US20210348166A1 - Immunotherapy of cancer - Google Patents

Immunotherapy of cancer Download PDF

Info

Publication number
US20210348166A1
US20210348166A1 US17/150,668 US202117150668A US2021348166A1 US 20210348166 A1 US20210348166 A1 US 20210348166A1 US 202117150668 A US202117150668 A US 202117150668A US 2021348166 A1 US2021348166 A1 US 2021348166A1
Authority
US
United States
Prior art keywords
human
cells
sdrna
immune
immunogenic composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/150,668
Inventor
Alexey WOLFSON
Alexey Eliseev
Taisia Shmushkovich
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Phio Pharmaceuticals Corp
Original Assignee
Phio Pharmaceuticals Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Phio Pharmaceuticals Corp filed Critical Phio Pharmaceuticals Corp
Priority to US17/150,668 priority Critical patent/US20210348166A1/en
Assigned to PHIO PHARMACEUTICALS CORP. reassignment PHIO PHARMACEUTICALS CORP. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: RXI PHARMACEUTICALS CORPORATION
Assigned to MIRIMMUNE, LLC reassignment MIRIMMUNE, LLC MERGER AND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: MIRIMMUNE INC., RXI MERGER SUB, LLC
Assigned to RXI PHARMACEUTICALS CORPORATION reassignment RXI PHARMACEUTICALS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MIRIMMUNE, LLC
Assigned to MIRIMMUNE INC. reassignment MIRIMMUNE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELISEEV, ALEXEY, SMUSHKOVICH, Taisia, WOLFSON, Alexey
Publication of US20210348166A1 publication Critical patent/US20210348166A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464429Molecules with a "CD" designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/17Immunomodulatory nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to immunogenic compositions, method of making immunogenic compositions, and methods of using immunogenic compositions for the treatment of cell proliferative disorders or infectious disease, including, for example, cancer and autoimmune disorders.
  • the invention provides cells that are treated with oligonucleotides specifically designed to modulate expression of target genes involved in tumor immune resistance mechanisms.
  • Immunotherapy is the “treatment of disease by inducing, enhancing, or suppressing an immune response”.
  • Immunotherapies designed to elicit or amplify an immune response are activation immunotherapies, while immunotherapies that reduce or suppress immune response are classified as suppression immunotherapies.
  • MAbs act through a mechanisms relevant to the body's own humoral immune response, by binding to key antigens involved in the tumor development and causing moderate forms of cell-mediated immunity, such as antibody-dependent cell-mediated cytotoxicity (ADCC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • TIL tumor-infiltrating lymphocytes
  • TAA tumor associated antigens
  • TIL-based therapeutic approaches are commonly referred to as “adoptive cell transfer” (ACT).
  • TAA tumor-associated antigens
  • TCR T-cell receptor
  • CAR chimeric antigen receptor
  • ACT methods may also be considered as passive immunotherapeutic approaches in that they act directly on the tumor cells without invoking an extended immune response.
  • ACT agents are capable of fully destroying the tumor cells, as opposed to the blockade of selected receptors and moderate cellular responses such as ADCC.
  • Active immunotherapeutic agents are often called therapeutic cancer vaccines, or just cancer vaccines. Many cancer vaccines are currently in clinical trials, and sipuleucell-T has recently become the first such vaccine approved by the United States FDA.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • Vaccines of this type currently in clinical trials are based both on autologous (e.g. OncoVAX, LipoNova) and allogeneic (e.g. Canvaxin, Onyvax-P, GVAX) tumor cell lines.
  • autologous e.g. OncoVAX, LipoNova
  • allogeneic e.g. Canvaxin, Onyvax-P, GVAX
  • DC-based cancer vaccines usually comprise DCs isolated from patients or generated ex vivo by culturing patient's hematopoietic progenitor cells or monocytes. DCs are further loaded with tumor antigens and sometimes combined with immune-stimulating agents, such as GM-CSF. A large number of DC-vaccines are now in clinical trials, and the first FDA-approved vaccine sipuleucell-T is based on DC.
  • immune resistance mechanisms One of the key physiologic functions of the immune system is to recognize and eliminate neoplastic cells, therefore an essential part of any tumor progression is the development of immune resistance mechanisms. Once developed, these mechanisms not only prevent the natural immune system from effecting the tumor growth, but also limit the efficacy of any immunotherapeutic approaches to cancer.
  • An important immune resistance mechanism involves immune-inhibitory pathways, sometimes referred to as immune checkpoints.
  • the immune-inhibitory pathways play particularly important role in the interaction between tumor cells and CD8+ cytotoxic T-lymphocytes, including ACT therapeutic agents.
  • important immune checkpoints are inhibitory receptors expressed on the T-cell surface, such as CTLA-4, PD1 and LAG3, among others.
  • Immunosuppression mechanisms also negatively affect the function of dendritic cells and, as a consequence, the efficacy of DC-based cancer vaccines. Immunosuppressive mechanisms can inhibit the ability of DC to present tumor antigens through the MHC class I pathway and to prime na ⁇ ve CD8+ T-cells for antitumor immunity.
  • ubiquitin ligase A20 and the broadly immune-suppressive protein SOCS1.
  • immune checkpoint blockade can lead to the breaking of immune self-tolerance, thereby inducing a novel syndrome of autoimmune/auto-inflammatory side effects, designated “immune related adverse events,” mainly including rash, colitis, hepatitis and endocrinopathies (Corsello, et al. J. Clin. Endocrinol. Metab., 2013, 98:1361).
  • Reported toxicity profiles of checkpoint inhibitors are different than the toxicity profiles reported for other classes of oncologic agents. Those involve inflammatory events in multiple organ systems, including skin, gastrointestinal, endocrine, pulmonary, hepatic, ocular, and nervous system. (Hodi, 2013 , Annals of Oncology, 24: Suppl, i7).
  • the immunotherapeutic cells of the invention prepared by treating cells with a combination oligonucleotide agents targeting genes associated with tumor or infections disease resistance mechanisms, as well as methods of producing such therapeutic cells and methods of treating disease with the produced therapeutic cells, satisfy this long felt need.
  • compositions comprising therapeutic cells obtained by treating cells ex vivo with oligonucleotides to modulate expression of target genes involved in immune suppression mechanisms.
  • the oligonucleotide agent may be an antisense oliogonucleotide (ASO), including locked nucleic acids (LNAs), methoxyethyl gapmers, and the like, or an siRNA, miRNA, miRNA-inhibitor, morpholino, PNA, and the like.
  • ASO antisense oliogonucleotide
  • LNAs locked nucleic acids
  • siRNA siRNA
  • miRNA miRNA
  • miRNA-inhibitor methoxyethyl gapmers
  • morpholino PNA
  • the oligonucleotides may be chemically modified, for example, including at least one 2-O-methyl modification, 2′-Fluro modification, and/or phosphorothioate modification.
  • the oligonucleotides may include one or more hydrophobic modification, for example, one or more sterol, cholesterol, vitamin D, Naphtyl, isobutyl, benzyl, indol, tryptophane, or phenyl hydrophobic modification.
  • the oligonucleotide may be a hydrophobically-modified siRNA-antisense hybrid.
  • the oligonucleotides may be used in combination with transmembrane delivery systems, such as delivery systems comprising lipids.
  • the cells are obtained and/or derived from a cancer or infectious disease patient, and may be, for example, tumor infiltrating lymphocytes (TIL) and/or T-cells, antigen presenting cells such as dendritic cells, natural killer cells, induced-pluripotent stem cells, stem central memory T-cells, and the like.
  • TIL tumor infiltrating lymphocytes
  • NK-cells antigen presenting cells such as dendritic cells, natural killer cells, induced-pluripotent stem cells, stem central memory T-cells, and the like.
  • the T-cells and NK-cells are preferably genetically engineered to express high-affinity T-Cell receptors (TCR) and/or chimeric antibody or antibody-fragment—T-Cell receptors (CAR).
  • the chimeric antibody/antibody fragment is preferably capable of binding to antigens expressed on tumor cells.
  • Immune cells may be engineered by transfection with plasmid, viral delivery vehicles, or mRNAs
  • the chimeric antibody or fragment is capable of binding CD19 receptors of B-cells and/or binding to antigens expressed on tumors, such as melanoma tumors.
  • antigens expressed on tumors such as melanoma tumors.
  • melanoma-expressed antigens include, for example, GD2, GD3, HMW-MAA, VEGF-R2, and the like.
  • Target genes identified herein for modification include: cytotoxic T-cell antigen 4 (CTLA4), programmed cell death protein 1 (PD1), tumor growth factor receptor beta (TGFR-beta), LAG3, TIM3, and adenosine A2a receptor; anti-apoptotic genes including, but not limited to: BAX, BAC, Casp8, and P53; A20 ubiquitine ligase (TNFAIP3, SOCS1 (suppressor of cytokine signaling), IDO (indolamine-2,3-dioxygenase; tryptophan-degrading enzyme), PD-L1 (CD274)(surface receptor, binder to PD1 on Tcells), Notch ligand Deltal (DLL1), Jagged 1, Jagged 2, FasL (pro-apoptotic surface molecule), CCL17, CCL22 (secreted chemokines that attract Treg cells), IL10 receptor (IL10RA), p38 (MAPK14), STAT3, TNFSF4
  • the engineered therapeutic cells are treated with RNAi agents designed to inhibit expression of one or more of the targeted genes.
  • the RNAi agent may comprise a guide sequence that hybridizes to a target gene and inhibits expression of the target gene through an RNA interference mechanism, where the target region is selected from the group listed in Table 1.
  • the RNA agent can be chemically modified, and preferably includes at least one 2′-O-methyl, 2′-O-Fluoro, and/or phosphorothioate modification, as well as at least one hydrophobic modification such as cholesterol, and the like.
  • the immunogenic compositions described herein are useful for the treatment of proliferative disorders, including cancers, and/or infectious disease and are produced by the ex-vivo treatment of cells with oligonucleotides to modulate the expression of target genes involved in tumor immune resistance mechanisms.
  • the ex vivo treatment of cells includes administering to the cells an oligonucleotide capable of targeting and inhibiting expression of a gene involved in a tumor suppressor mechanism, such as the genes listed in Table 1.
  • the oligonucleotide can be used in combination with a transmembrane delivery system that may comprise one or more of: lipid(s) and vector, such as a viral vector.
  • the invention includes a method of treating a cell proliferative disorder or infectious disease by administering to a subject in need thereof, an immunogenic composition comprising cells that have been treated with one or more oligonucleotide to modulate the expression of one or more target gene involved in tumor immune resistance mechanisms, for example, one or more of the target genes of Table 1.
  • the invention preferably includes immunogenic cells treated with a plurality of oligonucleotide agents targeting a combination of target genes described herein.
  • the combination may target a plurality of suppressor receptor genes, cytokine receptor genes, regulatory genes, and/or apoptotic factors in order to inhibit tumor immune resistance mechanisms.
  • the present invention is directed to novel immunotherapeutic cells, methods of generating the immunotherapeutic cells, and therapeutic methods employing such cells.
  • a new method of immune checkpoint inhibition is described herein, applicable to a broad variety of cell-based immunotherapies, including, but not limited to adaptive cell transfer, for example, based on TIL, TCR, CAR, and other cell types, as well as dendritic cell-based cancer vaccines.
  • Self-deliverable RNAi technology provides efficient transfection of short oligonucleotides in any cell type, including immune cells, providing increased efficacy of immunotherapeutic treatments.
  • the activated immune cells can be protected by preventing apoptosis via inhibition of key activators of the apoptotic pathway, such as BAC, BAX, Casp8, and P53, among others.
  • the activated immune cells modified by oligonucleotide transfer for a single therapeutic agent for administration to a subject providing a number of advantages as compared to separately administered combinations of vaccines and immunotherapeutics and separately administered checkpoint inhibitors. These advantages include lack of side effects associated with the checkpoint inhibitors in a single therapeutic agent (activated immune cells modified by oligonucleotides targeting immune resistance genes).
  • the claimed immunotherapeutic cells improves upon any known immunotherapeutic cells and methods of producing immunotherapeutic cells because it provides:
  • FIG. 1 is a schematic diagram showing the structure of an sdRNA molecule.
  • FIG. 2 is a graph showing sdRNA-induced silencing of GAPDH and MAP4K4 in HeLa cells.
  • FIG. 3 is a graph showing sdRNA-induced knock-down of multiple targets using sdRNA agents directed to three genes in NK-92 cells.
  • FIG. 4 is a graph showing the knock-down of gene expression in Human Primary T cells by sdRNA agents targeting TP53 and MAP4K4.
  • FIG. 5 is a graph showing sdRNA-induced knock-down of CTLA4 and PD1 in Human Primary T cells.
  • FIG. 6 is a graph showing the reduction of PDCD1 and CTLA-4 surface expression by sdRNA in Human Primary T cells.
  • FIG. 7 is a graph showing MAP4K4-cy3 sdRNA delivery into T and B cells in human PBMCs.
  • the invention is defined by the claims, and includes oligonucleotides specifically designed and selected to reduce and/or inhibit expression of suppressors of immune resistance (inhibitory oligonucleotides), compositions comprising cells modified by treatment with such inhibitory oligonucleotides, methods of making such compositions, and methods of using the compositions to treat proliferation and/or infectious diseases.
  • inhibitory oligonucleotides specifically designed and selected to reduce and/or inhibit expression of suppressors of immune resistance
  • compositions comprising cells modified by treatment with such inhibitory oligonucleotides
  • methods of making such compositions methods of using the compositions to treat proliferation and/or infectious diseases.
  • cells are treated with a combination of oligonucleotide agents, each agent particularly designed to interfere with and reduce the activity of a targeted immune suppressor.
  • the combination of oligonucleotide agents targets multiple immune suppressor genes selected from checkpoint inhibitor genes such as CTLA4, PD-1/PD-1L, BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3, B7-H4 receptors, and TGF beta type 2 receptor; cytokine receptors that inactivate immune cells, such as TGF-beta receptor A and IL-10 receptor; regulatory genes/transcription factors modulating cytokine production by immune cells, such as STAT-3 and P38, miR-155, miR-146a; and apoptotic factors involved in cascades leading to cell death, such as p53 and Cacp8.
  • checkpoint inhibitor genes such as CTLA4, PD-1/PD-1L, BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3, B7-H4 receptors, and TGF beta type 2 receptor
  • the oligonucleotide agent is a self-deliverable RNAi agent, which is a hydrophobically modified siRNA-antisense hybrid molecule, comprising a double-stranded region of about 13-22 base pairs, with or without a 3′-overhang on each of the sense and antisense strands, and a 3′ single-stranded tail on the antisense strand of about 2-9 nucleotides.
  • RNAi agent is a hydrophobically modified siRNA-antisense hybrid molecule, comprising a double-stranded region of about 13-22 base pairs, with or without a 3′-overhang on each of the sense and antisense strands, and a 3′ single-stranded tail on the antisense strand of about 2-9 nucleotides.
  • the oligonucleotide contains at least one 2′-O-Methyl modification, at least one 2′-O-Fluoro modification, and at least one phosphorothioate modification, as well as at least one hydrophobic modification selected from sterol, cholesterol, vitamin D, napthyl, isobutyl, benzyl, indol, tryptophane, phenyl, and the like hydrophobic modifiers (see FIG. 1 ).
  • the oligonucleotide may contain a plurality of such modifications.
  • Proliferative disease includes diseases and disorders characterized by excessive proliferation of cells and turnover of cellular matrix, including cancer, atherlorosclerosis, rheumatoid arthritis, psoriasis, idiopathic pulmonary fibrosis, scleroderma, cirrhosis of the liver, and the like.
  • Cancers include but are not limited to, one or more of: small cell lung cancer, colon cancer, breast cancer, lung cancer, prostate cancer, ovarian cancer, pancreatic cancer, melanoma, hematological malignancy such as chronic myeloid leukemia, and the like cancers where immunotherapeutic intervention to suppress tumor related immune resistance is needed.
  • Immune target genes can be grouped into at least four general categories: (1) checkpoint inhibitors; (2) cytokine receptors that inactivate immune cells, (3) anti-apoptotic genes; and (4) regulator genes, for example, transcription factors.
  • Immune Checkpoint inhibitors include immunotherapeutic agents that bind to certain checkpoint proteins, such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1) and its ligand PD-L1 to block and disable inhibitory proteins that prevent the immune system from attacking diseased cells such as cancer cells, liberating tumor-specific T cells to exert their effector function against tumor cells.
  • CTL-4 cytotoxic T lymphocyte antigen-4
  • PD-1 programmed death-1
  • Tumor related immune resistance genes include genes involved in checkpoint inhibition of immune response, such as CTLA-4 and PD-1/PD-L1; TGF-beta, LAG3, Tim3, adenosine A2a receptor;
  • Regulator genes include transcription factors and the like that modulate cytokine production by immune cells, and include p38, STAT3, microRNAs miR-155, miR-146a;
  • Anti-apoptotic genes include BAX, BAC, Casp8, P53 and the like; and combinations thereof.
  • Infectious diseases include, but are not limited to, diseases caused by pathogenic microorganisms, including, but not limited to, one or more of bacteria, viruses, parasites, or fungi, where immunotherapeutic intervention to suppress pathogen related immune resistance and/or overactive immune response.
  • Immunogenic composition includes cells treated with one or more oligonucleotide agent, wherein the cells comprise T-cells.
  • the T-cells may be genetically engineered, for example, to express high affinity T-cell receptors (TCR), chimeric antibody—T-cell receptors (CAR), where the chimeric antibody fragments are capable of binding to CD19 receptors of B-cells and/or to antigens expressed on tumor cells.
  • TCR T-cell receptors
  • CAR chimeric antibody—T-cell receptors
  • the chimeric antibody fragments bind antigens expressed on melanoma tumors, selected from GD2, GD3, HMW-MAA, and VEGF-R2.
  • Immunogenic compositions described herein include cells comprising antigen-presenting cells, dendritic cells, engineered T-cells, natural killer cells, stem cells, including induced pluripotent stem cells, and stem central memory T-cells.
  • the treated cell also comprises one or a plurality of oligonucleotide agents, preferably sdRNAi agents specifically targeting a gene involved in an immune suppression mechanism, where the oligonucleotide agent inhibits expression of said target gene.
  • the target gene is selected from A20 ubiquitin ligase such as TNFAIP3, SOCS1 (suppressor of cytokine signaling), Tyro3/Axl/Mer (suppressors of TLR signaling), IDO (indolamine-2,3-dioxygenase, tryptophan-degrading enzyme), PD-L1/CD274 (surface receptor, binds PD1 on T-cells), Notch ligand Delta (DLL1), Jagged 1, Jagged 2, FasL (pro-apoptotic surface molecule), CCL17, CCL22 (secreted chemokines that attract Treg cells), IL-10 receptor (IL10Ra), p38 (MAPK14), STAT3, TNFSF4 (OX40L), microRNA miR-155, miR-146a, anti-apoptotic genes, including but not limited to BAX, BAC, Casp8, and P53; and combinations thereof.
  • A20 ubiquitin ligase such
  • Particularly preferred target genes are those shown in Table 1.
  • Ex-vivo treatment includes cells treated with oligonucleotide agents that modulate expression of target genes involved in immune suppression mechanisms.
  • the oligonucleotide agent may be an antisense oligonucleotide, including, for example, locked nucleotide analogs, methyoxyethyl gapmers, cyclo-ethyl-B nucleic acids, siRNAs, miRNAs, miRNA inhibitors, morpholinos, PNAs, and the like.
  • the oligonucleotide agent is an sdRNAi agent targeting a gene involved in an immune suppression mechanism.
  • the cells treated in vitro by the oligonucleotide agent may be immune cells expanded in vitro, and can be cells obtained from a subject having a proliferative or infectious disease.
  • the cells or tissue may be treated in vivo, for example by in situ injection and/or intravenous injection.
  • Oligonucleotide or oligonucleotide agent refers to a molecule containing a plurality of “nucleotides” including deoxyribonucleotides, ribonucleotides, or modified nucleotides, and polymers thereof in single- or double-stranded form.
  • the term encompasses nucleotides containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides.
  • Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • Nucleotide as used herein to include those with natural bases (standard), and modified bases well known in the art.
  • the nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see, for example, PCT Publications No. WO 92/07065 and WO 93/15187.
  • Non-limiting examples of base modifications that can be introduced into nucleic acid molecules include, hypoxanthine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2,4,6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g. 6-methyluridine and pseudouridine), propyne, and others.
  • modified bases includes nucleotide bases other than adenine, guanine, cytosine, and uracil, modified for example, at the 1′ position or their equivalents.
  • deoxyribonucleotide encompasses natural and synthetic, unmodified and modified deoxyribonucleotides. Modifications include changes to the sugar moiety, to the base moiety and/or to the linkages between deoxyribonucleotide in the oligonucleotide.
  • RNA defines a molecule comprising at least one ribonucleotide residue.
  • ribonucleotide defines a nucleotide with a hydroxyl group at the 2′ position of a ⁇ -D-ribofuranose moiety.
  • RNA includes double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
  • Nucleotides of the RNA molecules described herein may also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • modified nucleotide refers to a nucleotide that has one or more modifications to the nucleoside, the nucleobase, pentose ring, or phosphate group.
  • modified nucleotides exclude ribonucleotides containing adenosine monophosphate, guanosine monophosphate, uridine monophosphate, and cytidine monophosphate and deoxyribonucleotides containing deoxyadenosine monophosphate, deoxyguanosine monophosphate, deoxythymidine monophosphate, and deoxycytidine monophosphate.
  • Modifications include those naturally-occurring that result from modification by enzymes that modify nucleotides, such as methyltransferases.
  • Modified nucleotides also include synthetic or non-naturally occurring nucleotides.
  • Synthetic or non-naturally occurring modifications in nucleotides include those with 2′ modifications, e.g., 2′-O-methyl, 2′-methoxyethoxy, 2′-fluoro, 2′-allyl, 2′-O-[2-(methylamino)-2-oxoethyl], 4′-thio, 4′-CH2-O-2′-bridge, 4′-(CH2) 2-O-2′-bridge, 2′-LNA, and 2′-O-(N-methylcarbamate) or those comprising base analogs.
  • amino is meant 2′-NH2 or 2′-O—NH2, which can be modified or unmodified.
  • modified groups are described, for example, in U.S. Pat. Nos. 5,672,695 and 6,248,878.
  • miRNA refers to a nucleic acid that forms a single-stranded RNA, which single-stranded RNA has the ability to alter the expression (reduce or inhibit expression; modulate expression; directly or indirectly enhance expression) of a gene or target gene when the miRNA is expressed in the same cell as the gene or target gene.
  • a miRNA refers to a nucleic acid that has substantial or complete identity to a target gene and forms a single-stranded miRNA.
  • miRNA may be in the form of pre-miRNA, wherein the pre-miRNA is double-stranded RNA.
  • the sequence of the miRNA can correspond to the full length target gene, or a subsequence thereof.
  • the miRNA is at least about 15-50 nucleotides in length (e.g., each sequence of the single-stranded miRNA is 15-50 nucleotides in length, and the double stranded pre-miRNA is about 15-50 base pairs in length).
  • the miRNA is 20-30 base nucleotides.
  • the miRNA is 20-25 nucleotides in length.
  • the miRNA is 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • Target gene includes genes known or identified as modulating the expression of a gene involved in an immune resistance mechanism, and can be one of several groups of genes, such as suppressor receptors, for example, CTLA4 and PD1; cytokine receptors that inactivate immune cells, for example, TGF-beta receptor, LAG3, Tim3, adenosine A2a receptor, and IL10 receptor; regulatory genes for example, STAT3, p38, mir155 and mir146a; and apoptosis factors involved in cascades leading to cell death, for example, P53, Casp8, BAX, BAC, and combinations thereof. See also preferred target genes listed in Table 1.
  • small interfering RNA defines a group of double-stranded RNA molecules, comprising sense and antisense RNA strands, each generally of about 1022 nucletides in length, optionally including a 3′ overhang of 1-3 nucleotides.
  • siRNA is active in the RNA interference (RNAi) pathway, and interferes with expression of specific target genes with complementary nucleotide sequences.
  • sdRNA refers to “self-deliverable” RNAi agents, that are formed as an asymmetric double-stranded RNA-antisense oligonucleotide hybrid.
  • the double stranded RNA includes a guide (sense) strand of about 19-25 nucleotides and a passenger (antisense) strand of about 10-19 nucleotides with a duplex formation that results in a single-stranded phosphorothiolated tail of about 5-9 nucleotides.
  • RNA sequences may be modified with stabilizing and hydrophobic modifications such as sterols, for example, cholesterol, vitamin D, naphtyl, isobutyl, benzyl, indol, tryptophane, and phenyl, which confer stability and efficient cellular uptake in the absence of any transfection reagent or formulation.
  • sterols for example, cholesterol, vitamin D, naphtyl, isobutyl, benzyl, indol, tryptophane, and phenyl
  • Immune response assays testing for IFN-induced proteins indicate sdRNAs produce a reduced immunostimulatory profile as compared other RNAi agents. See, for example, Byrne et al., December 2013 , J. Ocular Pharmacology and Therapeutics, 29(10): 855-864.
  • cells are obtained from subjects with proliferative disease such as cancer, or an infectious disease such as viral infection.
  • the obtained cells are treated directly as obtained or may be expanded in cell culture prior to treatment with oligonucleotides.
  • the cells may also be genetically modified to express receptors that recognize specific antigens expressed on the tumor cell surface (CAR) or intracellular tumor antigens presented on MHC class I (TCR).
  • CAR tumor cell surface
  • TCR MHC class I
  • siRNA Small interfering RNA
  • silencing RNA is a double stranded RNA molecule, generally 19-25 base pairs in length.
  • siRNA is used in RNA interference (RNAi), where it interferes with expression of specific genes with complementary nucleotide sequences.
  • Double stranded DNA can be generally used to define any molecule comprising a pair of complementary strands of RNA, generally a sense (passenger) and antisense (guide) strands, and may include single-stranded overhang regions.
  • dsRNA contrasted with siRNA, generally refers to a precursor molecule that includes the sequence of an siRNA molecule which is released from the larger dsRNA molecule by the action of cleavage enzyme systems, including Dicer.
  • sdRNA self-deliverable are a new class of covalently modified RNAi compounds that do not require a delivery vehicle to enter cells and have improved pharmacology compared to traditional siRNAs.
  • “Self-deliverable RNA” or sdRNA is a hydrophobically modified RNA interfering-antisense hybrid, demonstrated to be highly efficacious in vitro in primary cells and in vivo upon local administration. Robust uptake and/or silencing without toxicity has been demonstrated in several tissues including dermal, muscle, tumors, alveolar macrophages, spinal cord, and retina cells and tissues. In dermal layer and retina, intradermal and intra-vitreal injection of sdRNA at mg doses induced potent and long lasting silencing.
  • sdRNA is a superior functional genomics tool, enabling RNAi in primary cells and in vivo, it has a relatively low hit rate as compared to conventional siRNAs. While the need to screen large number of sequences per gene is not a limiting factor for therapeutic applications, it severely limits the applicability of sdRNA technology to functional genomics, where cost effective compound selection against thousands of genes is required. To optimize sdRNA structure, chemistry, targeting position, sequence preferences, and the like, a proprietary algorithm has been developed and utilized for sdRNA potency prediction. Availability of sdRNA reagents that are active in all cell types ex vivo and in vivo enables functional genomics and target stratification/validation studies.
  • SdRNA sequences were selected based on a proprietory selection algorithm, designed on the basis of a functional screen of over 500 sdRNA sequences in the luciferase reporter assay of HeLa cells. Regression analysis of was used to establish a correlation between the frequency of occurrence of specific nucleotide and modification at any specific position in sdRNA duplex and its functionality in gene suppression assay. This algorithm allows prediction of functional sdRNA sequences, defined as having over 70% knockdown at 1 ⁇ M concentration, with a probability over 40%.
  • Table 1 shows predictive gene targets identified using the proprietary algorithm and useful in the cellular immunotherapeutic compositions and methods described herein.
  • BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • Applic BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • ation of RNAi technology to functional genomics studies in prim BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • ary cells and in vivo is limited by requirements to formulate siRNAs into lipids or use of other cell delivery techniques.
  • the self-deliverable RNAi technology provides a method of directly transfecting cells with the RNAi agent, without the need for additional formulations or techniques.
  • the ability to transfect hard-to-transfect cell lines, high in vivo activity, and simplicity of use, are characteristics of the compositions and methods that present significant functional advantages over traditional siRNA-based techniques.
  • the sdRNAi technology allows direct delivery of chemically synthesized compounds to a wide range of primary cells and tissues, both ex-vivo and in vivo.
  • BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • B7-H3 and B7-H4 receptors B7-H3 and B7-H4 receptors
  • TGFbeta type 2 receptor TGFbeta type 2 receptor
  • siRNA molecules require a substantial reduction in size and the introduction of extensive chemical modifications which are not well tolerated by RNAi machinery, resulting in extremely low probability of finding active molecules (low hit rate).
  • the sdRNA technology allows efficient RNAi delivery to primary cells and tissues in vitro and in vivo, with demonstrated silencing efficiency in humans.
  • sdRNA are formed as hydrophobically-modified siRNA-antisense oligonucleotide hybrid structures, and are disclosed, for example in Byrne et al., December 2013, J. Ocular Pharmacology and Therapeutics, 29(10): 855-864.
  • the oligonucleotide agents preferably comprise one or more modification to increase stability and/or effectiveness of the therapeutic agent, and to effect efficient delivery of the oligonucleotide to the cells or tissue to be treated.
  • modifications include at least one
  • BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • 2′-O-methyl modification at least one 2′-O-Fluro modification, and at least one diphosphorothioate modification.
  • the oligonucleotide is modified to include one or more hydrophobic modification selected from sterol, cholesterol, vitamin D, naphtyl, isobutyl, benzyl, indol, tryptophane, and phenyl.
  • the hydrophobic modification is preferably a sterol.
  • the oligonucleotides may be delivered to the cells in combination with a transmembrane delivery system, preferably comprising lipids, viral vectors, and the like.
  • a transmembrane delivery system preferably comprising lipids, viral vectors, and the like.
  • the oligonucleotide agent is a self-delivery RNAi agent, that does not require any delivery agents.
  • This objective is accomplished by determining the appropriate genes to be targeted by the oligonucleotide in order to silence immune suppressor genes and using the proprietary algorithm to select the most appropriate target sequence.
  • the immunotherapeutic cell be modified to include multiple oligonucleotide agents targeting a variety of genes involved in immune suppression and appropriate for the selected target disease and genes.
  • a preferred immunotherapeutic cell is a T-Cell modified to knock-down both CTLA-4 and PD-1
  • oligonucleotides to related genes involved in immune suppression include varied combinations of the selected target sequences of Table 1.
  • BTLA B and T-lymphocyte attenuator
  • KIR killer immunoglobulin-like receptors
  • B and T-lymphocyte attenuator KIR (killer immunoglobulin-like receptors)
  • B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor Preferred BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor
  • therapeutic combinations include cells engineered to knock down gene expression of the following target genes:
  • the therapeutic compositions described herein are useful to treat a subject suffering from a proliferation disorder or infectious disease.
  • the immunotherapeutic composition is useful to treat disease characterized by suppression of the subjects immune mechanisms.
  • the sdRNA agents described herein are specifically designed to target genes involved in diseases-associated immune suppression pathways.
  • Methods of treating a subject comprise administering to a subject in need thereof, an immunogenic composition comprising an sdRNAi agent capable of inhibiting expression of genes involved in immune suppression mechanisms, for example, any of the genes listed in Table 1 or otherwise described herein.
  • Immunotherapeutic agents described herein were produced by treating cells with particular sdRNA agents designed to target and knock down specific genes involved in immune suppression mechanisms.
  • sdRNA agents designed to target and knock down specific genes involved in immune suppression mechanisms.
  • the following cells and cell lines have been successfully treated with sdRNA and were shown to knock down at least 70% of targeted gene expression in the specified human cells.
  • HeLa cells (ATCC CRM-CCL-2) were subcultured 24 hours before transfection and kept log phase.
  • the efficacy of several GAPDH sdRNAs was tested by qRT-PCR, including G13 sdRNA listed in the Table 1.
  • the total volume of medium for each oligo concentration point was calculated as [50 ⁇ l/well] ⁇ [number of replicates for each serum point]. Oligonucleotides were dispensed into a 96 well plate at 50 ⁇ l/well.
  • Cells were collected for transfection by trypsinization in a 50 ml tube, washed twice with medium containing 10% FBS without antibiotics, spun down at 200 ⁇ g for 5 minutes at room temperature and resuspended in EMEM medium containing twice the required amount of FBS for the experiment (6%) and without antibiotics. The concentration of the cells was adjusted to 120,000/ml to yield a final concentration of 6,000 cells/50 ⁇ l/well. The cells were dispensed at 50 ⁇ l/well into the 96-well plate with pre-diluted oligos and placed in the incubator for 48 hours.
  • RNA isolation was conducted according to the manufacturer's instructions, and the RNA was eluted with 100 ⁇ l RNase-free water, and used undiluted for one-step qRT-PCR.
  • NT cells Dilutions of non-transfected (NT) cells of 1:5 and 1:25 were prepared for the standard curve using RNase-free water. qRT-PCR was performed by dispensing 9 ⁇ l/well into a low profile PCR plate and adding 1 ⁇ l RNA/well from the earlier prepared RNA samples. After brief centrifugation, the samples were placed in the real-time cycler and amplified using the settings recommended by the manufacturer.
  • GAPDH gene expression was measured by qPCR, normalized to MAP4K4 and plotted as percent of expression in the presence of non-targeting sdRNA. The results were compared to the normalized according to the standard curve. As shown in FIG. 2 , several sdRNA agents targeting GAPDH or MAP4K4 significantly reduced their mRNA levels leading to more than 80-90% knock-down with 1 ⁇ M sdRNA. (See FIG. 2 ).
  • NK-92 cells were obtained from Conqwest and subjected to one-step RT-PCR analysis without RNA purification using the FastLane Cell Multiplex Kit (Qiagen, Cat. No. 216513). For transfection, NK-92 cells were collected by centrifugation and diluted with RPMI medium containing 4% FBS and IL2 1000 U/ml and adjusted to1,000,000 cells/ml.
  • sdRNA agents targeting MAP4K4, PPIB or GADPH were diluted separately in serum-free RPMI medium to 4 ⁇ M and individually aliquoted at 50 ⁇ l/well into a 96-well plate. The prepared cells were then added at 50 ⁇ l cells/well to the wells with either MAP4K4, PPIB or GAPDH sdRNAs. Cells were incubated for 24, 48, or 72 hours.
  • the plated transfected cells were washed once with 100 ⁇ l/well PBS and once with FCW buffer. After removal of supernatant, cell processing mix of 23.5 ⁇ l FCPL and 1.5 ⁇ l gDNA wipeout solution was added to each well and incubated for five minutes at room temperature. Lysates were then transferred to PCR strips and heated at 75° C. for five minutes.
  • MAP4K4-FAM MAP4K4-FAM
  • GAPDH-VIC human GAPDH-VIC
  • a volume of 9 ⁇ l/well of each reaction mix was dispensed into a low profile PCR plate.
  • One ⁇ l lysate per well was added from the previously prepared lysates.
  • the samples were amplified using the settings recommended by the manufacturer.
  • Results shown in FIG. 3 demonstrate significant silencing of each of the multiple targets, MAP4K4, PPIB, and GADPH by sdRNA agents transfected into NK-92 cells, including greater than 75% inhibition of expression of each target within 24 to 72 hours of incubation.
  • T-cells Primary human T-cells were obtained from AllCells (CA) and cultured in complete RPMI medium containing 1000 IU/ml IL2. Cells were activated with anti-CD3/CD28 Dynabeads (Gibco, 11131) according to the manufacturer's instructions for at least 4 days prior to the transfection. Cells were collected by brief vortexing to dislodge the beads from cells and separating them using the designated magnet.
  • sdRNA agents targeting TP53 or MAP4K4 were prepared by separately diluting the sdRNAs to 0.2-4 ⁇ M in serum-free RPMI per sample (well) and individually aliquoted at 100 ⁇ l/well of 96-well plate.
  • Cells were prepared in RPMI medium containing 4% FBS and IL2 2000 U/ml at 1,000,000 cells/ml and seeded at 100 ⁇ l/well into the 96-well plate with pre-diluted sdRNAs.
  • the plated transfected cells were washed once with 100 ⁇ l/well PBS and processed with FastLane Cell Multiplex Kit reagents essentially as described for the Example 4 and according to the manufacturer's instructions.
  • Taqman gene expression assays were used in the following combinations: human MAP4K4-FAM/GAPDH-VIC or human TP53-FAM (Taqman, Hs01034249_m1)/GAPDH-VIC.
  • a volume of 18 ⁇ l/well of each reaction mix was combined with 2 ⁇ l lysates per well from the previously prepared lysates. The samples were amplified as before (see Example 4).
  • Results shown in FIG. 4 demonstrate significant silencing of both MAP4K4 and TP53 by sdRNA agents transfected into T-cells, reaching 70-80% inhibition of gene expression with 1-2 ⁇ M sdRNA.
  • Melanomas utilize at least two particular pathways to suppress immune function of T-cells, and each involves both PD1 and CTLA4.
  • Melanoma tumors expressing the PD1 ligand, PD1L can be targeted with T-cells pretreated ex-vivo with sd-RNAi agents specifically designed to target PD1 and interfere with PD1 expression.
  • PD1 is also known as PDCD1, and particular targeting sequences and gene regions identified and predicted to be particularly functional in sdRNA mediated suppression, are shown in Table 1 for PDCD1 (NM_005018) and for CTLA4 (NM005214).
  • Treatment of melanoma tumors can be effected by providing to melanoma cells T-cells, such as tumor-infiltrating lymphocytes, pretreated ex-vivo with a combination of sdRNAs targeting PD1/PDCD1 and CTLA4, for example, targeting one or more of the twenty target sequences listed for PD1/PDCD1 and/or CTLA4.
  • T-cells such as tumor-infiltrating lymphocytes
  • a combination of sdRNAs targeting PD1/PDCD1 and CTLA4 for example, targeting one or more of the twenty target sequences listed for PD1/PDCD1 and/or CTLA4.
  • a combination of sdRNAs targeting PD1/PDCD1 and FASLG (NM_000639) and/or CTLA4 can increase T-cell toxicity in tumors expressing both PD1L and FAS.
  • T-cells used for the immunotherapy of melanoma can also be treated with sdRNA targeting other genes implicated in immunosuppression by the tumor.
  • receptors include, but are not limited to TGF-beta type 1 and 2 receptors, BTLA (binder of herpes virus entry indicator (HVEM) expressed on melanoma cells), and receptors of integrins expressed by myeloid derived suppressor cells (MDSC), such as CD11b, CD18, and CD29.
  • any combination of sdRNAs targeting PD1/PDCD1 and any one of know suppressing receptors may be helpful to reduce immune suppression and increase therapeutic efficacy.
  • T-cell or dendritic cell suppression may be modulated by various cytokines, such as IL10 and/or TGF beta. Suppressing corresponding receptors in T-cells and dendritic cells may be beneficial for their activity. For example, providing a combination of anti-PD1 with anti-IL10R sdRNAs is exptected to mitigate cytokine induced suppression of T-cells and dendritic cells, as compared with anti-PD1 alone.
  • sdRNA agents to suppress genes involved in apoptosis (programmed cell death), such as p53, Casp8 or other gene activating apoptosis may be beneficial to increase immune cell activity.
  • Combination of an anti-receptor sdRNAs with sdRNAs against pro-apoptotic genes can additionally reduce death of immune cells and thus increase their activity.
  • combination of anti-PD1 with anti-p53 sdRNAs may additionally protect T-cells from suppression by blocking activation of apoptosis.
  • sdRNA agents targeting PDCD1 and CTLA-4 were prepared by separately diluting the sdRNAs to 0.4-4 ⁇ M in serum-free RPMI per sample (well) and aliquoted at 100 ⁇ l/well of 96-well plate.
  • Cells were prepared in RPMI medium containing 4% FBS and IL2 2000 U/ml at 1,000,000 cells/ml and seeded at 100 ⁇ l/well into the 96-well plate with pre-diluted sdRNAs.
  • the transfected cells were washed once with 100 ⁇ l/well PBS and processed with FastLane Cell Multiplex Kit reagents essentially as described for the Example 4 and according to the manufacturer's instructions.
  • Taqman gene expression assays were used in the following combinations: human PDCD1-FAM (Taqman, Hs01550088_m1)/GAPDH-VIC or human CTLA4-FAM (Taqman, Hs03044418_m1)/GAPDH-VIC.
  • a volume of 18 ⁇ l/well of each reaction mix was combined with 2 ⁇ l lysates per well from the previously prepared lysates. The samples were amplified as before (see Example 4).
  • Results shown in FIG. 5 demonstrate significant silencing of PDCD1 and CTLA-4 by using combined sdRNA agents delivered to T-cells, obtaining greater than 60-70% inhibition of gene expression with 2 ⁇ M sdRNA.
  • sdRNA agents targeting CTLA-4 or PD1 were separately diluted to 5 ⁇ M in serum-free RPMI per sample (well) and aliquoted at 250 ⁇ l/well to 24-well plates. Cells mixed with magnetic beads were collected and adjusted to 500,000 cells in 250 ⁇ l RPMI medium containing 4% FBS and IL2 2000 IU/ml. Cells were seeded at 250 ⁇ l/well to the prepared plate containing pre-diluted sdRNAs. 24 hours later FBS was added to the cells to obtain 10% final concentration.
  • transfected cells were collected, separated from the activation beads using the magnet, as described in Example 5.
  • Cells were washed with PBS, spun down and resuspended in blocking buffer (PBS with 3% BSA) at 200,000 cells/50 ⁇ l/sample.
  • Antibody dilutions were prepared in the blocking buffer.
  • the antibodies were mixed in two combinations: anti-PD1/anti-CD3 (1:100 dilutions for both antibodies) and anti-CTLA4/anti-CD3 (10 ⁇ l/106 cells for anti-CTLA4; 1:100 for CD3).
  • the following antibodies were used: rabbit monoclonal [SP7] to CD3 (Abcam, ab16669); mouse monoclonal [BNI3] to CTLA4 (Abcam, ab33320) and mouse monoclonal [NAT105] to PD1 (Abcam, ab52587).
  • Cells were mixed with the diluted antibodies and incubated 30 minutes on ice. Cells were then washed twice with PBS containing 0.2% Tween-20 and 0.1% sodium azide.
  • sdRNA efficiently reduced surface expression of CTLA-4 and PD1 in activated Human Primary T cells.
  • PBMCs Human Peripheral Blood Mononuclear Cells
  • PBMCs were cultured in complete RPMI supplemented with 1.5% PHA solution and 500 U/ml IL2.
  • PBMCs were collected by centrifugation and diluted with RPMI medium containing 4% FBS and IL2 1000 U/ml and seeded to 24-well plate at 500,000 cells/well.
  • MAP4K4 sdRNA labeled with cy3 was added to the cells at 0.1 ⁇ M final concentration. After 72 hours of incubation, the transfected cells were collected, washed with PBS, spun down and diluted in blocking buffer (PBS with 3% BSA) at 200,000 cells/50 ⁇ l/sample.
  • Antibody dilutions were prepared in the blocking buffer as following: 1:100 final dilution anti-CD3 (Abcam, ab16669) and anti-CD19 at 10 ⁇ l/1,000,000 cells (Abcam, ab31947). Cells were mixed with the diluted antibodies and incubated 30 min on ice. Cells were then washed twice with PBS containing 0.2% Tween-20 and 0.1% sodium azide.
  • FIG. 7 shows efficient transfection over 97% of CD3-positive (t cells) and over 98% CD19-positive (B-cells) subsets in Human Peripheral Blood Mononuclear Cells (PBMCs).
  • PBMCs Human Peripheral Blood Mononuclear Cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Immunogenic modulators and compositions comprising oligonucleotide agents capable of inhibiting suppression of immune response by reducing expression of one or more gene involved with an immune response suppression mechanism.

Description

    CROSS REFERENCE
  • This application claims priority to U.S. Provisional Application No. 61/910,728, filed Dec. 2, 2013, herein incorporated by reference in its entirety.
  • TECHNICAL FIELD
  • The present invention relates to immunogenic compositions, method of making immunogenic compositions, and methods of using immunogenic compositions for the treatment of cell proliferative disorders or infectious disease, including, for example, cancer and autoimmune disorders.
  • More particularly, the invention provides cells that are treated with oligonucleotides specifically designed to modulate expression of target genes involved in tumor immune resistance mechanisms.
  • BACKGROUND
  • Immunotherapy is the “treatment of disease by inducing, enhancing, or suppressing an immune response”. Immunotherapies designed to elicit or amplify an immune response are activation immunotherapies, while immunotherapies that reduce or suppress immune response are classified as suppression immunotherapies.
  • Immunotherapy of cancer has become increasingly important in clinical practice over recent decades. The primary approach in today's standard of care is passive immunotherapy through the use of recombinant monoclonal antibodies (mAbs). MAbs act through a mechanisms relevant to the body's own humoral immune response, by binding to key antigens involved in the tumor development and causing moderate forms of cell-mediated immunity, such as antibody-dependent cell-mediated cytotoxicity (ADCC).
  • Another group of emerging immunotherapeutic approaches is based on the administration of cells capable of destroying tumor cells. The administered cells may be the patient's own tumor-infiltrating lymphocytes (TIL), isolated and expanded ex-vivo. In some cases, TIL are capable of recognizing a variety of tumor associated antigens (TAA), while in other cases TIL can be reactivated and expanded in vitro to recognize specific antigens. The TIL-based therapeutic approaches are commonly referred to as “adoptive cell transfer” (ACT).
  • Further developments of ACT involve genetic modifications of T-cells to express receptors that recognize specific tumor-associated antigens (TAA). Such modifications may induce the expression of a specific T-cell receptor (TCR) or of a chimeric antigen receptor (CAR) consisting of TAA-specific antibody fused to CD3/co-stimulatory molecule transmembrane and cytoplasmic domains.
  • The ACT methods may also be considered as passive immunotherapeutic approaches in that they act directly on the tumor cells without invoking an extended immune response. However, unlike mAbs, ACT agents are capable of fully destroying the tumor cells, as opposed to the blockade of selected receptors and moderate cellular responses such as ADCC.
  • There is ongoing development of numerous methods of active immunotherapy, which restore the ability of body's own immune system to generate antitumor response. Active immunotherapeutic agents are often called therapeutic cancer vaccines, or just cancer vaccines. Many cancer vaccines are currently in clinical trials, and sipuleucell-T has recently become the first such vaccine approved by the United States FDA.
  • There are several classes of cancer vaccines using different antigens and different mechanisms of generating cell-mediated immune response. One class of vaccines is based on peptide fragments of antigens selectively expressed by tumor cells. The peptides are administered alone or in combination with immune-stimulatory agents, which may include adjuvants and cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF).
  • Another class of cancer vaccines is based on modified (e.g. sub-lethally irradiated) tumor cells used as antigens, also in combinations with immunostimulatory agents. Vaccines of this type currently in clinical trials are based both on autologous (e.g. OncoVAX, LipoNova) and allogeneic (e.g. Canvaxin, Onyvax-P, GVAX) tumor cell lines.
  • Yet another class of cancer vaccines uses dendritic cells. By their nature, dendritic cells (DC) are “professional” antigen-presenting cells capable of generating of a strong antigen-dependent cell-mediated immune response and eliciting therapeutic T-cells in vivo. DC-based cancer vaccines usually comprise DCs isolated from patients or generated ex vivo by culturing patient's hematopoietic progenitor cells or monocytes. DCs are further loaded with tumor antigens and sometimes combined with immune-stimulating agents, such as GM-CSF. A large number of DC-vaccines are now in clinical trials, and the first FDA-approved vaccine sipuleucell-T is based on DC.
  • Mechanisms of Immunosuppression and Therapeutic Approaches to its Mitigation
  • One of the key physiologic functions of the immune system is to recognize and eliminate neoplastic cells, therefore an essential part of any tumor progression is the development of immune resistance mechanisms. Once developed, these mechanisms not only prevent the natural immune system from effecting the tumor growth, but also limit the efficacy of any immunotherapeutic approaches to cancer. An important immune resistance mechanism involves immune-inhibitory pathways, sometimes referred to as immune checkpoints. The immune-inhibitory pathways play particularly important role in the interaction between tumor cells and CD8+ cytotoxic T-lymphocytes, including ACT therapeutic agents. Among important immune checkpoints are inhibitory receptors expressed on the T-cell surface, such as CTLA-4, PD1 and LAG3, among others.
  • The importance of the attenuation of immune checkpoints has been recognized by the scientific and medical community. One way to mitigate immunosuppression is to block the immune checkpoints by specially designed agents. The CTLA-4-blocking-antibody, ipilimumab, has recently been approved by the FDA. Several molecules blocking PD1 are currently in clinical development.
  • Immunosuppression mechanisms also negatively affect the function of dendritic cells and, as a consequence, the efficacy of DC-based cancer vaccines. Immunosuppressive mechanisms can inhibit the ability of DC to present tumor antigens through the MHC class I pathway and to prime naïve CD8+ T-cells for antitumor immunity. Among the important molecules responsible for the immunosuppression mechanisms in DC are ubiquitin ligase A20 and the broadly immune-suppressive protein SOCS1.
  • The efficacy of immunotherapeutic approaches to cancer can be augmented by combining them with inhibitors of immune checkpoints. Numerous ongoing preclinical and clinical studies are exploring potential synergies between cancer vaccines and other immunotherapeutic agents and checkpoint blocking agents, for example, ipilimumab. Such combination approaches have the potential to result in significantly improved clinical outcomes.
  • However, there are a number of drawbacks of using cancer immunotherapeutic agents in combination with checkpoint inhibitors. For example, immune checkpoint blockade can lead to the breaking of immune self-tolerance, thereby inducing a novel syndrome of autoimmune/auto-inflammatory side effects, designated “immune related adverse events,” mainly including rash, colitis, hepatitis and endocrinopathies (Corsello, et al. J. Clin. Endocrinol. Metab., 2013, 98:1361).
  • Reported toxicity profiles of checkpoint inhibitors are different than the toxicity profiles reported for other classes of oncologic agents. Those involve inflammatory events in multiple organ systems, including skin, gastrointestinal, endocrine, pulmonary, hepatic, ocular, and nervous system. (Hodi, 2013, Annals of Oncology, 24: Suppl, i7).
  • In view of the above, there is a need for new cancer therapeutic agents that can be used in combination with checkpoint inhibitors as well as other classes of oncolytic agents without risk of adverse inflammatory events in multiple organ systems previously reported for checkpoint inhibitors. The immunotherapeutic cells of the invention, prepared by treating cells with a combination oligonucleotide agents targeting genes associated with tumor or infections disease resistance mechanisms, as well as methods of producing such therapeutic cells and methods of treating disease with the produced therapeutic cells, satisfy this long felt need.
  • SUMMARY OF EMBODIMENTS OF THE INVENTION
  • The efficacy of immunotherapeutic approaches to cell proliferation disorders and infectious diseases can be augmented by combining them with inhibitors of immune checkpoints. Numerous synergies between cancer vaccines and other immunotherapeutic agents and checkpoint blocking agents provide opportunities for combination approaches that may significantly improve clinical outcomes for example, in proliferative cell disorders and immune diseases.
  • Various embodiments of the inventions disclosed herein include compositions comprising therapeutic cells obtained by treating cells ex vivo with oligonucleotides to modulate expression of target genes involved in immune suppression mechanisms. The oligonucleotide agent may be an antisense oliogonucleotide (ASO), including locked nucleic acids (LNAs), methoxyethyl gapmers, and the like, or an siRNA, miRNA, miRNA-inhibitor, morpholino, PNA, and the like. The oligonucleotide is preferably a self-delivered (sd) RNAi agent. The oligonucleotides may be chemically modified, for example, including at least one 2-O-methyl modification, 2′-Fluro modification, and/or phosphorothioate modification. The oligonucleotides may include one or more hydrophobic modification, for example, one or more sterol, cholesterol, vitamin D, Naphtyl, isobutyl, benzyl, indol, tryptophane, or phenyl hydrophobic modification. The oligonucleotide may be a hydrophobically-modified siRNA-antisense hybrid. The oligonucleotides may be used in combination with transmembrane delivery systems, such as delivery systems comprising lipids.
  • In an embodiment, the cells are obtained and/or derived from a cancer or infectious disease patient, and may be, for example, tumor infiltrating lymphocytes (TIL) and/or T-cells, antigen presenting cells such as dendritic cells, natural killer cells, induced-pluripotent stem cells, stem central memory T-cells, and the like. The T-cells and NK-cells are preferably genetically engineered to express high-affinity T-Cell receptors (TCR) and/or chimeric antibody or antibody-fragment—T-Cell receptors (CAR). In an embodiment, the chimeric antibody/antibody fragment is preferably capable of binding to antigens expressed on tumor cells. Immune cells may be engineered by transfection with plasmid, viral delivery vehicles, or mRNAs.
  • In an embodiment, the chimeric antibody or fragment is capable of binding CD19 receptors of B-cells and/or binding to antigens expressed on tumors, such as melanoma tumors. Such melanoma-expressed antigens include, for example, GD2, GD3, HMW-MAA, VEGF-R2, and the like.
  • Target genes identified herein for modification include: cytotoxic T-cell antigen 4 (CTLA4), programmed cell death protein 1 (PD1), tumor growth factor receptor beta (TGFR-beta), LAG3, TIM3, and adenosine A2a receptor; anti-apoptotic genes including, but not limited to: BAX, BAC, Casp8, and P53; A20 ubiquitine ligase (TNFAIP3, SOCS1 (suppressor of cytokine signaling), IDO (indolamine-2,3-dioxygenase; tryptophan-degrading enzyme), PD-L1 (CD274)(surface receptor, binder to PD1 on Tcells), Notch ligand Deltal (DLL1), Jagged 1, Jagged 2, FasL (pro-apoptotic surface molecule), CCL17, CCL22 (secreted chemokines that attract Treg cells), IL10 receptor (IL10RA), p38 (MAPK14), STAT3, TNFSF4 (OX40L), MicroRNA miR-155, miR-146a, anti-apoptotic genes including but not limited to BAX, BAC, Casp8 and P53, and the like genes, and combinations thereof. Representative target sequences are listed in Table 1.
  • The engineered therapeutic cells are treated with RNAi agents designed to inhibit expression of one or more of the targeted genes. The RNAi agent may comprise a guide sequence that hybridizes to a target gene and inhibits expression of the target gene through an RNA interference mechanism, where the target region is selected from the group listed in Table 1. The RNA agent can be chemically modified, and preferably includes at least one 2′-O-methyl, 2′-O-Fluoro, and/or phosphorothioate modification, as well as at least one hydrophobic modification such as cholesterol, and the like.
  • The immunogenic compositions described herein are useful for the treatment of proliferative disorders, including cancers, and/or infectious disease and are produced by the ex-vivo treatment of cells with oligonucleotides to modulate the expression of target genes involved in tumor immune resistance mechanisms. The ex vivo treatment of cells includes administering to the cells an oligonucleotide capable of targeting and inhibiting expression of a gene involved in a tumor suppressor mechanism, such as the genes listed in Table 1. The oligonucleotide can be used in combination with a transmembrane delivery system that may comprise one or more of: lipid(s) and vector, such as a viral vector.
  • The invention includes a method of treating a cell proliferative disorder or infectious disease by administering to a subject in need thereof, an immunogenic composition comprising cells that have been treated with one or more oligonucleotide to modulate the expression of one or more target gene involved in tumor immune resistance mechanisms, for example, one or more of the target genes of Table 1.
  • The invention preferably includes immunogenic cells treated with a plurality of oligonucleotide agents targeting a combination of target genes described herein. The combination may target a plurality of suppressor receptor genes, cytokine receptor genes, regulatory genes, and/or apoptotic factors in order to inhibit tumor immune resistance mechanisms.
  • The present invention is directed to novel immunotherapeutic cells, methods of generating the immunotherapeutic cells, and therapeutic methods employing such cells.
  • A new method of immune checkpoint inhibition is described herein, applicable to a broad variety of cell-based immunotherapies, including, but not limited to adaptive cell transfer, for example, based on TIL, TCR, CAR, and other cell types, as well as dendritic cell-based cancer vaccines. Self-deliverable RNAi technology provides efficient transfection of short oligonucleotides in any cell type, including immune cells, providing increased efficacy of immunotherapeutic treatments. In addition, the activated immune cells can be protected by preventing apoptosis via inhibition of key activators of the apoptotic pathway, such as BAC, BAX, Casp8, and P53, among others.
  • The activated immune cells modified by oligonucleotide transfer for a single therapeutic agent for administration to a subject, providing a number of advantages as compared to separately administered combinations of vaccines and immunotherapeutics and separately administered checkpoint inhibitors. These advantages include lack of side effects associated with the checkpoint inhibitors in a single therapeutic agent (activated immune cells modified by oligonucleotides targeting immune resistance genes).
  • The claimed immunotherapeutic cells, method of producing immunotherapeutic cells by introduction of oligonucleotide molecules targeting immune resistance pathways, and methods of treating proliferative and infectious disease, improves upon any known immunotherapeutic cells and methods of producing immunotherapeutic cells because it provides:
      • 1) a single therapeutic composition providing a combination of checkpoint inhibitors and other immune resistance mechanism inhibitors;
      • 2) with reduced toxicity; and
      • 3) increased efficacy as compared with other compositions.
    BRIEF DESCRIPTION OF THE FIGURES
  • The foregoing features of embodiments will be more readily understood by reference to the following detailed description, taken with reference to the accompanying drawings, in which:
  • FIG. 1 is a schematic diagram showing the structure of an sdRNA molecule.
  • FIG. 2 is a graph showing sdRNA-induced silencing of GAPDH and MAP4K4 in HeLa cells.
  • FIG. 3 is a graph showing sdRNA-induced knock-down of multiple targets using sdRNA agents directed to three genes in NK-92 cells.
  • FIG. 4 is a graph showing the knock-down of gene expression in Human Primary T cells by sdRNA agents targeting TP53 and MAP4K4.
  • FIG. 5 is a graph showing sdRNA-induced knock-down of CTLA4 and PD1 in Human Primary T cells.
  • FIG. 6 is a graph showing the reduction of PDCD1 and CTLA-4 surface expression by sdRNA in Human Primary T cells.
  • FIG. 7 is a graph showing MAP4K4-cy3 sdRNA delivery into T and B cells in human PBMCs.
  • DETAILED DESCRIPTION OF SPECIFIC EMBODIMENTS
  • The invention is defined by the claims, and includes oligonucleotides specifically designed and selected to reduce and/or inhibit expression of suppressors of immune resistance (inhibitory oligonucleotides), compositions comprising cells modified by treatment with such inhibitory oligonucleotides, methods of making such compositions, and methods of using the compositions to treat proliferation and/or infectious diseases. In particular, cells are treated with a combination of oligonucleotide agents, each agent particularly designed to interfere with and reduce the activity of a targeted immune suppressor.
  • Preferably, the combination of oligonucleotide agents targets multiple immune suppressor genes selected from checkpoint inhibitor genes such as CTLA4, PD-1/PD-1L, BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3, B7-H4 receptors, and TGF beta type 2 receptor; cytokine receptors that inactivate immune cells, such as TGF-beta receptor A and IL-10 receptor; regulatory genes/transcription factors modulating cytokine production by immune cells, such as STAT-3 and P38, miR-155, miR-146a; and apoptotic factors involved in cascades leading to cell death, such as p53 and Cacp8.
  • Most preferably the oligonucleotide agent is a self-deliverable RNAi agent, which is a hydrophobically modified siRNA-antisense hybrid molecule, comprising a double-stranded region of about 13-22 base pairs, with or without a 3′-overhang on each of the sense and antisense strands, and a 3′ single-stranded tail on the antisense strand of about 2-9 nucleotides. The oligonucleotide contains at least one 2′-O-Methyl modification, at least one 2′-O-Fluoro modification, and at least one phosphorothioate modification, as well as at least one hydrophobic modification selected from sterol, cholesterol, vitamin D, napthyl, isobutyl, benzyl, indol, tryptophane, phenyl, and the like hydrophobic modifiers (see FIG. 1). The oligonucleotide may contain a plurality of such modifications.
  • Definitions
  • As used in this description and the accompanying claims, the following terms shall have the meanings indicated, unless the context requires otherwise:
  • Proliferative disease, as used herein, includes diseases and disorders characterized by excessive proliferation of cells and turnover of cellular matrix, including cancer, atherlorosclerosis, rheumatoid arthritis, psoriasis, idiopathic pulmonary fibrosis, scleroderma, cirrhosis of the liver, and the like. Cancers include but are not limited to, one or more of: small cell lung cancer, colon cancer, breast cancer, lung cancer, prostate cancer, ovarian cancer, pancreatic cancer, melanoma, hematological malignancy such as chronic myeloid leukemia, and the like cancers where immunotherapeutic intervention to suppress tumor related immune resistance is needed.
  • Immune target genes can be grouped into at least four general categories: (1) checkpoint inhibitors; (2) cytokine receptors that inactivate immune cells, (3) anti-apoptotic genes; and (4) regulator genes, for example, transcription factors.
  • Immune Checkpoint inhibitors (ICI), as used herein, include immunotherapeutic agents that bind to certain checkpoint proteins, such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1) and its ligand PD-L1 to block and disable inhibitory proteins that prevent the immune system from attacking diseased cells such as cancer cells, liberating tumor-specific T cells to exert their effector function against tumor cells.
  • Tumor related immune resistance genes, as used herein, include genes involved in checkpoint inhibition of immune response, such as CTLA-4 and PD-1/PD-L1; TGF-beta, LAG3, Tim3, adenosine A2a receptor;
  • Regulator genes, as used herein, include transcription factors and the like that modulate cytokine production by immune cells, and include p38, STAT3, microRNAs miR-155, miR-146a;
  • Anti-apoptotic genes, as used herein, include BAX, BAC, Casp8, P53 and the like; and combinations thereof.
  • Infectious diseases, as used herein, include, but are not limited to, diseases caused by pathogenic microorganisms, including, but not limited to, one or more of bacteria, viruses, parasites, or fungi, where immunotherapeutic intervention to suppress pathogen related immune resistance and/or overactive immune response.
  • Immunogenic composition, as used herein, includes cells treated with one or more oligonucleotide agent, wherein the cells comprise T-cells. The T-cells may be genetically engineered, for example, to express high affinity T-cell receptors (TCR), chimeric antibody—T-cell receptors (CAR), where the chimeric antibody fragments are capable of binding to CD19 receptors of B-cells and/or to antigens expressed on tumor cells. In one embodiment, the chimeric antibody fragments bind antigens expressed on melanoma tumors, selected from GD2, GD3, HMW-MAA, and VEGF-R2.
  • Immunogenic compositions described herein include cells comprising antigen-presenting cells, dendritic cells, engineered T-cells, natural killer cells, stem cells, including induced pluripotent stem cells, and stem central memory T-cells. The treated cell also comprises one or a plurality of oligonucleotide agents, preferably sdRNAi agents specifically targeting a gene involved in an immune suppression mechanism, where the oligonucleotide agent inhibits expression of said target gene.
  • In one embodiment, the target gene is selected from A20 ubiquitin ligase such as TNFAIP3, SOCS1 (suppressor of cytokine signaling), Tyro3/Axl/Mer (suppressors of TLR signaling), IDO (indolamine-2,3-dioxygenase, tryptophan-degrading enzyme), PD-L1/CD274 (surface receptor, binds PD1 on T-cells), Notch ligand Delta (DLL1), Jagged 1, Jagged 2, FasL (pro-apoptotic surface molecule), CCL17, CCL22 (secreted chemokines that attract Treg cells), IL-10 receptor (IL10Ra), p38 (MAPK14), STAT3, TNFSF4 (OX40L), microRNA miR-155, miR-146a, anti-apoptotic genes, including but not limited to BAX, BAC, Casp8, and P53; and combinations thereof.
  • Particularly preferred target genes are those shown in Table 1.
  • Ex-vivo treatment, as used herein, includes cells treated with oligonucleotide agents that modulate expression of target genes involved in immune suppression mechanisms. The oligonucleotide agent may be an antisense oligonucleotide, including, for example, locked nucleotide analogs, methyoxyethyl gapmers, cyclo-ethyl-B nucleic acids, siRNAs, miRNAs, miRNA inhibitors, morpholinos, PNAs, and the like. Preferably, the oligonucleotide agent is an sdRNAi agent targeting a gene involved in an immune suppression mechanism. The cells treated in vitro by the oligonucleotide agent may be immune cells expanded in vitro, and can be cells obtained from a subject having a proliferative or infectious disease. Alternatively, the cells or tissue may be treated in vivo, for example by in situ injection and/or intravenous injection.
  • Oligonucleotide or oligonucleotide agent, as used herein, refers to a molecule containing a plurality of “nucleotides” including deoxyribonucleotides, ribonucleotides, or modified nucleotides, and polymers thereof in single- or double-stranded form. The term encompasses nucleotides containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • Nucleotide, as used herein to include those with natural bases (standard), and modified bases well known in the art. The nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see, for example, PCT Publications No. WO 92/07065 and WO 93/15187. Non-limiting examples of base modifications that can be introduced into nucleic acid molecules include, hypoxanthine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2,4,6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g. 6-methyluridine and pseudouridine), propyne, and others. The phrase “modified bases” includes nucleotide bases other than adenine, guanine, cytosine, and uracil, modified for example, at the 1′ position or their equivalents.
  • As used herein, the term “deoxyribonucleotide” encompasses natural and synthetic, unmodified and modified deoxyribonucleotides. Modifications include changes to the sugar moiety, to the base moiety and/or to the linkages between deoxyribonucleotide in the oligonucleotide.
  • As used herein, the term “RNA” defines a molecule comprising at least one ribonucleotide residue. The term “ribonucleotide” defines a nucleotide with a hydroxyl group at the 2′ position of a □-D-ribofuranose moiety. The term RNA includes double-stranded RNA, single-stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, synthetic RNA, recombinantly produced RNA, as well as altered RNA that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Nucleotides of the RNA molecules described herein may also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally-occurring RNA.
  • As used herein, “modified nucleotide” refers to a nucleotide that has one or more modifications to the nucleoside, the nucleobase, pentose ring, or phosphate group. For example, modified nucleotides exclude ribonucleotides containing adenosine monophosphate, guanosine monophosphate, uridine monophosphate, and cytidine monophosphate and deoxyribonucleotides containing deoxyadenosine monophosphate, deoxyguanosine monophosphate, deoxythymidine monophosphate, and deoxycytidine monophosphate. Modifications include those naturally-occurring that result from modification by enzymes that modify nucleotides, such as methyltransferases.
  • Modified nucleotides also include synthetic or non-naturally occurring nucleotides. Synthetic or non-naturally occurring modifications in nucleotides include those with 2′ modifications, e.g., 2′-O-methyl, 2′-methoxyethoxy, 2′-fluoro, 2′-allyl, 2′-O-[2-(methylamino)-2-oxoethyl], 4′-thio, 4′-CH2-O-2′-bridge, 4′-(CH2) 2-O-2′-bridge, 2′-LNA, and 2′-O-(N-methylcarbamate) or those comprising base analogs. In connection with 2′-modified nucleotides as described for the present disclosure, by “amino” is meant 2′-NH2 or 2′-O—NH2, which can be modified or unmodified. Such modified groups are described, for example, in U.S. Pat. Nos. 5,672,695 and 6,248,878.
  • As used herein, “microRNA” or “miRNA” refers to a nucleic acid that forms a single-stranded RNA, which single-stranded RNA has the ability to alter the expression (reduce or inhibit expression; modulate expression; directly or indirectly enhance expression) of a gene or target gene when the miRNA is expressed in the same cell as the gene or target gene. In one embodiment, a miRNA refers to a nucleic acid that has substantial or complete identity to a target gene and forms a single-stranded miRNA. In some embodiments miRNA may be in the form of pre-miRNA, wherein the pre-miRNA is double-stranded RNA. The sequence of the miRNA can correspond to the full length target gene, or a subsequence thereof. Typically, the miRNA is at least about 15-50 nucleotides in length (e.g., each sequence of the single-stranded miRNA is 15-50 nucleotides in length, and the double stranded pre-miRNA is about 15-50 base pairs in length). In some embodiments the miRNA is 20-30 base nucleotides. In some embodiments the miRNA is 20-25 nucleotides in length. In some embodiments the miRNA is 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • Target gene, as used herein, includes genes known or identified as modulating the expression of a gene involved in an immune resistance mechanism, and can be one of several groups of genes, such as suppressor receptors, for example, CTLA4 and PD1; cytokine receptors that inactivate immune cells, for example, TGF-beta receptor, LAG3, Tim3, adenosine A2a receptor, and IL10 receptor; regulatory genes for example, STAT3, p38, mir155 and mir146a; and apoptosis factors involved in cascades leading to cell death, for example, P53, Casp8, BAX, BAC, and combinations thereof. See also preferred target genes listed in Table 1.
  • As used herein, small interfering RNA (siRNA), sometimes known as short interfering RNA or silencing RNA, defines a group of double-stranded RNA molecules, comprising sense and antisense RNA strands, each generally of about 1022 nucletides in length, optionally including a 3′ overhang of 1-3 nucleotides. siRNA is active in the RNA interference (RNAi) pathway, and interferes with expression of specific target genes with complementary nucleotide sequences.
  • As used herein, sdRNA refers to “self-deliverable” RNAi agents, that are formed as an asymmetric double-stranded RNA-antisense oligonucleotide hybrid. The double stranded RNA includes a guide (sense) strand of about 19-25 nucleotides and a passenger (antisense) strand of about 10-19 nucleotides with a duplex formation that results in a single-stranded phosphorothiolated tail of about 5-9 nucleotides.
  • The RNA sequences may be modified with stabilizing and hydrophobic modifications such as sterols, for example, cholesterol, vitamin D, naphtyl, isobutyl, benzyl, indol, tryptophane, and phenyl, which confer stability and efficient cellular uptake in the absence of any transfection reagent or formulation. Immune response assays testing for IFN-induced proteins indicate sdRNAs produce a reduced immunostimulatory profile as compared other RNAi agents. See, for example, Byrne et al., December 2013, J. Ocular Pharmacology and Therapeutics, 29(10): 855-864.
  • Cell-Based Immunotherapeutics
  • In general, cells are obtained from subjects with proliferative disease such as cancer, or an infectious disease such as viral infection. The obtained cells are treated directly as obtained or may be expanded in cell culture prior to treatment with oligonucleotides. The cells may also be genetically modified to express receptors that recognize specific antigens expressed on the tumor cell surface (CAR) or intracellular tumor antigens presented on MHC class I (TCR).
  • Oligonucleotide Agents
  • Antisense Oligonucleotides
  • Small interfering RNA (siRNA), sometimes known as short interfering RNA or silencing RNA, is a double stranded RNA molecule, generally 19-25 base pairs in length. siRNA is used in RNA interference (RNAi), where it interferes with expression of specific genes with complementary nucleotide sequences.
  • Double stranded DNA (dsRNA) can be generally used to define any molecule comprising a pair of complementary strands of RNA, generally a sense (passenger) and antisense (guide) strands, and may include single-stranded overhang regions. The term dsRNA, contrasted with siRNA, generally refers to a precursor molecule that includes the sequence of an siRNA molecule which is released from the larger dsRNA molecule by the action of cleavage enzyme systems, including Dicer.
  • sdRNA (self-deliverable) are a new class of covalently modified RNAi compounds that do not require a delivery vehicle to enter cells and have improved pharmacology compared to traditional siRNAs. “Self-deliverable RNA” or sdRNA is a hydrophobically modified RNA interfering-antisense hybrid, demonstrated to be highly efficacious in vitro in primary cells and in vivo upon local administration. Robust uptake and/or silencing without toxicity has been demonstrated in several tissues including dermal, muscle, tumors, alveolar macrophages, spinal cord, and retina cells and tissues. In dermal layer and retina, intradermal and intra-vitreal injection of sdRNA at mg doses induced potent and long lasting silencing.
  • While sdRNA is a superior functional genomics tool, enabling RNAi in primary cells and in vivo, it has a relatively low hit rate as compared to conventional siRNAs. While the need to screen large number of sequences per gene is not a limiting factor for therapeutic applications, it severely limits the applicability of sdRNA technology to functional genomics, where cost effective compound selection against thousands of genes is required. To optimize sdRNA structure, chemistry, targeting position, sequence preferences, and the like, a proprietary algorithm has been developed and utilized for sdRNA potency prediction. Availability of sdRNA reagents that are active in all cell types ex vivo and in vivo enables functional genomics and target stratification/validation studies.
  • Proprietary Algorithm
  • SdRNA sequences were selected based on a proprietory selection algorithm, designed on the basis of a functional screen of over 500 sdRNA sequences in the luciferase reporter assay of HeLa cells. Regression analysis of was used to establish a correlation between the frequency of occurrence of specific nucleotide and modification at any specific position in sdRNA duplex and its functionality in gene suppression assay. This algorithm allows prediction of functional sdRNA sequences, defined as having over 70% knockdown at 1 μM concentration, with a probability over 40%.
  • Table 1 shows predictive gene targets identified using the proprietary algorithm and useful in the cellular immunotherapeutic compositions and methods described herein.
  • Delivery of RNAi Agents
  • BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; Applic BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; ation of RNAi technology to functional genomics studies in prim BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; ary cells and in vivo is limited by requirements to formulate siRNAs into lipids or use of other cell delivery techniques. To circumvent delivery problems, the self-deliverable RNAi technology provides a method of directly transfecting cells with the RNAi agent, without the need for additional formulations or techniques. The ability to transfect hard-to-transfect cell lines, high in vivo activity, and simplicity of use, are characteristics of the compositions and methods that present significant functional advantages over traditional siRNA-based techniques. The sdRNAi technology allows direct delivery of chemically synthesized compounds to a wide range of primary cells and tissues, both ex-vivo and in vivo.
  • To enable BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; self-delivery, traditional siRNA molecules require a substantial reduction in size and the introduction of extensive chemical modifications which are not well tolerated by RNAi machinery, resulting in extremely low probability of finding active molecules (low hit rate). In contrast, the sdRNA technology allows efficient RNAi delivery to primary cells and tissues in vitro and in vivo, with demonstrated silencing efficiency in humans.
  • The general structure of sdRNA molecules is shown in FIG. 1. sdRNA are formed as hydrophobically-modified siRNA-antisense oligonucleotide hybrid structures, and are disclosed, for example in Byrne et al., December 2013, J. Ocular Pharmacology and Therapeutics, 29(10): 855-864.
  • Oligonucleotide Modifications: 2′-O-Methyl, 2′-O-Fluro, Phosphorothioate
  • The oligonucleotide agents preferably comprise one or more modification to increase stability and/or effectiveness of the therapeutic agent, and to effect efficient delivery of the oligonucleotide to the cells or tissue to be treated. Such modifications include at least one
  • BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; 2′-O-methyl modification, at least one 2′-O-Fluro modification, and at least one diphosphorothioate modification. Additionally, the oligonucleotide is modified to include one or more hydrophobic modification selected from sterol, cholesterol, vitamin D, naphtyl, isobutyl, benzyl, indol, tryptophane, and phenyl. The hydrophobic modification is preferably a sterol.
  • Delivery of Oligonucleotide Agents to Cells
  • The oligonucleotides may be delivered to the cells in combination with a transmembrane delivery system, preferably comprising lipids, viral vectors, and the like. Most preferably, the oligonucleotide agent is a self-delivery RNAi agent, that does not require any delivery agents.
  • Combination Therapy
  • Most preferred for this invention, e.g. particular combinations of elements and/or alternatives for specific needs. This objective is accomplished by determining the appropriate genes to be targeted by the oligonucleotide in order to silence immune suppressor genes and using the proprietary algorithm to select the most appropriate target sequence.
  • It is preferred that the immunotherapeutic cell be modified to include multiple oligonucleotide agents targeting a variety of genes involved in immune suppression and appropriate for the selected target disease and genes. For example, a preferred immunotherapeutic cell is a T-Cell modified to knock-down both CTLA-4 and PD-1
  • Additional combinations of oligonucleotides to related genes involved in immune suppression include varied combinations of the selected target sequences of Table 1.
  • BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; Preferred BTLA (B and T-lymphocyte attenuator), KIR (killer immunoglobulin-like receptors), B7-H3 and B7-H4 receptors and TGFbeta type 2 receptor; therapeutic combinations include cells engineered to knock down gene expression of the following target genes:
      • a) CTLA4 and PD1
      • b) STAT3 and p38
      • c) PD1 and BaxPD1, CTLA4, Lag-1, ILM-3, and TP53
      • d) PD1 and Casp8
      • e) PD1 and IL10R
  • The therapeutic compositions described herein are useful to treat a subject suffering from a proliferation disorder or infectious disease. In particular, the immunotherapeutic composition is useful to treat disease characterized by suppression of the subjects immune mechanisms. The sdRNA agents described herein are specifically designed to target genes involved in diseases-associated immune suppression pathways.
  • Methods of treating a subject comprise administering to a subject in need thereof, an immunogenic composition comprising an sdRNAi agent capable of inhibiting expression of genes involved in immune suppression mechanisms, for example, any of the genes listed in Table 1 or otherwise described herein.
  • EXAMPLES
  • The embodiments of the invention described above are intended to be merely exemplary; numerous variations and modifications will be apparent to those skilled in the art. All such variations and modifications are intended to be within the scope of the present invention as defined in any appended claims.
  • Example 1 Self-Deliverable RNAi Immunotherapeutic Agents
  • Immunotherapeutic agents described herein were produced by treating cells with particular sdRNA agents designed to target and knock down specific genes involved in immune suppression mechanisms. In particular, the following cells and cell lines have been successfully treated with sdRNA and were shown to knock down at least 70% of targeted gene expression in the specified human cells.
  • These studies demonstrated utility of these immunogenic agents to suppress expression of target genes in cells normally very resistant to transfection, and suggests the agents are capable of reducing expression of target cells in any cell type.
  • TABLE 2
    Cell Type Target Gene sdRNA target sequence % Knock Down
    Primary human T-cells TP53 (P53) GAGTAGGACATACCAGCTTA (SEQ ID NO: 1001) >70% 2 uM
    Primary human T-cells MAP4K4 AGAGTTCTGTGGAAGTCTA (SEQ ID NO: 1002) >70% 2 uM
    Jurkat T-Iymphoma cells MAP4K4 AGAGTTCTGTGGAAGTCTA (SEQ ID NO: 1003) 100% 1 uM 72h
    NK-92 cells MAP4K4 AGAGTTCTGTGGAAGTCTA (SEQ ID NO: 1004)  80% 2 uM 72h
    NK-92 cells PPIB ACAGCAAATTCCATCGTGT (SEQ ID NO: 1005) >75% 2 uM 72h
    NK-92 cells GADPH CTGGTAAAGTGGATATTGTT (SEQ ID NO: 1006) >90% 2 uM 72h
    HeLa Cells MAP4K4 AGAGTTCTGTGGAAGTCTA (SEQ ID NO: 1007) >80% 2 uM 72h
  • Example 2 Oligonucleotide Sequences for Inhibiting Expression of Target Genes
  • A number of human genes were selected as candidate target genes due to involvement in immune suppression mechanisms, including the following genes shown in Table 3:
  • TABLE 3
    BAX (NM_004324) BAK1 (NM_001188) CASP8 (NM_001228)
    ADORA2A (NM_000675) CTLA4 (NM_005214) LAG3 (NM002286)
    PDCD1 (NM_NM005018) TGFBR1 (NM-004612) HAVCR2 (NM_032782)
    CCL17 (NM_002987) CCL22 (NM_002990) DLL2 (NM_005618)
    FASLG (NM_000639) CD274 (NM_001267706) IDO1 (NM_002164)
    IL10RA (NM_001558) JAG1 (NM_000214) JAG2 (NM_002226)
    MAPK14 (NM_001315) SOCS1 (NM_003745) STAT3 (NM_003150)
    TNFA1P3 (NM_006290) TNFSF4 (NM_003326) TYRO2 (NM_006293)
    TP53 (NM_000546)
  • Each of the genes listed above was analyzed using a proprietary algorithm to identify preferred sdRNA targeting sequences and target regions for each gene for prevention of immunosuppression of antigen-presenting cells and T-cells. Results are shown in Table 1.
  • Example 3 Knock-Down of Target Gene (GAPDH) by sdRNA in HeLa Cells
  • HeLa cells (ATCC CRM-CCL-2) were subcultured 24 hours before transfection and kept log phase. The efficacy of several GAPDH sdRNAs was tested by qRT-PCR, including G13 sdRNA listed in the Table 1.
  • Solutions of GAPDH, MAP4K4 (positive control) and NTC (non-targeting control) sdRNA with twice the required concentration were prepared in serum-free EMEM medium, by diluting 100 μM oligonucleotides to 0.2-4 μM.
  • The total volume of medium for each oligo concentration point was calculated as [50 μl/well]×[number of replicates for each serum point]. Oligonucleotides were dispensed into a 96 well plate at 50 μl/well.
  • Cells were collected for transfection by trypsinization in a 50 ml tube, washed twice with medium containing 10% FBS without antibiotics, spun down at 200×g for 5 minutes at room temperature and resuspended in EMEM medium containing twice the required amount of FBS for the experiment (6%) and without antibiotics. The concentration of the cells was adjusted to 120,000/ml to yield a final concentration of 6,000 cells/50 μl/well. The cells were dispensed at 50 μl/well into the 96-well plate with pre-diluted oligos and placed in the incubator for 48 hours.
  • Gene Expression Analysis in HeLa Cells Using qRT-PCR
  • RNA was isolated from transfected HeLa cells using the PureLink™ Pro96 total RNA purification Kit (Ambion, Cat. No. 12173-011A), with Quanta qScript XLT One-Step RT-qPCR ToughMix, ROX (VWR, 89236672). The isolated RNA was analyzed for gene expression using the Human MAP4K4-FAM (Taqman Hs0377405 ml) and Human GAPDH-VIC (Applied Biosystems, Cat. No. 4326317E) gene expression assays.
  • The incubated plate was spun down and washed once with 100 μl/well PBS and lysed with 60 μl/well buffer provided in the kit. RNA isolation was conducted according to the manufacturer's instructions, and the RNA was eluted with 100 μl RNase-free water, and used undiluted for one-step qRT-PCR.
  • Dilutions of non-transfected (NT) cells of 1:5 and 1:25 were prepared for the standard curve using RNase-free water. qRT-PCR was performed by dispensing 9 μl/well into a low profile PCR plate and adding 1 μl RNA/well from the earlier prepared RNA samples. After brief centrifugation, the samples were placed in the real-time cycler and amplified using the settings recommended by the manufacturer.
  • GAPDH gene expression was measured by qPCR, normalized to MAP4K4 and plotted as percent of expression in the presence of non-targeting sdRNA. The results were compared to the normalized according to the standard curve. As shown in FIG. 2, several sdRNA agents targeting GAPDH or MAP4K4 significantly reduced their mRNA levels leading to more than 80-90% knock-down with 1 μM sdRNA. (See FIG. 2).
  • Example 4 Silencing of Multiple Targets by sdRNA in NK-92 Cells
  • NK-92 cells were obtained from Conqwest and subjected to one-step RT-PCR analysis without RNA purification using the FastLane Cell Multiplex Kit (Qiagen, Cat. No. 216513). For transfection, NK-92 cells were collected by centrifugation and diluted with RPMI medium containing 4% FBS and IL2 1000 U/ml and adjusted to1,000,000 cells/ml.
  • Multiple sdRNA agents targeting MAP4K4, PPIB or GADPH were diluted separately in serum-free RPMI medium to 4 μM and individually aliquoted at 50 μl/well into a 96-well plate. The prepared cells were then added at 50 μl cells/well to the wells with either MAP4K4, PPIB or GAPDH sdRNAs. Cells were incubated for 24, 48, or 72 hours.
  • At the specified timepoints, the plated transfected cells were washed once with 100 μl/well PBS and once with FCW buffer. After removal of supernatant, cell processing mix of 23.5 μl FCPL and 1.5 μl gDNA wipeout solution was added to each well and incubated for five minutes at room temperature. Lysates were then transferred to PCR strips and heated at 75° C. for five minutes.
  • To setup qRT-PCR, the lysates were mixed with QuantiTect reagents from the FastLane Cell Multiplex Kit and with primer probe mix for MAP4K4-FAM/GAPDH-VIC or PPIB-FAM/GAPDH-VIC. The following Taqman gene expression assays were used: human MAP4K4-FAM (Taqman, Hs00377405_m1), human PPIB-FAM (Taqman, Hs00168719_m1) and human GAPDH-VIC (Applied Biosystems, cat. No 4326317E).
  • A volume of 9 μl/well of each reaction mix was dispensed into a low profile PCR plate. One μl lysate per well was added from the previously prepared lysates. The samples were amplified using the settings recommended by the manufacturer.
  • Results shown in FIG. 3 demonstrate significant silencing of each of the multiple targets, MAP4K4, PPIB, and GADPH by sdRNA agents transfected into NK-92 cells, including greater than 75% inhibition of expression of each target within 24 to 72 hours of incubation.
  • Example 5 Silencing of TP53 and MAP4K4 by sdRNA in Human Primary T-Cells
  • Primary human T-cells were obtained from AllCells (CA) and cultured in complete RPMI medium containing 1000 IU/ml IL2. Cells were activated with anti-CD3/CD28 Dynabeads (Gibco, 11131) according to the manufacturer's instructions for at least 4 days prior to the transfection. Cells were collected by brief vortexing to dislodge the beads from cells and separating them using the designated magnet.
  • sdRNA agents targeting TP53 or MAP4K4 were prepared by separately diluting the sdRNAs to 0.2-4 μM in serum-free RPMI per sample (well) and individually aliquoted at 100 μl/well of 96-well plate. Cells were prepared in RPMI medium containing 4% FBS and IL2 2000 U/ml at 1,000,000 cells/ml and seeded at 100 μl/well into the 96-well plate with pre-diluted sdRNAs.
  • At the end of the transfection incubation period, the plated transfected cells were washed once with 100 μl/well PBS and processed with FastLane Cell Multiplex Kit reagents essentially as described for the Example 4 and according to the manufacturer's instructions. Taqman gene expression assays were used in the following combinations: human MAP4K4-FAM/GAPDH-VIC or human TP53-FAM (Taqman, Hs01034249_m1)/GAPDH-VIC. A volume of 18 μl/well of each reaction mix was combined with 2 μl lysates per well from the previously prepared lysates. The samples were amplified as before (see Example 4).
  • Results shown in FIG. 4 demonstrate significant silencing of both MAP4K4 and TP53 by sdRNA agents transfected into T-cells, reaching 70-80% inhibition of gene expression with 1-2 μM sdRNA.
  • Example 6 Immunotherapeutic Combination of sdRNAs for Treating Melanoma
  • Melanomas utilize at least two particular pathways to suppress immune function of T-cells, and each involves both PD1 and CTLA4. Melanoma tumors expressing the PD1 ligand, PD1L, can be targeted with T-cells pretreated ex-vivo with sd-RNAi agents specifically designed to target PD1 and interfere with PD1 expression. PD1 is also known as PDCD1, and particular targeting sequences and gene regions identified and predicted to be particularly functional in sdRNA mediated suppression, are shown in Table 1 for PDCD1 (NM_005018) and for CTLA4 (NM005214).
  • Treatment of melanoma tumors can be effected by providing to melanoma cells T-cells, such as tumor-infiltrating lymphocytes, pretreated ex-vivo with a combination of sdRNAs targeting PD1/PDCD1 and CTLA4, for example, targeting one or more of the twenty target sequences listed for PD1/PDCD1 and/or CTLA4. A combination of sdRNAs targeting PD1/PDCD1 and FASLG (NM_000639) and/or CTLA4, can increase T-cell toxicity in tumors expressing both PD1L and FAS.
  • In addition to and in combination with anti-CTLA-4 and anti-PD1 sdRNAs, T-cells used for the immunotherapy of melanoma can also be treated with sdRNA targeting other genes implicated in immunosuppression by the tumor. These receptors include, but are not limited to TGF- beta type 1 and 2 receptors, BTLA (binder of herpes virus entry indicator (HVEM) expressed on melanoma cells), and receptors of integrins expressed by myeloid derived suppressor cells (MDSC), such as CD11b, CD18, and CD29.
  • For tumors whose profile of expressed suppressive proteins is unknown, any combination of sdRNAs targeting PD1/PDCD1 and any one of know suppressing receptors may be helpful to reduce immune suppression and increase therapeutic efficacy.
  • Example 7 Combination of sdRNAs for Mitigating Immune Cell Suppression
  • T-cell or dendritic cell suppression may be modulated by various cytokines, such as IL10 and/or TGF beta. Suppressing corresponding receptors in T-cells and dendritic cells may be beneficial for their activity. For example, providing a combination of anti-PD1 with anti-IL10R sdRNAs is exptected to mitigate cytokine induced suppression of T-cells and dendritic cells, as compared with anti-PD1 alone.
  • Example 8 Combination of sdRNAs for Mitigating Immune Cell Suppression
  • When the mechanism of tumor suppression of immune cells may be not known, use of sdRNA agents to suppress genes involved in apoptosis (programmed cell death), such as p53, Casp8 or other gene activating apoptosis may be beneficial to increase immune cell activity. Combination of an anti-receptor sdRNAs with sdRNAs against pro-apoptotic genes can additionally reduce death of immune cells and thus increase their activity. For example, combination of anti-PD1 with anti-p53 sdRNAs may additionally protect T-cells from suppression by blocking activation of apoptosis.
  • Example 9 Silencing of CTLA-4 and PDCD1 by sdRNA in Human Primary T-Cells
  • Primary human T-cells were cultured and activated essentially as described in Example 5. sdRNA agents targeting PDCD1 and CTLA-4 were prepared by separately diluting the sdRNAs to 0.4-4 μM in serum-free RPMI per sample (well) and aliquoted at 100 μl/well of 96-well plate. Cells were prepared in RPMI medium containing 4% FBS and IL2 2000 U/ml at 1,000,000 cells/ml and seeded at 100 μl/well into the 96-well plate with pre-diluted sdRNAs.
  • 72 h later, the transfected cells were washed once with 100 μl/well PBS and processed with FastLane Cell Multiplex Kit reagents essentially as described for the Example 4 and according to the manufacturer's instructions. Taqman gene expression assays were used in the following combinations: human PDCD1-FAM (Taqman, Hs01550088_m1)/GAPDH-VIC or human CTLA4-FAM (Taqman, Hs03044418_m1)/GAPDH-VIC. A volume of 18 μl/well of each reaction mix was combined with 2 μl lysates per well from the previously prepared lysates. The samples were amplified as before (see Example 4).
  • Results shown in FIG. 5 demonstrate significant silencing of PDCD1 and CTLA-4 by using combined sdRNA agents delivered to T-cells, obtaining greater than 60-70% inhibition of gene expression with 2 μM sdRNA.
  • Example 10 Reduction of CTLA-4 and PDCD1 Surface Expression by sdRNA in Human Primary T-Cells
  • Primary human T-cells were cultured and activated essentially as described in Example 5.
  • sdRNA agents targeting CTLA-4 or PD1 were separately diluted to 5 μM in serum-free RPMI per sample (well) and aliquoted at 250 μl/well to 24-well plates. Cells mixed with magnetic beads were collected and adjusted to 500,000 cells in 250 μl RPMI medium containing 4% FBS and IL2 2000 IU/ml. Cells were seeded at 250 μl/well to the prepared plate containing pre-diluted sdRNAs. 24 hours later FBS was added to the cells to obtain 10% final concentration.
  • After 72 hours of incubation, the transfected cells were collected, separated from the activation beads using the magnet, as described in Example 5. Cells were washed with PBS, spun down and resuspended in blocking buffer (PBS with 3% BSA) at 200,000 cells/50 μl/sample.
  • Antibody dilutions were prepared in the blocking buffer. The antibodies were mixed in two combinations: anti-PD1/anti-CD3 (1:100 dilutions for both antibodies) and anti-CTLA4/anti-CD3 (10 μl/106 cells for anti-CTLA4; 1:100 for CD3). The following antibodies were used: rabbit monoclonal [SP7] to CD3 (Abcam, ab16669); mouse monoclonal [BNI3] to CTLA4 (Abcam, ab33320) and mouse monoclonal [NAT105] to PD1 (Abcam, ab52587). Cells were mixed with the diluted antibodies and incubated 30 minutes on ice. Cells were then washed twice with PBS containing 0.2% Tween-20 and 0.1% sodium azide.
  • Secondary antibodies were diluted in blocking buffer and mixed together resulting in a final dilution 1:500 for anti-mouse Cy5 (Abcam, ab97037) and 1:2000 for anti-rabbit Alexa-488 (Abcam, ab150077). Cells were mixed with the diluted antibodies at 1:1 ratio and incubated 30 minutes on ice. Cells were washed as before, and diluted in 500 μl PBS per tube. The data was acquired immediately on the Attune Acoustic Focusing Cytometer (Applied Biosystems).
  • As shown in FIG. 6, sdRNA efficiently reduced surface expression of CTLA-4 and PD1 in activated Human Primary T cells.
  • Example 11 MAP4K4 sdRNA Delivery into CD3- and CD19-Positive Subsets of Human Peripheral Blood Mononuclear Cells (PBMCs)
  • PBMCs were cultured in complete RPMI supplemented with 1.5% PHA solution and 500 U/ml IL2. For transfection, PBMCs were collected by centrifugation and diluted with RPMI medium containing 4% FBS and IL2 1000 U/ml and seeded to 24-well plate at 500,000 cells/well.
  • MAP4K4 sdRNA labeled with cy3 was added to the cells at 0.1 μM final concentration. After 72 hours of incubation, the transfected cells were collected, washed with PBS, spun down and diluted in blocking buffer (PBS with 3% BSA) at 200,000 cells/50 μl/sample.
  • Antibody dilutions were prepared in the blocking buffer as following: 1:100 final dilution anti-CD3 (Abcam, ab16669) and anti-CD19 at 10 μl/1,000,000 cells (Abcam, ab31947). Cells were mixed with the diluted antibodies and incubated 30 min on ice. Cells were then washed twice with PBS containing 0.2% Tween-20 and 0.1% sodium azide.
  • Secondary antibodies were diluted in the blocking buffer in a final dilution 1:500 for anti-mouse Cy5 (Abcam, ab97037) and 1:2000 for anti-rabbit Alexa-488 (Abcam, ab150077). Cells were mixed with the diluted antibodies at 1:1 ratio and incubated 30 min on ice. Cells were washed as before, and diluted in 500 μl PBS per tube. The data was acquired immediately on the Attune Acoustic Focusing Cytometer (Applied Biosystems).
  • FIG. 7 shows efficient transfection over 97% of CD3-positive (t cells) and over 98% CD19-positive (B-cells) subsets in Human Peripheral Blood Mononuclear Cells (PBMCs).
  • TABLE 1
    Targeting sequences and gene regions of genes targeted with sdRNAs to prevent
    immunosuppression of antigen-presenting cells and T-cells.
    SEQ ID SEQ ID
    Oligo_count Oligo_ID targeting sequence NO: Gene_region NO:
    Accession: NM_004324
    HUGO gene symbol: BAX
    1 BAX_NM_004324_ GAATTGCTCAAGTTCATTGA 1 CCTCCACTGCCTCTGGAATTGCTCAAG 21
    human_835 TTCATTGATGACCCTCTG
    2 BAX_NM_004324_ TTCATCCAGGATCGAGCAGG 2 CTTTTGCTTCAGGGTTTCATCCAGGAT 22
    human_ 57 CGAGCAGGGCGAATGGGG
    3 BAX_NM_004324_ ATCATCAGATGTGGTCTATA 3 TCTCCCCATCTTCAGATCATCAGATGT 23
    human_684 GGTCTATAATGCGTTTTC
    4 BAX_NM_004324_ TACTTTGCCAGCAAACTGGT 4 GTTGTCGCCCTTTTCTACTTTGCCAGCA 24
    human_412 AACTGGTGCTCAAGGCC
    5 BAX_NM_004324_ GGTTGGGTGAGACTCCTCAA 5 ATCCAAGACCAGGGTGGTTGGGTGAG 25
    human_538 ACTCCTCAAGCCTCCTCAC
    6 BAX_NM_004324_ CTACTTTGCCAGCAAACTGG 6 GGTTGTCGCCCTTTTCTACTTTGCCAGC 26
    human_411 AAACTGGTGCTCAAGGC
    7 BAX_NM_004324_ GCGTTTTCCTTACGTGTCTG 7 GATGTGGTCTATAATGCGTTTTCCTTA 27
    human_706 CGTGTCTGATCAATCCCC
    8 BAX_NM_004324_ TACGTGTCTGATCAATCCCC 8 ATAATGCGTTTTCCTTACGTGTCTGATC 28
    human_7 16 AATCCCCGATTCATCTA
    9 BAX_NM_004324_ TCAGGGTTTCATCCAGGATC 9 AGGGGCCCTTTTGCTTCAGGGTTTCAT 29
    human_ 50 CCAGGATCGAGCAGGGCG
    10 BAX_NM_004324_ TGACGGCAACTTCAACTGGG 10 AGCTGACATGTTTTCTGACGGCAACTT 30
    human_ 72 CAACTGGGGCCGGGTTGT
    11 BAX_NM_004324_ CAGCTGACATGTTTTCTGAC 11 TCTTTTTCCGAGTGGCAGCTGACATGT 31
    human_356 TTTCTGACGGCAACTTCA
    12 BAX_NM_004324_ AGCTGACATGTTTTCTGACG 12 CTTTTTCCGAGTGGCAGCTGACATGTT 32
    human_357 TTCTGACGGCAACTTCAA
    13 BAX_NM_004324_ CACTGTGACCTTGACTTGAT 13 AGTGACCCCTGACCTCACTGTGACCTT 33
    human_776 GACTTGATTAGTGCCTTC
    14 BAX_NM_004324_ TCCTTACGTGTCTGATCAAT 14 GTCTATAATGCGTTTTCCTTACGTGTCT 34
    human_712 GATCAATCCCCGATTCA
    15 BAX_NM_004324_ GATCAGAACCATCATGGGCT 15 CAAGGTGCCGGAACTGATCAGAACCA 35
    human_465 TCATGGGCTGGACATTGGA
    16 BAX_NM_004324_ CTTCTGGAGCAGGTCACAGT 16 TCTGGGACCCTGGGCCTTCTGGAGCA 36
    human_642 GGTCACAGTGGTGCCCTCT
    17 BAX_NM_004324_ TGAGCAGATCATGAAGACAG 17 GGGGCCCACCAGCTCTGAGCAGATCA 37
    human_117 TGAAGACAGGGGCCCTTTT
    18 BAX_NM_004324_ TATAATGCGTTTTCCTTACG 18 TCATCAGATGTGGTCTATAATGCGTTT 38
    human_700 TCCTTACGTGTCTGATCA
    19 BAX_NM_004324_ CCCATCTTCAGATCATCAGA 19 CAGTGGTGCCCTCTCCCCATCTTCAGA 39
    human_673 TCATCAGATGTGGTCTAT
    20 BAX_NM_004324_ AGGTGCCGGAACTGATCAGA 20 AGGCCCTGTGCACCAAGGTGCCGGAA 40
    human_452 CTGATCAGAACCATCATGG
    Accession: NM_001188
    HUGO gene symbol: BAK1
    1 BAK1_NM_001188 TGGTTTGTTATATCAGGGAA 41 ACAGGGCTTAGGACTTGGTTTGTTA 61
    _human_1813 TATCAGGGAAAAGGAGTAGG
    2 BAK1_NM_001188 TGGTACGAAGATTCTTCAAA 42 TGTTGGGCCAGTTTGTGGTACGAAG 62
    _human_911 ATTCTTCAAATCATGACTCC
    3 BAK1_NM_001188 TTATATCAGGGAAAAGGAGT 43 TTAGGACTTGGTTTGTTATATCAGG 63
    _human_1820 GAAAAGGAGTAGGGAGTTCA
    4 BAK1_NM_001188 TCCCTTCCTCTCTCCTTATA 44 GTCCTCTCAGTTCTCTCCCTTCCTCTC 64
    _human_1678 TCCTTATAGACACTTGCT
    5 BAK1_NM_001188 TCAAATCATGACTCCCAAGG 45 TGGTACGAAGATTCTTCAAATCATG 65
    _human_926 ACTCCCAAGGGTGCCCTTTG
    6 BAK1_NM_001188 TGTTATATCAGGGAAAAGGA 46 GCTTAGGACTTGGTTTGTTATATCA 66
    _human_1818 GGGAAAAGGAGTAGGGAGTT
    7 BAK1_NM_001188 ACGAAGATTCTTCAAATCAT 47 GGGCCAGTTTGTGGTACGAAGATTC 67
    _human_915 TTCAAATCATGACTCCCAAG
    8 BAK1_NM_001188 GGTACGAAGATTCTTCAAAT 48 GTTGGGCCAGTTTGTGGTACGAAGA 68
    _human_912 TTCTTCAAATCATGACTCCC
    9 BAK1_NM_001188 GAAGTTCTTGATTCAGCCAA 49 GGGGGTCAGGGGGGAGAAGTTCTT 69
    _human_2086 GATTCAGCCAAATGCAGGGAG
    10 BAK1_NM_001188 CCTATGAGTACTTCACCAAG 50 CCACGGCAGAGAATGCCTATGAGTA 70
    _human_620 CTTCACCAAGATTGCCACCA
    11 BAK1_NM_001188 TATCAGGGAAAAGGAGTAGG 51 GGACTTGGTTTGTTATATCAGGGAA 71
    _human_1823 AAGGAGTAGGGAGTTCATCT
    12 BAK1_NM_001188 CTCTCCTTATAGACACTTGC 52 GTTCTCTCCCTTCCTCTCTCCTTATAG 72
    _human_1687 ACACTTGCTCCCAACCCA
    13 BAK1_NM_001188 ACTTGGTTTGTTATATCAGG 53 ACTACAGGGCTTAGGACTTGGTTTG 73
    _human_1810 TTATATCAGGGAAAAGGAGT
    14 BAK1_NM_001188 AAGATCAGCACCCTAAGAGA 54 ATTCAGCTATTCTGGAAGATCAGCA 74
    _human_1399 CCCTAAGAGATGGGACTAGG
    15 BAK1_NM_001188 GTTTGAGAGTGGCATCAATT 55 GATTGCCACCAGCCTGTTTGAGAGT 75
    _human_654 GGCATCAATTGGGGCCGTGT
    16 BAK1_NM_001188 GACTATCAACACCACTAGGA 56 TCTAAGTGGGAGAAGGACTATCAAC 76
    _human_1875 ACCACTAGGAATCCCAGAGG
    17 BAK1_NM_001188 AGCTTTAGCAAGTGTGCACT 57 CCTCAAGAGTACAGAAGCTTTAGCA 77
    _human_1043 AGTGTGCACTCCAGCTTCGG
    18 BAK1_NM_001188 TTCATCTGGAGGGTTCTAAG 58 AAAAGGAGTAGGGAGTTCATCTGG 78
    _human_1846 AGGGTTCTAAGTGGGAGAAGG
    19 BAK1_NM_001188 AAGTTCTTGATTCAGCCAAA 59 GGGGTCAGGGGGGAGAAGTTCTTG 79
    _human_2087 ATTCAGCCAAATGCAGGGAGG
    20 BAK1_NM_001188 GTTATATCAGGGAAAAGGAG 60 CTTAGGACTTGGTTTGTTATATCAG 80
    _human_1819 GGAAAAGGAGTAGGGAGTTC
    Accession: NM_001228
    HUGO gene symbol: CASP8
    1 CASP8_NM_001228 TTAAATCATTAGGAATTAAG 121 TCTGCTTGGATTATTTTAAATCATTAG 141
    _human_2821 GAATTAAGTTATCTTTAA
    2 CASP8_NM_001228 GAATTAAGTTATCTTTAAAA 122 ATTTTAAATCATTAGGAATTAAGTTAT 142
    _human_2833 CTTTAAAATTTAAGTATC
    3 CASP8_NM_001228 AACTTTAATTCTCTTTCAAA 123 TGTTAATATTCTATTAACTTTAATTCT 143
    _human_2392 CTTTCAAAGCTAAATTCC
    4 CASP8_NM_001228 GACTGAAGTGAACTATGAAG 124 TATTCTCACCATCCTGACTGAAGTGA 144
    _human_1683 ACTATGAAGTAAGCAACAA
    5 CASP8_NM_001228 ATATTCTCCTGCCTTTTAAA 125 GGGAATATTGAGATTATATTCTCCTG 145
    _human_281 CCTTTTAAAAAGATGGACT
    6 CASP8_NM_001228 AGTTATCTTTAAAATTTAAG 126 AATCATTAGGAATTAAGTTATCTTTA 146
    _human_2839 AAATTTAAGTATCTTTTTT
    7 CASP8_NM_001228 TAGATTTTCTACTTTATTAA 127 TATTTACTAATTTTCTAGATTTTCTACT 147
    _human_2164 TTATTAATTGTTTTGCA
    8 CASP8_NM_001228 CTGTGCCCAAATCAACAAGA 128 CATCCTGAAAAGAGTCTGTGCCCAAA 148
    _human_888 TCAACAAGAGCCTGCTGAA
    9 CASP8_NM_001228 AGCTGGTGGCAATAAATACC 129 TTTGGGAATGTTTTTAGCTGGTGGCA 149
    _human_2283 ATAAATACCAGACACGTAC
    10 CASP8_NM_001228 TCCTACCGAAACCCTGCAGA 130 GTGAATAACTGTGTTTCCTACCGAAA 150
    _human_1585 CCCTGCAGAGGGAACCTGG
    11 CASP8_NM_001228 TATAAGAGCTAAAGTTAAAT 131 TGTTTTGCACTTTTTTATAAGAGCTAA 151
    _human_2200 AGTTAAATAGGATATTAA
    12 CASP8_NM_001228 CACTATGTTTATTTACTAAT 132 ACTATTTAGATATAACACTATGTTTAT 152
    _human_2140 TTACTAATTTTCTAGATT
    13 CASP8_NM_001228 ATTGTTATCTATCAACTATA 133 GGGCTTATGATTCAGATTGTTATCTA 153
    _human_2350 TCAACTATAAGCCCACTGT
    14 CASP8_NM_001228 TAACTGTGTTTCCTACCGAA 134 GATGGCCACTGTGAATAACTGTGTTT 154
    _human_1575 CCTACCGAAACCCTGCAGA
    15 CASP8_NM_001228 TAATTCTCTTTCAAAGCTAA 135 ATATTCTATTAACTTTAATTCTCTTTCA 155
    _human_2397 AAGCTAAATTCCACACT
    16 CASP8_NM_001228 TATATGCTTGGCTAACTATA 136 TGCTTTTATGATATATATATGCTTGGC 156
    _human_2726 TAACTATATTTGCTTTTT
    17 CASP8_NM_001228 CTCTGCTTGGATTATTTTAA 137 CATTTGCTCTTTCATCTCTGCTTGGAT 157
    _human_2805 TATTTTAAATCATTAGGA
    18 CASP8_NM_001228 ATGCTTGGCTAACTATATTT 138 TTTTATGATATATATATGCTTGGCTAA 158
    _human_2729 CTATATTTGCTTTTTGCT
    19 CASP8_NM_001228 ATAAGAGCTAAAGTTAAATA 139 GTTTTGCACTTTTTTATAAGAGCTAAA 159
    _human_2201 GTTAAATAGGATATTAAC
    20 CASP8_NM_001228 ATCTTTAAAATTTAAGTATC 140 ATTAGGAATTAAGTTATCTTTAAAATT 160
    _human_2843 TAAGTATCTTTTTTCAAA
    Accession: NM_000675
    HUGO gene symbol: ADORA2A
    1 ADORA2A_NM_00067 TAACTGCCTTTCCTTCTAAA 161 GTGAGAGGCCTTGTCTAACTGCC 181
    5_human_2482 TTTCCTTCTAAAGGGAATGTTT
    2 ADORA2A_NM_00067 TTCCTTCTAAAGGGAATGTT 162 CTTGTCTAACTGCCTTTCCTTCTAA 182
    5_human_2491 AGGGAATGTTTTTTTCTGAG
    3 ADORA2A_NM_00067 GCCTTTCCTTCTAAAGGGAA 163 AGGCCTTGTCTAACTGCCTTTCCT 183
    5_human_2487 TCTAAAGGGAATGTTTTTTTC
    4 ADORA2A_NM_00067 TTTTCTGAGATAAAATAAAA 164 CTAAAGGGAATGTTTTTTTCTGAG 184
    5_human_2512 ATAAAATAAAAACGAGCCACA
    5 ADORA2A_NM_00067 CATCTCTTGGAGTGACAAAG 165 TCTCAGTCCCAGGGCCATCTCTTG 185
    5_human_2330 GAGTGACAAAGCTGGGATCAA
    6 ADORA2A_NM_00067 CATGGTGTACTTCAACTTCT 166 GGTCCCCATGAACTACATGGTGT 186
    5_human_987 ACTTCAACTTCTTTGCCTGTGT
    7 ADORA2A_NM_00067 CTAACTGCCTTTCCTTCTAA 167 AGTGAGAGGCCTTGTCTAACTGC 187
    5_human_2481 CTTTCCTTCTAAAGGGAATGTT
    8 ADORA2A_NM_00067 CTGATGATTCATGGAGTTTG 168 TGGAGCAGGAGTGTCCTGATGAT 188
    5_human_1695 TCATGGAGTTTGCCCCTTCCTA
    9 ADORA2A_NM_00067 CTCAGAGTCCTCTGTGAAAA 169 CCTGGTTTCAGGAGACTCAGAGT 189
    5_human_264 CCTCTGTGAAAAAGCCCTTGGA
    10 ADORA2A_NM_00067 AACGAGCCACATCGTGTTTT 170 CTGAGATAAAATAAAAACGAGCC 190
    5_human_2531 ACATCGTGTTTTAAGCTTGTCC
    11 ADORA2A_NM_00067 TCCTTCTAAAGGGAATGTTT 171 TTGTCTAACTGCCTTTCCTTCTAAA 191
    5_human_2492 GGGAATGTTTTTTTCTGAGA
    12 ADORA2A_NM_00067 CATGAACTACATGGTGTACT 172 TGAGGATGTGGTCCCCATGAACT 192
    5_human_978 ACATGGTGTACTTCAACTTCTT
    13 ADORA2A_NM_00067 AACTGCCTTTCCTTCTAAAG 173 TGAGAGGCCTTGTCTAACTGCCTT 193
    5_human_2483 TCCTTCTAAAGGGAATGTTTT
    14 ADORA2A_NM_00067 CAGATGTTTCATGCTGTGAG 174 TGGGTTCTGAGGAAGCAGATGTT 194
    5_human_1894 TCATGCTGTGAGGCCTTGCACC
    15 ADORA2A_NM_00067 CCCATGAACTACATGGTGTA 175 TTTGAGGATGTGGTCCCCATGAA 195
    5_human_976 CTACATGGTGTACTTCAACTTC
    16 ADORA2A_NM_00067 AGGCAGCAAGAACCTTTCAA 176 CGCAGCCACGTCCTGAGGCAGCA 196
    5_human_1384 AGAACCTTTCAAGGCAGCTGGC
    17 ADORA2A_NM_00067 GTCCTGATGATTCATGGAGT 177 GGATGGAGCAGGAGTGTCCTGAT 197
    5_human_1692 GATTCATGGAGTTTGCCCCTTC
    18 ADORA2A_NM_00067 GTACTTCAACTTCTTTGCCT 178 CATGAACTACATGGTGTACTTCAA 198
    5_human_993 CTTCTTTGCCTGTGTGCTGGT
    19 ADORA2A_NM_00067 TGTAAGTGTGAGGAAACCCT 179 TTTTTCCAGGAAAAATGTAAGTGT 199
    5_human_2167 GAGGAAACCCTTTTTATTTTA
    20 ADORA2A_NM_00067 CCTACTTTGGACTGAGAGAA 180 TGAGGGCAGCCGGTTCCTACTTT 200
    5_human_1815 GGACTGAGAGAAGGGAGCCCCA
    Accession: NM_005214
    HUGO gene symbol: CTLA4
    1 CTLA4_NM_005214_ TGATTCTGTGTGGGTTCAAA 201 TCTATATAAAGTCCTTGATTCTGT 221
    human_61 GTGGGTTCAAACACATTTCAA
    2 CTLA4_NM_005214_ TTATTTGTTTGTGCATTTGG 202 GCTATCCAGCTATTTTTATTTGTTT 222
    human_909 GTGCATTTGGGGGGAATTCA
    3 CTLA4_NM_005214_ TGATTACATCAAGGCTTCAA 203 TCTTAAACAAATGTATGATTACAT 223
    human_1265 CAAGGCTTCAAAAATACTCAC
    4 CTLA4_NM_005214_ GATGTGGGTCAAGGAATTAA 204 GGGATGCAGCATTATGATGTGGG 224
    human_1094 TCAAGGAATTAAGTTAGGGAAT
    5 CTLA4_NM_005214_ CCTTTTATTTCTTAAACAAA 205 AAGTTAAATTTTATGCCTTTTATTT 225
    human_1241 CTTAAACAAATGTATGATTA
    6 CTLA4_NM_005214_ GATTACATCAAGGCTTCAAA 206 CTTAAACAAATGTATGATTACATC 226
    human_1266 AAGGCTTCAAAAATACTCACA
    7 CTLA4_NM_005214_ TCTGTGTGGGTTCAAACACA 207 TATAAAGTCCTTGATTCTGTGTGG 227
    human_65 GTTCAAACACATTTCAAAGCT
    8 CTLA4_NM_005214_ TTGATAGTATTGTGCATAGA 208 TATATATATTTTAATTTGATAGTAT 228
    human_1405 TGTGCATAGAGCCACGTATG
    9 CTLA4_NM_005214_ TGCCTTTTATTTCTTAAACA 209 TCAAGTTAAATTTTATGCCTTTTAT 229
    human_1239 TTCTTAAACAAATGTATGAT
    10 CTLA4_NM_005214_ TCCATGAAAATGCAACAACA 210 TTTAACTCAATATTTTCCATGAAA 230
    human_1912 ATGCAACAACATGTATAATAT
    11 CTLA4_NM_005214_ TTATTTCTTAAACAAATGTA 211 TAAATTTTATGCCTTTTATTTCTTA 231
    human_1245 AACAAATGTATGATTACATC
    12 CTLA4_NM_005214_ TTAATGGTTTGAATATAAAC 212 GTTTTTGTGTATTTGTTAATGGTTT 232
    human_1449 GAATATAAACACTATATGGC
    13 CTLA4_NM_005214_ ATGTGGGTCAAGGAATTAAG 213 GGATGCAGCATTATGATGTGGGT 233
    human_1095 CAAGGAATTAAGTTAGGGAATG
    14 CTLA4_NM_005214_ AGCCGAAATGATCTTTTCAA 214 GTATGAGACGTTTATAGCCGAAA 234
    human_1208 TGATCTTTTCAAGTTAAATTTT
    15 CTLA4_NM_005214_ GTTTGAATATAAACACTATA 215 GTGTATTTGTTAATGGTTTGAATA 235
    human_1455 TAAACACTATATGGCAGTGTC
    16 CTLA4_NM_005214_ TATGCCTTTTATTTCTTAAA 216 TTTCAAGTTAAATTTTATGCCTTTT 236
    human_1237 ATTTCTTAAACAAATGTATG
    17 CTLA4_NM_005214_ TTCCATGAAAATGCAACAAC 217 TTTTAACTCAATATTTTCCATGAA 237
    human_1911 AATGCAACAACATGTATAATA
    18 CTLA4_NM_005214_ CATCTCTCTTTAATATAAAG 218 CATTTGGGGGGAATTCATCTCTCT 238
    human_937 TTAATATAAAGTTGGATGCGG
    19 CTLA4_NM_005214_ GGAATTCATCTCTCTTTAAT 219 TTTGTGCATTTGGGGGGAATTCAT 239
    human_931 CTCTCTTTAATATAAAGTTGG
    20 CTLA4_NM_005214_ ATCTATATAAAGTCCTTGAT 220 TCTGGGATCAAAGCTATCTATATA 240
    human_45 AAGTCCTTGATTCTGTGTGGG
    Accession: NM_002286
    HUGO gene symbol: LAG3
    1 LAG3_NM_002286 GACTTTACCCTTCGACTAGA 241 ACTGGAGACAATGGCGACTTTACC 261
    _human_1292 CTTCGACTAGAGGATGTGAGC
    2 LAG3_NM_002286 CAACGTCTCCATCATGTATA 242 CTACAGAGATGGCTTCAACGTCTC 262
    _human_1096 CATCATGTATAACCTCACTGT
    3 LAG3_NM_002286 GTCCTTTCTCTGCTCCTTTT 243 TTTCTCATCCTTGGTGTCCTTTCTCT 263
    _human_1721 GCTCCTTTTGGTGACTGGA
    4 LAG3_NM_002286 TCCAGTATCTGGACAAGAAC 244 GCTTTGTGAGGTGACTCCAGTATC 264
    _human_1465 TGGACAAGAACGCTTTGTGTG
    5 LAG3_NM_002286 ATTTTCTGCCTTAGAGCAAG 245 GTGGCGACCAAGACGATTTTCTGC 265
    _human_1795 CTTAGAGCAAGGGATTCACCC
    6 LAG3_NM_002286 TTTCACCTTTGGAGAAGACA 246 ACTGGAGCCTTTGGCTTTCACCTTT 266
    _human_1760 GGAGAAGACAGTGGCGACCA
    7 LAG3_NM_002286 CATTTTGAACTGCTCCTTCA 247 AGCCTCCGACTGGGTCATTTTGAA 267
    _human_904 CTGCTCCTTCAGCCGCCCTGA
    8 LAG3_NM_002286 TCATCACAGTGACTCCCAAA 248 CTGTCACATTGGCAATCATCACAGT 268
    _human_1398 GACTCCCAAATCCTTTGGGT
    9 LAG3_NM_002286 GCTTTCACCTTTGGAGAAGA 249 TGACTGGAGCCTTTGGCTTTCACCT 269
    _human_1758 TTGGAGAAGACAGTGGCGAC
    10 LAG3_NM_002286 CTTTGGCTTTCACCTTTGGA 250 TTTGGTGACTGGAGCCTTTGGCTTT 270
    _human_1753 CACCTTTGGAGAAGACAGTG
    11 LAG3_NM_002286 ATTTTGAACTGCTCCTTCAG 251 GCCTCCGACTGGGTCATTTTGAACT 271
    _human_905 GCTCCTTCAGCCGCCCTGAC
    12 LAG3_NM_002286 CACATTGGCAATCATCACAG 252 GCTCAATGCCACTGTCACATTGGC 272
    _human_1387 AATCATCACAGTGACTCCCAA
    13 LAG3_NM_002286 TTTCTGACCTCCTTTTGGAG 253 ACTGCCCCCTTTCCTTTTCTGACCTC 273
    _human_301 CTTTTGGAGGGCTCAGCGC
    14 LAG3_NM_002286 CGACTGGGTCATTTTGAACT 254 ATCTCTCAGAGCCTCCGACTGGGT 274
    _human_895 CATTTTGAACTGCTCCTTCAG
    15 LAG3_NM_002286 TACTTCACAGAGCTGTCTAG 255 CTTGGAGCAGCAGTGTACTTCACA 275
    _human_1625 GAGCTGTCTAGCCCAGGTGCC
    16 LAG3_NM_002286 ATTGGCAATCATCACAGTGA 256 CAATGCCACTGTCACATTGGCAATC 276
    _human_1390 ATCACAGTGACTCCCAAATC
    17 LAG3_NM_002286 CTGTTTCTCATCCTTGGTGT 257 GCAGGCCACCTCCTGCTGTTTCTCA 277
    _human_1703 TCCTTGGTGTCCTTTCTCTG
    18 LAG3_NM_002286 TTGTGAGGTGACTCCAGTAT 258 CCTGGGGAAGCTGCTTTGTGAGGT 278
    _human_1453 GACTCCAGTATCTGGACAAGA
    19 LAG3_NM_002286 TTTGGCTTTCACCTTTGGAG 259 TTGGTGACTGGAGCCTTTGGCTTTC 279
    _human_1754 ACCTTTGGAGAAGACAGTGG
    20 LAG3_NM_002286 TGGAGACAATGGCGACTTTA 260 TGACCTCCTGGTGACTGGAGACAA 280
    _human_1279 TGGCGACTTTACCCTTCGACT
    Accession: NM_005018
    HUGO gene symbol: PDCD1
    1 PDCD1_NM_005018 TATTATATTATAATTATAAT 281 CCTTCCCTGTGGTTCTATTATATTAT 301
    _human_2070 AATTATAATTAAATATGAG
    2 PDCD1_NM_005018 TCTATTATATTATAATTATA 282 CCCCTTCCCTGTGGTTCTATTATATT 302
    _human_2068 ATAATTATAATTAAATATG
    3 PDCD1_NM_005018 CATTCCTGAAATTATTTAAA 283 GCTCTCCTTGGAACCCATTCCTGAA 303
    _human_1854 ATTATTTAAAGGGGTTGGCC
    4 PDCD1_NM_005018 CTATTATATTATAATTATAA 284 CCCTTCCCTGTGGTTCTATTATATT 304
    _human_2069 ATAATTATAATTAAATATGA
    5 PDCD1_NM_005018 AGTTTCAGGGAAGGTCAGAA 285 CTGCAGGCCTAGAGAAGTTTCAGG 305
    _human_1491 GAAGGTCAGAAGAGCTCCTGG
    6 PDCD1_NM_005018 TGTGGTTCTATTATATTATA 286 GGGATCCCCCTTCCCTGTGGTTCTA 306
    _human_2062 TTATATTATAATTATAATTA
    7 PDCD1_NM_005018 TGTGTTCTCTGTGGACTATG 287 CCCCTCAGCCGTGCCTGTGTTCTCT 307
    _human_719 GTGGACTATGGGGAGCTGGA
    8 PDCD1_NM_005018 CCCATTCCTGAAATTATTTA 288 GAGCTCTCCTTGGAACCCATTCCTG 308
    _human_1852 AAATTATTTAAAGGGGTTGG
    9 PDCD1_NM_005018 TGCCACCATTGTCTTTCCTA 289 TGAGCAGACGGAGTATGCCACCAT 309
    _human_812 TGTCTTTCCTAGCGGAATGGG
    10 PDCD1_NM_005018 AAGTTTCAGGGAAGGTCAGA 290 CCTGCAGGCCTAGAGAAGTTTCAG 310
    _human_1490 GGAAGGTCAGAAGAGCTCCTG
    11 PDCD1_NM_005018 CTGTGGTTCTATTATATTAT 291 AGGGATCCCCCTTCCCTGTGGTTCT 311
    _human_2061 ATTATATTATAATTATAATT
    12 PDCD1_NM_005018 TTCTATTATATTATAATTAT 292 CCCCCTTCCCTGTGGTTCTATTATA 312
    _human_2067 TTATAATTATAATTAAATAT
    13 PDCD1_NM_005018 TTTCAGGGAAGGTCAGAAGA 293 GCAGGCCTAGAGAAGTTTCAGGG 313
    _human_1493 AAGGTCAGAAGAGCTCCTGGCT
    14 PDCD1_NM_005018 CTTGGAACCCATTCCTGAAA 294 ACCCTGGGAGCTCTCCTTGGAACC 314
    _human_1845 CATTCCTGAAATTATTTAAAG
    15 PDCD1_NM_005018 TCCCTGTGGTTCTATTATAT 295 ACAAGGGATCCCCCTTCCCTGTGG 315
    _human_2058 TTCTATTATATTATAATTATA
    16 PDCD1_NM_005018 CCTGTGGTTCTATTATATTA 296 AAGGGATCCCCCTTCCCTGTGGTTC 316
    _human_2060 TATTATATTATAATTATAAT
    17 PDCD1_NM_005018 TGGAACCCATTCCTGAAATT 297 CCTGGGAGCTCTCCTTGGAACCCA 317
    _human_1847 TTCCTGAAATTATTTAAAGGG
    18 PDCD1_NM_005018 CCTTCCCTGTGGTTCTATTA 298 GGGACAAGGGATCCCCCTTCCCTG 318
    _human_2055 TGGTTCTATTATATTATAATT
    19 PDCD1_NM_005018 TTCCCTGTGGTTCTATTATA 299 GACAAGGGATCCCCCTTCCCTGTG 319
    _human_2057 GTTCTATTATATTATAATTAT
    20 PDCD1_NM_005018 CACAGGACTCATGTCTCAAT 300 CAGGCACAGCCCCACCACAGGACT 320
    _human_1105 CATGTCTCAATGCCCACAGTG
    Accession: NM_004612
    HUGO gene symbol: TGFBR1
    1 TGFBR1_NM_004612 CCTGTTTATTACAACTTAAA 321 GTTAATAACATTCAACCTGTTTAT 341
    _human_5263 TACAACTTAAAAGGAACTTCA
    2 TGFBR1_NM_004612 CCATTGGTGGAATTCATGAA 322 TTGCTCGACGATGTTCCATTGGTG 342
    _human_1323 GAATTCATGAAGATTACCAAC
    3 TGFBR1_NM_004612 TTTTCCTTATAACAAAGACA 323 TTTAGGGATTTTTTTTTTTCCTTAT 343
    _human_6389 AACAAAGACATCACCAGGAT
    4 TGFBR1_NM_004612 TGTATTACTTGTTTAATAAT 324 TTTTTATAGTTGTGTTGTATTACTT 344
    _human_3611 GTTTAATAATAATCTCTAAT
    5 TGFBR1_NM_004612 TTATTGAATCAAAGATTGAG 325 TGCTGAAGATATTTTTTATTGAAT 345
    _human_3882 CAAAGATTGAGTTACAATTAT
    6 TGFBR1_NM_004612 TTCTTACCTAAGTGGATAAA 326 GTTACAATTATACTTTTCTTACCTA 346
    _human_3916 AGTGGATAAAATGTACTTTT
    7 TGFBR1_NM_004612 ATGTTGCTCAGTTACTCAAA 327 TAAAGTATGGGTATTATGTTGCTC 347
    _human_5559 AGTTACTCAAATGGTACTGTA
    8 TGFBR1_NM_004612 ATATTTGTACCCCAAATAAC 328 GGTACTGTATTGTTTATATTTGTA 348
    _human_5595 CCCCAAATAACATCGTCTGTA
    9 TGFBR1_NM_004612 TGTAAATGTAAACTTCTAAA 329 TTATGCAATCTTGTTTGTAAATGT 349
    _human_5222 AAACTTCTAAAAATATGGTTA
    10 TGFBR1_NM_004612 AGAATGAGTGACATATTACA 330 AACCAAAGTAATTTTAGAATGAG 350
    _human_3435 TGACATATTACATAGGAATTTA
    11 TGFBR1_NM_004612 CCATTTCTAAGCCTACCAGA 331 GTTGTTGTTTTTGGGCCATTTCTA 351
    _human_3709 AGCCTACCAGATCTGCTTTAT
    12 TGFBR1_NM_004612 ATATTCCAAAAGAATGTAAA 332 ATTGTATTTGTAGTAATATTCCAA 352
    _human_5826 AAGAATGTAAATAGGAAATAG
    13 TGFBR1_NM_004612 TTACTTCCAATGCTATGAAG 333 TATAATAACTGGTTTTTACTTCCA 353
    _human_3146 ATGCTATGAAGTCTCTGCAGG
    14 TGFBR1_NM_004612 TCTTTATCTGTTCAAAGACT 334 TGTAAGCCATTTTTTTCTTTATCTG 354
    _human_2675 TTCAAAGACTTATTTTTTAA
    15 TGFBR1_NM_004612 GTCTAAGTATACTTTTAAAA 335 CATTTTAATTGTGTTGTCTAAGTA 355
    _human_2529 TACTTTTAAAAAATCAAGTGG
    16 TGFBR1_NM_004612 ATCTTTGGACATGTACTGCA 336 GAGATACTAAGGATTATCTTTGG 356
    _human_5079 ACATGTACTGCAGCTTCTTGTC
    17 TGFBR1_NM_004612 GTGTTGTATTACTTGTTTAA 337 TTTGTTTTTATAGTTGTGTTGTATT 357
    _human_3607 ACTTGTTTAATAATAATCTC
    18 TGFBR1_NM_004612 TGCTGTAGATGGCAACTAGA 338 CATGCCATATGTAGTTGCTGTAGA 358
    _human_5994 TGGCAACTAGAACCTTTGAGT
    19 TGFBR1_NM_004612 TCTTTCACTTATTCAGAACA 339 GTATACTATTATTGTTCTTTCACTT 359
    _human_2177 ATTCAGAACATTACATGCCT
    20 TGFBR1_NM_004612 GTATTTGTAGTAATATTCCA 340 TTTAAATTGTATATTGTATTTGTA 360
    _human_5814 GTAATATTCCAAAAGAATGTA
    Accession: NM_032782
    HUGO gene symbol: HAVCR2
    1 HAVCR2_NM_032782 CTCATAGCAAAGAGAAGATA 361 TTTTCAAATGGTATTCTCATAGCA 381
    _human_937 AAGAGAAGATACAGAATTTAA
    2 HAVCR2_NM_032782 GTATTCTCATAGCAAAGAGA 362 TTTAATTTTCAAATGGTATTCTCAT 382
    _human_932 AGCAAAGAGAAGATACAGAA
    3 HAVCR2_NM_032782 TTGCTTGTTGTGTGCTTGAA 363 TGTATTGGCCAAGTTTTGCTTGTT 383
    _human_2126 GTGTGCTTGAAAGAAAATATC
    4 HAVCR2_NM_032782 TATTCGTGGACCAAACTGAA 364 TCTGACCAACTTCTGTATTCGTGG 384
    _human_2171 ACCAAACTGAAGCTATATTTT
    5 HAVCR2_NM_032782 ATTGTGGAGTAGACAGTTGG 365 GCTACTGCTCATGTGATTGTGGA 385
    _human_158 GTAGACAGTTGGAAGAAGTACC
    6 HAVCR2_NM_032782 GTTGTGTGCTTGAAAGAAAA 366 GGCCAAGTTTTGCTTGTTGTGTGC 386
    _human_2132 TTGAAAGAAAATATCTCTGAC
    7 HAVCR2_NM_032782 TGTTGTGTGCTTGAAAGAAA 367 TGGCCAAGTTTTGCTTGTTGTGTG 387
    _human_2131 CTTGAAAGAAAATATCTCTGA
    8 HAVCR2_NM_032782 CCCTAAACTTAAATTTCAAG 368 TTGACAGAGAGTGGTCCCTAAAC 388
    _human_2313 TTAAATTTCAAGACGGTATAGG
    9 HAVCR2_NM_032782 ACATCCAGATACTGGCTAAA 369 GATGTGAATTATTGGACATCCAG 389
    _human_489 ATACTGGCTAAATGGGGATTTC
    10 HAVCR2_NM_032782 CATTTTCAGAAGATAATGAC 370 GGAGCAGAGTTTTCCCATTTTCAG 390
    _human_1272 AAGATAATGACTCACATGGGA
    11 HAVCR2_NM_032782 CACATTGGCCAATGAGTTAC 371 TCTAACACAAATATCCACATTGGC 391
    _human_785 CAATGAGTTACGGGACTCTAG
    12 HAVCR2_NM_032782 TGCTTGTTGTGTGCTTGAAA 372 GTATTGGCCAAGTTTTGCTTGTTG 392
    _human_2127 TGTGCTTGAAAGAAAATATCT
    13 HAVCR2_NM_032782 GAGTAGACAGTTGGAAGAAG 373 GCTCATGTGATTGTGGAGTAGAC 393
    _human_164 AGTTGGAAGAAGTACCCAGTCC
    14 HAVCR2_NM_032782 TTGTTGTGTGCTTGAAAGAA 374 TTGGCCAAGTTTTGCTTGTTGTGT 394
    _human_2130 GCTTGAAAGAAAATATCTCTG
    15 HAVCR2_NM_032782 CGGCGCTTTAATTTTCAAAT 375 TCTGGCTCTTATCTTCGGCGCTTT 395
    _human_911 AATTTTCAAATGGTATTCTCA
    16 HAVCR2_NM_032782 TTTGGCACAGAAAGTCTAAA 376 TGAAAGCATAACTTTTTTGGCACA 396
    _human_1543 GAAAGTCTAAAGGGGCCACTG
    17 HAVCR2_NM_032782 GATCTGTCTTGCTTATTGTT 377 AGACGGTATAGGCTTGATCTGTC 397
    _human_2346 TTGCTTATTGTTGCCCCCTGCG
    18 HAVCR2_NM_032782 GGTGTGTATTGGCCAAGTTT 378 GAAGTGCATTTGATTGGTGTGTA 398
    _human_2107 TTGGCCAAGTTTTGCTTGTTGT
    19 HAVCR2_NM_032782 CCCATTTTCAGAAGATAATG 379 ATGGAGCAGAGTTTTCCCATTTTC 399
    _human_1270 AGAAGATAATGACTCACATGG
    20 HAVCR2_NM_032782 TGGCACAGAAAGTCTAAAGG 380 AAAGCATAACTTTTTTGGCACAGA 400
    _human_1545 AAGTCTAAAGGGGCCACTGAT
    Accession: NM_002987
    HUGO gene symbol: CCL17
    1 CCL17_NM_002987 AAATACCTGCAAAGCCTTGA 401 GTGAAGAATGCAGTTAAATACCTGC 421
    _human_385 AAAGCCTTGAGAGGTCTTGA
    2 CCL17_NM_002987 TTTTGTAACTGTGCAGGGCA 402 CAGGGATGCCATCGTTTTTGTAACT 422
    _human_318 GTGCAGGGCAGGGCCATCTG
    3 CCL17_NM_002987 AGAGTGAAGAATGCAGTTAA 403 GACCCCAACAACAAGAGAGTGAAG 423
    _human_367 AATGCAGTTAAATACCTGCAA
    4 CCL17_NM_002987 AAGCCTTGAGAGGTCTTGAA 404 AGTTAAATACCTGCAAAGCCTTGAG 424
    _human_396 AGGTCTTGAAGCCTCCTCAC
    5 CCL17_NM_002987 AATACCTGCAAAGCCTTGAG 405 TGAAGAATGCAGTTAAATACCTGCA 425
    _human_386 AAGCCTTGAGAGGTCTTGAA
    6 CCL17_NM_002987 TGCAGTTAAATACCTGCAAA 406 CAAGAGAGTGAAGAATGCAGTTAA 426
    _human_378 ATACCTGCAAAGCCTTGAGAG
    7 CCL17_NM_002987 CAACAACAAGAGAGTGAAGA 407 CATCTGTTCGGACCCCAACAACAAG 427
    _human_357 AGAGTGAAGAATGCAGTTAA
    8 CCL17_NM_002987 CTGAATTCAAAACCAGGGTG 408 CTGCTGATGGGAGAGCTGAATTCAA 428
    _human_55 AACCAGGGTGTCTCCCTGAG
    9 CCL17_NM_002987 ATACCTGCAAAGCCTTGAGA 409 GAAGAATGCAGTTAAATACCTGCAA 429
    _human_387 AGCCTTGAGAGGTCTTGAAG
    10 CCL17_NM_002987 TTCCCCTTAGAAAGCTGAAG 410 ACTTCAAGGGAGCCATTCCCCTTAG 430
    _human_254 AAAGCTGAAGACGTGGTACC
    11 CCL17_NM_002987 GGAGAGCTGAATTCAAAACC 411 CACCGCCTGCTGATGGGAGAGCTG 431
    _human_49 AATTCAAAACCAGGGTGTCTC
    12 CCL17_NM_002987 GCAGTTAAATACCTGCAAAG 412 AAGAGAGTGAAGAATGCAGTTAAA 432
    _human_379 TACCTGCAAAGCCTTGAGAGG
    13 CCL17_NM_002987 GAAGAATGCAGTTAAATACC 413 CAACAACAAGAGAGTGAAGAATGC 433
    _human_372 AGTTAAATACCTGCAAAGCCT
    14 CCL17_NM_002987 ATGCAGTTAAATACCTGCAA 414 ACAAGAGAGTGAAGAATGCAGTTA 434
    _human_377 AATACCTGCAAAGCCTTGAGA
    15 CCL17_NM_002987 CATTCCCCTTAGAAAGCTGA 415 GTACTTCAAGGGAGCCATTCCCCTT 435
    _human_252 AGAAAGCTGAAGACGTGGTA
    16 CCL17_NM_002987 AGAGCTGAATTCAAAACCAG 416 CCGCCTGCTGATGGGAGAGCTGAAT 436
    _human_51 TCAAAACCAGGGTGTCTCCC
    17 CCL17_NM_002987 GATGGGAGAGCTGAATTCAA 417 GTGTCACCGCCTGCTGATGGGAGA 437
    _human_45 GCTGAATTCAAAACCAGGGTG
    18 CCL17_NM_002987 TGATGGGAGAGCTGAATTCA 418 AGTGTCACCGCCTGCTGATGGGAGA 438
    _human_44 GCTGAATTCAAAACCAGGGT
    19 CCL17_NM_002987 ACTTTGAGCTCACAGTGTCA 419 GCTCAGAGAGAAGTGACTTTGAGCT 439
    _human_16 CACAGTGTCACCGCCTGCTG
    20 CCL17_NM_002987 GAGTGAAGAATGCAGTTAAA 420 ACCCCAACAACAAGAGAGTGAAGA 440
    _human_368 ATGCAGTTAAATACCTGCAAA
    Accession: NM_002990
    HUGO gene symbol: CCL22
    1 CCL22_NM_002990 GTATTTGAAAACAGAGTAAA 441 GCTGGAGTTATATATGTATTTGAA 461
    _human_2083 AACAGAGTAAATACTTAAGAG
    2 CCL22_NM_002990 CAATAAGCTGAGCCAATGAA 442 GGTGAAGATGATTCTCAATAAGC 462
    _human_298 TGAGCCAATGAAGAGCCTACTC
    3 CCL22_NM_002990 TACTTAAGAGGCCAAATAGA 443 TGAAAACAGAGTAAATACTTAAG 463
    _human_2103 AGGCCAAATAGATGAATGGAAG
    4 CCL22_NM_002990 ATGTATTTGAAAACAGAGTA 444 AAGCTGGAGTTATATATGTATTTG 464
    _human_2081 AAAACAGAGTAAATACTTAAG
    5 CCL22_NM_002990 TTCATACAGCAAGTATGGGA 445 TTGAGAAATATTCTTTTCATACAG 465
    _human_2496 CAAGTATGGGACAGCAGTGTC
    6 CCL22_NM_002990 CTGCAGACAAAATCAATAAA 446 GAGCCCAGAAAGTGGCTGCAGAC 466
    _human_1052 AAAATCAATAAAACTAATGTCC
    7 CCL22_NM_002990 TGCAGACAAAATCAATAAAA 447 AGCCCAGAAAGTGGCTGCAGACA 467
    _human_1053 AAATCAATAAAACTAATGTCCC
    8 CCL22_NM_002990 GGCCAAATAGATGAATGGAA 448 AGTAAATACTTAAGAGGCCAAAT 468
    _human_2112 AGATGAATGGAAGAATTTTAGG
    9 CCL22_NM_002990 AATAAGCTGAGCCAATGAAG 449 GTGAAGATGATTCTCAATAAGCT 469
    _human_299 GAGCCAATGAAGAGCCTACTCT
    10 CCL22_NM_002990 AAGAGGCCAAATAGATGAAT 450 ACAGAGTAAATACTTAAGAGGCC 470
    _human_2108 AAATAGATGAATGGAAGAATTT
    11 CCL22_NM_002990 AAATAGATGAATGGAAGAAT 451 AATACTTAAGAGGCCAAATAGAT 471
    _human_2116 GAATGGAAGAATTTTAGGAACT
    12 CCL22_NM_002990 AAACAGAGTAAATACTTAAG 452 TATATATGTATTTGAAAACAGAGT 472
    _human_2091 AAATACTTAAGAGGCCAAATA
    13 CCL22_NM_002990 AGCTGGAGTTATATATGTAT 453 TGACTTGGTATTATAAGCTGGAG 473
    _human_2067 TTATATATGTATTTGAAAACAG
    14 CCL22_NM_002990 ACCTTTGACTTGGTATTATA 454 ATGGTGTGAAAGACTACCTTTGA 474
    _human_2047 CTTGGTATTATAAGCTGGAGTT
    15 CCL22_NM_002990 AACCTTCAGGGATAAGGAGA 455 TGGCGTGGTGTTGCTAACCTTCA 475
    _human_238 GGGATAAGGAGATCTGTGCCGA
    16 CCL22_NM_002990 GTGAAAGACTACCTTTGACT 456 AATTCATGCTATGGTGTGAAAGA 476
    _human_2037 CTACCTTTGACTTGGTATTATA
    17 CCL22_NM_002990 CTATGGTGTGAAAGACTACC 457 ACAATCAAATTCATGCTATGGTGT 477
    _human_2030 GAAAGACTACCTTTGACTTGG
    18 CCL22_NM_002990 CACTACGGCTGGCTAATTTT 458 ATTACAGGTGTGTGCCACTACGG 478
    _human_1682 CTGGCTAATTTTTGTATTTTTA
    19 CCL22_NM_002990 GGAGTTATATATGTATTTGA 459 TTGGTATTATAAGCTGGAGTTATA 479
    _human_2071 TATGTATTTGAAAACAGAGTA
    20 CCL22_NM_002990 ATATCAATACAGAGACTCAA 460 CCAAAAGGCAGTTACATATCAAT 480
    _human_1111 ACAGAGACTCAAGGTCACTAGA
    Accession: NM_005618
    HUGO gene symbol: DLL1
    1 DLL1_NM_005618 CTGTTTTGTTAATGAAGAAA 481 TATTTGAGTTTTTTACTGTTTTGTTA 501
    _human_3246 ATGAAGAAATTCCTTTTTA
    2 DLL1_NM_005618 TTGTATATAAATGTATTTAT 482 TGTGACTATATTTTTTTGTATATAAA 502
    _human_3193 TGTATTTATGGAATATTGT
    3 DLL1_NM_005618 TGTTTTGTTAATGAAGAAAT 483 ATTTGAGTTTTTTACTGTTTTGTTAA 503
    _human_3247 TGAAGAAATTCCTTTTTAA
    4 DLL1_NM_005618 AATTTTGGTAAATATGTACA 484 GTTTTTTATAATTTAAATTTTGGTAA 504
    _human_3141 ATATGTACAAAGGCACTTC
    5 DLL1_NM_005618 AAATTTTATGAATGACAAAA 485 ATATTTTTCCAAAATAAATTTTATGA 505
    _human_3293 ATGACAAAAAAAAAAAAAA
    6 DLL1_NM_005618 TTTATGGAATATTGTGCAAA 486 TTGTATATAAATGTATTTATGGAATA 506
    _human_3208 TTGTGCAAATGTTATTTGA
    7 DLL1_NM_005618 TTACTGTTTTGTTAATGAAG 487 TGTTATTTGAGTTTTTTACTGTTTTGT 507
    _human_3243 TAATGAAGAAATTCCTTT
    8 DLL1_NM_005618 TTCTTGAATTAGAAACACAA 488 TTATGAGCCAGTCTTTTCTTGAATTA 508
    _human_2977 GAAACACAAACACTGCCTT
    9 DLL1_NM_005618 CAGTTGCTCTTAAGAGAATA 489 CCGTTGCACTATGGACAGTTGCTCTT 509
    _human_2874 AAGAGAATATATATTTAAA
    10 DLL1_NM_005618 CAACTTCAAAAGACACCAAG 490 CGGACTCGGGCTGTTCAACTTCAAA 510
    _human_2560 AGACACCAAGTACCAGTCGG
    11 DLL1_NM_005618 TCCAAAATAAATTTTATGAA 491 TTTTTAAAATATTTTTCCAAAATAAA 511
    _human_3285 TTTTATGAATGACAAAAAA
    12 DLL1_NM_005618 GAACTGAATTACGCATAAGA 492 TATATTTAAATGGGTGAACTGAATT 512
    _human_2909 ACGCATAAGAAGCATGCACT
    13 DLL1_NM_005618 GGATTTTGTGACAAACCAGG 493 TGTGATGAGCAGCATGGATTTTGTG 513
    _human_1173 ACAAACCAGGGGAATGCAAG
    14 DLL1_NM_005618 TACTGTTTTGTTAATGAAGA 494 GTTATTTGAGTTTTTTACTGTTTTGTT 514
    _human_3244 AATGAAGAAATTCCTTTT
    15 DLL1_NM_005618 TTTGGTAAATATGTACAAAG 495 TTTTATAATTTAAATTTTGGTAAATA 515
    _human_3144 TGTACAAAGGCACTTCGGG
    16 DLL1_NM_005618 CCAAAATAAATTTTATGAAT 496 TTTTAAAATATTTTTCCAAAATAAAT 516
    _human_3286 TTTATGAATGACAAAAAAA
    17 DLL1_NM_005618 ATAATTTAAATTTTGGTAAA 497 TGATGTTCGTTTTTTATAATTTAAAT 517
    _human_3133 TTTGGTAAATATGTACAAA
    18 DLL1_NM_005618 AAATGGGTGAACTGAATTAC 498 AGAGAATATATATTTAAATGGGTGA 518
    _human_2901 ACTGAATTACGCATAAGAAG
    19 DLL1_NM_005618 TTCGGGTCTATGTGACTATA 499 TATGTACAAAGGCACTTCGGGTCTA 519
    _human_3168 TGTGACTATATTTTTTTGTA
    20 DLL1_NM_005618 ACTGTTTTGTTAATGAAGAA 500 TTATTTGAGTTTTTTACTGTTTTGTTA 520
    _human_3245 ATGAAGAAATTCCTTTTT
    Accession: NM_000639
    HUGO gene symbol: FASLG
    1 FASLG_NM_000639 TAGCTCCTCAACTCACCTAA 521 GGTTCAAAATGTCTGTAGCTCCTC 541
    _human_1154 AACTCACCTAATGTTTATGAG
    2 FASLG_NM_000639 ATGTTTTCCTATAATATAAT 522 TGTCAGCTACTAATGATGTTTTCC 542
    _human_1771 TATAATATAATAAATATTTAT
    3 FASLG_NM_000639 TTTTCCTATAATATAATAAA 523 CAGCTACTAATGATGTTTTCCTAT 543
    _human_1774 AATATAATAAATATTTATGTA
    4 FASLG_NM_000639 TTCCTATAATATAATAAATA 524 GCTACTAATGATGTTTTCCTATAA 544
    _human_1776 TATAATAAATATTTATGTAGA
    5 FASLG_NM_000639 TGCATTTGAGGTCAAGTAAG 525 GAGGGTCTTCTTACATGCATTTGA 545
    _human_1086 GGTCAAGTAAGAAGACATGAA
    6 FASLG_NM_000639 ATTGATTGTCAGCTACTAAT 526 TAGTGCTTAAAAATCATTGATTGT 546
    _human_1750 CAGCTACTAATGATGTTTTCC
    7 FASLG_NM_000639 AAATGAAAACATGTAATAAA 527 ATGTGCATTTTTGTGAAATGAAAA 547
    _human_1820 CATGTAATAAAAAGTATATGT
    8 FASLG_NM_000639 ATTGTGAAGTACATATTAGG 528 AGAGAGAATGTAGATATTGTGAA 548
    _human_1659 GTACATATTAGGAAAATATGGG
    9 FASLG_NM_000639 GCTTTCTGGAGTGAAGTATA 529 CTATGGAATTGTCCTGCTTTCTGG 549
    _human_667 AGTGAAGTATAAGAAGGGTGG
    10 FASLG_NM_000639 CATTTGGTCAAGATTTTGAA 530 GGAAAATATGGGTTGCATTTGGT 550
    _human_1692 CAAGATTTTGAATGCTTCCTGA
    11 FASLG_NM_000639 GGCTTATATAAGCTCTAAGA 531 TCTCAGACGTTTTTCGGCTTATAT 551
    _human_986 AAGCTCTAAGAGAAGCACTTT
    12 FASLG_NM_000639 ACCAGTGCTGATCATTTATA 532 GCAGTGTTCAATCTTACCAGTGCT 552
    _human_911 GATCATTTATATGTCAACGTA
    13 FASLG_NM_000639 CCATTTAACAGGCAAGTCCA 533 GCTGAGGAAAGTGGCCCATTTAA 553
    _human_598 CAGGCAAGTCCAACTCAAGGTC
    14 FASLG_NM_000639 AAGTACATATTAGGAAAATA 534 AATGTAGATATTGTGAAGTACAT 554
    _human_1665 ATTAGGAAAATATGGGTTGCAT
    15 FASLG_NM_000639 TGTGTGTGTGTATGACTAAA 535 GTGTGTGTGTGTGTGTGTGTGTG 555
    _human_1625 TGTATGACTAAAGAGAGAATGT
    16 FASLG_NM_000639 AAGAGGGAGAAGCATGAAAA 536 CTGGGCTGCCATGTGAAGAGGGA 556
    _human_1238 GAAGCATGAAAAAGCAGCTACC
    17 FASLG_NM_000639 GTGTATGACTAAAGAGAGAA 537 TGTGTGTGTGTGTGTGTGTATGA 557
    _human_1632 CTAAAGAGAGAATGTAGATATT
    18 FASLG_NM_000639 GTATTTCCAGTGCAATTGTA 538 CCTAACACAGCATGTGTATTTCCA 558
    _human_1581 GTGCAATTGTAGGGGTGTGTG
    19 FASLG_NM_000639 CAACTCTAATAGTGCTTAAA 539 ATGCTTCCTGACAATCAACTCTAA 559
    _human_1726 TAGTGCTTAAAAATCATTGAT
    20 FASLG_NM_000639 GTGTGTGTGTATGACTAAAG 540 TGTGTGTGTGTGTGTGTGTGTGT 560
    human_1626 GTATGACTAAAGAGAGAATGTA
    Accession: NM_001267706
    HUGO gene symbol: CD274
    1 CD274_NM_001267706 ACCTGCATTAATTTAATAAA 561 ATTGTCACTTTTTGTACCTGCATTA 581
    _human_3222 ATTTAATAAAATATTCTTAT
    2 CD274_NM_001267706 AACTTGCCCAAACCAGTAAA 562 GCAAACAGATTAAGTAACTTGCC 582
    _human_1538 CAAACCAGTAAATAGCAGACCT
    3 CD274_NM_001267706 ATTTGCTCACATCTAGTAAA 563 ACTTGCTGCTTAATGATTTGCTCA 583
    _human_1218 CATCTAGTAAAACATGGAGTA
    4 CD274_NM_001267706 CCTTTGCCATATAATCTAAT 564 TTTATTCCTGATTTGCCTTTGCCAT 584
    _human_1998 ATAATCTAATGCTTGTTTAT
    5 CD274_NM_001267706 ATATAGCAGATGGAATGAAT 565 ATTTTAGTGTTTCTTATATAGCAG 585
    _human_2346 ATGGAATGAATTTGAAGTTCC
    6 CD274_NM_001267706 GCCTTTGCCATATAATCTAA 566 ATTTATTCCTGATTTGCCTTTGCCA 586
    _human_1997 TATAATCTAATGCTTGTTTA
    7 CD274_NM_001267706 GATTTGCCTTTGCCATATAA 567 ATTATATTTATTCCTGATTTGCCTT 587
    _human_1992 TGCCATATAATCTAATGCTT
    8 CD274_NM_001267706 AATTTTCATTTACAAAGAGA 568 CTTAATAATCAGAGTAATTTTCAT 588
    _human_1905 TTACAAAGAGAGGTCGGTACT
    9 CD274_NM_001267706 AGTGTTTCTTATATAGCAGA 569 ATTTTTATTTATTTTAGTGTTTCTT 589
    _human_2336 ATATAGCAGATGGAATGAAT
    10 CD274_NM_001267706 GCTTTCTGTCAAGTATAAAC 570 GAACTTTTGTTTTCTGCTTTCTGTC 590
    _human_2656 AAGTATAAACTTCACTTTGA
    11 CD274_NM_001267706 CATTTGGAAATGTATGTTAA 571 TCTAAAGATAGTCTACATTTGGAA 591
    _human_2235 ATGTATGTTAAAAGCACGTAT
    12 CD274_NM_001267706 TTATTTTAGTGTTTCTTATA 572 CTTTGCTATTTTTATTTATTTTAGT 592
    _human_2329 GTTTCTTATATAGCAGATGG
    13 CD274_NM_001267706 GTGGTAGCCTACACACATAA 573 CAGCTTTACAATTATGTGGTAGCC 593
    _human_1433 TACACACATAATCTCATTTCA
    14 CD274_NM_001267706 ATGAGGAGATTAACAAGAAA 574 GGAGCTCATAGTATAATGAGGAG 594
    _human_1745 ATTAACAAGAAAATGTATTATT
    15 CD274_NM_001267706 CAATTTTGTCGCCAAACTAA 575 TTGTAGTAGATGTTACAATTTTGT 595
    _human_1183 CGCCAAACTAAACTTGCTGCT
    16 CD274_NM_001267706 TATATAGCAGATGGAATGAA 576 TATTTTAGTGTTTCTTATATAGCA 596
    _human_2345 GATGGAATGAATTTGAAGTTC
    17 CD274_NM_001267706 AAATGCCACTAAATTTTAAA 577 CTGTCTTTTCTATTTAAATGCCACT 597
    _human_2069 AAATTTTAAATTCATACCTT
    18 CD274_NM_001267706 TCTTTCCCATAGCTTTTCAT 578 TTTGTTTCTAAGTTATCTTTCCCAT 598
    _human_2414 AGCTTTTCATTATCTTTCAT
    19 CD274_NM_001267706 TATATTCATGACCTACTGGC 579 GATATTTGCTGTCTTTATATTCAT 599
    _human_129 GACCTACTGGCATTTGCTGAA
    20 CD274_NM_001267706 GTCCAGTGTCATAGCATAAG 580 TATTATTACAATTTAGTCCAGTGT 600
    _human_1783 CATAGCATAAGGATGATGCGA
    Accession: NM_002164
    HUGO gene symbol: IDO1
    1 IDO1_NM_002164 ATTCTGTCATAATAAATAAA 601 AAAAAAAAAAGATATATTCTGTCA 621
    _human_1896 TAATAAATAAAAATGCATAAG
    2 IDO1_NM_002164 TATCTTATCATTGGAATAAA 602 AAGTTTTGTAATCTGTATCTTATCA 622
    _human_1532 TTGGAATAAAATGACATTCA
    3 IDO1_NM_002164 GTGATGGAGACTGCAGTAAA 603 TTTTGTTCTCATTTCGTGATGGAGA 623
    _human_578 CTGCAGTAAAGGATTCTTCC
    4 IDO1_NM_002164 TTCTGTCATAATAAATAAAA 604 AAAAAAAAAGATATATTCTGTCAT 624
    _human_1897 AATAAATAAAAATGCATAAGA
    5 IDO1_NM_002164 CTTGTAGGAAAACAACAAAA 605 AATACCTGTGCATTTCTTGTAGGAA 625
    _human_1473 AACAACAAAAGGTAATTATG
    6 IDO1_NM_002164 ATAAAATGACATTCAATAAA 606 TATCTTATCATTGGAATAAAATGAC 626
    _human_1547 ATTCAATAAATAAAAATGCA
    7 IDO1_NM_002164 CGTAAGGTCTTGCCAAGAAA 607 GGTCATGGAGATGTCCGTAAGGTC 627
    _human_412 TTGCCAAGAAATATTGCTGTT
    8 IDO1_NM_002164 TCTTGTAGGAAAACAACAAA 608 AAATACCTGTGCATTTCTTGTAGGA 628
    _human_1472 AAACAACAAAAGGTAATTAT
    9 IDO1_NM_002164 AACTGGAGGCACTGATTTAA 609 ACTGGAAGCCAAAGGAACTGGAG 629
    _human_1248 GCACTGATTTAATGAATTTCCT
    10 IDO1_NM_002164 CAATACAAAAGACCTCAAAA 610 GTTTTACCAATAATGCAATACAAAA 630
    _human_1440 GACCTCAAAATACCTGTGCA
    11 IDO1_NM_002164 TGCTTCTGCAATCAAAGTAA 611 GGTGGAAATAGCAGCTGCTTCTGC 631
    _human_636 AATCAAAGTAATTCCTACTGT
    12 IDO1_NM_002164 AATGACATTCAATAAATAAA 612 TTATCATTGGAATAAAATGACATTC 632
    _human_1551 AATAAATAAAAATGCATAAG
    13 IDO1_NM_002164 ATCATTGGAATAAAATGACA 613 TGTAATCTGTATCTTATCATTGGAA 633
    _human_1538 TAAAATGACATTCAATAAAT
    14 IDO1_NM_002164 ACCAATAATGCAATACAAAA 614 ACTATGCAATGTTTTACCAATAATG 634
    _human_1430 CAATACAAAAGACCTCAAAA
    15 IDO1_NM_002164 ATCTGTATCTTATCATTGGA 615 ACTAGAAGTTTTGTAATCTGTATCT 635
    _human_1527 TATCATTGGAATAAAATGAC
    16 IDO1_NM_002164 ATCTTATCATTGGAATAAAA 616 AGTTTTGTAATCTGTATCTTATCAT 636
    _human_1533 TGGAATAAAATGACATTCAA
    17 IDO1_NM_002164 CAGCTGCTTCTGCAATCAAA 617 TATTGGTGGAAATAGCAGCTGCTT 637
    _human_632 CTGCAATCAAAGTAATTCCTA
    18 IDO1_NM_002164 GCAATACAAAAGACCTCAAA 618 TGTTTTACCAATAATGCAATACAAA 638
    _human_1439 AGACCTCAAAATACCTGTGC
    19 IDO1_NM_002164 TCCTACTGTATTCAAGGCAA 619 TGCAATCAAAGTAATTCCTACTGTA 639
    _human_657 TTCAAGGCAATGCAAATGCA
    20 IDO1_NM_002164 CAGAGCCACAAACTAATACT 620 CATTACCCATTGTAACAGAGCCAC 640
    _human_1398 AAACTAATACTATGCAATGTT
    Accession: NM_001558
    HUGO gene symbol: IL10RA
    1 IL10RA_NM_001558_ TTGTTCATTTATTTATTGGA 641 CTTTATTTATTTATTTTGTTCATTT 661
    human_3364 ATTTATTGGAGAGGCAGCAT
    2 IL10RA_NM_001558_ TTATTCCAATAAATTGTCAA 642 AGTGATACATGTTTTTTATTCCAA 662
    human_3626 TAAATTGTCAAGACCACAGGA
    3 IL10RA_NM_001558_ TATTTTCTGGACACTCAAAC 643 AGATCTTAAGGTATATATTTTCTG 663
    human_2395 GACACTCAAACACATCATAAT
    4 IL10RA_NM_001558_ TTTATTGGAGAGGCAGCATT 644 TATTTTGTTCATTTATTTATTGGAG 664
    human_3375 AGGCAGCATTGCACAGTGAA
    5 IL10RA_NM_001558_ ACCTTGGAGAAGTCACTTAT 645 GTTTCCAGTGGTATGACCTTGGA 665
    human_3469 GAAGTCACTTATCCTCTTGGAG
    6 IL10RA_NM_001558_ TTATTTATTTATTTTGTTCA 646 GTTCCCTTGAAAGCTTTATTTATTT 666
    human_3351 ATTTTGTTCATTTATTTATT
    7 IL10RA_NM_001558_ CTCTTTCCTGTATCATAAAG 647 TCTCCCTCCTAGGAACTCTTTCCT 667
    human_2108 GTATCATAAAGGATTATTTGC
    8 IL10RA_NM_001558_ CTGAGGAAATGGGTATGAAT 648 GGATGTGAGGTTCTGCTGAGGAA 668
    human_3563 ATGGGTATGAATGTGCCTTGAA
    9 IL10RA_NM_001558_ GAATGTGCCTTGAACACAAA 649 TGAGGAAATGGGTATGAATGTGC 669
    human_3579 CTTGAACACAAAGCTCTGTCAA
    10 IL10RA_NM_001558_ GGACACTCAAACACATCATA 650 AGGTATATATTTTCTGGACACTCA 670
    human_2403 AACACATCATAATGGATTCAC
    11 IL10RA_NM_001558_ CTGTATCATAAAGGATTATT 651 CCTAGGAACTCTTTCCTGTATCAT 671
    human_2115 AAAGGATTATTTGCTCAGGGG
    12 IL10RA_NM_001558_ TCACTTCCGAGAGTATGAGA 652 TGAAAGCATCTTCAGTCACTTCCG 672
    human_563 AGAGTATGAGATTGCCATTCG
    13 IL10RA_NM_001558_ TCTCTGGAGCATTCTGAAAA 653 TCTCAGCCCTGCCTTTCTCTGGAG673
    human_3197 CATTCTGAAAACAGATATTCT
    14 IL10RA_NM_001558_ TTATGCCAGAGGCTAACAGA 654 AAGCTGGCTTGTTTCTTATGCCAG 674
    human_2987 AGGCTAACAGATCCAATGGGA
    15 IL10RA_NM_001558_ AGTGGCATTGACTTAGTTCA 655 AGGGGCCAGGATGACAGTGGCA 675
    human_1278 TTGACTTAGTTCAAAACTCTGAG
    16 IL10RA_NM_001558_ TTTCTGGACACTCAAACACA 656 TCTTAAGGTATATATTTTCTGGAC 676
    human_2398 ACTCAAACACATCATAATGGA
    17 IL10RA_NM_001558_ GCATTGCACAGTGAAAGAAT 657 TTTATTGGAGAGGCAGCATTGCA 677
    human_3390 CAGTGAAAGAATTCTGGATATC
    18 IL10RA_NM_001558_ GACCTTGGAGAAGTCACTTA 658 TGTTTCCAGTGGTATGACCTTGGA 678
    human_3468 GAAGTCACTTATCCTCTTGGA
    19 IL10RA_NM_001558_ TCACGTTCACACACAAGAAA 659 AGGTGCCGGGAAACTTCACGTTC 679
    human_610 ACACACAAGAAAGTAAAACATG
    20 IL10RA_NM_001558_ ACTTTGCTGTTTCCAGTGGT 660 GAAATTCTAGCTCTGACTTTGCTG 680
    human_3446 TTTCCAGTGGTATGACCTTGG
    Accession: NM_000214
    HUGO gene symbol: JAG1
    1 JAG1_NM_000214 TATTTGATTTATTAACTTAA 681 ATTAATCACTGTGTATATTTGATTT 701
    _human_4799 ATTAACTTAATAATCAAGAG
    2 JAG1_NM_000214 GAAAAGTAATATTTATTAAA 682 TTGGCAATAAATTTTGAAAAGTAA 702
    _human_5658 TATTTATTAAATTTTTTTGTA
    3 JAG1_NM_000214 ACTTTGTATAGTTATGTAAA 683 AATGTCAAAAGTAGAACTTTGTAT 703
    _human_4752 AGTTATGTAAATAATTCTTTT
    4 JAG1_NM_000214 GAATACTTGAACCATAAAAT 684 TCTAATAAGCTAGTTGAATACTTGA 704
    _human_5418 ACCATAAAATGTCCAGTAAG
    5 JAG1_NM_000214 TCTTGGCAATAAATTTTGAA 685 TCTTTGATGTGTTGTTCTTGGCAAT 705
    _human_5641 AAATTTTGAAAAGTAATATT
    6 JAG1_NM_000214 TTTCTGCTTTAGACTTGAAA 686 TGTTTGTTTTTTGTTTTTCTGCTTTA 706
    _human_5150 GACTTGAAAAGAGACAGGC
    7 JAG1_NM_000214 TATATTTATTGACTCTTGAG 687 GATCATAGTTTTATTTATATTTATT 707
    _human_4526 GACTCTTGAGTTGTTTTTGT
    8 JAG1_NM_000214 TATGATGACGTACAAGTAGT 688 TTTGTATATTGGTTTTATGATGACG 708
    _human_4566 TACAAGTAGTTCTGTATTTG
    9 JAG1_NM_000214 GTGTTGTTCTTGGCAATAAA 689 AAATGCATCTTTGATGTGTTGTTCT 709
    _human_5634 TGGCAATAAATTTTGAAAAG
    10 JAG1_NM_000214 CTGATCTAAAAGGGAATAAA 690 CCTTTTTCCATGCAGCTGATCTAAA 710
    _human_173 AGGGAATAAAAGGCTGCGCA
    11 JAG1_NM_000214 TACGACGTCAGATGTTTAAA 691 GATGGAATTTTTTTGTACGACGTCA 711
    _human_5031 GATGTTTAAAACACCTTCTA
    12 JAG1_NM_000214 AATAATCAAGAGCCTTAAAA 692 TTGATTTATTAACTTAATAATCAAG 712
    _human_4817 AGCCTTAAAACATCATTCCT
    13 JAG1_NM_000214 GTATGAAAACATGGAACAGT 693 TTATTAAATTTTTTTGTATGAAAAC 713
    _human_5685 ATGGAACAGTGTGGCCTCTT
    14 JAG1_NM_000214 TGGTTTTATGATGACGTACA 694 GTTGTTTTTGTATATTGGTTTTATG 714
    _human_4560 ATGACGTACAAGTAGTTCTG
    15 JAG1_NM_000214 TTCTGCTTTAGACTTGAAAA 695 GTTTGTTTTTTGTTTTTCTGCTTTAG 715
    _human_5151 ACTTGAAAAGAGACAGGCA
    16 JAG1_NM_000214 CTTGGCAATAAATTTTGAAA 696 CTTTGATGTGTTGTTCTTGGCAATA 716
    _human_5642 AATTTTGAAAAGTAATATTT
    17 JAG1_NM_000214 TTTAATCTACTGCATTTAGG 697 GATTTGATTTTTTTTTTTAATCTACT 717
    _human_5377 GCATTTAGGGAGTATTCTA
    18 JAG1_NM_000214 TGTATAGTTATGTAAATAAT 698 TCAAAAGTAGAACTTTGTATAGTTA 718
    _human_4756 TGTAAATAATTCTTTTTTAT
    19 JAG1_NM_000214 ATTTATATTTATTGACTCTT 699 TTAGATCATAGTTTTATTTATATTTA 719
    _human_4523 TTGACTCTTGAGTTGTTTT
    20 JAG1_NM_000214 CTTTTCACCATTCGTACATA 700 TGTAAATTCTGATTTCTTTTCACCAT 720
    _human_5325 TCGTACATAATACTGAACC
    Accession: NM_002226
    HUGO gene symbol: JAG2
    1 JAG2_NM_002226 CGTTTCTTTAACCTTGTATA 721 AATGTTTATTTTCTACGTTTCTTTAA 741
    _human_4266 CCTTGTATAAATTATTCAG
    2 JAG2_NM_002226 TAAATGAATGAACGAATAAA 722 GGCAGAACAAATGAATAAATGAAT 742
    _human_5800 GAACGAATAAAAATTTTGACC
    3 JAG2_NM_002226 TCATTCATTTATTCCTTTGT 723 GGTCAAAATTTTTATTCATTCATTT 743
    _human_5450 ATTCCTTTGTTTTGCTTGGT
    4 JAG2_NM_002226 GTAAATGTGTACATATTAAA 724 TGAAAGTGCATTTTTGTAAATGTGT 744
    _human_5021 ACATATTAAAGGAAGCACTC
    5 JAG2_NM_002226 ACCCACGAATACGTATCAAG 725 AGTATAAAATTGCTTACCCACGAAT 745
    _human_5398 ACGTATCAAGGTCTTAAGGA
    6 JAG2_NM_002226 GTTTTATAAAATAGTATAAA 726 AAACAGCTGAAAACAGTTTTATAA 746
    _human_5371 AATAGTATAAAATTGCTTACC
    7 JAG2_NM_002226 CAACTGAGTCAAGGAGCAAA 727 TGAGGGGTAGGAGGTCAACTGAG 747
    _human_5691 TCAAGGAGCAAAGCCAAGAACC
    8 JAG2_NM_002226 ATGTGTACATATTAAAGGAA 728 AGTGCATTTTTGTAAATGTGTACAT 748
    _human_5025 ATTAAAGGAAGCACTCTGTA
    9 JAG2_NM_002226 TTCTTTAACCTTGTATAAAT 729 GTTTATTTTCTACGTTTCTTTAACCT 749
    _human_4269 TGTATAAATTATTCAGTAA
    10 JAG2_NM_002226 ATTTTCTACGTTTCTTTAAC 730 AAAAACCAAATGTTTATTTTCTACG 750
    _human_4258 TTTCTTTAACCTTGTATAAA
    11 JAG2_NM_002226 CAGTTTTATAAAATAGTATA 731 TAAAACAGCTGAAAACAGTTTTAT 751
    _human_5369 AAAATAGTATAAAATTGCTTA
    12 JAG2_NM_002226 GCACAGGCAGAACAAATGAA 732 GAGTGAGGCTGCCTTGCACAGGCA 752
    _human_5780 GAACAAATGAATAAATGAATG
    13 JAG2_NM_002226 TCAGGCTGAAAACAATGGAG 733 ATTATTCAGTAACTGTCAGGCTGA 753
    _human_4302 AAACAATGGAGTATTCTCGGA
    14 JAG2_NM_002226 TAAAATTGCTTACCCACGAA 734 TTTTATAAAATAGTATAAAATTGCT 754
    _human_5387 TACCCACGAATACGTATCAA
    15 JAG2_NM_002226 GTCAGGCTGAAAACAATGGA 735 AATTATTCAGTAACTGTCAGGCTG 755
    _human_4301 AAAACAATGGAGTATTCTCGG
    16 JAG2_NM_002226 AAATGTGTACATATTAAAGG 736 AAAGTGCATTTTTGTAAATGTGTAC 756
    _human_5023 ATATTAAAGGAAGCACTCTG
    17 JAG2_NM_002226 CAGTAACTGTCAGGCTGAAA 737 CTTGTATAAATTATTCAGTAACTGT 757
    _human_4293 CAGGCTGAAAACAATGGAGT
    18 JAG2_NM_002226 GTATTCTCGGATAGTTGCTA 738 GCTGAAAACAATGGAGTATTCTCG 758
    _human_4321 GATAGTTGCTATTTTTGTAAA
    19 JAG2_NM_002226 TCTCACACAAATTCACCAAA 739 AGGCGGAGAAGTTCCTCTCACACA 759
    _human_3994 AATTCACCAAAGATCCTGGCC
    20 JAG2_NM_002226 TTGTTTTGCTTGGTCATTCA 740 CATTCATTTATTCCTTTGTTTTGCTT 760
    _human_5466 GGTCATTCAGAGGCAAGGT
    Accession: NM_001315
    HUGO gene symbol: MAPK14
    1 MAPK14_NM_001315 TCATGCGAAAAGAACCTACA 761 ATTTCAGTCCATCATTCATGCGAAAA 781
    _human_670 GAACCTACAGAGAACTGCG
    2 MAPK14_NM_001315 AAATGTCAGAAGCTTACAGA 762 CTGAACAACATTGTGAAATGTCAGA 782
    _human_833 AGCTTACAGATGACCATGTT
    3 MAPK14_NM_001315 AAACATATGAAACATGAAAA 763 GAACTGCGGTTACTTAAACATATGA 783
    _human_707 AACATGAAAATGTGATTGGT
    4 MAPK14_NM_001315 CAGTTCCTTATCTACCAAAT 764 ACAGATGACCATGTTCAGTTCCTTAT 784
    _human_863 CTACCAAATTCTCCGAGGT
    5 MAPK14_NM_001315 TCCTGGTACAGACCATATTA 765 TGGAAGAACATTGTTTCCTGGTACA 785
    _human_1150 GACCATATTAACCAGCTTCA
    6 MAPK14_NM_001315 TTCCTTATCTACCAAATTCT 766 GATGACCATGTTCAGTTCCTTATCTA 786
    _human_866 CCAAATTCTCCGAGGTCTA
    7 MAPK14_NM_001315 TTCCTGGTACAGACCATATT 767 CTGGAAGAACATTGTTTCCTGGTAC 787
    _human_1149 AGACCATATTAACCAGCTTC
    8 MAPK14_NM_001315 AAGTATATACATTCAGCTGA 768 ATTCTCCGAGGTCTAAAGTATATAC 788
    _human_896 ATTCAGCTGACATAATTCAC
    9 MAPK14_NM_001315 CATTACAACCAGACAGTTGA 769 ATGCTGAACTGGATGCATTACAACC 789
    _human_1076 AGACAGTTGATATTTGGTCA
    10 MAPK14_NM_001315 AGGGACCTAAAACCTAGTAA 770 GCTGACATAATTCACAGGGACCTAA 790
    _human_926 AACCTAGTAATCTAGCTGTG
    11 MAPK14_NM_001315 CTCTGGAGGAATTCAATGAT 771 TTACACCTGCAAGGTCTCTGGAGGA 791
    _human_765 ATTCAATGATGTGTATCTGG
    12 MAPK14_NM_001315 TAAACATATGAAACATGAAA 772 AGAACTGCGGTTACTTAAACATATG 792
    _human_706 AAACATGAAAATGTGATTGG
    13 MAPK14_NM_001315 GATCTGAACAACATTGTGAA 773 CATCTCATGGGGGCAGATCTGAACA 793
    _human_815 ACATTGTGAAATGTCAGAAG
    14 MAPK14_NM_001315 TCAGTTCCTTATCTACCAAA 774 TACAGATGACCATGTTCAGTTCCTTA 794
    _human_862 TCTACCAAATTCTCCGAGG
    15 MAPK14_NM_001315 ATAATTCACAGGGACCTAAA 775 ATACATTCAGCTGACATAATTCACA 795
    _human_917 GGGACCTAAAACCTAGTAAT
    16 MAPK14_NM_001315 CGAGGTCTAAAGTATATACA 776 ATCTACCAAATTCTCCGAGGTCTAA 796
    _human_887 AGTATATACATTCAGCTGAC
    17 MAPK14_NM_001315 GAAATGTCAGAAGCTTACAG 777 TCTGAACAACATTGTGAAATGTCAG 797
    _human_832 AAGCTTACAGATGACCATGT
    18 MAPK14_NM_001315 AGCTGTTGACTGGAAGAACA 778 GATGCATAATGGCCGAGCTGTTGAC 798
    _human_1125 TGGAAGAACATTGTTTCCTG
    19 MAPK14_NM_001315 AAATTCTCCGAGGTCTAAAG 779 AGTTCCTTATCTACCAAATTCTCCGA 799
    _human_879 GGTCTAAAGTATATACATT
    20 MAPK14_NM_001315 AATGTGATTGGTCTGTTGGA 780 CATATGAAACATGAAAATGTGATTG 800
    _human_725 GTCTGTTGGACGTTTTTACA
    Accession: NM_003745
    HUGO gene symbol: SOCS1
    1 SOCS1_NM_003745 CTGCTGTGCAGAATCCTATT 801 TCTGGCTTTATTTTTCTGCTGTGCAGAA 821
    _human_1141 TCCTATTTTATATTTTT
    2 SOCS1_NM_003745 GCTGTGCAGAATCCTATTTT 802 TGGCTTTATTTTTCTGCTGTGCAGAATC 822
    _human_1143 CTATTTTATATTTTTTA
    3 SOCS1_NM_003745 TTAAAGTCAGTTTAGGTAAT 803 CCTATTTTATATTTTTTAAAGTCAGTTT 823
    _human_1170 AGGTAATAAACTTTATT
    4 SOCS1_NM_003745 CTGTGCAGAATCCTATTTTA 804 GGCTTTATTTTTCTGCTGTGCAGAATCC 824
    _human_1144 TATTTTATATTTTTTAA
    5 SOCS1_NM_003745 GTTTACATATACCCAGTATC 805 CTCCTACCTCTTCATGTTTACATATACC 825
    _human_1076 CAGTATCTTTGCACAAA
    6 SOCS1_NM_003745 ATTTTGTTATTACTTGCCTG 806 CTGGGATGCCGTGTTATTTTGTTATTA 826
    _human_837 CTTGCCTGGAACCATGTG
    7 SOCS1_NM_003745 TAACTGGGATGCCGTGTTAT 807 CCGTGCACGCAGCATTAACTGGGATG 827
    _human_819 CCGTGTTATTTTGTTATTA
    8 SOCS1_NM_003745 TGTTATTACTTGCCTGGAAC 808 GATGCCGTGTTATTTTGTTATTACTTGC 828
    _human_841 CTGGAACCATGTGGGTA
    9 SOCS1_NM_003745 TTTCTGCTGTGCAGAATCCT 809 GTCTCTGGCTTTATTTTTCTGCTGTGCA 829
    _human_1138 GAATCCTATTTTATATT
    10 SOCS1_NM_003745 CGTGTTATTTTGTTATTACT 810 CATTAACTGGGATGCCGTGTTATTTTG 830
    _human_831 TTATTACTTGCCTGGAAC
    11 SOCS1_NM_003745 TTTTAAAGTCAGTTTAGGTA 811 ATCCTATTTTATATTTTTTAAAGTCAGT 831
    _human_1168 TTAGGTAATAAACTTTA
    12 SOCS1_NM_003745 TGCTGTGCAGAATCCTATTT 812 CTGGCTTTATTTTTCTGCTGTGCAGAAT 832
    _human_1142 CCTATTTTATATTTTTT
    13 SOCS1_NM_003745 GGATGCCGTGTTATTTTGTT 813 ACGCAGCATTAACTGGGATGCCGTGTT 833
    _human_825 ATTTTGTTATTACTTGCC
    14 SOCS1_NM_003745 TTTAAAGTCAGTTTAGGTAA 814 TCCTATTTTATATTTTTTAAAGTCAGTT 834
    _human_1169 TAGGTAATAAACTTTAT
    15 SOCS1_NM_003745 TAAAGTCAGTTTAGGTAATA 815 CTATTTTATATTTTTTAAAGTCAGTTTA 835
    _human_1171 GGTAATAAACTTTATTA
    16 SOCS1_NM_003745 TCTGCTGTGCAGAATCCTAT 816 CTCTGGCTTTATTTTTCTGCTGTGCAGA 836
    _human_1140 ATCCTATTTTATATTTT
    17 SOCS1_NM_003745 ATATACCCAGTATCTTTGCA 817 CCTCTTCATGTTTACATATACCCAGTAT 837
    _human_1082 CTTTGCACAAACCAGGG
    18 SOCS1_NM_003745 AGAATCCTATTTTATATTTT 818 ATTTTTCTGCTGTGCAGAATCCTATTTT 838
    _human_1150 ATATTTTTTAAAGTCAG
    19 SOCS1_NM_003745 GGTTGTTGTAGCAGCTTAAC 819 CCTCTGGGTCCCCCTGGTTGTTGTAGC 839
    _human_1011 AGCTTAACTGTATCTGGA
    20 SOCS1_NM_003745 CCCAGTATCTTTGCACAAAC 820 TCATGTTTACATATACCCAGTAT 840
    _human_1087 CTTTGCACAAACCAGGGGTTGG
    Accession: NM_003150
    HUGO gene symbol: STAT3
    1 STAT3_NM_003150 ATATTGCTGTATCTACTTTA 841 TTTTTTTTTTTTGGTATATTGCTGT 861
    _human_4897 ATCTACTTTAACTTCCAGAA
    2 STAT3_NM_003150 TGTTTGTTAAATCAAATTAG 842 GTTTCTGTGGAATTCTGTTTGTTA 862
    _human_4325 AATCAAATTAGCTGGTCTCTG
    3 STAT3_NM_003150 TTTATCTAAATGCAAATAAG 843 TGTGGGTGATCTGCTTTTATCTAA 863
    _human_2730 ATGCAAATAAGGATGTGTTCT
    4 STAT3_NM_003150 ATTTTCCTTTGTAATGTATT 844 TTTATAAATAGACTTATTTTCCTTT 864
    _human_3615 GTAATGTATTGGCCTTTTAG
    5 STAT3_NM_003150 TATCAGCACAATCTACGAAG 845 GAGTCGAATGTTCTCTATCAGCAC 865
    _human_453 AATCTACGAAGAATCAAGCAG
    6 STAT3_NM_003150 AGCTTAACTGATAAACAGAA 846 CTTCAGTACATAATAAGCTTAACT 866
    _human_4477 GATAAACAGAATATTTAGAAA
    7 STAT3_NM_003150 GTTGTTGTTGTTCTTAGACA 847 CAGCTTTTTGTTATTGTTGTTGTTG 867
    _human_2870 TTCTTAGACAAGTGCCTCCT
    8 STAT3_NM_003150 GTTGTTGTTCTTAGACAAGT 848 CTTTTTGTTATTGTTGTTGTTGTTC 868
    _human_2873 TTAGACAAGTGCCTCCTGGT
    9 STAT3_NM_003150 TCTGTATTTAAGAAACTTAA 849 TATCAGCATAGCCTTTCTGTATTT 869
    _human_3096 AAGAAACTTAAGCAGCCGGGC
    10 STAT3_NM_003150 TTATTTTCCTTTGTAATGTA 850 TTTTTATAAATAGACTTATTTTCCT 870
    _human_3613 TTGTAATGTATTGGCCTTTT
    11 STAT3_NM_003150 TAACTGATAAACAGAATATT 851 AGTACATAATAAGCTTAACTGATA 871
    _human_4481 AACAGAATATTTAGAAAGGTG
    12 STAT3_NM_003150 ACATTCTGGGCACAAACACA 852 GATCCCGGAAATTTAACATTCTGG 872
    _human_1372 GCACAAACACAAAAGTGATGA
    13 STAT3_NM_003150 GTGATCTGCTTTTATCTAAA 853 AATGAGTGAATGTGGGTGATCTG 873
    _human_2720 CTTTTATCTAAATGCAAATAAG
    14 STAT3_NM_003150 CAGACCCGTCAACAAATTAA 854 GCAGAATCTCAACTTCAGACCCGT 874
    _human_1044 CAACAAATTAAGAAACTGGAG
    15 STAT3_NM_003150 GGAGCTGTTTAGAAACTTAA 855 GGAGGAGAGAATCGTGGAGCTG 875
    _human_1148 TTTAGAAACTTAATGAAAAGTGC
    16 STAT3_NM_003150 ACCATTGGGTTTAAATCATA 856 GTGAGACTTGGGCTTACCATTGG 876
    _human_4523 GTTTAAATCATAGGGACCTAGG
    17 STAT3_NM_003150 GGAGAATCTAAGCATTTTAG 857 AATAGGAAGGTTTAAGGAGAATC 877
    _human_3573 TAAGCATTTTAGACTTTTTTTT
    18 STAT3_NM_003150 CCTTGCTGACATCCAAATAG 858 CATTGCACTTTTTAACCTTGCTGA 878
    _human_2987 CATCCAAATAGAAGATAGGAC
    19 STAT3_NM_003150 AAATTAAGAAATAATAACAA 859 CCTAGGTTTCTTTTTAAATTAAGA 879
    _human_3041 AATAATAACAATTAAAGGGCA
    20 STAT3_NM_003150 TTTTAAATTAAGAAATAATA 860 AAGCCCTAGGTTTCTTTTTAAATT 880
    _human_3037 AAGAAATAATAACAATTAAAG
    Accession: NM_006290
    HUGO gene symbol: TNFAIP3
    1 TNFAIP3_NM_006290 AGCTTGAACTGAGGAGTAAA 881 ACTTCTAAAGAAGTTAGCTTGAAC 901
    _human_3451 TGAGGAGTAAAAGTGTGTACA
    2 TNFAIP3_NM_006290 CCTTTGCAACATCCTCAGAA 882 AATACACATATTTGTCCTTTGCAA 902
    _human_916 CATCCTCAGAAGGCCAATCAT
    3 TNFAIP3_NM_006290 TTCTTTCCAAAGATACCAAA 883 ACGAATCTTTATAATTTCTTTCCAA 903
    _human_4422 AGATACCAAATAAACTTCAG
    4 TNFAIP3_NM_006290 TTATTTTATTACAAACTTCA 884 TGTAATTCACTTTATTTATTTTATT 904
    _human_3688 ACAAACTTCAAGATTATTTA
    5 TNFAIP3_NM_006290 TATTTATACTTATTATAAAA 885 GTGAAAAAAAGTAATTATTTATAC 905
    _human_4536 TTATTATAAAAAGTATTTGAA
    6 TNFAIP3_NM_006290 CATTTCAGACAAAATGCTAA 886 AAGGCCAATCATTGTCATTTCAGA 906
    _human_949 CAAAATGCTAAGAAGTTTGGA
    7 TNFAIP3_NM_006290 ATGAAGGAGAAGCTCTTAAA 887 GATCCTGAAAATGAGATGAAGGA 907
    _human_1214 GAAGCTCTTAAAAGAGTACTTA
    8 TNFAIP3_NM_006290 ATTTTGTGTTGATCATTATT 888 AGTTGATATCTTAATATTTTGTGT 908
    _human_4489 TGATCATTATTTCCATTCTTA
    9 TNFAIP3_NM_006290 TTCATCGAGTACAGAGAAAA 889 TTTTGCACACTGTGTTTCATCGAG 909
    _human_2204 TACAGAGAAAACAAACATTTT
    10 TNFAIP3_NM_006290 TTACTGGGAAGACGTGTAAC 890 AAAAATTAGAATATTTTACTGGGA 910
    _human_3394 AGACGTGTAACTCTTTGGGTT
    11 TNFAIP3_NM_006290 TCATTGAAGCTCAGAATCAG 891 ACTGCCAGAAGTGTTTCATTGAA 911
    _human_2355 GCTCAGAATCAGAGATTTCATG
    12 TNFAIP3_NM_006290 TTCCATTCTTAATGTGAAAA 892 TGTGTTGATCATTATTTCCATTCTT 912
    _human_4508 AATGTGAAAAAAAGTAATTA
    13 TNFAIP3_NM_006290 TGAAGGATACTGCCAGAAGT 893 TGGAAGCACCATGTTTGAAGGAT 913
    _human_2332 ACTGCCAGAAGTGTTTCATTGA
    14 TNFAIP3_NM_006290 CACAAGAGTCAACATTAAAA 894 ATAAATGTAACTTTTCACAAGAGT 914
    _human_4650 CAACATTAAAAAATAAATTAT
    15 TNFAIP3_NM_006290 AATTATTTATACTTATTATA 895 AATGTGAAAAAAAGTAATTATTTA 915
    _human_4533 TACTTATTATAAAAAGTATTT
    16 TNFAIP3_NM_006290 TTCGTGCTTCTCCTTATGAA 896 CATATTCATCGATGTTTCGTGCTT 916
    _human_3907 CTCCTTATGAAACTCCAGCTA
    17 TNFAIP3_NM_006290 TATTTTATTACAAACTTCAA 897 GTAATTCACTTTATTTATTTTATTA 917
    _human_3689 CAAACTTCAAGATTATTTAA
    18 TNFAIP3_NM_006290 TATTACAAACTTCAAGATTA 898 TCACTTTATTTATTTTATTACAAAC 918
    _human_3694 TTCAAGATTATTTAAGTGAA
    19 TNFAIP3_NM_006290 CTCTTAAAGTTGATATCTTA 899 TGTTTTCATCTAATTCTCTTAAAGT 919
    _human_4467 TGATATCTTAATATTTTGTG
    20 TNFAIP3_NM_006290 TTCCAAAGATACCAAATAAA 900 ATCTTTATAATTTCTTTCCAAAGAT 920
    _human_4426 ACCAAATAAACTTCAGTGTT
    Accession: NM_003326
    HUGO gene symbol: TNFSF4
    1 TNFSF4_NM_003326 AATTTGACTTAGCCACTAAC 921 GAGATCAGAATTTTAAATTTGACT 941
    _human_2984 TAGCCACTAACTAGCCATGTA
    2 TNFSF4_NM_003326 GATATTAATAATATAGTTAA 922 GAGAGTATTAATATTGATATTAAT 942
    _human_3422 AATATAGTTAATAGTAATATT
    3 TNFSF4_NM_003326 CTGTGAATGCACATATTAAA 923 TGCTTACAGTGTTATCTGTGAATG 943
    _human_3119 CACATATTAAATGTCTATGTT
    4 TNFSF4_NM_003326 GTTTTCTATTTCCTCTTAAG 924 GGATTTTTTTTTCCTGTTTTCTATT 944
    _human_2208 TCCTCTTAAGTACACCTTCA
    5 TNFSF4_NM_003326 AAATAGCACTAAGAAGTTAT 925 ATTCAATCTGATGTCAAATAGCAC 945
    _human_1727 TAAGAAGTTATTGTGCCTTAT
    6 TNFSF4_NM_003326 CCAATCCCGATCCAAATCAT 926 AATGCTTAAGGGATTCCAATCCC 946
    _human_3311 GATCCAAATCATAATTTGTTCT
    7 TNFSF4_NM_003326 CTATTTAGAGAATGCTTAAG 927 TTAGTTAGATATTTTCTATTTAGA 947
    _human_3286 GAATGCTTAAGGGATTCCAAT
    8 TNFSF4_NM_003326 CAGTTTGCATATTGCCTAAA 928 AGGTTAAATTGATTGCAGTTTGCA 948
    _human_1222 TATTGCCTAAATTTAAACTTT
    9 TNFSF4_NM_003326 CTCGAATTCAAAGTATCAAA 929 TATCACATCGGTATCCTCGAATTC 949
    _human_326 AAAGTATCAAAGTACAATTTA
    10 TNFSF4_NM_003326 ATCTGTGAATGCACATATTA 930 TATGCTTACAGTGTTATCTGTGAA 950
    _human_3117 TGCACATATTAAATGTCTATG
    11 TNFSF4_NM_003326 TTTGTGGGAAAAGAATTGAA 931 TATACATGGCAGAGTTTTGTGGG 951
    _human_2938 AAAAGAATTGAATGAAAAGTCA
    12 TNFSF4_NM_003326 ATTGACCATGTTCTGCAAAA 932 ATTTCACTTTTTGTTATTGACCATG 952
    _human_2537 TTCTGCAAAATTGCAGTTAC
    13 TNFSF4_NM_003326 GATTCTTCATTGCAAGTGAA 933 GGTGGACAGGGCATGGATTCTTC 953
    _human_776 ATTGCAAGTGAAGGAGCCTCCC
    14 TNFSF4_NM_003326 GATGTCAAATAGCACTAAGA 934 TATCAAATTCAATCTGATGTCAAA 954
    _human_1721 TAGCACTAAGAAGTTATTGTG
    15 TNFSF4_NM_003326 GTATACAGGGAGAGTGAGAT 935 AAGAGAGATTTTCTTGTATACAG 955
    _human_1459 GGAGAGTGAGATAACTTATTGT
    16 TNFSF4_NM_003326 GTTGCTATGAGTCAAGGAGT 936 AATGTCTATGTTCTTGTTGCTATG 956
    _human_3152 AGTCAAGGAGTGTAACCTTCT
    17 TNFSF4_NM_003326 TAGTTGAAATGTCCCCTTAA 937 GTATCCCCTTATGTTTAGTTGAAA 957
    _human_1882 TGTCCCCTTAACTTGATATAA
    18 TNFSF4_NM_003326 CTCTGTGCCAAACCTTTTAT 938 GATGATTTGTAACTTCTCTGTGCC 958
    _human_1980 AAACCTTTTATAAACATAAAT
    19 TNFSF4_NM_003326 CTCTGTCTAGAAATACCATA 939 ATGAAAAATAATGATCTCTGTCTA 959
    _human_1770 GAAATACCATAGACCATATAT
    20 TNFSF4_NM_003326 GGTTTCAAGAAATGAGGTGA 940 CACAGAAACATTGCTGGTTTCAA 960
    _human_1680 GAAATGAGGTGATCCTATTATC
    Accession: NM_006293
    HUGO gene symbol: TYRO3
    1 TYRO3_NM_006293 AGTTGCTGTTTAAAATAGAA 961 CATTTCCAAGCTGTTAGTTGCTGTT 981
    _human_3927 TAAAATAGAAATAAAATTGA
    2 TYRO3_NM_006293 CTGTTTAAAATAGAAATAAA 962 CCAAGCTGTTAGTTGCTGTTTAAAA 982
    _human_3932 TAGAAATAAAATTGAAGACT
    3 TYRO3_NM_006293 GGCATCAGCGATGAACTAAA 963 ACATTGGACAGCTTGGGCATCAGC 983
    _human_1731 GATGAACTAAAGGAAAAACTG
    4 TYRO3_NM_006293 AATATCCTAAGACTAACAAA 964 GCTACCAAATCTCAAAATATCCTAA 984
    _human_3699 GACTAACAAAGGCAGCTGTG
    5 TYRO3_NM_006293 GTTGCTGTTTAAAATAGAAA 965 ATTTCCAAGCTGTTAGTTGCTGTTT 985
    _human_3928 AAAATAGAAATAAAATTGAA
    6 TYRO3_NM_006293 AAAATAGAAATAAAATTGAA 966 TGTTAGTTGCTGTTTAAAATAGAAA 986
    _human_3938 TAAAATTGAAGACTAAAGAC
    7 TYRO3_NM_006293 CTGTGAAGCTCACAACCTAA 967 GAGCACCATGTTTTCCTGTGAAGC 987
    _human_842 TCACAACCTAAAAGGCCTGGC
    8 TYRO3_NM_006293 TTGAAGACTAAAGACCTAAA 968 AAAATAGAAATAAAATTGAAGACT 988
    _human_3953 AAAGACCTAAAAAAAAAAAAA
    9 TYRO3_NM_006293 TCCTAAGACTAACAAAGGCA 969 CCAAATCTCAAAATATCCTAAGACT 989
    _human_3703 AACAAAGGCAGCTGTGTCTG
    10 TYRO3_NM_006293 GGACATTTCCAAGCTGTTAG 970 GGTCCTAGCTGTTAGGGACATTTC 990
    _human_3909 CAAGCTGTTAGTTGCTGTTTA
    11 TYRO3_NM_006293 ATGTTTCCATGGTTACCATG 971 AGGAGTGGGGTGGTTATGTTTCCA 991
    _human_3190 TGGTTACCATGGGTGTGGATG
    12 TYRO3_NM_006293 TAGTTGCTGTTTAAAATAGA 972 ACATTTCCAAGCTGTTAGTTGCTGT 992
    _human_3926 TTAAAATAGAAATAAAATTG
    13 TYRO3_NM_006293 AAAATTGAAGACTAAAGACC 973 GTTTAAAATAGAAATAAAATTGAA 993
    _human_3949 GACTAAAGACCTAAAAAAAAA
    14 TYRO3_NM_006293 AGCTGTTAGGGACATTTCCA 974 CATGGGGCGGGTCCTAGCTGTTAG 994
    _human_3900 GGACATTTCCAAGCTGTTAGT
    15 TYRO3_NM_006293 GAGGACGTGTATGATCTCAT 975 CCTCCGGAGTGTATGGAGGACGTG 995
    _human_2511 TATGATCTCATGTACCAGTGC
    16 TYRO3_NM_006293 TTTTAGGTGAGGGTTGGTAA 976 CCTTGTAATATTCCCTTTTAGGTGA 996
    _human_3400 GGGTTGGTAAGGGGTTGGTA
    17 TYRO3_NM_006293 AGCTGACATCATTGCCTCAA 977 TGTGAAGATGCTGAAAGCTGACAT 997
    _human_1895 CATTGCCTCAAGCGACATTGA
    18 TYRO3_NM_006293 AAATCTCAAAATATCCTAAG 978 TCTGAGCACGCTACCAAATCTCAA 998
    _human_3690 AATATCCTAAGACTAACAAAG
    19 TYRO3_NM_006293 AAGCTGTTAGTTGCTGTTTA 979 GTTAGGGACATTTCCAAGCTGTTA 999
    _human_3919 GTTGCTGTTTAAAATAGAAAT
    20 TYRO3_NM_006293 TCCTTGTAATATTCCCTTTT 980 AGTCACAAAGAGATGTCCTTGTAA 1000
    _human_3384 TATTCCCTTTTAGGTGAGGGT
    Accession: NM_000546
    HUGO gene symbol: TP53
    1 TP53_NM_000546_ TGTTTGGGAGATGTAAGAAA 81 TTTTACTGTGAGGGATGTTTGGG 101
    human_1630 AGATGTAAGAAATGTTCTTGCA
    2 TP53_NM_000546_ GCATTGTGAGGGTTAATGAA 82 CCTACCTCACAGAGTGCATTGTGA 102
    human_1808 GGGTTAATGAAATAATGTACA
    3 TP53_NM_000546_ TCGATCTCTTATTTTACAAT 83 TATCCCATTTTTATATCGATCTCTT 103
    human_2538 ATTTTACAATAAAACTTTGC
    4 TP53_NM_000546_ TGTGAGGGTTAATGAAATAA 84 CCTCACAGAGTGCATTGTGAGGG 104
    human_1812 TTAATGAAATAATGTACATCTG
    5 TP53_NM_000546_ GAGTATTTGGATGACAGAAA 85 GGAAATTTGCGTGTGGAGTATTT 105
    human_812 GGATGACAGAAACACTTTTCGA
    6 TP53_NM_000546_ GGATGTTTGGGAGATGTAAG 86 GGTTTTTACTGTGAGGGATGTTTG 106
    human_1627 GGAGATGTAAGAAATGTTCTT
    7 TP53_NM_000546_ GAAATGTTCTTGCAGTTAAG 87 GTTTGGGAGATGTAAGAAATGTT 107
    human_1646 CTTGCAGTTAAGGGTTAGTTTA
    8 TP53_NM_000546_ ATGTACATCTGGCCTTGAAA 88 AGGGTTAATGAAATAATGTACAT 108
    human_1831 CTGGCCTTGAAACCACCTTTTA
    9 TP53_NM_000546_ AGAAATGTTCTTGCAGTTAA 89 TGTTTGGGAGATGTAAGAAATGT 109
    human_1645 TCTTGCAGTTAAGGGTTAGTTT
    10 TP53_NM_000546_ GGTGAACCTTAGTACCTAAA 90 GTCTGACAACCTCTTGGTGAACCT 110
    human_2015 TAGTACCTAAAAGGAAATCTC
    11 TP53_NM_000546_ TAACTTCAAGGCCCATATCT 91 CTGTTGAATTTTCTCTAACTTCAA 111
    human_1753 GGCCCATATCTGTGAAATGCT
    12 TP53_NM_000546_ CTTATCCGAGTGGAAGGAAA 92 GCCCCTCCTCAGCATCTTATCCGA 112
    human_782 GTGGAAGGAAATTTGCGTGTG
    13 TP53_NM_000546_ ATGATCTGGATCCACCAAGA 93 CATCTCTTGTATATGATGATCTGG 113
    human_2086 ATCCACCAAGACTTGTTTTAT
    14 TP53_NM_000546_ AATTTTCTCTAACTTCAAGG 94 TGTCCCTCACTGTTGAATTTTCTCT 114
    human_1744 AACTTCAAGGCCCATATCTG
    15 TP53_NM_000546_ TCTCTTATTTTACAATAAAA 95 CCATTTTTATATCGATCTCTTATTT 115
    human_2542 TACAATAAAACTTTGCTGCC
    16 TP53_NM_000546_ TTATTTTACAATAAAACTTT 96 TTTTATATCGATCTCTTATTTTACA 116
    human_2546 ATAAAACTTTGCTGCCACCT
    17 TP53_NM_000546_ GCCTTGAAACCACCTTTTAT 97 AATAATGTACATCTGGCCTTGAAA 117
    human_1842 CCACCTTTTATTACATGGGGT
    18 TP53_NM_000546_ TATATCGATCTCTTATTTTA 98 TTTATATCCCATTTTTATATCGATC 118
    human_2534 TCTTATTTTACAATAAAACT
    19 TP53_NM_000546_ CCTTAGTACCTAAAAGGAAA 99 CAACCTCTTGGTGAACCTTAGTAC 119
    human_2021 CTAAAAGGAAATCTCACCCCA
    20 TP53_NM_000546_ CATTGTGAGGGTTAATGAAA 100 CTACCTCACAGAGTGCATTGTGA 120
    human_1809 GGGTTAATGAAATAATGTACAT

Claims (25)

1. An immune modulator comprising an sdRNA, wherein the sdRNA comprises a guide strand and a passenger strand, wherein the guide strand is about 19-25 nucleotides long, and the passenger strand is about 10-19 nucleotides long, wherein the sdRNA includes a double stranded region and a single stranded region, wherein the single stranded region includes 5-9 phosphorothioate modifications, wherein the sdRNA is chemically modified, including at least one 2′-O-methyl modification or 2′-fluoro modification, and wherein the sdRNA is capable of suppressing expression of HAVCR2.
2. An immunogenic composition comprising an sdRNA, wherein the sdRNA comprises a guide strand and a passenger strand, wherein the guide strand is about 19-25 nucleotides long, and the passenger strand is about 10-19 nucleotides long, wherein the sdRNA includes a double stranded region and a single stranded region, wherein the single stranded region includes 5-9 phosphorothioate modifications, wherein the sdRNA is chemically modified, including at least one 2′-O-methyl modification or 2′-fluoro modification, wherein the sdRNA is capable of suppressing expression of HAVCR2, and wherein the immunogenic composition further comprises immune cells modified by the sdRNA to suppress expression of HAVCR2.
3. The immunogenic composition of claim 2, wherein the immune cells within the composition are further modified to suppress expression of a plurality of immune checkpoint genes.
4. The immunogenic composition of claim 2, wherein said cells are modified to suppress expression of at least one immune checkpoint gene, and at least one gene selected from the group consisting of: an anti-apoptosis gene, a cytokine receptor gene, and a regulator gene.
5-8. (canceled)
9. The immunogenic composition of claim 3, wherein said modified cells further comprise one or more sdRNA capable of suppressing expression of at least one target gene selected from the group consisting of: CTLA4, PD1/PDCD1, TGFBR1, TGFRBR2, IL10RA, TP53, BAX, BAK1, CASP8, ADORA2A, LAG3, CCL17, CCL22, DLL2, FASLG, CD274, IDO1, ILIORA, JAG1, JAG2, MAPK14, SOCS1, STAT3, TNFAIP3, TYRO3, BTLA, KIR, B7-H3 receptor, and B7-H4 receptor.
10. The immunogenic composition of claim 9, wherein said cells are selected from the group consisting of T-cells, NK-cells, antigen-presenting cells, dendritic cells, stem cells, induced pluripotent stem cells, and/or stem central memory T-cells.
11. The immunogenic composition of claim 9, wherein said cells are T-cells comprising one or more transgene expressing high affinity T-cell receptors (TCR) and/or chimeric antibody-T-cell receptors (CAR).
12. (canceled)
13. The immunogenic composition of claim 9, wherein said cells comprise one or more sdRNA targeting one or more anti-apoptotic genes selected from the group consisting of BAX, BAC, TP53, and Casp8.
14. (canceled)
15. The immunogenic composition of claim 9, wherein said sdRNA comprises at least one 2′-O-methyl modification and/or at least one 2′-O-Fluoro modification, and at least one phosphorothioate modification.
16. The immunogenic composition of claim 9, wherein said sdRNA comprises at least one hydrophobic modification.
17. The immunogenic composition of claim 9, wherein said sdRNA is modified to comprises at least one cholesterol molecule.
18. The immunogenic composition of claim 9, wherein said sdRNA targets a sequence selected from SEQ ID NOs: 1-360 and 381-1000.
19. A method of producing the immunogenic composition of claim 3, said method comprising transforming at least one cell with one or more sdRNA capable of targeting and suppressing expression of one or more immune target genes, wherein at least one sdRNA interferes with HAVCR2 expression.
20. The method of claim 19, wherein the at least one cell comprises an sdRNA that inhibits expression of one or more genes selected from the group consisting of: CTLA4, PD1/PDCD1, TGFBR1, TGFRBR2, IL10RA, TP53, BAX, BAK1, CASP8, ADOARA2A, LAG3, CCL17, CCL22, DLL2, FASLG, CD274, IDO1, ILIORA, JAG1, JAG2, MAPK14, SOCS1, STAT3, TNFAIP3, TYRO3, BTLA, KIR, B7-H3 receptor, and B7-H4 receptor.
21. The method of claim 19, wherein said cells are selected from the group consisting of T-cells, NK-cells, antigen-presenting cells, dendritic cells, stem cells, induced pluripotent stem cells, and/or stem central memory T-cells.
22-24. (canceled)
25. A method for treating a subject having proliferative disease or infectious disease, the method comprising administering to the subject the immune modulator of claim 1.
26. The method of claim 25, wherein said proliferative disease is cancer.
27. The method of claim 25, wherein said infectious disease is a pathogen infection.
28. (canceled)
29. The immune modulator of claim 1, wherein at least one sdRNA targets a sequence selected from SEQ ID NOs: 361-380.
30. The immunogenic composition of claim 2, wherein at least one sdRNA targets a sequence selected from SEQ ID NOs: 361-380.
US17/150,668 2013-12-02 2021-01-15 Immunotherapy of cancer Abandoned US20210348166A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/150,668 US20210348166A1 (en) 2013-12-02 2021-01-15 Immunotherapy of cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361910728P 2013-12-02 2013-12-02
PCT/US2014/068244 WO2015084897A2 (en) 2013-12-02 2014-12-02 Immunotherapy of cancer
US201615100536A 2016-05-31 2016-05-31
US17/150,668 US20210348166A1 (en) 2013-12-02 2021-01-15 Immunotherapy of cancer

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/068244 Continuation WO2015084897A2 (en) 2013-12-02 2014-12-02 Immunotherapy of cancer
US15/100,536 Continuation US10934550B2 (en) 2013-12-02 2014-12-02 Immunotherapy of cancer

Publications (1)

Publication Number Publication Date
US20210348166A1 true US20210348166A1 (en) 2021-11-11

Family

ID=53274274

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/100,536 Active US10934550B2 (en) 2013-12-02 2014-12-02 Immunotherapy of cancer
US17/150,668 Abandoned US20210348166A1 (en) 2013-12-02 2021-01-15 Immunotherapy of cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/100,536 Active US10934550B2 (en) 2013-12-02 2014-12-02 Immunotherapy of cancer

Country Status (6)

Country Link
US (2) US10934550B2 (en)
EP (1) EP3079707A4 (en)
JP (3) JP6772062B2 (en)
CN (2) CN106061488B (en)
RU (1) RU2744194C2 (en)
WO (1) WO2015084897A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11396654B2 (en) 2008-09-22 2022-07-26 Phio Pharmaceuticals Corp. Neutral nanotransporters
US11584933B2 (en) 2010-03-24 2023-02-21 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US11667915B2 (en) 2009-02-04 2023-06-06 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US11926828B2 (en) 2014-09-05 2024-03-12 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting TYR or MMP1

Families Citing this family (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104975020B (en) 2008-02-11 2020-01-17 菲奥医药公司 Modified RNAi polynucleotides and uses thereof
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
US20110268761A1 (en) 2010-03-02 2011-11-03 Hapten Pharmaceuticals, Llc Effective Sensitizing Dose of a Gelled Immunomodulating Topical Composition
WO2011119887A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Rna interference in dermal and fibrotic indications
RU2615143C2 (en) 2010-03-24 2017-04-04 Адвирна Self-delivered rnai compounds of reduced size
EP4385568A2 (en) 2010-04-06 2024-06-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of cd274/pd-l1 gene
EP3079707A4 (en) * 2013-12-02 2017-10-18 RXi Pharmaceuticals Corporation Immunotherapy of cancer
CA2947270A1 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic acids targeting mdm2 or mycn
EP3261669B1 (en) 2015-02-25 2022-08-03 Memorial Sloan Kettering Cancer Center Use of inactivated nonreplicating modified vaccinia virus ankara (mva)as monoimmunotherapy or in combination with immune checkpoint blocking agents for solid tumors
US10548930B2 (en) 2015-04-17 2020-02-04 Memorial Sloan Kettering Cancer Center Use of MVA or MVAΔE3L as immunotherapeutic agents against solid tumors
EP3317399B1 (en) * 2015-06-30 2024-06-26 Cellectis Methods for improving functionality in nk cell by gene inactivation using specific endonuclease
JP6983752B2 (en) 2015-07-06 2021-12-17 フィオ ファーマシューティカルズ コーポレーションPhio Pharmaceuticals Corp. Nucleic acid molecule targeting superoxide dismutase 1 (SOD1)
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
CN108472314A (en) 2015-07-31 2018-08-31 明尼苏达大学董事会 The cell and therapy of modification
JP7028764B2 (en) * 2015-09-02 2022-03-02 アルナイラム ファーマシューティカルズ, インコーポレイテッド Programmed cell death 1 Ligand 1 (PD-L1) iRNA composition and its usage
WO2017070151A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
KR20170054262A (en) 2015-11-09 2017-05-17 사회복지법인 삼성생명공익재단 SOCS suppressed stem cell with increased immunosuppression and use thereof
WO2017082562A1 (en) * 2015-11-09 2017-05-18 사회복지법인 삼성생명공익재단 Stem cell with suppressed socs and improved immunosuppressive ability and use thereof
US10982215B2 (en) 2015-12-09 2021-04-20 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting programmed cell death 1 ligand 1 (PD-L1) and methods of use thereof
CN107034193B (en) * 2016-02-03 2020-06-05 北京马力喏生物科技有限公司 Therapeutic compositions for the treatment of B-cell leukemia and B-cell lymphoma
SG11201807051VA (en) 2016-02-25 2018-09-27 Memorial Sloan Kettering Cancer Center Replication competent attenuated vaccinia viruses with deletion of thymidine kinase with and without the expression of human flt3l or gm-csf for cancer immunotherapy
CN109152827B (en) 2016-02-25 2023-07-21 纪念斯隆凯特琳癌症中心 Recombinant MVA or MVA delta E3L expressing human FLT3L and use thereof as immunotherapeutic agent against solid tumors
WO2018009972A1 (en) * 2016-07-14 2018-01-18 Peter Maccallum Cancer Institute Chimeric antigen receptor modified t cells
JP2018052930A (en) * 2016-09-21 2018-04-05 千葉県 Novel alkylating agent
AU2017336094A1 (en) * 2016-09-29 2019-04-18 Immunitybio, Inc. HLA class I-deficient NK-92 cells with decreased immunogenicity
CN110191952A (en) * 2016-10-07 2019-08-30 瑟卡尔纳制药有限公司 The new method for the treatment of cancer
KR101971322B1 (en) * 2016-10-17 2019-04-23 사회복지법인 삼성생명공익재단 Methods for Selecting Improved Stem Cell Using SOCS Inhibition
AU2017346885B2 (en) 2016-10-18 2021-10-21 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US11332713B2 (en) 2016-11-16 2022-05-17 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
CN107058315B (en) * 2016-12-08 2019-11-08 上海优卡迪生物医药科技有限公司 Strike the siRNA for subtracting people PD-1, recombinant expression CAR-T carrier and its construction method and application
US11242509B2 (en) 2017-05-12 2022-02-08 Memorial Sloan Kettering Cancer Center Vaccinia virus mutants useful for cancer immunotherapy
WO2019006418A2 (en) 2017-06-30 2019-01-03 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
EP3664817A4 (en) * 2017-08-07 2021-09-22 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
CR20200251A (en) 2017-11-17 2020-07-17 Iovance Biotherapeutics Inc Til expansion from fine needle aspirates and small biopsies
EP3714041A1 (en) 2017-11-22 2020-09-30 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
CN111356476B (en) * 2017-12-22 2023-03-10 江苏恒瑞医药股份有限公司 LAG-3 antibody pharmaceutical composition and application thereof
SG11202006541UA (en) 2018-01-08 2020-08-28 Iovance Biotherapeutics Inc Processes for generating til products enriched for tumor antigen-specific t-cells
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
WO2019136459A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
CN112040986A (en) 2018-03-15 2020-12-04 Ksq治疗公司 Gene regulatory compositions and methods for improved immunotherapy
AU2019244479A1 (en) 2018-03-30 2020-11-12 Les Hopitaux Universitaires De Geneve Micro RNA expression constructs and uses thereof
CN110904045A (en) * 2018-09-17 2020-03-24 中国科学院动物研究所 Modified T cells, methods of making and uses thereof
AU2019377422A1 (en) 2018-11-05 2021-05-27 Iovance Biotherapeutics, Inc. Treatment of NSCLC patients refractory for anti-PD-1 antibody
JP2022506586A (en) 2018-11-05 2022-01-17 アイオバンス バイオセラピューティクス,インコーポレイテッド Process for the production of tumor-infiltrating lymphocytes and their use in immunotherapy
SG11202104630PA (en) 2018-11-05 2021-06-29 Iovance Biotherapeutics Inc Selection of improved tumor reactive t-cells
EP3877511A1 (en) 2018-11-05 2021-09-15 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathway inhibitors
JP2022514023A (en) 2018-12-19 2022-02-09 アイオバンス バイオセラピューティクス,インコーポレイテッド Methods and Uses for Expanding Tumor-Infiltrating Lymphocytes Using Manipulated Cytokine Receptor Pairs
EP3931310A1 (en) 2019-03-01 2022-01-05 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
AU2020252372A1 (en) * 2019-04-02 2021-10-28 Board Of Regents, The University Of Texas System Combinations of transcription inhibitors and immune checkpoint inhibitors for treatment of disease
US20220249559A1 (en) 2019-05-13 2022-08-11 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
CN114174512A (en) * 2019-05-20 2022-03-11 奥利克斯医药有限公司 Asymmetric siRNA inhibiting PD-1 expression
CN110354260B (en) * 2019-08-19 2020-06-26 启辰生生物科技(珠海)有限公司 Immunopotentiator, pharmaceutical composition and application thereof
WO2021064235A1 (en) * 2019-10-04 2021-04-08 Secarna Pharmaceuticals Gmbh & Co. Kg Oligonucleotide based ex vivo cell therapy
EP4055167A2 (en) 2019-11-08 2022-09-14 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides targeting bromodomain containing protein 4 (brd4) for immunotherapy
CA3157344A1 (en) * 2019-11-20 2021-05-27 Cartherics Pty. Ltd. Method for providing immune cells with enhanced function
US20230220341A1 (en) 2019-12-11 2023-07-13 lovance Biotherapeutics, Inc. Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same
EP4085136A1 (en) 2019-12-31 2022-11-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides with improved systemic delivery
KR20230016623A (en) * 2020-04-30 2023-02-02 세카나 파머씨티컬스 지엠비에이치 엔 씨오. 케이지 PD-1-specific antisense oligonucleotides and their use in therapy
EP4146794A1 (en) 2020-05-04 2023-03-15 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
JP2023524108A (en) 2020-05-04 2023-06-08 アイオバンス バイオセラピューティクス,インコーポレイテッド Selection of improved tumor-reactive T cells
JP7450292B2 (en) * 2020-07-03 2024-03-15 レモネックス インコーポレイテッド RNA and nucleic acid carriers containing it
US20230372397A1 (en) 2020-10-06 2023-11-23 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
TW202241468A (en) 2020-12-11 2022-11-01 美商艾歐凡斯生物治療公司 Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with braf inhibitors and/or mek inhibitors
EP4262827A1 (en) 2020-12-17 2023-10-25 Iovance Biotherapeutics, Inc. Treatment of cancers with tumor infiltrating lymphocytes
JP2023554395A (en) 2020-12-17 2023-12-27 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment with tumor-infiltrating lymphocyte therapy in combination with CTLA-4 and PD-1 inhibitors
WO2022170219A1 (en) 2021-02-05 2022-08-11 Iovance Biotherapeutics, Inc. Adjuvant therapy for cancer
BR112023021665A2 (en) 2021-04-19 2023-12-19 Iovance Biotherapeutics Inc METHOD FOR TREATING A CANCER, AND, COMPOSITION
TW202327631A (en) 2021-07-28 2023-07-16 美商艾歐凡斯生物治療公司 Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
WO2023015264A1 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Immunotherapy of cancer utilizing natural killer cells treated with chemically modified oligonucleotides
CN118043459A (en) 2021-08-04 2024-05-14 菲奥医药公司 Chemically modified oligonucleotides
CA3235824A1 (en) 2021-10-27 2023-05-04 Frederick G. Vogt Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
AU2022388729A1 (en) 2021-11-10 2024-05-16 Iovance Biotherapeutics, Inc. Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
EP4282963A1 (en) 2022-05-23 2023-11-29 Eberhard Karls Universität Tübingen, Medizinische Fakultät Nucleic acid modified biological cell with expansion-dependent gene expression
WO2024030758A1 (en) 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
WO2024033896A1 (en) * 2022-08-12 2024-02-15 Biorchestra Co., Ltd. Oligonucleotides targeting indoleamine 2,3-dioxygenase and uses thereof
WO2024064769A1 (en) * 2022-09-21 2024-03-28 Phio Pharmaceuticals Corp. Induction of stem-like activated t cells
WO2024098024A1 (en) 2022-11-04 2024-05-10 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof

Family Cites Families (258)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4201860A (en) 1978-05-09 1980-05-06 Bristol-Myers Company Purine derivatives
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4426330A (en) 1981-07-20 1984-01-17 Lipid Specialties, Inc. Synthetic phospholipid compounds
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US5643889A (en) 1984-07-11 1997-07-01 Temple University-Of The Commonwealth System Of Pennsylvania Cholesterol conjugates of 2'5'-oligoadenylate derivatives and antiviral uses thereof
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5750666A (en) 1988-05-26 1998-05-12 Competitve Technologies, Inc. Polynucleotide phosphorodithioate compounds
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5527528A (en) 1989-10-20 1996-06-18 Sequus Pharmaceuticals, Inc. Solid-tumor treatment method
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5495009A (en) 1989-10-24 1996-02-27 Gilead Sciences, Inc. Oligonucleotide analogs containing thioformacetal linkages
WO1991006556A1 (en) 1989-10-24 1991-05-16 Gilead Sciences, Inc. 2' modified oligonucleotides
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5514786A (en) 1990-01-11 1996-05-07 Isis Pharmaceuticals, Inc. Compositions for inhibiting RNA activity
US6395492B1 (en) 1990-01-11 2002-05-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US6399754B1 (en) 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US6153737A (en) 1990-01-11 2000-11-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5914396A (en) 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US6358931B1 (en) 1990-01-11 2002-03-19 Isis Pharmaceuticals, Inc. Compositions and methods for modulating RNA
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5658731A (en) 1990-04-09 1997-08-19 Europaisches Laboratorium Fur Molekularbiologie 2'-O-alkylnucleotides as well as polymers which contain such nucleotides
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
ATE154246T1 (en) 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc NUCLEASE RESISTANT PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECTE AND MODULATE GENE EXPRESSION
WO1992003464A1 (en) 1990-08-28 1992-03-05 Microprobe Corporation Solid support synthesis of 3'-tailed oligonucleotides via a linking molecule
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
EP0549686A4 (en) 1990-09-20 1995-01-18 Gilead Sciences Inc Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
CA2093664C (en) 1990-10-12 2003-07-29 Fritz Eckstein Modified ribozymes
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5607923A (en) 1991-10-15 1997-03-04 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating cytomegalovirus having phosphorothioate linkages of high chiral purity
US5661134A (en) 1991-10-15 1997-08-26 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating Ha-ras or Ki-ras having phosphorothioate linkages of high chiral purity
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5856455A (en) 1991-12-24 1999-01-05 Isis Pharmaceuticals, Inc. Gapped 2'-modified oligonucleotides
IL104461A (en) 1992-01-22 2001-05-20 Hoechst Ag Oligonucleotide analogs, their preparation and pharmaceutical compositions containing them
US5652094A (en) 1992-01-31 1997-07-29 University Of Montreal Nucleozymes
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5817781A (en) 1992-06-01 1998-10-06 Gilead Sciences, Inc. Modified internucleoside linkages (II)
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US6346614B1 (en) 1992-07-23 2002-02-12 Hybridon, Inc. Hybrid oligonucleotide phosphorothioates
JP3351476B2 (en) 1993-01-22 2002-11-25 三菱化学株式会社 Phospholipid derivatives and liposomes containing the same
HU9501974D0 (en) 1993-03-31 1995-09-28 Sterling Winthrop Inc Oligonucleotides with amide linkages replacing phosphodiester linkages
CN1124980A (en) 1993-03-31 1996-06-19 海布里顿公司 Modified oligonucleotides having improved anti-influenza activity
ES2128535T3 (en) 1993-05-12 1999-05-16 Novartis Ag NUCLEOSIDES AND OLIGONUCLEOTIDES WITH 2'-ETER GROUPS.
FR2705099B1 (en) 1993-05-12 1995-08-04 Centre Nat Rech Scient Phosphorothioate triester oligonucleotides and process for their preparation.
US6015886A (en) 1993-05-24 2000-01-18 Chemgenes Corporation Oligonucleotide phosphate esters
US5580972A (en) 1993-06-14 1996-12-03 Nexstar Pharmaceuticals, Inc. Purine nucleoside modifications by palladium catalyzed methods
US5428149A (en) 1993-06-14 1995-06-27 Washington State University Research Foundation Method for palladium catalyzed carbon-carbon coulping and products
US5534259A (en) 1993-07-08 1996-07-09 Liposome Technology, Inc. Polymer compound and coated particle composition
US5532130A (en) 1993-07-20 1996-07-02 Dyad Pharmaceutical Corporation Methods and compositions for sequence-specific hybridization of RNA by 2'-5' oligonucleotides
US5614621A (en) 1993-07-29 1997-03-25 Isis Pharmaceuticals, Inc. Process for preparing oligonucleotides using silyl-containing diamino phosphorous reagents
US5417978A (en) 1993-07-29 1995-05-23 Board Of Regents, The University Of Texas System Liposomal antisense methyl phosphonate oligonucleotides and methods for their preparation and use
PT748382E (en) 1993-09-02 2003-03-31 Ribozyme Pharm Inc NUCLEIC ENZYMAL ACIDS CONTAINING NON-NUCLEOTIDES
CA2170869C (en) 1993-09-03 1999-09-14 Phillip Dan Cook Amine-derivatized nucleosides and oligonucleosides
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
JPH09504297A (en) 1993-10-27 1997-04-28 リボザイム・ファーマシューティカルズ・インコーポレーテッド 2'-amide and 2'-peptide modified oligonucleotides
AU687492B2 (en) 1993-11-16 1998-02-26 Genta Incorporated Synthetic oligomers having phosphonate internucleosidyl linkages of undefined chirality mixed with non-phosphonate internucleosidyl linkages
US5646126A (en) 1994-02-28 1997-07-08 Epoch Pharmaceuticals Sterol modified oligonucleotide duplexes having anticancer activity
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5777153A (en) 1994-07-08 1998-07-07 Gilead Sciences, Inc. Cationic lipids
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5591721A (en) 1994-10-25 1997-01-07 Hybridon, Inc. Method of down-regulating gene expression
US5681940A (en) 1994-11-02 1997-10-28 Icn Pharmaceuticals Sugar modified nucleosides and oligonucleotides
US5948767A (en) 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5830430A (en) 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
ATE285477T1 (en) 1995-06-07 2005-01-15 Inex Pharmaceutical Corp PRODUCTION OF LIPID-NUCLIC ACID PARTICLES USING A HYDROPHOBIC LIPID-NUCLIC ACID COMPLEX INTERMEDIATE PRODUCT AND FOR USE IN GENE TRANSFER
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
FR2738151B1 (en) 1995-09-04 1997-09-26 Rhone Poulenc Rorer Sa ANTAGONISTS OF THE ONCOGENIC ACTIVITY OF THE MDM2 PROTEIN, AND THEIR USE IN THE TREATMENT OF CANCERS
US5734041A (en) 1995-10-20 1998-03-31 Mcgill University Preparation of chiral phosphorothioate oligomers
US5705621A (en) 1995-11-17 1998-01-06 Isis Pharmaceuticals, Inc. Oligomeric phosphite, phosphodiester, Phosphorothioate and phosphorodithioate compounds and intermediates for preparing same
US5684143A (en) 1996-02-21 1997-11-04 Lynx Therapeutics, Inc. Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
US6331617B1 (en) 1996-03-21 2001-12-18 University Of Iowa Research Foundation Positively charged oligonucleotides as regulators of gene expression
US6444806B1 (en) 1996-04-30 2002-09-03 Hisamitsu Pharmaceutical Co., Inc. Conjugates and methods of forming conjugates of oligonucleotides and carbohydrates
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US5789416B1 (en) 1996-08-27 1999-10-05 Cv Therapeutics Inc N6 mono heterocyclic substituted adenosine derivatives
US6111085A (en) 1996-09-13 2000-08-29 Isis Pharmaceuticals, Inc. Carbamate-derivatized nucleosides and oligonucleosides
US6977244B2 (en) 1996-10-04 2005-12-20 Board Of Regents, The University Of Texas Systems Inhibition of Bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
US7789841B2 (en) 1997-02-06 2010-09-07 Exogen, Inc. Method and apparatus for connective tissue treatment
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
AU7499198A (en) 1997-05-21 1998-12-11 Johns Hopkins University, The Gene expression profiles in normal and cancer cells
DE19727932A1 (en) 1997-07-01 1999-01-07 Invitek Gmbh Method and test kit for the detection of malignant tumors
US6013786A (en) 1997-08-22 2000-01-11 Hybridon, Inc. MDM2-specific antisense oligonucleotides
US6028183A (en) 1997-11-07 2000-02-22 Gilead Sciences, Inc. Pyrimidine derivatives and oligonucleotides containing same
AU1507199A (en) 1997-12-15 1999-07-05 Yamanouchi Pharmaceutical Co., Ltd. Novel pyrimidine-5-carboxamide derivatives
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6020475A (en) 1998-02-10 2000-02-01 Isis Pharmeuticals, Inc. Process for the synthesis of oligomeric compounds
JP2003525017A (en) 1998-04-20 2003-08-26 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleic acid molecules with novel chemical composition that can regulate gene expression
WO1999060012A1 (en) 1998-05-21 1999-11-25 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US20040241845A1 (en) 1998-05-22 2004-12-02 Luc Desgroseillers Mammalian staufen and use thereof
US6277967B1 (en) 1998-07-14 2001-08-21 Isis Pharmaceuticals, Inc. Carbohydrate or 2′-modified oligonucleotides having alternating internucleoside linkages
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6271358B1 (en) 1998-07-27 2001-08-07 Isis Pharmaceuticals, Inc. RNA targeted 2′-modified oligonucleotides that are conformationally preorganized
US20040009938A1 (en) 1998-08-07 2004-01-15 Muthiah Manoharan Methods of enhancing renal uptake of oligonucleotides
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
US6020483A (en) 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6455586B1 (en) 1998-12-15 2002-09-24 Leonard L. Kaplan Topical immunomodulating compositions for treatment of aids, Hepatitis B & C, other infectious diseases, and cancer
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US6465628B1 (en) 1999-02-04 2002-10-15 Isis Pharmaceuticals, Inc. Process for the synthesis of oligomeric compounds
US6207819B1 (en) 1999-02-12 2001-03-27 Isis Pharmaceuticals, Inc. Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
NZ513402A (en) 1999-02-12 2003-06-30 Sankyo Co Novel nucleosides and oligonucleotide analogues
US6121437A (en) 1999-03-16 2000-09-19 Isis Pharmaceuticals, Inc. Phosphate and thiophosphate protecting groups
KR100816572B1 (en) 1999-04-28 2008-03-24 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Anti-VEGF antibody and a pharmaceutical composition comprising the same
GB9925459D0 (en) 1999-10-27 1999-12-29 Plant Bioscience Ltd Gene silencing
DE10160151A1 (en) 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
US7829693B2 (en) 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
US20050032733A1 (en) 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
EP1272630A2 (en) 2000-03-16 2003-01-08 Genetica, Inc. Methods and compositions for rna interference
AU2001249622B2 (en) 2000-03-30 2007-06-07 Massachusetts Institute Of Technology RNA sequence-specific mediators of RNA interference
AU2001256865A1 (en) 2000-05-11 2001-11-20 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Myc targets
EP1303495B1 (en) 2000-07-24 2010-05-26 Krenitsky Pharmaceuticals, Inc. Substituted 5-alkynyl pyrimidines having neurotrophic activity
US20020132788A1 (en) 2000-11-06 2002-09-19 David Lewis Inhibition of gene expression by delivery of small interfering RNA to post-embryonic animal cells in vivo
HU230458B1 (en) 2000-12-01 2016-07-28 Europäisches Laboratorium für Molekularbiologie (EMBL) Rna interference mediating small rna molecules
FR2818642B1 (en) 2000-12-26 2005-07-15 Hoechst Marion Roussel Inc NOVEL DERIVATIVES OF PURINE, PROCESS FOR PREPARING THEM, THEIR USE AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND THEIR NEW USE
JP2004532616A (en) 2000-12-28 2004-10-28 ジョンソン・アンド・ジョンソン・リサーチ・ピー・ティー・ワイ・リミテッド Double-stranded RNA-mediated gene suppression
US7906492B2 (en) 2001-01-16 2011-03-15 Sloan-Kettering Institute For Cancer Research Therapy-enhancing glucan
US20030077829A1 (en) 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
EP1627061B1 (en) 2001-05-18 2009-08-12 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING CHEMICALLY MODIFIED SHORT INTERFERING NUCLEIC ACID (siNA)
US20070032441A1 (en) 2001-05-18 2007-02-08 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (sina)
WO2003070884A2 (en) 2002-02-20 2003-08-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF MDR P-GLYCOPROTEIN GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
DE10133858A1 (en) 2001-07-12 2003-02-06 Aventis Pharma Gmbh Synthetic double-stranded oligonucleotides for targeted inhibition of gene expression
AU2002329667A1 (en) 2001-07-30 2003-02-17 Uta Griesenbach Specific inhibition of gene expression by small double stranded rnas
EP2325193A3 (en) 2001-11-02 2012-05-02 Insert Therapeutics, Inc. Methods and compositions for therapeutic use of RNA interference
WO2003064625A2 (en) 2002-02-01 2003-08-07 Sequitur, Inc. Oligonucleotide compositions with enhanced efficiency
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
AU2003202376B2 (en) 2002-02-01 2008-10-30 Mcgill University Oligonucleotides comprising alternating segments and uses thereof
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
WO2003099840A1 (en) 2002-05-24 2003-12-04 Isis Pharmaceuticals, Inc. Oligonucleotides having modified nucleoside units
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
AU2003260370B2 (en) 2002-08-05 2008-05-22 Silence Therapeutics Gmbh Further novel forms of interfering RNA molecules
WO2004027030A2 (en) 2002-09-18 2004-04-01 Isis Pharmaceuticals, Inc. Efficient reduction of target rna’s by single- and double-stranded oligomeric compounds
CA2881743A1 (en) 2002-09-25 2004-04-08 University Of Massachusetts In vivo gene silencing by chemically modified and stable sirna
AU2003304386A1 (en) 2002-10-30 2005-02-25 The Center For Blood Research, Inc. Methods for treating and preventing apoptosis-related diseases using rna interfering agents
EP1562970A4 (en) 2002-11-01 2006-04-12 Univ Pennsylvania COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF HIF-1 ALPHA
US9150605B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
JP2006507841A (en) 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA
WO2004065600A2 (en) 2003-01-17 2004-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference by palindromic or modified rna molecules
DE10302421A1 (en) 2003-01-21 2004-07-29 Ribopharma Ag New double-stranded interfering RNA, useful for inhibiting hepatitis C virus, has one strand linked to a lipophilic group to improve activity and eliminate the need for transfection auxiliaries
US20040167090A1 (en) 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
DK2216407T3 (en) 2003-03-07 2016-03-29 Alnylam Pharmaceuticals Inc therapeutic compositions
WO2004090105A2 (en) 2003-04-02 2004-10-21 Dharmacon, Inc. Modified polynucleotides for use in rna interference
ES2702942T3 (en) 2003-04-17 2019-03-06 Alnylam Pharmaceuticals Inc Modified RNAi agents
US20060178327A1 (en) 2003-05-30 2006-08-10 Yeung Wah Hin A Inhibition of gene expression by delivery of specially selected double stranded or other forms of small interfering RNA precursors enabling the formation and function of small interfering RNA in vivo and in vitro
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US8309704B2 (en) 2003-06-02 2012-11-13 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
DK3604537T3 (en) 2003-06-13 2022-02-28 Alnylam Europe Ag Double-stranded ribonucleic acid with increased efficiency in an organism
US20050106594A1 (en) 2003-08-22 2005-05-19 Andrew Ellington In vitro selection of aptamer beacons
EP2123759B1 (en) 2004-02-06 2014-01-15 Thermo Fisher Scientific Biosciences Inc. Stabilized RNAS as transfection controls and silencing reagents
US20060069050A1 (en) 2004-02-17 2006-03-30 University Of Massachusetts Methods and compositions for mediating gene silencing
US20080071068A1 (en) 2004-02-20 2008-03-20 Hideki Oba Cytoplasmic Localization Dna and Rna
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
KR101147147B1 (en) 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
WO2005096781A2 (en) 2004-04-06 2005-10-20 University Of Massachusetts Methods and compositions for treating gain-of-function disorders using rna interference
WO2006019430A2 (en) 2004-04-20 2006-02-23 Nastech Pharmaceutical Company Inc. Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
EP1765415A4 (en) 2004-06-03 2010-03-24 Isis Pharmaceuticals Inc Oligomeric compounds that facilitate risc loading
US20060211766A1 (en) 2004-09-30 2006-09-21 Kaplan Leonard L Gelled immunomodulating topical compositions and a method of treating warts and other human papilloma virus skin infections
EP1796732B1 (en) 2004-10-01 2013-10-30 Novartis Vaccines and Diagnostics, Inc. Modified small interfering rna molecules and methods of use
US20100069620A1 (en) 2004-12-02 2010-03-18 Rxi Pharmaceuticals Corp. Novel compositions of chemically modified small interfering rna
CA2589084A1 (en) 2004-12-15 2006-06-22 Merck & Co., Inc. Inhibitors of akt activity
US20060142228A1 (en) 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
WO2006078979A2 (en) * 2005-01-20 2006-07-27 Nature Technology Corp. Vectors and methods for genetic immunization
WO2006113679A2 (en) 2005-04-15 2006-10-26 Board Of Regents, The University Of Texas System Delivery of sirna by neutral lipid compositions
CA2604333A1 (en) * 2005-04-15 2006-10-19 Cenix Bioscience Gmbh Human marker genes and agents for cardiovascular disorders and artherosclerosis
WO2006128141A2 (en) 2005-05-27 2006-11-30 Sirna Therapeutics, Inc. Rna interference mediated inhibition of stromal cell-derived factor-1 (sdf-1) gene expression using short interfering nucleic acid (sina)
PT1888033E (en) 2005-06-09 2014-05-15 Meda Ab Method and composition for treating inflammatory disorders
CA2612200A1 (en) * 2005-06-15 2006-12-21 London Health Sciences Centre Research Inc. Method of cancer treatment using sirna silencing
WO2007030167A1 (en) 2005-09-02 2007-03-15 Nastech Pharmaceutical Company Inc. Modification of double-stranded ribonucleic acid molecules
US8603991B2 (en) 2005-11-18 2013-12-10 Gradalis, Inc. Individualized cancer therapy
WO2007069068A2 (en) 2005-12-16 2007-06-21 Diatos Cell penetrating peptide conjugates for delivering nucleic acids into cells
US7825099B2 (en) 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
US20070231392A1 (en) 2006-01-23 2007-10-04 Ernst Wagner CHEMICALLY MODIFIED POLYCATION POLYMER FOR siRNA DELIVERY
EP1986609A2 (en) 2006-01-26 2008-11-05 University of Massachusetts Rna silencing agents for use in therapy and nanotransporters for efficient delivery of same
CN101432022A (en) * 2006-02-28 2009-05-13 肿瘤疗法科学股份有限公司 Methods for damaging cells using effector functions of anti-epha4 antibodies
US20100035973A1 (en) * 2006-07-17 2010-02-11 Nationwide Children's Hospital, Inc. Disruption of programmed death 1 (pd-1) ligand to adjuvant adeno-associated virus vector vaccines
US8129340B2 (en) 2006-09-08 2012-03-06 Institut Gustave Roussy Inhibitors of protein phosphatase 1, GADD34 and protein phosphatase 1/GADD34 complex, preparation and uses thereof
JP2010502970A (en) 2006-09-08 2010-01-28 アンスティテュ・グスターブ・ルシ Compounds that modulate cell surface exposure of calreticulin, KDEL receptor and / or ERp57 and their use to evaluate the efficacy of cancer treatment
CA2663601C (en) 2006-09-22 2014-11-25 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
US20090010948A1 (en) * 2006-12-15 2009-01-08 The University Of Hong Kong Anti-tumor vaccines delivered by dendritic cells devoid of interleukin-10
RU2478400C2 (en) 2006-12-27 2013-04-10 Эмори Юниверсити Compositions and methods of treating infections and tumours
WO2008109353A1 (en) 2007-03-02 2008-09-12 Mdrna, Inc. Nucleic acid compounds for inhibiting map2k gene expression and uses thereof
WO2008109105A2 (en) 2007-03-06 2008-09-12 Flagship Ventures Methods and compositions for improved therapeutic effects with sirna
WO2009020344A2 (en) 2007-08-06 2009-02-12 Postech Acad Ind Found Small interfering rnas (sirnas) controlling multiple target genes and method for preparing the same
CN105018492B (en) 2007-08-27 2018-08-24 北京强新生物科技有限公司 Composition of asymmetric aiRNA and application thereof
EP2205740A2 (en) 2007-10-02 2010-07-14 Rxi Pharmaceuticals Corp. Tripartite rnai constructs
EP2231168A4 (en) 2007-10-03 2012-01-04 Quark Pharmaceuticals Inc Novel sirna structures
CA2910760C (en) 2007-12-04 2019-07-09 Muthiah Manoharan Targeting lipids
KR100949791B1 (en) 2007-12-18 2010-03-30 이동기 Novel siRNA Structure for Minimizing Off-target Effects and Relaxing Saturation of RNAi Machinery and the Use Thereof
CN104975020B (en) 2008-02-11 2020-01-17 菲奥医药公司 Modified RNAi polynucleotides and uses thereof
US20110218334A1 (en) 2008-07-11 2011-09-08 Alnylam Pharmaceuticals, Inc. PHOSPHOROTHIOATE OLIGONUCLEOTIDES AND NON-NUCLEOSIDIC PHOSPHOROTHIOATES AS DELIVERY AGENTS FOR iRNA AGENTS
WO2010008582A2 (en) 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Phagocytic cell drug delivery system
US20110251258A1 (en) 2008-07-24 2011-10-13 Rxi Pharmaceuticals Corporation Rnai constructs and uses thereof
WO2010027830A2 (en) 2008-08-25 2010-03-11 Excaliard Pharmaceuticals, Inc. Methods for modulation of connective tissue growth factor expression by administering antisense oligonucleotides through a delivery system so as to reduce scarring from wound healing and threat fibrotic diseases
NZ591416A (en) 2008-08-25 2012-11-30 Excaliard Pharmaceuticals Inc Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP2949752B1 (en) 2008-09-22 2017-12-20 RXi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
EP3067359A1 (en) 2008-09-23 2016-09-14 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
US8822425B2 (en) 2008-11-14 2014-09-02 Antisense Pharma Gmbh Dosage of oligonucleotides suitable for the treatment of tumors
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
EP2361306A1 (en) 2008-12-04 2011-08-31 OPKO Ophthalmics, LLC Compositions and methods for selective inhibition of pro-angiogenic vegf isoforms
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
WO2011002776A1 (en) 2009-06-29 2011-01-06 Nitric Biotherapeutics, Inc. Pharmaceutical formulations for iontophoretic delivery of an immunomodulator
CA2782728A1 (en) 2009-12-04 2011-06-09 Opko Ophthalmics, Llc Compositions and methods for inhibition of vegf
US20110268761A1 (en) 2010-03-02 2011-11-03 Hapten Pharmaceuticals, Llc Effective Sensitizing Dose of a Gelled Immunomodulating Topical Composition
WO2011119887A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Rna interference in dermal and fibrotic indications
EP3578183B1 (en) * 2010-03-24 2021-09-08 Phio Pharmaceuticals Corp. Rna interference in ocular indications
RU2615143C2 (en) 2010-03-24 2017-04-04 Адвирна Self-delivered rnai compounds of reduced size
JP2013531981A (en) 2010-06-11 2013-08-15 アンチセンス・ファーマ・ゲーエムベーハー Method for selective oligonucleotide modification
US20120046186A1 (en) 2010-08-20 2012-02-23 Pelham Robert J Gene Expression Markers for Prediction of Response to Platinum-Based Chemotherapy Drugs
EP2648735A4 (en) * 2010-12-06 2014-07-30 Univ British Columbia Granzyme b inhibitor compositions, methods and uses for promoting wound healing
WO2012112079A1 (en) 2011-02-14 2012-08-23 Vitaspero, Inc. IMPROVING CELLULAR IMMUNOTHERAPEUTIC VACCINES EFFICACY WITH GENE SUPPRESSION IN DENDRITIC CELLS AND T-LYMPHOCYTES USING SiRNA
WO2013101436A1 (en) 2011-12-07 2013-07-04 Duke University Methods of identifying and using mdm2 inhibitors
RU2497514C1 (en) * 2012-04-06 2013-11-10 Аббясов Рушан Хамидуллович Immunomodulator
US20140072613A1 (en) * 2012-09-10 2014-03-13 Cynthia Lander Compositions and Methods for Treating Cutaneous Scarring
NO3036326T3 (en) 2013-08-20 2018-03-03
EP3079707A4 (en) * 2013-12-02 2017-10-18 RXi Pharmaceuticals Corporation Immunotherapy of cancer
CN105960265A (en) 2013-12-04 2016-09-21 阿克赛医药公司 Methods for treatment of wound healing utilizing chemically modified oligonucleotides
CN103820454B (en) 2014-03-04 2016-03-30 上海金卫生物技术有限公司 The method of CRISPR-Cas9 specific knockdown people PD1 gene and the sgRNA for selectively targeted PD1 gene
CA2947270A1 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic acids targeting mdm2 or mycn
JP2017514908A (en) 2014-05-01 2017-06-08 アールエックスアイ ファーマシューティカルズ コーポレーション Methods for the treatment of disorders in the front of the eye utilizing nucleic acid molecules
US10900039B2 (en) 2014-09-05 2021-01-26 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting Tyr or MMP1
JP2018502837A (en) 2014-12-03 2018-02-01 アールエックスアイ ファーマシューティカルズ コーポレーション Method for treating alopecia areata using a gene regulatory approach
CN105734058B (en) * 2014-12-09 2019-11-08 中国辐射防护研究院 SiRNA, recombinant vector and the purposes of the HAVCR2 gene of silencing people
LT3277814T (en) 2015-04-03 2020-10-26 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
US20160319278A1 (en) 2015-04-03 2016-11-03 University Of Massachusetts Fully stabilized asymmetric sirna
HUE057431T2 (en) 2015-04-03 2022-05-28 Univ Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
JP6983752B2 (en) 2015-07-06 2021-12-17 フィオ ファーマシューティカルズ コーポレーションPhio Pharmaceuticals Corp. Nucleic acid molecule targeting superoxide dismutase 1 (SOD1)
US20190029974A1 (en) 2015-09-11 2019-01-31 Rxi Pharmaceuticals Corporation Methods for treating skin disorders and conditions utilizing haptens
WO2017070151A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
EP3609531A2 (en) * 2017-04-10 2020-02-19 immatics biotechnologies GmbH Peptides and combination of peptides for use in immunotherapy against leukemias and other cancers
WO2018195355A1 (en) 2017-04-19 2018-10-25 Rxi Pharmaceuticals Corporation Topical delivery of nucleic acid compounds
EP3664817A4 (en) * 2017-08-07 2021-09-22 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
CN112040986A (en) * 2018-03-15 2020-12-04 Ksq治疗公司 Gene regulatory compositions and methods for improved immunotherapy
US20220220469A1 (en) * 2019-05-20 2022-07-14 The Broad Institute, Inc. Non-class i multi-component nucleic acid targeting systems
CA3219148A1 (en) * 2021-05-17 2022-11-24 Frederick G. Vogt Pd-1 gene-edited tumor infiltrating lymphocytes and uses of same in immunotherapy

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11396654B2 (en) 2008-09-22 2022-07-26 Phio Pharmaceuticals Corp. Neutral nanotransporters
US11667915B2 (en) 2009-02-04 2023-06-06 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US11584933B2 (en) 2010-03-24 2023-02-21 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US11926828B2 (en) 2014-09-05 2024-03-12 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting TYR or MMP1

Also Published As

Publication number Publication date
WO2015084897A2 (en) 2015-06-11
RU2016126488A3 (en) 2018-12-06
CN113151180A (en) 2021-07-23
US10934550B2 (en) 2021-03-02
CN106061488A (en) 2016-10-26
RU2744194C2 (en) 2021-03-03
EP3079707A2 (en) 2016-10-19
JP2023067871A (en) 2023-05-16
JP6772062B2 (en) 2020-10-21
EP3079707A4 (en) 2017-10-18
RU2016126488A (en) 2018-12-06
JP2016540511A (en) 2016-12-28
CN106061488B (en) 2021-04-09
JP2021019608A (en) 2021-02-18
WO2015084897A3 (en) 2015-09-03
US20160304873A1 (en) 2016-10-20

Similar Documents

Publication Publication Date Title
US20210348166A1 (en) Immunotherapy of cancer
Soldevilla et al. 2-fluoro-RNA oligonucleotide CD40 targeted aptamers for the control of B lymphoma and bone-marrow aplasia
JP2020074788A (en) SINGLE STRANDED RNAi AGENT INCLUDING INTERNAL NON-NUCLEIC ACID SPACER
JP2021512090A (en) Compositions and Methods for Delivering Drugs to Immune Cells
US11639496B2 (en) Reducing fratricide of immune cells expressing NKG2D-based receptors
WO2013049307A9 (en) Enhanced immune memory development by aptamer targeted mtor inhibition of t cells
JP4517061B2 (en) Efficient production method for dumbbell-shaped DNA
WO2010026660A1 (en) Rna capable of inhibiting expression of zfat gene
JP2023515671A (en) Methods and compositions for sensitizing tumor cells to immunotherapy
JP2023521345A (en) Compositions and methods for enhancing activation and cytolytic activity of CD8+ T cells by disruption of the SAGA (SPT-ADA-GCN5-acetyltransferase) complex
JP5249998B2 (en) Osteopontin siRNA
WO2019221280A1 (en) Immune checkpoint inhibitor
JP5098033B2 (en) siRNA, recombinant vector expressing this siRNA, NR4A2 gene expression inhibitor, IL-17 gene expression inhibitor, and CD4 positive T cell proliferation inhibitor
JP2022541212A (en) SiRNA sequences targeting the expression of the human genes JAK1 or JAK3 for therapeutic use
KR102397455B1 (en) Asymmetric siRNA Inhibiting Expression of PD-1
JP4589794B2 (en) Osteopontin siRNA
EP4124656A1 (en) Pd-l1 micrornas
JP2020515618A (en) Enhances immunotherapy by decreasing beta-catenin expression
CN113337505B (en) Application of artificially constructed antisense nucleotide fragment Ri111 in polarization of Th1 cells
JP2008211985A (en) Il-17 production inhibitor and method for screening the same
WO2024047115A1 (en) THERAPEUTIC USE OF THE miR155 SNP rs377265631
WO2023227354A1 (en) Nucleic acid modified biological cell with expansion-dependent gene expression
WO2023225512A2 (en) Methods for optimizing t cell immunotherapeutic effector and memory function
WO2022266538A2 (en) Compositions and methods for targeting, editing or modifying human genes
JP2004350607A (en) Telomerase inhibitor

Legal Events

Date Code Title Description
AS Assignment

Owner name: PHIO PHARMACEUTICALS CORP., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:RXI PHARMACEUTICALS CORPORATION;REEL/FRAME:056971/0080

Effective date: 20181114

Owner name: MIRIMMUNE, LLC, MASSACHUSETTS

Free format text: MERGER AND CHANGE OF NAME;ASSIGNORS:MIRIMMUNE INC.;RXI MERGER SUB, LLC;REEL/FRAME:056966/0494

Effective date: 20170109

Owner name: RXI PHARMACEUTICALS CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MIRIMMUNE, LLC;REEL/FRAME:056966/0401

Effective date: 20170330

Owner name: MIRIMMUNE INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOLFSON, ALEXEY;ELISEEV, ALEXEY;SMUSHKOVICH, TAISIA;SIGNING DATES FROM 20160526 TO 20160527;REEL/FRAME:056964/0697

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION