Non-limiting examples and nucleases include zinc finger meganucleases and nucleases. The nuclease may comprise heterologous DNA binding domains and cleavage (e.g., zinc finger nucleases; mega-nuclease DNA binding domains with heterologous cleavage domains) or, alternatively, the DNA binding domain a naturally occurring nuclease can be altered to bind to a selected target site (for example, a meganuclease that was designed to bind to a different site than the cognate recognition site). For example, engineering endonuclease homing with tailored DNA binding specificities has been described, see Chames et al. (2005) Nuc / eic Acids Res 33 (20): e178; Arnould et al. (2006) j- Mol. Biol, 355: 443-458 and Grizot et al (2009) Nuc / eic Acids Res July 7 and publication. In addition, ZFP engineering was also 'described. See, for example, US patents 6,534,261; 6,607,882; 6,824,978; 6,979,539; 6,933,113; 7,163,824; and 7,013,219. . In certain embodiments, the nuclease is a meganuclease ( Donuclease homing). Naturally occurring meganucleases recognize 15-40 5 base pair cleavage sites and are commonly grouped into four families. the LAGLIDADG family, the GIY-YIG family, the His-Cyst box family and the HNH family. Examples of homing endonucleases include l-Scel, l-Ceul, Pl-Pspl, Pl-Sce, l-ScelV, l-Csml, l-Panl, I-Scell, | -Ppol, l-Scelll, I-Crel, 1 -Tevl, 1-Tevll and 1-TevIIl. Its recognition strings are known. See, - also US patent 5,420,032; U.S. Patent No. 6,833,252; Belfort et al. (1997) Nuc / eic Acids Res. 25: 3379-3388; Dujon et al. (1989) Gene 82: 115- "118: Perler et al. (1994) Nucleic Acids Res, 22, 1125-1127; Jasin (1996) Trends Genet. 12: 224-228; Gimble et al. (1996) J- Mol. Biol. 263: 163-180; Argast et al. (1998) J. Mol. Biol. 280: 345-353 and the New England Biolabs catalog. 15 DNA binding domains of naturally occurring meganucleases, mainly from the family LAGLIDADG, were used to promote site-specific genome modification in plants, yeast, Drosophila, mammalian cells and mice, but this approach was limited to modifying homologous genes that preserve the meganuclease recognition sequence (Monet et al. (1999), Biochem. Biophysics. Res. Co / nmon. 255: 88-93) or for pre-engineered genomes in which a sequence of recognition has been introduced (Route et al. (1994), Mol. Ce //. Biol. 14: 8096-106: Chilton et al, (2003), P / ant Physio / ogy. 133: 956-65; Puchta et al. (1996), Proc. Natl. Acad. Sci USA 93: 5055-60; Rong et al. 25 (2002), Genes Dev. 16: 1568-8 1; Gouble et al. (2006), J- Gene Med. 8 (5): 616-622). Thus, attempts have been made to prepare meganucleases designed to present new binding specificity at clinically or biotechnologically relevant sites (Porteus et al. (2005), Nat. Biotechnol. 23: 967-73; Sussman et al- (2004 ), J. Mol. Biol. 342: 31-41; Epinat et 30 al. (2003), Nuc / eic Acids Res. 31: 2952-62; Chevalier et al. (2002) Molec. Ce // 10: 895 -905; Epinat et al. (2003) Nucleic Acids Res. 31: 2952-2962; Ash-worth et al. (2006) Nature 441: 656-659; Paques et al. (2007) Current Gene Therapy 7: 49-66; US patent publication 20070117128; 20060206949: 20060153826; 20060078552; and 20040002092). In addition, li- domains. binding to naturally occurring or projected meganuclease DNAs were further operably linked with a cleavage domain from a heterologous nuclease (e.g., Fokl). In other embodiments, the nuclease is a zinc finger (ZFN) nuclease. ZFNS comprise a zinc finger protein that has been designed to bind to a target site in a gene of choice and cleavage domain or a cleavage half-domain. · 10 As noted above, zinc finger binding domains can be designed to bind to a sequence of choice. See, for example, 'Beerli et al. (2002) Nature Biotechnol. 20: 135-141; Pabo et al. (2001) Ann. Rev. Biochem. 70: 313-340; Isalan et al. (2001) Nature Biotechnol. 19: 656-660; Segal et al. (2001) Curr. Opin. Biotechnol. 12: 632-637; Choo et al. 15 (2000) Curr. Opin. Struct. Biol. 10: 411-416. A zinc finger binding domain may have a new binding specificity, compared to a naturally occurring zinc finger protein. Engineering methods include, among others, rational design and various types of selection. The rational design includes, for example, use of databases comprising triplet (or quadruplet) nucleotide sequences and functional zinc finger amino acid sequences, where each triplet or quadruple nucleotide sequence is associated with one or more sequences zinc finger amino acid that binds to the particular triplet or quadruplet sequence. See, for example, US co-ownership patents. 6,453,242 and 6,534,261, incorporated by reference here in their entirety. Exemplary selection methods, including phage display and two-hybrid systems, are described in US patents 5,789,538; 5,925-523; 6,007,988; 6,013,453; 6,410,248; 6,140,466; 6,200,759; and 6,242,568; as well as WO 98/37186: WO 98/53057; WO 00/27878; WO 30 01/88197 and GB 2,338,237. In addition, the improvement of binding specificity for zinc finger binding domains has been described, for example, WO 02/077227 of co-ownership. The selection of target sites; ZFNS and methods for design and construction of fusion proteins (and polynucleotides encoding them) are known to those skilled in the art and described in detail in US Patent Application Publication20050064474 and 20060188987, incorporated by reference in your wholes here. In addition, as disclosed in this and other references, zinc finger domains and / or multi-finger zinc finger proteins can be linked together using any appropriate linker sequence, including, for example, linkers of 5 or more amino acids in length. See, for example, - 10 US patents 6,479,626; 6,903,185; and 7,153,949 for exemplary linker sequences 6 or more amino acids in length. The proteins described here can include any combination of appropriate ligands between the individual zinc fingers of the protein. Nucleases such as ZFNs and / or meganucleases still comprise a nuclease (cleavage domain, half-cleavage domain). As noted above, the cleavage domain can be heterologous to the DNA binding domain, for example, a zinc finger DNA binding domain and a nuclease cleavage domain or a DNA binding domain and cleavage domain of a different nuclease. Heterologous cleavage domains can be obtained from any endonuclease or exonucleosis. Examples of endonucleases from which a cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases. See, for example, 2002-2003 Catalog, New England Biolabs, Beverly, MA: and Belfort et al. (1997) Nuc / eic Acids Res. 25: 3379-25 2588. Additional enzymes that cleave DNA are known (for example, Sl Nuclease; bean sprout nuclease; pancreatic DNase I; micrococcal nuclease; yeast endonuclease; see also Linn et al. (eds.) Nuc / aces, Cold Spring Harbor Laboratory Press, 1993). One or more of these enzymes (or functional fragments thereof) can be used as a source of cleavage domains and half-domains of cleavages. Similarly, a cleavage half-domain can be derived from any nuclease or portion thereof, as set out below, which requires dimerization for cleavage activity. In general, two fusion proteins are required for cleavage if the fusion proteins match. comprise half domains of cleavages. Alternatively, a simple protein comprising two half-domains of cleavages can be used. The two cleavage half-domains can be derived from the same endonuclease (or functional fragments thereof), or each cleavage half-domain can be derived from a different endonuclease (or functional fragment thereof). In addition, the target sites for the two fusion proteins are preferably arranged, with respect to each other, of nio- 10 than the binding of the two fusion proteins to their respective target sites places the cleavage half domains in one spatial orientation to each other that allows the cleavage half domains to form a functional cleavage domain, for example, by demerization. Thus, in certain embodiments, the edges near the target sites are separated by 15 5-8 nucleotides or 15-18 nucleotides. However, any integral number of nucleotides or nucleotide pairs can intervene between two target sites (for example , from 2 to 50 pairs of nucleotides or more). In general, the cleavage site is among the target sites. Restriction endonucleases (restriction enzymes) are present in many species and are capable of specific binding to the DNA sequence (at a recognition site), and cleave DNA at or near the binding site. Certain restriction enzymes (for example, type IIS) cleave DNA on sites removed from the recognition site and have separable binding and cleavage domains. For example, the 11S type of the Fok I 25 enzyme catalyzes double-stranded DNA cleavage, in 9 nucleotides from its recognition site on one strand and 13 nucleotides from its recognition site on the other. See, for example, US Patents 5,356,802; 5.436.1 "0 and 5,487-994; as well as Li et al. (1992) Proc. Natl. Acad. Sci. USA 89: 4275- 4279; Li et al. (1993) Proc. Natl. Acad. Sci. USA 90: 2764-2768; Kim et al. 30 (1994a) Proc. Natl. Acad. Sci. USA 91: 883-887; Kim et al. (1994b) J. Biol, Chem. 269: 31,978-31,982. Thus, in one embodiment, fusion proteins comprise the cleavage domain (or cleavage half-domain) of at least one Type IIS restriction enzyme and one or more zinc finger binding domains, which may or may not be designed. . An exemplary restriction enzyme type llS, whose cleavage domain is separable from the binding domain, is Fok I. This particular enzyme is active as a dimer. Bitinaite et al. (1998) Proc. Natl. Acad. Sci. USA 95: 10,570-10,575. Thus, for the purposes of the present invention, the portion of the Fok I enzyme used in disclosed fusion proteins is considered to be a half-cleavage domain. Thus, for double-stranded targeted cleavage and / or targeted replacement of cell sequences using - 10 zinc-Fok I finger fusions, two fusion proteins, each comprising a Fokl half-cleavage domain, can be used to reconstitute a domain catalytically active cleavage, Alternatively, a single polypeptide molecule containing a zinc finger binding domain and two Fok I cleavage half domains can still be used. Parameters 15 for targeted cleavage and targeted sequence change using zinc-Fok I finger fusions are provided elsewhere in this invention. A cleavage domain or cleavage half-domain can be any portion of a protein that retains cleavage activity, or that retains the ability to multimerize (e.g., dimerize) to form a functional cleavage domain. Exemplary type IIS restriction enzymes are described in International Publication WO 071014275, incorporated here in their entirety. Additional restriction enzymes still contain separable binding domains and cleavages, and these are contemplated by the present invention - See, for example, Roberts et al- (2003) Nuc / eic Acids Res. 31: 418-420. In certain embodiments, the cleavage domain comprises one or more projected cleavage half-domains (still referred to as dimerization domain mutants) that minimize or prevent homodimerization, as described, for example, in patent publication US 30 20050064474 and 20060188987 and North American Order 11 / 805,850 (deposited on May 23, 2007), the disclosures of which are incorporated by reference in their entirety here. Amino acid residues in the positions "Fok I sions 446, 447, 479, 483, 484, 486, 487, 490, 491, 496, 498, 499, 500, 531, 534, 537, and 538 are all targets for influencing Fok dimerization" I cleavage half-domains. Examples of Fok I 5 projected cleavage half-domains that form mandatory heterodimers include a pair in which a first cleavage half-domain includes mutations in amino acid residues at positions 490 and 538 of Fok I and a second cleavage half-domain includes mutations in amino acid residues 486 and 499. Thus, in one embodiment, a mutation in 490 replaces Glu "10 (E) with Lys (K); the 538 mutation replaces lso (I) with Lys (K); the 486 mutation replaced Gln (Q) with Glu (E): and the 499 mutation replaces lso ( I) with Lys (K) Specifically, the projected cleavage half-domains described here were prepared by mutation positions 490 (E ~ K) and 538 (I ~ K) in a cleavage half-domain to produce a half-projected cleavage domain designated "E490K: 1538K" and by mutation of positions 486 (Q ~ E) and 499 (I ~ L) in another cleavage half-domain to produce a designated half-domain of cleavage designated "Q486E : 1499L ". The projected cleavage half-domains described here are mandatory heterodimer mutants in which abnormal cleavage is minimized or abolished. 20 See, for example. Provisional Order Example US 60 / 808,486 (deposited May 25, 2006), the disclosures of which have been incorporated by reference in their entirety for all purposes- Projected half-domains described here can be prepared using any appropriate method, for example, by genesis targeting the wild type cleavage half-domain site (Fok l) as described in US Patent Publication Example 5 20050064474 and US Patent Publication Example 38 2007 / 0305346 and 2008/0131962 and provisional patent application US 61 / 337,769, filed on February 8, 2010 and 61 / 403,916, filed on September 23, 2010. 30 Nuclease expression constructs can be readily designed using methods known in the technical. See, for example, US Patent Publications 20030232410; 20050208489; 20050026157; 20050064474; 20060188987; 20060063231: and International publication WO 07/014275. In certain embodiments, nuclease expression is under the control of an inducible promoter, for example, the galactokinase promoter that is activated (de-repressed) in the presence of raffinose and / or galactose and dammed in the presence of glucose. In particular, the galactokinase promoter is induced and the nucleases expressed in successive changes in the carbon source (for example, from glucose to raffinose to galactose). Other non-limiting examples of inducible promoters include CUPl, MET15, PHO5, and tet-responsive promoters. - 10 Release The proteins (eg, ZFPS), polynucleotides that encode them and compositions comprising the proteins and / or polynucleotides described herein can be released to a target cell by any appropriate means. Suitable cells include, but are not limited to, eukaryotic and prokaryotic cells and cell lines. Non-limiting examples of said cells or cell lines generated from said cells include COS, CHO (for example, CHO-S, CHO-KI, CHO-DG44, CHO-DUXBII, CHO-DUKX, CHOKISV) , VERO, MDCK, Nl38, V79, B14AF28-G3, BHK, Hak, NSO, SP2 / 0-Ag14, HeLa, HEK293 (e.g. HEK293-F, 20 HEK293-H, HEK293-T), and perC6 cells as well as insect cells like Spodoptera fugiperda (Sf), or fungi cells like Saccharomyces, Pi- chia and Schizosaccharomyces. In certain embodiments, the cell line is a CHO-KI, MDCK or HEK293 cell line. Suitable primary cells include peripheral blood mononuclear cells (PBMC), and other subsets of blood cells such as, inter alia, T cells. CD4 + or CD8 + T cells. Suitable cells also include stem cells such as, for example, embryonic stem cells, induced pluripotent stem cells, hematopoietic stem cells, neuronal stem cells and mesenchymal stem cells. Protein delivery methods comprising zinc finger proteins as described herein are described, for example, in US patents 6,453,242; 6,503,717; 6,534,261; 6-599,692; 6,607 882: 6,689,558; 6,824,978; 6,933,113; 6,979,539; 7,013,219; and 7,163,824, the revelations of which are incorporated here in their entirety. The zinc finger proteins as described herein can further be released using vectors containing sequences that encode one or more of the 5 zinc finger proteins. Any vector system can be used including, but not limited to, plasmid vectors, retroviral vectors, lentiviral vectors, adenovirus vectors, poxvirus vectors; herpesvirus and adeno-associated vectors, etc. See also US patents 6,534-261; 6,607,882; 6,824,978; 6,933,113: 6,979,539: 7,013,219; and 7,163,824, incor- porated by reference here in their entirety. Besides that. it will be apparent that any of these vectors may comprise one or more sequences that encode zinc finger protein. Thus, when one or more ZFPS are introduced into the cell, the ZFPs can be conducted on the same or different vectors. When multiple vectors are used, each vector can comprise a sequence encoding one or more ZFPs. Conventional viral and non-viral gene transfer methods can be used to introduce nucleic acids encoding projected ZFPS into target cells (eg, mammalian cells) and tissues. These methods can also be used to deliver nucleic acids that encode ZFPs to cells in vitro. In certain embodiments, nucleic acids encoding ZFPS are administered for gene therapy uses in vivo or ex vivo. Non-viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer. Viral vector release systems include DNA and RNA viruses, which have episomal or integrated genomes after release to the cell. For a review of gene therapy procedures, see Anderson, Science 256: 808-813 (1992); Nabel & Felgner, TIBTECH 11: 211-217 (1993); Mitani & Caskey, TIBTECH 11: 162-166 (1993): Dillon, T / BTECH 11: 167-175 (1993); Miller, Nature 357: 455-460 (1992); Van 30 Brunt, Biotechno / ogy 6 (10): 1149-1154 (1988); Vigne, Restorative Neuro / ogy and Neuroscience 8: 35-36 (1995); Kremer & Perricaudet, British Medical Bul- etin 51 (1): 31-44 (1995); Haddada et al., In Current Topics in Microbio / ogy and lmmuno / ogy Doerfler and Bõhm (eds.) (1995): and Yu et al., Gene Therapy 1: 13-26 (1994). Methods for non-viral release of nucleic acids include electroporation, lipofection, microinjection, biolistics, virosomes, liposomes, i-5 munoliposomes, polycation or Iipid conjugates: nucleic acid, naked DNA, artificial virions, and agent-enhanced DNA uptake . Sonoporation using, for example, the Sonitron 2000 system (Rich-Mar) can also be used to release nucleic acids. Additional examples of nucleic acid - 10 delivery systems include those provided by Amaxa® Biosystems (Cologne, Germany), Maxcyte, lnc. (Rockville, Maryland), BTX Molecular Delivery Systems (Hollison, MA) and Copernicus Therapeutics lnc, (see, for example, US6008336). Lipofection is described in, for example, US 5,049,386, US 4,946. 787; and US 4,897,355) and lipofection reagents are sold commercially (for example, Transfectam '"and Lipofectin"). Cationic and neutral lipids that are suitable for polypucleotide lipofection by efficient receptor recognition include those of Felgner, WO 91/17424, WO 91/16024. The release can be to cells (ex vivo administration) or target sites (in vivo administration). 20 The preparation of lipid: nucleic acid complexes, including liposomes targeted as immunolipid complexes, is well known in the art (see, for example, Crystal, Science 270: 404-410 (1995): Blaese et al., Cancer Gene Ther. 2: 291-297 (1995); Behr et al., Bioconjugate Chem. 5: 382-389 (1994): Remy et al., Bioconjugate Chem. 5: 647-654 (1994): Gao 25 et al., Gene Therapy 2: 710-722 (1995); Ahmad et al., Cancer Res. 52: 4817- 4820 (1992); US patents 4,186,183, 4,217-344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4-774,085, 4,837,028, and 4,946,787). Additional delivery methods include using nucleic acid packaging to be released in En-30 GenelC (EDVS) delivery vehicles. These EDVS are specifically released to target tissues using bispecific antibodies where one arm of the antibody has specificity for the target tissue and the other has specificity for the EDV. The anti- The body brings the EDVs to the surface of the target cell and then the EDV is brought into the cell by endocytosis. Once in the cell, the contents are released (see, '"MacOiarmid et al (2009) Nature Biotechno / ogy vol 27 (7) p. 643). The use of systems based on viral RNA or DNA for the release of nucleic acids encoding engineered ZFPS have the advantage of highly evolved processes to target a virus to specific cells in the body and trafficking the viral load to the nucleus. Viral vectors can be administered directly to patients (in vivo) or can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo). Conventional virus-based systems for ZFP release include, but are not limited to, retroviral vectors, lentiviruses, adenovirals, adeno-associated, vaccinia virus and herpes simplex for gene transfer. Integration of the host genome is possible with methods of gene transfer by retrovirus, lentivirus, and adeno-associated viruses, 15 generally resulting in long-term expression of the inserted transgene. In addition, high transduction efficiencies have been observed in many different types of target cells and tissues. The tropism of a retrovirus can be altered by incorporating foreign envelope proteins, expanding the potential target population of target cells. Lentiviral vectors are retroviral vectors that are capable of transducing or infecting cells not dividing and typically produce high viral titers. The selection of the retroviral gene transfer system depends on the target tissue. Retroviral vectors are comprised of long-acting cis terminal repeats with a packaging capacity of up to 6-25 10 kb of the foreign sequence. The minimum cis-acting LTRS are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene in the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based on murine leukemia virus (MuLV), gibbon monkey leukemia virus (GaLV), simian immunodeficiency virus (SlV), human immunodeficiency virus (HlV), and combinations of same (see, for example, Buchscher et al., J. Virol- 66: 2731-2739 (1992); Johann et al., J- Virol. 66: 1635-1640 (1992); Sommerfelt et al., Virol. 176: 58-59 (1990); Wilson et al., J. Virol. 63: 2374-2378 (1989); Miller et al., J. Virol. 65: 2220-2224 (1991); "PCT / US94 / 05700). In applications where transient expression is preferred, 5 adenoviral-based systems can be used. Adrenoviral-based vectors are capable of very high transduction efficiency in many cell types and do not require cell division.With said vectors, high titer and high levels of expression were obtained.This vector can be produced in large quantities in a relatively simple system.Vector vectors - 10 adeno-associated ("AAV") are still used to transduce cells with target nucleic acids, for example, in vitro production of nucleic acids and peptides, and for in vivo and ex vivo gene therapy procedures (see, for example, West et al., Viro / ogy 160: 38- 47 (1987); US patent 4,797,368; WO 93/24641; Kotin, Human Gene Therapy 5: 793-801 (1994); 15 Muzyczka, J. C / in. Invest. 94: 1351 (1994). Construction of matching AAV vectors are described in a number of publications, including US patent 5,173,414; Tratschin et al., Mol. Ce //. Biol. 5: 3251-3260 (1985); Tratschin, et al., Mol. Cell. Biol. 4: 2072-2081 (1984); Hermonat & Muzyczka, PNAS 81: 6466-6470 (1984); and Samulski et al., J. Vi / o /. 63: 03822-3828 (1989). 20 At least six viral vector approaches are currently available for gene transfer in clinical studies, which use approaches that involve complementation of defective vectors by genes inserted in helper cell lines to generate the transduction agent. pLASN and MFG-S are examples of retroviral vectors that have been used in clinical studies (Dunbar et al., B / ood 85: 3048-305 (1995): Kohn etal., Nat. Med. 1: 1017-102 (1995 ); Malech etal., PNAS 94:22 12133-12138 (1997)). PA317 / pLASN was the first therapeutic vector used in a gene therapy study. (Blaese et al., Science 270: 475-480 (1995)). Transduction efficiencies of 50% or greater were observed for packaged vectors of those with MFG-S. (Ellem et al., / Mmuno / lmmunother. 44 (1): 10-20 (1997); Dranoff et al., Hum. Gene Ther. 1: 111-2 (1997). Recombinant adeno-associated virus vectors ( rAAV) are a promising alternative for gene delivery systems based on defective and non-pathogenic adeno-associated parvovirus type 2 viruses. "All vectors are derived from a plasmid that retains only AAV 145 bp of inverted terminal repeats flanking the cassette of expression are transgene. Efficient gene transfer and stable transgene release due to integration into the transduced cell genomes are key characteristics for this vector system. (Wagner et al., Lancet 351: 9117 1702-3 (1998), Kearns et al., Gene Ther. 9: 748-55 (1996)). Other AAV serotypes, including AAVI, AAV3, AAV4, AAV5, AAV6 and AAV8, can also be - 10 used in accordance with the present invention. Replication-deficient recombinant adenoviral vectors (Ad) can be produced in high titer and readily infect a number of different cell types. Most adenovirus vectors are designed so that a transgene replaces the Ad Ela, Elb, and / or E3 genes; 15 subsequently the defective replication vector is propagated in human 293 cells that provide trans deleted gene function. Ad vectors can transduce various types of tissues in vivo, including differentiated cells not dividing like those found in liver, kidney, and muscle. Conventional Ad vehicles have great driving capacity. An example of the use of an Ad vector in a clinical study involved polynucleotide therapy for anti-tumor immunization with intramuscular injection (Sterman et al., Hum. Gene Ther. 7: 1083-9 (1998)) - Additional examples of the use of adenovirus vectors for transfer in clinical studies include Rosenecker et al., Infection 24: 15-10 (1996); Sterman et al., Hum. Gene 25 Ther. 9: 7 1083-1089 (1998); Welsh et al., Hum. Gene Ther. 2: 205-18 (1995); Alvarez et al., Hum. Gene Ther. 5: 597-613 (1997); Topf et al., Gene Ther. 5: 507-513 (1998); Sterman et al., Hum. Gene Ther. 7: 1083-1089 (1998) - Packaging cells are used to form viral particles that are capable of infecting a host cell. Said cells include 293 cells, which package adenovirus, and uj2 cells or PA317 cells, which package retrovirus. Viral vectors used in gene therapy are generally generated by a producing cell line that packages a nucleic acid vector into a viral particle. Vectors typically contain minimal viral sequences required for packaging and integration. subsequent integration into a host (if applicable), other viral sequences being replaced by an expression cassette encoding the protein to be expressed. The missing viral functions are provided in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only have terminal repeat sequences (ITR) from the AAV genome that are required for packaging and integration into the host genome. Viral DNA is packaged in a line- - 10 cell gem, which contains a helper plasmid that encodes the other AAV genes, called rep and cap, but lacking ITR sequences. The cell line is still infected with adenovirus as a helper. The helper virus promotes AAV vector replication and expression of MV genes from the helper plasmid. The helper plasmid is not packaged in significant quantities due to the lack of ITR sequences. Contamination with adenovirus can be reduced by, for example, heat treatment to which adenovirus is more sensitive than AAV. In many gene therapy applications, it is desirable that the gene therapy vector be delivered with a high degree of specificity to a particular type of tissue. Thus, a viral vector can be modified to have specificity for a particular cell type expressed as a ligand as a fusion protein with a viral coating protein on the external surface of the virus. The ligand is chosen because it has an affinity for a receptor known to be present in the cell type of interest. For example, Han 25 et al., Proc. Natl. Acad. Sci. USA 92: 9747-9751 (1995), reported that Moloney's murine leukemine virus can be modified to express human gp70-fused metabolite, and the recombinant virus infects certain human breast cancer cells that express growth factor receptor - human epidermal. This principle can be extended to other pairs of target-virus cells, where the target cell expresses a receptor and the virus expresses a fusion protein comprising a ligand for the cell surface receptor. For example, filamentous phage can be designed to show antibody fragments (for example, FAB or Fv) containing specific binding affinity for virtually any es- cellular receptor. harvested. Although the above description applies mainly to viral vectors, the same principles can be applied to non-viral vectors. Said ».5 vectors can be designed to contain specific absorption sequences that favor the absorption of specific target cells. Gene therapy vectors can be released in vivo by administration to an individual patient, typically by systemic administration (eg, intravenous, intraperitoneal, intramuscular, subdermal infusion) - 10 ca, or intracranial) or topical application, as described below. Alternatively, vectors can be released to cells ex vivo, as explan- cells. from an individual patient (for example, lymphocytes, bone marrow aspirates, tissue biopsy) or hematopoietic stem cells from a universal donor, followed by reimplantation of cells in a patient, usually after selection for cells that incorporated the vector. Cell transfection ex vivo for diagnosis, research, or gene therapy (for example, via re-infusion of transfected cells into the host organism) is well known to those skilled in the art. In a preferred modality, the cells are isolated from the subject organism, transfected with a ZFP nucleic acid (gene or CDNA), and reinfused back into the subject organism (for example, patient). Various cell types suitable for ex vivo transfection are well known to those skilled in the art (see, for example, Freshney et al., Cu / ture of Animal Cel / s, A Manual of Basic Technique (3rd ed. 1994)) and the references cited here for 25 a discussion of how to isolate and grow patient cells). Suitable cells include, among other eukaryotic and prokaryotic cells and / or cell lines. Non-limiting examples of said cells or cell lines generated from said cells include COS, CHO (for example, CHO-S, CHO-KI, CHO-DG44, CHO-DUXBII, CHO-30 DUKX, CHOKISV), VERO, MDCK, Nl38, V79, B14AF28-G3, BHK, Hak, NSO, SP2 / 0-Ag14, HeLa, HEK293 (e.g. HEK293-F, HEK293-H, HEK293-T), and perC6 cells as well insect cells like Spodoptera fugiperda (Sf), or fungi cells like Saccharomyces, Pichia and Schizo- saccharomyces. In certain embodiments, the cell line is a CHO-KI, MDCK or HEK293 cell line. In addition, primary cells can be isolated and used ex vivo for re-introduction into the subject to be treated after treatment with ZFNs. Suitable primary cells include peripheral blood mononuclear cells (PBMC), and other subsets of blood cells such as, among others, CD4 + T cells or CD8 + T cells. The cells still include stem cells such as, for example, embryonic stem cells, induced pluripotent stem cells, stem cells - 10 hematopoietic, neuronal stem cells and mesenchymal stem cells In one embodiment, stem cells are used in ex vivo procedures for cell transfection and gene therapy. The advantage of using stem cells is that they can be differentiated into other cell types in vitro, or they can be introduced into a mammal (like the donor of the 15 cells) where they will graft into the bone marrow. Methods for differentiating CD34 + cells in vitro into important immune cell types using cytokines such as GM-CSF, lFN-y and TNF-a are known (see, lnaba et al., J. Exp-Med. 176: 1693-1702 (1992)). Stem cells are isolated from transduction and differentiation 20 using known methods. For example, stem cells are isolated from bone marrow cells by panning the bone marrow cells with antibodies that bind to unwanted cells, such as CD4 + and CD8 + (T cells), CD45 "(pan8 cells), GR-l (granulocytes), and lad (differentiated antigen presenting cells) (see, lnaba et al., J. Exp. Med. 176: 1693-25 1702 (1992)) Stem cells that have been modified can still be used in some modalities. For example, stem cells that have been made resistant to apoptosis can be used as therapeutic compositions where the stem cells still contain the ZFP TFS of the invention. Resistance to apoptosis can see, for example, inactivating BAX and / or BAK using BAF or BAK-specific ZFNS (see, US patent application 12 / 456,043) in stem cells, or those that are disrupted in a caspase, new- using caspase-6 specific ZFNS for example. These cells can be transfected into the ZFP TFs that are known to regulate PDl Mutant F or seivative type. Vectors (for example, retroviruses, adenoviruses, liposomes, 5 etc.) containing therapeutic ZFP nucleic acids can also be administered directly to an organism for cell transduction in vivo. Alternatively, naked DNA can be administered. Administration is by any means normally used to introduce a molecule in ultimate contact with blood or tissue cells including, among others, - injection, infusion, topical application and electroporation. Appropriate methods for administering said nucleic acids are available and well known to those skilled in the art, and although more than one route can be used to administer a particular composition, a particular route can generally provide a more immediate and effective reaction than another way. 15 Methods for introducing DNA into hematopoietic stem cells are disclosed, for example, in US patent 5,928,638. Useful vectors for introducing transgenes into hematopoietic stem cells, for example, CD34 "cells, include adenovirus type 35. Vectors suitable for introducing transgenes into immune cells (eg, T cells) include non-integrating Ientivirus vectors, See, for example, Ory et al. (1996) Proc-Natl-Acad. Sci. USA 93: 11382-11388; Dull et al- (1998) J- Virol- 72: 8463 8471; Zuffery et al- (1998) J. Virol- 72: 9873-9880; Follenzi et al. (2000) Nature Genetics 25: 217-222. Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition- Thus, there is a wide variety of appropriate formulations of pharmaceutical compositions available, as described below (see, for example, Remington's Pharmaceutical Sciences, 17 'ed., 1989). 30 Applications. Developed compositions and methods can be used for any application where it is desired to modulate the exp resection of one or more PDls genes. In particular, these methods and compositions can be used where modulation of a PD1 allele is desired, including but not limited to. therapeutic applications and research. The methods and compositions can be used to treat chronic infectious diseases like HIV / AIDS and HCV. In addition, the methods and compositions can be used to treat cancers such as melanoma, ovarian cancer, colorectal cancer / cdon, renal cell carcinoma, plasmacytoma / myeloma, breast cancer and lung cancer. Diseases and conditions in which PDl suppresses ZFP TFS or ZFNS can be used as therapeutic agents include, among others, diseases - 10 chronic infectious diseases and cancer. Diseases and conditions in which the activation of a PDl gene can be useful as a therapeutic treatment include "autoimmune diseases such as systemic lupus erythematosus (SLE) - Polynucleotides encoding ZFP TFS or ZFNs can be used as the therapies themselves, or can be incorporated in vectors for release.15 Methods and compositions comprising ZFP-TFS that suppress a PDl allele, and / or PDF-specific ZFNS can also be used in conjunction with other therapies designed to treat a chronic infectious disease or cancer . These ZFPs or ZFNs (or polynucleotides that encode these ZFPS or ZFNs) can be administered concomitantly (for example, in pharmaceutical compositions) or can be administered sequentially in any order. Any type of cancer can be treated, including, but not limited to, lung cancer, pancreatic cancer, liver cancer, bone cancer, breast cancer, colorectal cancer, ovarian cancer, leukemia, lymphoma, brain cancer and the like. Similarly, ZFP TFS designed to activate a PD1 allele can be used with other therapies designed to treat an autoimmune disease. Methods and compositions for treatment further include cellular compositions in which a mutant copy of the PD1 allele within cells isolated from a patient has been modified to a wild type PD1 allele using a PDl-specific ZFN. These ex vivo modified cells are then reintroduced into the patient. In addition, methods and compositions comprising modified stem cells are still imagined. For example, stem cell compositions in which a mutant copy of the PD1 den- allele. stem cell was modified to a wild-type PD1 allele using a PDl-specific ZFN. In other embodiments, 5 stem cell compositions are provided in which a wild type PD1 allele within the stem cells has been modified using PD1-specific ZFNS. These compositions can be used in conjunction with other therapies. These compositions can be administered concomitantly (for example, in the same pharmaceutical compositions) or can be administered alone. 10 in any order. The methods and compositions of the invention are still useful for designing and implementing in vitro and in vivo models, for example, animal models of chronic infection, cancer or autoimmunity, which allows the study of these disorders and other useful therapeutic discoveries. The following examples refer to exemplary embodiments of the present invention in which the nuclease comprises a ZFN. It will be appreciated that this is for the purpose of exemplification only and that other nucleases may be used, for example, homing endonucleases (meganuclases) with projected DNA binding domains and / or naturally occurring fusions of projected homing endonucleases ( meganuclases) in the DNA binding domains and heterologous cleavage domains. EXAMPLES Example 1: Identification of Persistently Specific Biologically Active ZFNs for PDl 25 ZFNS were assembled against the human PDl gene and were tested by ELISA and CELI assays as described in Miller et al. (2007) Nat. Biotechnol. 25: 778-785 and US patent publication 20050064474 and international patent publication WO2005 / 014791. Specific examples of ZFPs are described in Table 1. The first column in this Table is like an internal reference name (number) for a ZFP. Table 2 lists target binding sites in PD1. "F" refers to the finger and the number following "F" refers to which zinc finger (for example zk% h p / o, "Fl" refers to finger 1). Table 1: zinc finger protein directed to Hunan PDl SBS #: T * SÜ Fl; n F3 F4 F5 F ¢ s 4qDsLsv Í% D $ RKX R-DDLTR RsDHLm n (SEQ TD XO. l (SEQ ID (seq rn 7ü): CSEQ ID NO: N / A ) NO-3) 4) Sj RSDIJLTR rsdhi rr DRSALSR DRSAL ^ R (SEQ IDNO i (SEQ JD KD: (SEQ ID (SEQ IDNO N'A 4) Zj 9) xo: 8L_ RSDDLSK I RNDHRKy DRSALSR DRSALík (SEQ JD (SEQ ID '= (SEQ Ü) N'n ""' i 'S; J; I = I NO: J2) XO: 9} RSDHLSE QS, AqRKN) 7ÊÍ s ísL'QID i (SFQ ID ísFQ ID Nj'Á i N: "A i NO: Í4) NO: 13) NO: JS) i. r :: if) RSANLÍIK RSDHLSQ TSSXRKI DRSNLSR ISEQID (SEQ ID (SEQ ID (SEQ ID NO: lS) No: 19i ¥ 0: 20)) ('SEQ ID' = (SEQ ID (SEO ID!! -KCk22) i Nc) t18) MJ: ZO), Noàj); RhDHÚN TRPYLKR DRSALAR i,: "" ') =:; (SEQ JID (SFQ ID tsEQjD 1 * NA z i i ' E i bXj: 3) l NO: R) NO.25) NO: 9) i 'Qg ^ jr * H RSDSLSY "j HNDSRKN ranSllr RSDHLTQ i n23" (SEQ: n i (SÉÕ! D I (SEQ K) ÍSEQ ID (SEQ ID! Mià 't Nc "i): NCk2) NOS) RSDELTR i RNhNLR-T Noà6) No: 5" t DRSAÚR. P L "RPSTLHR l TRPVLKR 2.SD35 (SEQ ilj I t $ I ".Q7D (SEQ ID (SEQ IIJ (SEQ ID l N / À i l i1f T NCJ: 27)! NO: 29} NO: 25Í NÒ: 9'Í AND '' ^ "") =, '' TNWFILRT {SEQ ID ÍKPVLKR isEQID DkSALAR {SEQ ID NiÀ Noi9Í = fél'è NO: Z5) NO: 30} RS'JELTR l R1W.L1 "L TRPYLKR DRSALÀ, R (SEQ! D (SEQ JD ISEQ ID {SEQID N! '^ NO- ZB} XO: 3!) NC): 2S) No: 9j, l NO 2 ) Woa-R RSAQLAT TRWLW DRSALAR 2% 11 l RPSTLHR (& EQ Ib (SEQ m I.SÉQ ID (5EQ ID NO'A l (sü2 ED í NOz27b NC lB) No: 32í XQ: 2SI NC): 9) '"" ": = RSDELTR 1" SEQ ID RCTIILYL {S [Q II) TRPVLSR (SEQ ID DRSALÀR (SEQ ID Ne'À NO: 28) NO: 25) NO: 9) NiiÈ I) DRSALAR: RSDELTR TRPVLKR (SEQ ID (SEQ [D NO: 28) NO: 25) ': o! 92jdj !: RSDELTR TRPVLKR DRSALAR I; "°" l =! NO: 27) (SEQ ID NO: 28) (SEQID NO: 2S) = "" ^ l 25015! RPSTLHR RSDELTR TRPVLKR (SEQ LD (SEQ ID | (SEQ TD NO: 27) i 25016 i RKFARPS NO: 28) RNFSRSD HPHHRMC I TRPVLKR NO: 25), 'si :, l "' ^ (SEQ ID (SEQ ID i (SEQ ID l (SEQ ID NO: 36) NO: 37) _ NO: 38) NO: 25) D ( SI] N., A RSDELTR RMGRLST I TRPVLKR ÊJS! (SEQ ID (SEQ ID NO: 28) RSDELTR gjg;) NO: 25) TRPVLMR, "o, g; i" '^ (SEQ ID (SEQ JD l (SEQ ID NO: 28) = C Y NO: 41) =: I "" "^ DRSALAR l RSDELTR TRPVLKR: "°"! = NO: 27) (SEQ ID NO: 28) (SEQ ID NO: 42) 1 (SEQ ID NO: 25)): "0" i = RSDELTR (SEQ ID = "1 TRPVLKR (SEQID NO: 28) NO: 25)) 'j) RSDELTR TRFNLKR} (SEQ ID (SEQID NO: 28) NO: 25) RSDELTR TRPVLKR (SEQ ID (SEQ ID NO: 28) NO: Z5) RSDELTR (TRPVLKR SEQ ID NO: (SEQID 28) NO: 25) RSDELTR RPMHL'I'N I TRPVLKR (SEQ ID (SEQ ID NO: 28 / NO: 49) NO: 25) | 25031 I RNAALTR RSDELTR RSPHLYH ' TRPVLKR (SEQID (SEQ LD (SEQ ID j (SEQ ID NO: 45) NO: 28) NO: 50) NO: 25G | "°" i = "O: 4S) RSDELTR (SEQ ID NO: 28) RCEALHH (SEQ ID NO: 5J) TRPVLKR (SE.Q ID NO: 25) DRSAQAR I = I "'^ DRSALAR j 25034 i RjNAALTR RSDELTR RCEALHH' TRPVLKR (SEQ ID (SEQ ID (SEQ ID (SEQ ID NO: 45) NO: 28) NO: Sl) _ NOQS) ':' O! 9:] "" '^ TRPVÜR DRSALAR! RSDEL.TR RSPHLYH '25036 RNAALTR (SEQ ID (SEQ ID (SEQ ID (SEQ ID NO: 45) NO: 28) NO: 50) NO: 25) D' "ÍRj" "" ^ RSDELTR RLPALLS TRPVLKR '25040 RNAAI.TR (SEQ ID (SEQ ID (SEQ ID (SEQ ID (SEQ ID (:' oS) "I" "'^ NO: 45 NO: 28) NO-.53) NO: 25) RSDELTR DRSALAR i HNAALTR RTYNRTQ ' TRPVLKR 2504! (SEQ ID (SEQ ID (SEQ ID (SEQ ID (SEQ ID I N / A NO: 54) NO: 28) NO: 55) NO: 25) NO: 9) Table 2: ZFN target sites in the human PDl gene sBsse Target site 12942 ccAGGGCGCCTGTGCGAtctgc.atgcct (SE.Q ID NO: 5Ú) 12946 caGTCGTCIUGGCGGTGctacaactggg (SEQ ID NO: 57) 12947 caGTCGGgggggggggggggggggggggggggggg ID NO: 58) 12971 ctGTGGACTATGCGGAGCrGgamF ¢ a (SEQ ID NO: 59) 12972 cTt7T "GGACTATc3C £ GI'LC7CI '% a = ca (SEQ ID NO: 59) 18759 caGTCGTCTGGGCGGTGct (SF-Q ID h'23) gcAGGGCGCCrGTGGGAÊclgcatgcct (SEQ ID NO: 56) 25005 caGTCGTCTGGGCGGTGct (SFQ ID NO: 60) 25006 caGTCGTCTGGGCGGTGct (SEQ ID NO: 60) 25010! caGTCGTCTCíGGCGGTGct (seq id no: 60) 15011 caGTCGTCTCGGCGGTGet {: Seq id no: 60) 250P caGTCGTCTGGGCGGTGct (SE.Q ID NO: 60 ') F "" "25013" caGTCGTCTGGGQ-ID 2 GTGct (60) caGTCGTCTGGGCGGTGct (SEQ ID NO: 60) "r" 25015 caGTCGTCTGGGCGGTGct (SEQ ID NO: 60) 250! 6 caGTCGTCTGGGCGG1 "Gct (SEQ ID> '0:60)! 25017 caGTCGTCTGGGCGTTG' (3gTcI'cggcggTgã & Q id ¥ õ: ãj "| I 25022! 25023 cmj'T (tgTcTà3gcggTgct (SEQ JD NO: 60)! 1! '25025 cagTcgrcTgggccgTgct (SEO ID NO: KO)]] I 2'027 caGTCGTCTGGGCGGTGct (SFQ ID NO: 60) 25028 caCírcgTCTGGGCGGTG ¢ l (SEQ ID NO: 60) 25029 ¢ ac "7" rc: gTc ~: T'cKgcgg'rgct (SEQ ID NO: 60) 250TCCGGT'G .GGTGct (SEQ ID NO: 60) 2503i CaGTCGTCTG · GGCGGTGct (SEQ ID NO: 60)! ís6ii caGTC-G'T "« '^ G'GCGGTGct (SEQ ID NO: 60) 25034 caGTCC3TCTGGGCGGTGct (SEQ ID NO: 60) 25036 caGTCGTCTGGGCGGT'Cct (SEQ ID NO: 60) 25040 caGTCGTCTGGGCCGTGGCGTGG "041 caGTCGTCTGGGCGGTGct (SEQ ID b'O: 6th) | Initial in vitro activity tests were performed on samples nucleofected cell strips as described (above. Briefly, plasmids encoding ZFP-FOKI fusions were introduced into K562 cells by transfection using the Amaxa '"Nucleofection kit as specified by the manufacturer. For transfection, two million K562 cells were mixed 5 con7 varying amounts of each zinc finger nuclease expression plasmid and 100 L Amaxa '"Solution V. The cells were transfected in an Amaxa Nucleofector jjTM using T-16 program. Lmmediately after transfection, the cells were divided into two flasks different and grown in RPMI medium (lnvitrogen) supplemented with 10 ° / o FBS in 5 ° / 0 CO2 - 10 to 30 ° C or 37 ° C for four days. From this initial in vitro screening, two leading ZFN pairs were identified and submitted for elaboration to try to improve their efficiency These pairs direct to exons 1 and 5 of the PDl gene, respectively. The elaborated (improved) proteins were tested in a time experiment. o-course, essentially as described above. The results are summarized in Table 3 below. Table 3: PDl NHEJ ° /) NHEJ Target l Pair ZFN I Day 3 Day 7 Day 9 exon 1 i 12942/12946: 8 7 5 exon 1 _ i 12942/12947 i 10 6 6 exon 5 12934/12971 11 6 1- 5 t exon5 12934/12972 _1_ 7.5 2) As shown in Table 3, treatment of cells with ZFNS against exon 5 causes the loss of a greater proportion of cells edited 20 in the population genome, while the signal of genome editing in cells treated with ZFNs designed against exon 1 is much more stable. To determine ZFN activity at the PDl site, Cel-l-based Surveyor "" Nuclease assays were performed essentially as per the manufacturer's instructions (Trangenomic SURVEYOR'm), cells were collected and chromosomal DNA prepared using a Quickextract kit "" according to the manufacturer's instructions (Epicentre®). The appropriate region of the PD1 site was PCR amplified using Accuprime "High-fidelity DNA polymerase (lnvitrogen). PCR reactions were heated to 94 ° C, 0 and gradually cooled to room temperature - Approximately 20Ong of ringed DNA was mixed with 0.33µL of Cel-l enzyme and incubated for 20 minutes at 42 ° C. Reaction products were analyzed by electrophoresis on polyacrylamide gel in lX Tris-borate-EDTA buffer. The constructs were also tested on primary PBMC samples that were kept at 30 ° C or 37 ° C for 3 or 10 days (see Table 4). Briefly, PBMC were obtained from AllCells and were grown - 10 in RPMI + 10 ° / o FBS + 1 ° /, L-Glutamine (30 mg / mL) + IL-2 (1ng / mL, Sigma) and activated with anti-CD3 / CD28 beads according to the protocol of manufacturer (Dynal). The cells were seeded in 3E5 cell / mL in 1mL of volume in a 24-well plate. Adenoviral vectors were constructed containing the ZFN 15 pairs of interest as described (see, US patent publication 20080159996) and were added two days later in a MOl of 10, 30, or 100 (MOl calculated on the basis of an infectious title). The cells were collected 3 or 10 days after exposure to the virus and gene modification efficiency was determined using a 20 SURVEYOR '"Cel-l-based Nuclease assay, performed as described in the international patent publication WO 07 / 014275. See also Oleykowski et al. (1998) Nucleic Acids Res. 26: 4597-4602; Qui et al. (2004) BioTechniques 36: 702-707; Yeung et al. (2005) 81oTechniques 38: 749- 758. For the ZFN pairs shown in Table 4, each ZFN was tested 25 in combination with ZFN 12942. Activity is measured by percent NHEJ activity as measured by the Cel-l based SURVEYORtm Nuclease assay described above. Additional pairs of PDF-specific ZFNs were further tested for activity on primary PBMCs as described above, and the results are shown in Table 4. In the data shown in Table 4, the specific number for PDl 12942 was always paired with the second ZFN listed in Table 4 to form an active pair (that is, ZFN 12942 has been paired each of ZFN 12947 to 25041). See also Figure 1 (samples as shown in Table 4). Table 4: Activity of PDl ZFNS 37 ° C 37 ° C 30 ° C 30 ° C 12942+ Day 3 Day 10 Day 3 Day 10 Sample na (Percent (Percent (Percent (Percent Figure 1 NHEJ) NHEJ) NHEJ) NHEJ) 12947 2.1 1.6 4.2 2.3 1 18 759 4.7 2.2 4.9 4.3 2 25005 4.4 2.3 4.5 2.4 3 25006 2.9 8.1 5, 2 9.9 4 25010 4.9 1.8 5.0 2.8 5 25011 3.1 9.2 3.1 12.8 6 25012 5.9 8.5 8.2 14.7 7 25013 3, 7 0.6 4.0 1.8 8 25014 10.7 6.6 8.3 9.6 9 25015 3.9 3.9 5.3 7.3 10 25016 7.3 12.8 7.7 13 , 6 11 25017 7.7 9.6 6.1 15.0 - 12 25022 3.6 5.2 4.2 9.0 13 25023 3.1 8.3 7.1 7.8 7.8 14 25025 8.8 10.6 6.5 7.6 - 15 25027 6.0 9.5 5.9 6.6 16 25028 4.3 5.2 4.8 6.2 - 17 25029 8.1 12.8 7.6 14.3 18 25030 7.6 9.6 5.4 10.7 19 25031 9.4 14.5 3.8 15.3 20 —— - 25032 6.7 4.2 6.6 6.0 6.0 25034 4.9 4.7 6.2 4.1 22 25036 8.3 4.2 6.9 9.7 23 25040 6.1 3.6 3.6 4.6 24 25041 7.9 11.2 5, 2 4,5 25 To assess the local effects of NHEJ activity directed at ZFN at the molecular level, CD8 + cells were treated with the specific ZFN pair exon 12942 and 12947- Briefly, CD8 + cells were purchased from AllCells and were grown in RPMI + 10 ° / o FBS + 1 ° / j L-Glutamine (30 mg / mL) "IL-2 (30 µg / mL, Sigma) and left to stand for 4-24 hours. The pIasmids were constructed containing the ZFN pairs of in-. teresse as described above and 1e6 cells / nucleofection were used with the Amaxa "Nucleofection kit as specified by the manufacturer. Cells 5 were activated 12-24 hours after nucleofection with anti-Cl) 3lCO28 beads according to the manufacturer's protocol (Dynal). Cells were collected 3 or 10 days after nucleofection and gene modification efficiency was determined using a Cel-l-based SURVYORtm Nuclease assay, performed as described in the publication - 10 of international patent WO 07/014275. See also Oleykowski et al. (1998) Nuc / eic Acids res. 26: 4597-4602; Qui et al. (2004) BioTechniques 36: 702-707; Yeung et al. (2005) Bio7 "echniques 38: 749-758. PCR products were cloned and transfected in E. co / i. Antibiotic resistant subclones were grown, the plasmids isolated 15 and then subjected to sequence analysis to observe any sequence changes that occurred as a result of NHEJ (see, Figure 2). As can be seen from the figure, a variety of insertions and deletions were observed in the vicinity of the ZFN cleavage site. These ZFNs were further tested in the yeast system as described in US publication 20090111119. Example 2: Ex vivo activity of PD1-specific ZFNs in mice gos- To be in tune with the concept of deleting PDl in vivo, mouse specific PDL ZFNS were prepared as described above and then tested ex vivo. The sequence characteristics of the zinc finger domains, as well as their binding specificities are shown below in Tables 5 and 6. Table 5: specific zinc finger designs for murine PD1 ! SBS # Fl F2 F3 F4 F5 F6 D6íÇbT; Ci RSAhLTR ÍsciSíSí QSGDLTR QSSDLRR 14534 (SEQ ro (SEQ ID (SEQID (SEQ ID (SEQID N / A NO22) NO: J7) NO: 61) no: 62L NO: 63) 1453 + DDWNISQ RSANLTR TSGSLSR QSGDLTR QSSDLRR Fokl (SEQ ID (SEQID (SEQID (SEQ ID (SEQ ID (SEQ LD KK NO22) NO: l7) NO: 61) YO: 62) NO: 63) N'A) QSSHLTR RSDNLRE DRSNLSR TSSNRKTR 14536 (SEQ ID (SEQ ID (SEQ ID (SEQ ID (SEQID! Nô: 64) NQ: 65) NO: 20) NO: l9) NO: 66) NO:)! = QSSHLTR RSDNLRE DRSKLSR RSDSLSK QSAKRTT i TSSNRKT Fòkl (SEQ ID (SEQID {SEQ ID (SEQ ID (SEQ ID (SEQ ID 'EL NQS4) NO: 65) KO: 20) NO: 19l NO: 66) NO:) QSGDLTR RSDNLSE ERANRNS DRSDLSR QSSDLRR I 14 $ 45 (SEQ LD (SEQ ID (SEQ ID ('SEQ ID (SEQ ID N / A I I no: 62L_ "0:67) NO: 68) NO: 69) No: 63j l! 4545- QSGDLTR RSDNLSE ERANRK'S DRSDLSR QSSDLRR 'Fokf (SEQID (SEQ ID (SEQ ID (SEQ ID (SEQ ID X.'A i KK NO: 62) NO: 67) NO: 68) NO: 69) NO: 63) DRSHIAR RSDDLSR QSANRTK RSDTLSE AXSNRIK 14546 (SEQID GEQ ID (SEQID (SEQ ID (SEQ ID N / A NO: 70) NO: 7l) NO: 72) Noi73) NO: 74) DRSHLAR RSDD'-SR QSANRTK RsDns' ANSNI 14546- (SEQ LD {SEQ ID (, SEQ ID Ifohel GEQID (SEQID N / A NO: 70) No: 7i) NQ: 72) NO: 73) NO: 74) Table 6: Connection specifics for finger designs of zinc-specific for SBS PDl # Target site 14534 gtGCTGCAGTTGAGCrGg = Kagggt (SEQ ID NO: 75) 14534- FOKIKK gúGCTGCAGTTGAGCTGgcaamggg (SEQ ID NO: 75) 14536 ccCAAGTGAATGACCAGGAGGGCCAGG ID NO: 76) 14545 caGCTGCCCAACAGGCAtgacttccaca (SEQ ID NO: 77) 14545- Fokl KK mGCTGCCCAACAGGCAtgacWcaca (SEQ ID NO: 77) 14346 atGATCTGGAAGCGGCCa = tggacgg (SEQ ID NO: 78) AGG (GC) 78) On day 1, splenocytes collected from transgenic Pmel TCR / Rag1 - / - mice were processed in a single cell suspension and resuspended in complete medium (RPMI-1640, 10% Fetal bovine serum, O, 1mM non-amino acids o essentials, 1mM sodium pyruvate, 2mM L-glutamine, 50 µg / ml gentamicin sulfate, 50 µM 2-mercaptoethanol) with 25 µ1 lnvitrogen Mouse T-Activator CD3 / CD28 Dynabeads / 10 'cells. The cells were plated in 24-well plates at 2 X 106 cells / ml (2ml / well). On day 3, the cells were collected, separated from beads using a magnet and counted. Splenocyte transfection was performed using the protocol provided with the Amaxa '"Nucleofection kit. Briefly, 1 x 10 and 7 viable cells were resuspended in 100 µl of Amaxa Nucleofector solution and transfected with 4 µg of plasmid containing ZFN 14546 and 14545 pair, or 4 µg of mut PDl Fokl plasmid containing ZFN 14546-Fok! EL and 14545-Fok I KK, 2.5 µg of Amaxa pmax GFP vector, or a control without DNA, using Amaxa Nucleofector (X-Ol program). The cells were grown in 2 ml of fully complemented medium 30 ° Amaxa Mouse T Cell Nucleofector - 10 C after transfection. The next day, one ml of Amaxa Mouse T celi Nucleofector medium was removed and replaced with one ml of complete rhein supplemented with 10 U / ml IL-2. Two days later the cells were collected and counted. Two million cells from each group were plated in wells coated with anti-C03 in a 24-well plate. On the next day, the cells were collected and stained with anti-POl PE: anti-CD3 PE-Cy7, and anti-CO8 APC. The cells were analyzed on a BD FACSCalibur. Plots show ° / 0 of PDl positive CD3 + CD8 + cells in each group and median PDl fluorescence for each group (see, Figure 3). The data show that the expression of PDl is reduced in cells that received the 20 PDF-specific ZFNs, even in the presence of anti-CD3 stimulation. To verify that the reduction in PDl expression was evident at later time points, the cells were still collected at 72 hours after anti-CD3 stimulation, instead of 24 hours as described above. The cells were collected and stained with anti-PD1 PE: anti-CD3 PE-25 Cy7, and anti-CD8 APC. The cells were analyzed on a BD FACSCalibur. The data are shown in Figure 4. The upper histograms show ° / 0 of PDl-positive CD3 + CD8 + cells in each group and the median PDl fluorescence for each group. The lower plots show the frequency of cells that express PDI / CFSE. Importantly, mut 30 PDl Fokl and wt PDl Fokl show a higher frequency of PD1 "'g CFSE'i" (dividing cells) than control groups, and demonstrate that PDl expression is still reduced in cells treated with the specific ZFNS for for PDl still 72 hours after anti-CD3 stimulation. Example 3: Activity of human PDl-specific ZFNS in TILS and human PDL-specific ZFNS were tested on human tumor infiltrating lymphocytes (TILS) in the presence of tumors and evaluated 5 as described above and using methods known in the art (see, for example, Yoshino et al., (1992) Cancer Research 52: 775-781). PD1-specific ZFNs were activated using anti-CD3 antibodies as described above, so the cells were transduced with Ad5 / F35 adenovirus expressing PDl-specific ZFNS. The cells were expanded with - 10 IL2 and then restimulated with anti-CD3 antibodies or with tumors and evaluated 24 hours after stimulation. The results are shown in Table "7 'below. Table 7: expression and viability of PDl in TILS | CD3 stimulation | Tumor extracts! I gfp 12942 EL / 12942 / GFP 12942 Elj 12942/12947 KK 12947 I 12947 KK 12947 I Expression of PDl I 32.2% 31.5% 14.1% 22.9% 13.8% 7.5% IP / o feasibility TIL I 30.3 ° / o 34.2% 45.6% 18% 32% 47.9% I ° / 0 viability N / AN / AN / A cell 45.7 ° / o 33.1% 19.6% tumor ° / 0 PDl + cells divided 1.6% 0.9% 0.3% I 1.1 ° /, I 0.3% 0. 1 ° / oPD1 - cells divided- 3.9% 3 0 ° 6 2 1 ° / 0 I 3 O ° / jl 1 4 ° / 0 0.8% of The data in Table 7 show that when cells are stimulated by anti-CD3 antibodies, reduced PDl expression, through the action of PDl-specific ZFNs, increases the reduced cell viability. When transduced cells are treated with tumors, the same phenomenon is observed - mediated ZFN reduces in PD1 and leads to an increase in TIL viability. Still the data show that a reduction in the expression of 20 PDl in the transduced TILs leads to a reduction in tumor cell viability. Example 4: Purification of PDl edited on primary human T cells The PD1-specific ZFN pairs that have been further elaborated. were tested on CD4 + T cells as described above in Example 1. As seen in Figure 5, up to 44% editing was observed with some 5 pairs. In this experiment, 'Positive Control' indicates cutting using the pair 25025/12942 ZFN in CD8 + T cells previously performed under different experimental conditions. A leading pair, 25029/12942 was chosen to further use for the isolation of cells modified by PD1. Briefly, in these experiments, CD4 + T cells were treated with mRNA encoding PD1-specific ZFNS, cultured under "30 degree" conditions and then stimulated with a first exposure to anti-CD3 / CD8 beads ( Dynal) as described above in Example 1, which stimulates strong expression of ZFN transgenes and promotes cleavage at the PD1 site (see, US patent publication 15 20080311095) .After the first stimulation, the cells were re-stimulated and subjected to a purification procedure, by FACs or by affinity chromatography. Briefly, CD4 + T cells were treated with CCR5-specific ZFNS (see, US Patent Publication 20080159996) or 20 PDl-specific ZFNS. The cells were collected and analyzed for editing PDl by the Cel-l assay (described above), i) after the first stimulation, ii) after the second stimulation but before any purification, iii) after cell classification for CD25 + (an activation marker), PD1 (- ) using standard methodology, or iv) after affinity chromatography using a matrix made with anti-PD1 antibody or anti-C025 antibody. As shown in Figure 6, using the cell classification technique (where cells were isolated that were positive for CD25, but negative for PD1), up to 56% of the recovered cells demonstrated to be modified as assessed by the Cel-1 assay. PD1 (-) cells purified by the affinity chromatography technique (where the cells were subjected to affinity matrices using anti-PD1 antibodies, anti-CD25 antibodies, or both) presented in general PCl modification up to 42% as assessed by Cel-l analysis. The cells that were purified by cell classification were still analyzed according to their PDl site by sequencing, and the results are shown below in Table 8. As can be seen 5. From the Table, the target percentage change ('° / 0 NHEJ') predicted by the Cel-l analysis is similar to that found by the sequencing analysis. In this Table, the label 'Sample' corresponds to those shown in Figure 6. Table 8: Percentage of PDl modification in CD25 + I cells Sample I ° / 0 NHEJ i ° /) NHEJ por! Number I Insights! Deletions I by Cel I i sequencing! modified * I 4! 43) _ 62] 54de87) 3de54 j51de54 L_ 8 l 56 I 81, i 65de80 i 4de65 j61de65 13 I 42 59 43de73 1de43 j42de43 *: 'Modified number' indicates the number of sequences in the sequence group that was observed at be modified. For example, in sample 4, 54 sequences out of 87 analyzed were modified. PDF-specific ZFNS were further tested on CD8 + T cells. In this experiment, mRNAs encoding PDF-specific ZFNS were produced using the Ribomax Large Scale RNA Production-15 tionT7 kit (Promega), followed by RNeasy mini kit (Qiagen), both according to the manufacturer's protocols. Variant amounts of mRNAs were used to transduce the cells using the Amaxa Nucleofection delivery system as described above, and the PD1 percentage modification was analyzed by the Cel1 assay. 20 As shown in Figure 7, the amount of modification observed, as described as '° /) NHEJ', is related to the amount of mR-NA used, with lower amounts of mRNA input resulting in lower percentages of target modification. These results demonstrate that the 25 PDl-specific ZFNS described here are able to modify the PDl site in cell lines and in primary T cells. All patents, patent applications and publications mentioned those here are hereby incorporated into their totalities. Although the invention was provided in some detail by way of illustration and example for the purpose of clarifying understanding, it will be apparent to those skilled in the art that various changes and modifications can be practiced without departing from the spirit or scope of the invention. Thus, the previous descriptions and examples should not be interpreted as Limiting.
权利要求:
Claims (16) [1] 1. Zinc finger protein, characterized by the fact that it binds to a target site in a PD1 gene, in which the target site is selected from the group consisting of SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID 5 NO: 59 and SEQ ID NO: 60. [2] 2. Fusion protein, characterized by the fact that it comprises a zinc finger protein, as defined in claim 1, and a functional domain. [3] 3. Fusion protein according to claim 2, characterized by the fact that the functional domain comprises a transcriptional regulatory domain. [4] 4. Fusion protein according to claim 3, characterized by the fact that the transcriptional regulatory domain is an activation domain or a repression domain. [5] 5. Fusion protein according to claim 2, characterized by the fact that the functional domain comprises a cleavage domain or half a cleavage domain. [6] 6. Polynucleotide, characterized in that it comprises a zinc finger protein, as defined in claim 1, or a fusion protein, as defined in any of claims 2 to 5. [7] 7. Method for modulating expression of a PD1 gene in a cell, characterized by the fact that it comprises: introducing at least one polynucleotide into the cell, as defined in claim 6, under conditions such that the expression of the PD1 gene is modulated . [8] 8. Method according to claim 7, characterized in that the modulation comprises activation, repression or inactivation by cleavage of the PD1 gene. [9] 9. Method according to claim 8, characterized by the fact that inactivation occurs via union of the non-homologous end (NHEJ) following the cleavage of the PD1 gene. [10] 10. Method according to claim 8, characterized by the fact that the modulation comprises inactivation by cleavage and also in that the method comprises introducing an exogenous sequence flanked by sequences having homology to the PD1 gene in the cell, in which the exogenous sequence it is integrated into the PD1 gene by homologous recombination. 5 [11] Method according to any one of claims 8 to 10, characterized in that the cell is a primary cell or a stem cell. [12] 12. Method according to claim 14, characterized by the fact that the primary cell is a peripheral blood mononuclear cell (PBMC), a CD4 + T cell or a CD8 + T cell or the stem cell is a -embryonic stem, an induced pluripotent stem cell, a hematopoietic stem cell, a neuronal stem cell or a mesenchymal stem cell. [13] 13. Use of a polynucleotide, as defined in claim 6, characterized in that it is for the preparation of a drug to treat an individual having a disease or disorder with the characteristic of aberrant expression or regulation of a PD1 gene, in which the expression of the PD1 gene is modulated according to the method as defined in any one of claims 8 to 12, thereby treating the disease or disorder. [14] 14. Use according to claim 13, characterized in that the disease or disorder is a cancer, a viral infection or an autoimmune disease. [15] 15. Use according to claim 13 or 14, characterized by the fact that it also comprises administering additional antiviral or anti-cancer therapies to the individual. [16] 16. Invention, characterized by the fact that in any form of its embodiments or in any applicable category of claim, for example, product or process or use encompassed by the matter initially described, revealed or illustrated in the patent application.
类似技术:
公开号 | 公开日 | 专利标题 US10500229B2|2019-12-10|Targeted disruption of the MHC cell receptor US10046028B2|2018-08-14|Methods and compositions for modulating PD1 US10858416B2|2020-12-08|Methods and compositions for modification of a HLA locus US11235026B2|2022-02-01|Methods and compositions for modulating PD1
同族专利:
公开号 | 公开日 ES2658691T3|2018-03-12| CA2817355C|2019-10-15| AU2011329759B2|2016-06-02| WO2012068380A2|2012-05-24| EP3034084A1|2016-06-22| EP3311822A1|2018-04-25| JP5976661B2|2016-08-24| WO2012068380A3|2012-07-26| EP2640399A4|2014-08-06| CA2817355A1|2012-05-24| US20110136895A1|2011-06-09| CN103282029B|2017-03-22| AU2011329759A1|2013-05-30| EP3311822B1|2020-05-13| EP2640399A2|2013-09-25| IL226196A|2018-01-31| AU2016204975B2|2018-08-09| ES2801749T3|2021-01-13| KR101921548B1|2018-11-26| US8563314B2|2013-10-22| US20140030240A1|2014-01-30| JP2014501506A|2014-01-23| US20160220634A1|2016-08-04| IL226196D0|2013-07-31| PT2640399T|2018-02-19| EP3034084B1|2018-08-01| DK2640399T3|2018-03-05| NO2640399T3|2018-05-19| US10046028B2|2018-08-14| KR20130140054A|2013-12-23| US9402879B2|2016-08-02| AU2016204975A1|2016-08-04| EP2640399B1|2017-12-20| CN103282029A|2013-09-04|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US789538A|1904-11-11|1905-05-09|Colin E Ham|Dumb-bell.| US4217344A|1976-06-23|1980-08-12|L'oreal|Compositions containing aqueous dispersions of lipid spheres| US4235871A|1978-02-24|1980-11-25|Papahadjopoulos Demetrios P|Method of encapsulating biologically active materials in lipid vesicles| US4186183A|1978-03-29|1980-01-29|The United States Of America As Represented By The Secretary Of The Army|Liposome carriers in chemotherapy of leishmaniasis| US4261975A|1979-09-19|1981-04-14|Merck & Co., Inc.|Viral liposome particle| US4485054A|1982-10-04|1984-11-27|Lipoderm Pharmaceuticals Limited|Method of encapsulating biologically active materials in multilamellar lipid vesicles | US4501728A|1983-01-06|1985-02-26|Technology Unlimited, Inc.|Masking of liposomes from RES recognition| US4946787A|1985-01-07|1990-08-07|Syntex Inc.|N--dialkyloxy)- and N--dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor| US4897355A|1985-01-07|1990-01-30|Syntex Inc.|N[ω,-dialkyloxy]- and N-[ω,-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor| US5049386A|1985-01-07|1991-09-17|Syntex Inc.|N-ω,-dialkyloxy)- and N--dialkenyloxy)Alk-1-YL-N,N,N-tetrasubstituted ammonium lipids and uses therefor| US4797368A|1985-03-15|1989-01-10|The United States Of America As Represented By The Department Of Health And Human Services|Adeno-associated virus as eukaryotic expression vector| US4774085A|1985-07-09|1988-09-27|501 Board of Regents, Univ. of Texas|Pharmaceutical administration systems containing a mixture of immunomodulators| US5422251A|1986-11-26|1995-06-06|Princeton University|Triple-stranded nucleic acids| US4837028A|1986-12-24|1989-06-06|Liposome Technology, Inc.|Liposomes with enhanced circulation time| US5176996A|1988-12-20|1993-01-05|Baylor College Of Medicine|Method for making synthetic oligonucleotides which bind specifically to target sites on duplex DNA molecules, by forming a colinear triplex, the synthetic oligonucleotides and methods of use| US5264618A|1990-04-19|1993-11-23|Vical, Inc.|Cationic lipids for intracellular delivery of biologically active molecules| AU7979491A|1990-05-03|1991-11-27|Vical, Inc.|Intracellular delivery of biologically active substances by means of self-assembling lipid complexes| US5173414A|1990-10-30|1992-12-22|Applied Immune Sciences, Inc.|Production of recombinant adeno-associated virus vectors| US5420032A|1991-12-23|1995-05-30|Universitge Laval|Homing endonuclease which originates from chlamydomonas eugametos and recognizes and cleaves a 15, 17 or 19 degenerate double stranded nucleotide sequence| US5436150A|1992-04-03|1995-07-25|The Johns Hopkins University|Functional domains in flavobacterium okeanokoities restriction endonuclease| US5356802A|1992-04-03|1994-10-18|The Johns Hopkins University|Functional domains in flavobacterium okeanokoites restriction endonuclease| US5487994A|1992-04-03|1996-01-30|The Johns Hopkins University|Insertion and deletion mutants of FokI restriction endonuclease| US5792632A|1992-05-05|1998-08-11|Institut Pasteur|Nucleotide sequence encoding the enzyme I-SceI and the uses thereof| US5587308A|1992-06-02|1996-12-24|The United States Of America As Represented By The Department Of Health & Human Services|Modified adeno-associated virus vector capable of expression from a novel promoter| JP4012243B2|1994-01-18|2007-11-21|ザスクリップスリサーチインスティチュート|Zinc finger protein derivatives and methods therefor| US6140466A|1994-01-18|2000-10-31|The Scripps Research Institute|Zinc finger protein derivatives and methods therefor| US6242568B1|1994-01-18|2001-06-05|The Scripps Research Institute|Zinc finger protein derivatives and methods therefor| US5877302A|1994-03-23|1999-03-02|Case Western Reserve University|Compacted nucleic acids and their delivery to cells| US5585245A|1994-04-22|1996-12-17|California Institute Of Technology|Ubiquitin-based split protein sensor| AU698152B2|1994-08-20|1998-10-22|Gendaq Limited|Improvements in or relating to binding proteins for recognition of DNA| US5789538A|1995-02-03|1998-08-04|Massachusetts Institute Of Technology|Zinc finger proteins with high affinity new DNA binding specificities| US5928638A|1996-06-17|1999-07-27|Systemix, Inc.|Methods for gene transfer| US5925523A|1996-08-23|1999-07-20|President & Fellows Of Harvard College|Intraction trap assay, reagents and uses thereof| GB9703369D0|1997-02-18|1997-04-09|Lindqvist Bjorn H|Process| GB2338237B|1997-02-18|2001-02-28|Actinova Ltd|In vitro peptide or protein expression library| US6342345B1|1997-04-02|2002-01-29|The Board Of Trustees Of The Leland Stanford Junior University|Detection of molecular interactions by reporter subunit complementation| GB9710807D0|1997-05-23|1997-07-23|Medical Res Council|Nucleic acid binding proteins| GB9710809D0|1997-05-23|1997-07-23|Medical Res Council|Nucleic acid binding proteins| US6479653B1|1997-08-26|2002-11-12|Ariad Gene Therapeutics, Inc.|Compositions and method for regulation of transcription| US6410248B1|1998-01-30|2002-06-25|Massachusetts Institute Of Technology|General strategy for selecting high-affinity zinc finger proteins for diverse DNA target sites| WO1999045132A1|1998-03-02|1999-09-10|Massachusetts Institute Of Technology|Poly zinc finger proteins with improved linkers| US6140815A|1998-06-17|2000-10-31|Dover Instrument Corporation|High stability spin stand platform| US6140081A|1998-10-16|2000-10-31|The Scripps Research Institute|Zinc finger binding domains for GNN| GB9824544D0|1998-11-09|1999-01-06|Medical Res Council|Screening system| US6534261B1|1999-01-12|2003-03-18|Sangamo Biosciences, Inc.|Regulation of endogenous gene expression in cells using zinc finger proteins| US7013219B2|1999-01-12|2006-03-14|Sangamo Biosciences, Inc.|Regulation of endogenous gene expression in cells using zinc finger proteins| US7070934B2|1999-01-12|2006-07-04|Sangamo Biosciences, Inc.|Ligand-controlled regulation of endogenous gene expression| US6453242B1|1999-01-12|2002-09-17|Sangamo Biosciences, Inc.|Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites| US7030215B2|1999-03-24|2006-04-18|Sangamo Biosciences, Inc.|Position dependent recognition of GNN nucleotide triplets by zinc fingers| PL354286A1|1999-08-23|2003-12-29|Dana-Farber Cancer Institutedana-Farber Cancer Institute|Pd-1, a receptor for b7-4, and uses therefor| AT353365T|1999-08-23|2007-02-15|Dana Farber Cancer Inst Inc|NEW B7-4 MOLECULES AND THEIR USES| US6599692B1|1999-09-14|2003-07-29|Sangamo Bioscience, Inc.|Functional genomics using zinc finger proteins| AT309536T|1999-12-06|2005-11-15|Sangamo Biosciences Inc|METHODS OF USING RANDOMIZED ZINCFINGER PROTEIN LIBRARIES FOR IDENTIFYING GENERAL FUNCTIONS| WO2001059450A2|2000-02-08|2001-08-16|Sangamo Biosciences, Inc.|Cells expressing zinc finger protein for drug discovery| US20020061512A1|2000-02-18|2002-05-23|Kim Jin-Soo|Zinc finger domains and methods of identifying same| AT353361T|2000-04-28|2007-02-15|Sangamo Biosciences Inc|TARGETED MODIFICATION OF THE CHROMATIN STRUCTURE| AU5587001A|2000-04-28|2001-11-12|Sangamo Biosciences Inc|Databases of regulatory sequences; methods of making and using same| US20030044787A1|2000-05-16|2003-03-06|Joung J. Keith|Methods and compositions for interaction trap assays| JP2002060786A|2000-08-23|2002-02-26|Kao Corp|Germicidal stainproofing agent for hard surface| US6919204B2|2000-09-29|2005-07-19|Sangamo Biosciences, Inc.|Modulation of gene expression using localization domains| US6946292B2|2000-10-06|2005-09-20|Kyowa Hakko Kogyo Co., Ltd.|Cells producing antibody compositions with increased antibody dependent cytotoxic activity| US6794136B1|2000-11-20|2004-09-21|Sangamo Biosciences, Inc.|Iterative optimization in the design of binding proteins| AU1792902A|2000-11-28|2002-06-11|Sangamo Biosciences Inc|Modulation of gene expression using insulator binding proteins| US7067317B2|2000-12-07|2006-06-27|Sangamo Biosciences, Inc.|Regulation of angiogenesis with zinc finger proteins| US7262054B2|2002-01-22|2007-08-28|Sangamo Biosciences, Inc.|Zinc finger proteins for DNA binding and gene regulation in plants| GB0108491D0|2001-04-04|2001-05-23|Gendaq Ltd|Engineering zinc fingers| AT513040T|2001-08-17|2011-07-15|Toolgen Inc|ZINC FINGER DOMAIN LIBRARIES| EP1421177A4|2001-08-20|2006-06-07|Scripps Research Inst|Zinc finger binding domains for cnn| EP1425421A2|2001-09-07|2004-06-09|Everygene AB|Polymorphisms of pd-1| CA2474486C|2002-01-23|2013-05-14|The University Of Utah Research Foundation|Targeted chromosomal mutagenesis using zinc finger nucleases| DE60316124T3|2002-03-15|2018-03-22|Cellectis|HYBRID AND SINGLE CHAIN MEGANUCLEASES AND ITS APPLICATIONS| US20030232410A1|2002-03-21|2003-12-18|Monika Liljedahl|Methods and compositions for using zinc finger endonucleases to enhance homologous recombination| US7361635B2|2002-08-29|2008-04-22|Sangamo Biosciences, Inc.|Simultaneous modulation of multiple genes| CA2497913C|2002-09-05|2014-06-03|California Institute Of Technology|Use of chimeric nucleases to stimulate gene targeting| US20060153826A1|2003-01-28|2006-07-13|Sylvain Arnould|Use of meganucleases for inducing homologous recombination ex vivo and in toto in vertebrate somatic tissues and application thereof| US7972854B2|2004-02-05|2011-07-05|Sangamo Biosciences, Inc.|Methods and compositions for targeted cleavage and recombination| US7888121B2|2003-08-08|2011-02-15|Sangamo Biosciences, Inc.|Methods and compositions for targeted cleavage and recombination| JP4555292B2|2003-08-08|2010-09-29|サンガモバイオサイエンシズインコーポレイテッド|Methods and compositions for targeted cleavage and recombination| US8409861B2|2003-08-08|2013-04-02|Sangamo Biosciences, Inc.|Targeted deletion of cellular DNA sequences| DE602005011943D1|2004-04-08|2009-02-05|Sangamo Biosciences Inc|COMPOSITIONS FOR THE TREATMENT OF NEUROPATHIC AND NEURODEEGENERATIVE DISEASES| AU2005233550B2|2004-04-08|2010-11-18|Sangamo Therapeutics, Inc.|Treatment of neuropathic pain with zinc finger proteins| CA2579677A1|2004-09-16|2006-03-30|Sangamo Biosciences, Inc.|Compositions and methods for protein production| EP2327771A1|2005-03-15|2011-06-01|Cellectis|I-crel meganuclease variants with modified specificity, method of preparation and uses thereof| WO2007014181A2|2005-07-25|2007-02-01|Johns Hopkins University|Site-specific modification of the human genome using custom-designed zinc finger nucleases| JP2009502170A|2005-07-26|2009-01-29|サンガモバイオサイエンシズインコーポレイテッド|Targeted integration and expression of foreign nucleic acid sequences| ES2425022T3|2005-10-18|2013-10-10|Precision Biosciences|Rationally designed meganucleases with sequence specificity and altered DNA binding affinity| WO2007049095A1|2005-10-25|2007-05-03|Cellectis|Laglidadg homing endonuclease variants having mutations in two functional subdomains and use thereof| CA2651494C|2006-05-25|2015-09-29|Sangamo Biosciences, Inc.|Engineered cleavage half-domains| AT536371T|2006-05-25|2011-12-15|Sangamo Biosciences Inc|PROCESS AND COMPOSITIONS FOR GENERIC ACTIVATION| US20100035973A1|2006-07-17|2010-02-11|Nationwide Children's Hospital, Inc.|Disruption of programmed death 1 ligand to adjuvant adeno-associated virus vector vaccines| WO2008153742A2|2007-05-23|2008-12-18|Sangamo Biosciences, Inc.|Methods and compositions for increased transgene expression| AU2008305590A1|2007-09-27|2009-04-02|Sangamo Therapeutics, Inc.|Genomic editing in zebrafish using zinc finger nucleases| EP2188384B1|2007-09-27|2015-07-15|Sangamo BioSciences, Inc.|Rapid in vivo identification of biologically active nucleases| CA2715080C|2007-09-28|2021-09-28|Intrexon Corporation|Therapeutic gene-switch constructs and bioreactors for the expression of biotherapeutic molecules, and uses thereof| US8345045B2|2008-03-04|2013-01-01|Microsoft Corporation|Shader-based extensions for a declarative presentation framework| EP3594333A1|2010-02-08|2020-01-15|Sangamo Therapeutics, Inc.|Engineered cleavage half-domains| US9405700B2|2010-11-04|2016-08-02|Sonics, Inc.|Methods and apparatus for virtualization in an integrated circuit|EP2206723A1|2009-01-12|2010-07-14|Bonas, Ulla|Modular DNA-binding domains| US20110239315A1|2009-01-12|2011-09-29|Ulla Bonas|Modular dna-binding domains and methods of use| JP2013513389A|2009-12-10|2013-04-22|リージェンツオブザユニバーシティオブミネソタ|DNA modification mediated by TAL effectors| KR101953237B1|2010-05-17|2019-02-28|상가모 테라퓨틱스, 인코포레이티드|Novel dna-binding proteins and uses thereof| CA2849920A1|2011-10-06|2013-04-11|Sangamo Biosciences, Inc.|Methods and compositions for regulating hiv infection| US10704021B2|2012-03-15|2020-07-07|Flodesign Sonics, Inc.|Acoustic perfusion devices| RU2725542C2|2013-05-13|2020-07-02|Селлектис|Methods for constructing high-activity t-cells for immunotherapy| US10648001B2|2012-07-11|2020-05-12|Sangamo Therapeutics, Inc.|Method of treating mucopolysaccharidosis type I or II| WO2014059173A2|2012-10-10|2014-04-17|Sangamo Biosciences, Inc.|T cell modifying compounds and uses thereof| CN105120656A|2012-12-21|2015-12-02|塞尔克蒂斯股份有限公司|Potatoes with reduced cold-induced sweetening| US10113162B2|2013-03-15|2018-10-30|Cellectis|Modifying soybean oil composition through targeted knockout of the FAD2-1A/1B genes| KR102192599B1|2013-04-05|2020-12-18|다우 아그로사이언시즈 엘엘씨|Methods and compositions for integration of an exogenous sequence within the genome of plants| SG11201508528TA|2013-05-02|2015-11-27|Anaptysbio Inc|Antibodies directed against programmed death-1 | US9873894B2|2013-05-15|2018-01-23|Sangamo Therapeutics, Inc.|Methods and compositions for treatment of a genetic condition| US20150017136A1|2013-07-15|2015-01-15|Cellectis|Methods for engineering allogeneic and highly active t cell for immunotherapy| EP3039136B8|2013-08-28|2020-12-16|Sangamo Therapeutics, Inc.|Compositions for linking dna-binding domains and cleavage domains| WO2015057976A1|2013-10-17|2015-04-23|Sangamo Biosciences, Inc.|Delivery methods and compositions for nuclease-mediated genome engineering in hematopoietic stem cells| WO2015057980A1|2013-10-17|2015-04-23|Sangamo Biosciences, Inc.|Delivery methods and compositions for nuclease-mediated genome engineering| WO2015074806A1|2013-11-20|2015-05-28|Mann+Hummel Gmbh|Filter element having a filter bellows| CN106030310B|2013-12-13|2019-01-04|通用医疗公司|Soluble high-molecular amountTAU type and its application| US9725710B2|2014-01-08|2017-08-08|Flodesign Sonics, Inc.|Acoustophoresis device with dual acoustophoretic chamber| ES2879373T3|2014-03-18|2021-11-22|Sangamo Therapeutics Inc|Methods and compositions for the regulation of zinc finger protein expression| WO2015164748A1|2014-04-24|2015-10-29|Sangamo Biosciences, Inc.|Engineered transcription activator like effectorproteins| CA2947622A1|2014-05-13|2015-11-19|Sangamo Biosciences, Inc.|Methods and compositions for prevention or treatment of a disease| EP3151846A4|2014-06-05|2017-12-27|Sangamo BioSciences, Inc.|Methods and compositions for nuclease design| EP3158072B1|2014-06-20|2021-01-13|Cellectis|Potatoes with reduced granule-bound starch synthase| WO2016014794A1|2014-07-25|2016-01-28|Sangamo Biosciences, Inc.|Methods and compositions for modulating nuclease-mediated genome engineering in hematopoietic stem cells| WO2016014837A1|2014-07-25|2016-01-28|Sangamo Biosciences, Inc.|Gene editing for hiv gene therapy| WO2016019144A2|2014-07-30|2016-02-04|Sangamo Biosciences, Inc.|Gene correction of scid-related genes in hematopoietic stem and progenitor cells| SI3194570T1|2014-09-16|2022-01-31|Sangamo Therapeutics, Inc.|Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells| KR20180020125A|2015-03-27|2018-02-27|프레지던트 앤드 펠로우즈 오브 하바드 칼리지|Modified T cells and methods for their manufacture and use| EP3294342A4|2015-05-08|2018-11-07|President and Fellows of Harvard College|Universal donor stem cells and related methods| AU2016291778B2|2015-07-13|2021-05-06|Sangamo Therapeutics, Inc.|Delivery methods and compositions for nuclease-mediated genome engineering| GB2595063B|2015-07-31|2022-03-09|Univ Minnesota|Modified cells and methods of therapy| US10837024B2|2015-09-17|2020-11-17|Cellectis|Modifying messenger RNA stability in plant transformations| EP3380622A4|2015-11-23|2019-08-07|Sangamo Therapeutics, Inc.|Methods and compositions for engineering immunity| EA201891212A1|2015-12-18|2019-01-31|Сангамо Терапьютикс, Инк.|ADDRESS DISORGANIZATION OF GKGS CELL RECEPTOR| CA3008413A1|2015-12-18|2017-06-22|Sangamo Therapeutics, Inc.|Targeted disruption of the t cell receptor| CN109152847A|2016-01-15|2019-01-04|桑格摩生物治疗股份有限公司|For treating the method and composition of neurological disease| WO2017134601A1|2016-02-02|2017-08-10|Cellectis|Modifying soybean oil composition through targeted knockout of the fad3a/b/c genes| AU2016391970A1|2016-02-02|2018-07-26|Sangamo Therapeutics, Inc.|Compositions for linking DNA-binding domains and cleavage domains| US11214789B2|2016-05-03|2022-01-04|Flodesign Sonics, Inc.|Concentration and washing of particles with acoustics| AU2017302551A1|2016-07-26|2019-01-31|The General Hospital Corporation|Variants of CRISPR from Prevotella and Francisella 1 | SG10201913948PA|2016-08-24|2020-03-30|Sangamo Therapeutics Inc|Engineered target specific nucleases| US10960085B2|2016-09-07|2021-03-30|Sangamo Therapeutics, Inc.|Modulation of liver genes| JP2019532080A|2016-10-18|2019-11-07|リージェンツ オブ ザ ユニバーシティ オブ ミネソタ|Tumor infiltrating lymphocytes and methods of treatment| BR112019007210A2|2016-10-20|2019-08-13|Sangamo Therapeutics Inc|Methods and Compositions for the Treatment of Fabry Disease| AU2017347928A1|2016-10-31|2019-05-09|Sangamo Therapeutics, Inc.|Gene correction of scid-related genes in hematopoietic stem and progenitor cells| MA49863A|2016-11-01|2020-06-17|Anaptysbio Inc|ANTIBODIES DIRECTED AGAINST PROGRAMMED DEATH 1 | US20200040345A1|2016-12-08|2020-02-06|Case Western Reserve University|Methods and compositions for enhancing functional myelin production| JP2020530307A|2017-06-30|2020-10-22|インティマ・バイオサイエンス,インコーポレーテッド|Adeno-associated virus vector for gene therapy| AU2018364660A1|2017-11-09|2020-05-14|Sangamo Therapeutics, Inc.|Genetic modification of cytokine inducible SH2-containing proteingene| KR20200089334A|2017-12-14|2020-07-24|프로디자인 소닉스, 인크.|Acoustic transducer driver and controller| JP2022500052A|2018-09-18|2022-01-04|サンガモ セラピューティクス, インコーポレイテッド|Programmed cell death 1specific nuclease| KR20210146986A|2019-04-02|2021-12-06|상가모 테라퓨틱스, 인코포레이티드|Methods of treating beta-thalassemia| US20210130828A1|2019-11-01|2021-05-06|Sangamo Therapeutics, Inc.|Gin recombinase variants|
法律状态:
2020-09-15| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2020-09-15| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. | 2021-03-16| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]| 2021-03-30| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]| 2021-11-23| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US12/927,557|US8563314B2|2007-09-27|2010-11-17|Methods and compositions for modulating PD1| US12/927,557|2010-11-17| PCT/US2011/061201|WO2012068380A2|2010-11-17|2011-11-17|Methods and compositions for modulating pd1| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|