US20190142967A1 - Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer - Google Patents

Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer Download PDF

Info

Publication number
US20190142967A1
US20190142967A1 US16/253,056 US201916253056A US2019142967A1 US 20190142967 A1 US20190142967 A1 US 20190142967A1 US 201916253056 A US201916253056 A US 201916253056A US 2019142967 A1 US2019142967 A1 US 2019142967A1
Authority
US
United States
Prior art keywords
tumor
human
cell
antibody
receptor
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
US16/253,056
Inventor
Daniel Hicklin
Kenneth Nelson Wills
Cynthia Seidel-Dugan
William Winston
Philipp Steiner
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.)
Epicentrx Inc
Original Assignee
Trieza Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Trieza Therapeutics Inc filed Critical Trieza Therapeutics Inc
Priority to US16/253,056 priority Critical patent/US20190142967A1/en
Assigned to Trieza Therapeutics, Inc. reassignment Trieza Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STEINER, PHILIPP
Assigned to Trieza Therapeutics, Inc. reassignment Trieza Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WINSTON, WILLIAM, SEIDEL-DUGAN, CYNTHIA, HICKLIN, DANIEL, WILLS, KENNETH NELSON
Publication of US20190142967A1 publication Critical patent/US20190142967A1/en
Assigned to EPICENTRX, INC. reassignment EPICENTRX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Trieza Therapeutics, Inc.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5418IL-7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5428IL-10
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04MTELEPHONIC COMMUNICATION
    • H04M3/00Automatic or semi-automatic exchanges
    • H04M3/42Systems providing special services or facilities to subscribers
    • H04M3/42025Calling or Called party identification service
    • H04M3/42034Calling party identification service
    • H04M3/42059Making use of the calling party identifier
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04MTELEPHONIC COMMUNICATION
    • H04M3/00Automatic or semi-automatic exchanges
    • H04M3/42Systems providing special services or facilities to subscribers
    • H04M3/42221Conversation recording systems
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04MTELEPHONIC COMMUNICATION
    • H04M3/00Automatic or semi-automatic exchanges
    • H04M3/42Systems providing special services or facilities to subscribers
    • H04M3/436Arrangements for screening incoming calls, i.e. evaluating the characteristics of a call before deciding whether to answer it
    • H04M3/4365Arrangements for screening incoming calls, i.e. evaluating the characteristics of a call before deciding whether to answer it based on information specified by the calling party, e.g. priority or subject
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04MTELEPHONIC COMMUNICATION
    • H04M3/00Automatic or semi-automatic exchanges
    • H04M3/42Systems providing special services or facilities to subscribers
    • H04M3/54Arrangements for diverting calls for one subscriber to another predetermined subscriber
    • H04M3/541Arrangements for diverting calls for one subscriber to another predetermined subscriber based on information specified by the calling party
    • 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
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10332Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10371Demonstrated in vivo effect
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/24Vectors characterised by the absence of particular element, e.g. selectable marker, viral origin of replication
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/30Vector systems having a special element relevant for transcription being an enhancer not forming part of the promoter region
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the invention described herein relates generally to the fields of immunology, virology, molecular biology, and more specifically to oncolytic adenoviruses having therapeutic applications.
  • Cancer is a leading cause of death in the United States and elsewhere. Depending on the type of cancer, it is typically treated with surgery, chemotherapy, and/or radiation. These treatments often fail, and it is clear that new therapies are necessary, to be used alone or in combination with current standards of care.
  • viruses that can preferentially target tumor cells for destruction are being used experimentally as vectors for the delivery of immune-stimulating cargo.
  • the propagation of a lasting anti-tumor host immune response in combination with the destruction of tumor cells is described in, e.g., Lichty et al., 2014, Nature Reviews Cancer, 14: 559-567.
  • Adenoviruses are medium-sized (90-100 nm), non-enveloped icosahedral viruses, which have double stranded linear DNA of about 36 kilobase pairs in a protein capsid.
  • the viral capsid has fiber structures that participate in attachment of the virus to the target cell.
  • the knob domain of the fiber protein binds to the receptor of the target cell (e.g., CD46 or Coxsackie and adenovirus receptor (CAR)), secondly, the virus interacts with an integrin molecule and thirdly, the virus is endocytosed into the target cell.
  • the viral genome is transported from endosomes into the nucleus and the replication machinery of the target cell is utilized also for viral purposes.
  • the adenoviral genome has early (E1-E4), intermediate (IX and IVa2) and late genes (L1-L5), which are transcribed in sequential order.
  • Early gene products affect defense mechanisms, cell cycle and cellular metabolism of the host cell.
  • Intermediate and late genes encode structural viral proteins for production of new virions.
  • Ad-5 Adenovirus serotype 5
  • Ad-5 Ad5 vectors E1 and/or E3 regions were deleted enabling insertion of foreign DNA to the vectors.
  • deletions of other regions as well as further mutations have provided extra properties to viral vectors. Indeed, various modifications of adenoviruses have been suggested for achieving efficient anti-tumor effects.
  • Adenoviral vectors mediate gene transfer at a high efficacy compared to other vector systems, and they are currently the most frequently used vectors for cancer gene therapy.
  • a non-replicating p53 expressing adenoviral vector and a replication selective virus (H101) have received regulatory approval in China.
  • Several attempts to achieve tumor-selective control through the insertion of tumor selective promoter elements upstream of the E1 or other adenovirus critical promoters have had variable levels of success, but ultimately were limited by “leaky” gene expression of viral proteins in non-tumor cells and by reduced ability to propagate and lyse tumor cells compared to wild-type virus infections.
  • a pharmaceutical composition comprising an effective amount of a recombinant adenoviral vector comprising: a transgene insertion site located between the start site of adenoviral E1b-19K and the start site of adenoviral E1b-55K, wherein a first DNA sequence and a second DNA sequence are each inserted into the transgene insertion site; wherein the first DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, and wherein the second DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL
  • the adenoviral vector comprises an IRES element or encodes a self-cleaving 2A peptide sequence between the first DNA sequence and the second DNA sequence.
  • the vector comprises a modified E3 region.
  • the vector comprises an intact E3 region.
  • the vector comprises a third DNA sequence inserted into the E3 region, wherein the third DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, or a human OX40 ligand.
  • the chimeric human IL-12 polypeptide comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide. In another embodiment, the chimeric human IL-12 polypeptide comprises a sequence as set forth in SEQ ID NO:46.
  • the adenoviral vector comprises a nucleic acid sequence at least 95% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 85% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 75% identical in an E3 region to vector d1327.
  • the pharmaceutical composition is formulated for systemic administration.
  • the pharmaceutical composition is formulated for systemic administration.
  • a pharmaceutical composition comprising an effective amount of a recombinant adenoviral vector comprising: a first transgene insertion site located between the start site of adenoviral E1b-19K and the start site of adenoviral E1b-55K; a second transgene insertion site located in adenoviral E3 region; a first DNA sequence, present in the first transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand; and a second DNA sequence, present in the second transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti
  • the pharmaceutical composition further comprises a third DNA sequence inserted into a third transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand.
  • a third DNA sequence inserted into a third transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand.
  • At least one of the first DNA sequence, the second DNA sequence, and the third DNA sequence independently comprises an IRES element and/or a self-cleaving 2A peptide.
  • at least one E1a regulatory sequence Pea3 binding site, or a functional portion thereof of the adenoviral vector is modified or deleted.
  • a sequence between two Pea3 sites of the adenoviral vector is deleted.
  • the adenoviral vector comprises a modified E3 region.
  • the adenoviral vector comprises a nucleic acid sequence at least 95% identical in an E3 region to vector d1327.
  • the adenoviral vector comprises a nucleic acid sequence at least 85% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 75% identical in an E3 region to vector d1327.
  • the chimeric human IL-12 polypeptide comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide.
  • a method for treating a tumor in a human subject in need thereof comprising administering to the human with the tumor a therapeutic amount of the pharmaceutical composition of the first aspect by systemic or intratumoral administration.
  • method for treating a tumor in a human subject in need thereof comprising administering to the human with the tumor a therapeutic amount of the pharmaceutical composition of the second aspect by systemic or intratumoral administration.
  • FIG. 1 is a cartoon illustrating transcription factor binding regulation of conformational structure/activity of E1a enhancer region.
  • FIG. 2 is an illustration of a vector map demonstrating an adenoviral vector expressing two immunomodulatory polypeptides.
  • FIG. 3 is an illustration of a vector map demonstrating an adenoviral vector expressing at least one immunomodulatory polypeptide at an E4 site.
  • FIG. 4 is an illustration of the TRZ200 virus genome: an E1b 19k empty, E3-deleted virus.
  • FIG. 5 is an illustration of a vector map showing the plasmid comprising the TRZ6 hIL-12 plasmid.
  • FIG. 6A shows the activity of various oncolytic viruses compared to the empty virus (“d19k”), 38 days after cell implantation (primary tumor). Black bars represent virus alone and hatched bars represent virus+anti-PD-L1 antibody.
  • FIG. 6B is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), each of five viruses alone (CTLA-4, IL-12, IL-7, CD70, and IL-10) or combined with anti-PD-L1 antibody.
  • FIG. 6C is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), showing three viruses—OX40L, CD40L, and GM-CSF alone or combined with anti-PD-L1 antibody.
  • FIG. 6D shows virus buffer and control IgG only (left) and virus buffer and anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6E shows d19k (empty virus) and control IgG only (left) and d19k and anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6F shows CTLA-4 virus with control IgG (left) or anti-PD-L1 (right); the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6G shows IL-12 virus with control IgG (left) or anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6H shows GM-C S F virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6I shows IL-7 virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6J shows CD40L virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6K shows L10 trap virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6L shows OX40L virus with control IgG (left) or anti-PD-L1 antibody.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7A shows virus buffer only.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7B shows empty virus only (TRZ000).
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7C shows TRZ010 (IL-10trap)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7D shows TRZ011 (OX40 ligand)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7E shows TRZ009 (CD70)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7F shows TRZ007 (IL-7)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7G shows TRZ002 (IL-12)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7H shows TRZ004 (GM-CSF)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7I shows TRZ003 (flagellin)+empty virus.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7J shows TRZ002+TRZ010.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7K shows TRZ002+TRZ007.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7L shows TRZ007+TRZ010.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7M shows TRZ011+TRZ004.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7N shows TRZ009+TRZ003.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7O shows TRZ002+TRZ009.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7P shows TRZ007+TRZ009.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7Q shows TRZ007+TRZ004.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7R shows TRZ002+TRZ011.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7S shows TRZ010+TRZ004.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7T shows TRZ002+TRZ004.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7U shows virus buffer and anti-PD-L1.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7V shows TRZ002+empty virus+anti-PD-L1.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7W shows TRZ009+anti-PD-L1.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7X shows TRZ007+empty virus+anti-PD-L1.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7Y shows TRZ002+TRZ007+anti-PD-L1.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7Z shows TRZ007+TRZ009+anti-PD-L1.
  • FIG. 8A is schematic representations of the rationale for modifications of the E1a Enhancer region of adenoviral vectors, in which Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics.
  • FIG. 8A is an illustration of a wild-type adenoviral genome (top) and the TAV-255 adenoviral construct in which residues -305 to -255 are deleted (bottom).
  • FIG. 8B is schematic representations of the rationale for modifications of the E1a Enhancer region of adenoviral vectors, in which Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics.
  • FIG. 8B is an illustration of the potential method of action of the TAV-255 construct, in which the deletion removes Pea3 sites II and III, which moves Pea3 sites IV and V closer to the promoter as distal control elements.
  • FIG. 9A is a cartoon of the contrast between the single transgene constructs (top) used in the virus mixing examples, and the dual transgene vectors used to make the E1/E3 transgene adenoviruses.
  • FIG. 9B shows the results of mice injected i.t. with Empty virus+/ ⁇ anti-PD-L1 or with TRZ-409 (IL-12/IL-7 dual transgene)+/ ⁇ anti-PD-L1.
  • the tumor volume of the injected tumor is shown in the left panel, and the tumor volume of the contralateral tumor is shown in the right panel.
  • the left arrow indicates virus injection and the right arrow indicates anti-PD-L1 injection.
  • TRZ-409 injection reduced tumor volume significantly compared to empty virus.
  • the combination with anti-PD-L1 was slightly more efficacious in this study.
  • FIG. 9C shows the results of a second experiment using the inverse of TRZ-409, TRZ-403, in which the IL-7 transgene occupies the E1 region and the IL-12 gene occupies the E3 region (which has a stronger promoter than the E1).
  • TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1.
  • TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1.
  • Plasma levels of IL-12 and IFN- ⁇ ( FIG. 9D ) for all mice injected with TRZ-403 show that expression of the transgenes is well tolerated. The study was extended for all mice having tumors over 500 mm 3 .
  • mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days).
  • mice having primary tumors injected with TRZ-403 (IL-7+IL-12), TRZ-403+anti-PD-L1, TRZ-403+control IgG, or TRZ-409 (IL-12+1L-7), and controls including untreated mice and mice injected with empty vector TRZ-d19K with anti-PD-L1, anti-PD-1, or a control IgG.
  • mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days). Both TRZ-403 and TRZ-409 showed efficacy over the controls; all mice receiving control injections were deceased by day 56 of the study.
  • FIG. 9F compares a mixture of two single transgene viruses with a dual transgene virus having the same transgenes. As can be seen in the primary tumor (left panel) TRZ-403 showed the most efficacy in reducing tumor volume, followed by the mixture of IL-7 and IL-12 single transgene viruses. The single transgene IL-12 virus showed a small amount of efficacy by comparison. In the contralateral tumor, however (right panel) only the dual transgene virus, TRZ-403, reduced the tumor volume, showing significant superiority over the mixture of viruses.
  • the left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows).
  • FIG. 7AA shows TRZ002+TRZ009+anti-PD-L1.
  • replicating virus is meant to include a virus that undergoes the process of intracellular viral multiplication, consisting of the synthesis of proteins, nucleic acids, and sometimes lipids, and their assembly into a new infectious particle.
  • adenovirus refers to any of a group of DNA-containing viruses (small infectious agents) that cause conjunctivitis and upper respiratory tract infections in humans.
  • Adenoviral vectors are described in Peng, Z., “Current Status of Gendicine in China: Recombinant Human Ad-p53 Agent for Treatment of Cancers,” Hum Gene Ther 16:1016-1027 (2005); No authors listed, “The End of the Beginning: Oncolytic Virotherapy Achieves Clinical Proof-of-concept,” Mol Ther 13:237-238 (2006); Vile et al., “The Oncolytic Virotherapy Treatment Platform for Cancer: Unique Biological and Biosafety Points to Consider,” Cancer Gene Ther 9:1062-1067 (2002); Harrison et al., “Wild-type Adenovirus Decreases Tumor Xenograft Growth, but Despite Viral Persistence Complete Tumor Responses are Rarely Achieved—Deletion of the Viral Elb-19-kD
  • Adenoviral positions referenced herein are to positions in Adenovirus type 5 (GenBank 10 accession #M73260; the virus is available from the American Type Culture Collection, Rockville, Md., U.S.A., under accession number VR-5). It will be understood that corresponding positions can be identified in other adenovirus vectors by alignment using BLAST 2.0 under default settings (Altschul et al., J. Mol. Biol. 215:403-410 (1990)). Software for performing BLAST analyses is publicly available on the Web through the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov/).
  • control elements or “regulatory sequences” can also be incorporated into the vector-construct.
  • control elements refers collectively to promoter regions, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites (“IRES”), enhancers, and the like, which collectively provide for the replication, transcription, and translation of a coding sequence(s) in a recipient cell. Not all of these control elements need always be present so long as the selected coding sequence is capable of being replicated, transcribed, and translated in an appropriate host cell.
  • promoter region is used herein in its ordinary sense to refer to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding RNA polymerase and initiating transcription of a downstream (3′-direction) coding sequence.
  • Transcriptional control signals in eukaryotes comprise “promoter” and “enhancer” elements.
  • Promoter and enhancer elements have been isolated from a variety of eukaryotic sources, including genes in yeast, insect, and mammalian cells, and viruses. Analogous control elements, i.e., promoters, are also found in prokaryotes. Such elements may vary in their strength and specificity. For example, promoters may be “constitutive” or “inducible.”
  • a constitutive promoter is a promoter that directs expression of a gene throughout the development and life of an organism.
  • constitutive promoters that are widely used for inducing expression of transgenes include the cytomegalovirus (CMV) early promoter, those derived from any of the several actin genes, which are known to be expressed in most cells types (U.S. Pat. No. 6,002,068 to Privalle et al., which is hereby incorporated by reference in its entirety), and the ubiquitin promoter, which is a gene product known to accumulate in many cell types.
  • CMV cytomegalovirus
  • 3′ polyadenylation regions can be present to provide for proper maturation of the mRNA transcripts.
  • the 3′ polyadenylation region will preferably be from the adenovirus sequence downstream of the inserted transgene, but the native 3′-untranslated region of the immunomodulatory gene may be used, or an alternative polyadenylation signal from, for example, SV40, particularly including a splice site, which provides for more efficient expression, could also be used.
  • the 3′-untranslated region derived from a gene highly expressed in a particular cell type could be fused with the immunomodulatory gene.
  • Inhibitors “Inhibitors,” “activators,” and “modulators” of expression or of activity are used to refer to inhibitory, activating, or modulating molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., ligands, agonists, antagonists, and their homologs and mimetics.
  • the term “modulator” includes inhibitors and activators.
  • Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or protease inhibitor activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein, e.g., antagonists.
  • Activators are agents that, e.g., induce or activate the expression of a described target protein or bind to, stimulate, increase, open, activate, facilitate, enhance activation or protease inhibitor activity, sensitize or up regulate the activity of described target protein (or encoding polynucleotide), e.g., agonists.
  • Modulators include naturally occurring and synthetic ligands, antagonists and agonists (e.g., small chemical molecules, antibodies and the like that function as either agonists or antagonists).
  • Such assays for inhibitors and activators include, e.g., applying putative modulator compounds to cells expressing the described target protein and then determining the functional effects on the described target protein activity, as described above.
  • Samples or assays comprising described target protein that are treated with a potential activator, inhibitor, or modulator are compared to control samples without the inhibitor, activator, or modulator to examine the extent of effect.
  • Control samples (untreated with modulators) are assigned a relative activity value of 100%.
  • Inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%.
  • Activation of the described target protein is achieved when the activity value relative to the control is 110%, optionally 150%, optionally 200, 300%, 400%, 500%, or 1000-3000% or higher.
  • Antibody refers to a polypeptide comprising a framework region from an immunoglobulin gene or fragments thereof that specifically binds and recognizes an antigen.
  • the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and mu constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
  • Naturally occurring immunoglobulins have a common core structure in which two identical light chains (about 24 kD) and two identical heavy chains (about 55 or 70 kD) form a tetramer.
  • the amino-terminal portion of each chain is known as the variable (V) region and can be distinguished from the more conserved constant (C) regions of the remainder of each chain.
  • V variable
  • C constant
  • Within the variable region of the light chain is a C-terminal portion known as the J region.
  • Within the variable region of the heavy chain there is a D region in addition to the J region.
  • Most of the amino acid sequence variation in immunoglobulins is confined to three separate locations in the V regions known as hypervariable regions or complementarity determining regions (CDRs) which are directly involved in antigen binding.
  • CDRs complementarity determining regions
  • the CDRs are held in place by more conserved framework regions (FRs). Proceeding from the amino-terminus, these regions are designated FR1, FR2, FR3, and FR4, respectively.
  • FR1, FR2, FR3, and FR4 Proceeding from the amino-terminus, these regions are designated FR1, FR2, FR3, and FR4, respectively.
  • the locations of CDR and FR regions and a numbering system have been defined by, e.g., Kabat et al. (Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, U.S. Government Printing Office (1991)).
  • An exemplary immunoglobulin (antibody) structural unit comprises a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these light and heavy chains respectively.
  • Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to V H -C H1 by a disulfide bond.
  • the F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′2 dimer into an Fab′ monomer.
  • the Fab′ monomer is essentially Fab with part of the hinge region (see FUNDAMENTAL IMMUNOLOGY (Paul ed., 3d ed. 1993). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al., Nature 348:552-554 (1990)).
  • Monoclonal antibodies refer to antibodies derived from a single clone.
  • Techniques for the production of single chain antibodies can be adapted to produce antibodies to polypeptides of this invention.
  • transgenic mice, or other organisms such as other mammals may be used to express humanized antibodies.
  • phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g., McCafferty et al., Nature 348:552-554 (1990); Marks et al., Biotechnology 10:779-783 (1992)).
  • a “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • a “humanized” antibody is an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol., 44:65-92 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994).
  • the terms “treat” and “treating” in the context of the administration of a therapy refers to a treatment/therapy from which a subject receives a beneficial effect, such as the reduction, decrease, attenuation, diminishment, stabilization, remission, suppression, inhibition or arrest of the development or progression of cancer, or a symptom thereof.
  • the treatment/therapy that a subject receives results in at least one or more of the following effects: (i) the reduction or amelioration of the severity of cancer and/or a symptom associated therewith; (ii) the reduction in the duration of a symptom associated with cancer; (iii) the prevention in the recurrence of a symptom associated with cancer; (iv) the regression of cancer and/or a symptom associated therewith; (v) the reduction in hospitalization of a subject; (vi) the reduction in hospitalization length; (vii) the increase in the survival of a subject; (viii) the inhibition of the progression of cancer and/or a symptom associated therewith; (ix) the enhancement or improvement the therapeutic effect of another therapy; (x) a reduction or elimination in the cancer cell population; (xi) a reduction in the growth of a tumor or neoplasm; (xii) a decrease in tumor size; (xiii) a reduction in the formation of a tumor; (xiv) eradication
  • the treatment/therapy that a subject receives does not cure cancer, but prevents the progression or worsening of the disease. In certain embodiments, the treatment/therapy that a subject receives does not prevent the onset/development of cancer, but may prevent the onset of cancer symptoms.
  • the term “in combination” in the context of the administration of (a) therapy(ies) to a subject refers to the use of more than one therapy.
  • the use of the term “in combination” does not restrict the order in which therapies are administered to a subject.
  • a first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region, e.g., of the entire polypeptide sequences or specific region, if indicated), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be “substantially identical.”
  • sequence comparison For sequence comparison, one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of, e.g., a full-length sequence or from 20 to 600, about 50 to about 200, or about 100 to about 150 amino acids or nucleotides in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well-known in the art.
  • Optimal alignment of sequences for comparison are conducted by a BLAST 2.0 algorithm, which is described in Altschul et al. (1990) J Mol. Biol. 215:403-410.
  • HSPs high scoring sequence pairs
  • Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0).
  • M forward score for a pair of matching residues; always >0
  • N penalty score for mismatching residues; always ⁇ 0.
  • a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W word length
  • E expectation
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • compositions and methods of treating cancer in a subject are provided.
  • an immune stimulatory transgene from the adenovirus for example, including but not limited to an E1b 19K deleted region in an exemplary vector such as TAV-255 (e.g., TAV-255 ⁇ 19)
  • the immune system will be primed to fight cancer systemically in the entire body of the cancer patient as long as the metastatic tumor cells express the same tumor antigens as the primary tumor cells. Since metastases are derived from the primary tumors and genetically very similar to the primary tumor cells, metastatic tumor growth will be inhibited, in some embodiments, to the same extent as the primary tumor.
  • adenoviruses as described herein.
  • a cell transformed with any one of the recombinant adenoviruses described herein is provided.
  • a method of selectively expressing a peptide in a target cell comprises contacting the target cell with any one of the recombinant adenoviruses described herein.
  • the recombinant adenovirus comprises a E1a regulatory sequence deletion mutant operably linked to a nucleotide sequence encoding a peptide, e.g., a peptide associated with viral replication or with cancer.
  • adenoviral therapy that utilize the oncolytic adenoviruses of the instant invention as adenoviral vectors that express one, two, or more recombinant immunomodulatory genes.
  • the oncolytic adenovirus contains a heterologous gene that encodes a therapeutic protein, incorporated within the viral genome, such that the heterologous gene is expressed within an infected cell.
  • a therapeutic protein refers to a protein that provides one or more therapeutic benefit when expressed in a given cell. In particular, the therapeutic benefit includes recruitment of the host immune system to the tumor.
  • E1a is the first protein produced by an adenovirus upon infection of a cell, activating other adenoviral promoters and facilitating infected cells to enter cell division. Rendering expression of this protein under tumor-selective control is an effective means of limiting expression of viral proteins and oncolysis to tumor cells.
  • Normal cells require mitogenic growth signals (GS) before they can move from a quiescent state into an active proliferative state.
  • Tumor cells are able to generate many of their own growth signals or mimic normal growth signals, and transcription factors such as E2F1 and Pea3 are commonly overexpressed in tumor cells at levels that can cooperate in forming conformational structures optimal for driving E1a transcription during adenovirus infection and replication.
  • GS mitogenic growth signals
  • E1a enhancer region Small deletions selectively targeting the binding sites for E2F1 and Pea3 sites in the E1a enhancer region are an alternative and less disruptive method than complete replacement of the E1a enhancer region with a transcriptionally restricted promoter element. Cooperative binding and transcriptionally optimized conformation of the E1a enhancer region could still take place due to the over-abundance of transcription factors found in tumor cells, while in normal, non-dividing cells, the disruption of binding sites would further inhibit the ability to form optimized conformations in the limiting level of mitogenic growth signals.
  • Pea3 binding sites The five Pea3 transcription factor binding sites, also known as E1AF (or originally as EF-1A-enhancer binding factor to the E1a core motif) have differential effects on the production of E1a mRNA levels as demonstrated by specific deletions of individual and paired sites.
  • the murine Pea3 sites are described herein as Pea3 sites I, II, III, IV, and V.
  • the main binding sites for Pea3 are sites I and III, while sites II, IV, and V are slightly degenerate versions.
  • Pea3 binds cooperatively between sites II and III, IV and V, and II and I, and this cooperative binding activates E1a transcription. (see, e.g., Hearing (J. Virol 65, 1991, Mol Cell Biol 9, 1989, and Nuc Acids Res 20, 1992).
  • Pea3 itself is a dimer of both ⁇
  • ⁇ subunits where the ⁇ subunit makes the primary DNA contact and the ⁇ subunit forms a heteromultimeric complex with the ⁇ subunit both in solution or on a dimeric binding site.
  • binding at sites I and III, followed by the cooperative binding at site II would cause conformational changes that serve to bring this protein/DNA complex closer to the activation transcription factor (ATF) binding site and TATA box for full activation of E1a mRNA expression.
  • ATF activation transcription factor
  • the two lower affinity sites, Pea3 IV and Pea3 5 do not appear to contribute much to activation under these normal circumstances, as they may be too far away or unoccupied.
  • site II causes the greatest reduction in transcriptional activity, pointing to the importance of the cooperative binding effect it has for sites III and I in activating transcription, presumably through a conformational change. Deletion of either site III or I had much less of a reduction, since presumably you could still have cooperative binding between site II and the remaining site III or I. Deletion of both sites I and III (but not II) reduces transcription to the levels seen with Pea3 site II deletion alone, and the combination of deleting both sites III and II, or sites II and I also results in similar (but not greater) levels of reduction.
  • tumor cells may have enough E1AF to bind the lower affinity binding sites IV and V, along with site I, and potentially lead to a conformational change needed to activate transcription of E1a in tumor but not normal cells with this deleted virus.
  • the present invention provides replicating adenoviruses.
  • the replicating vectors of the instant invention contain recombinant (e.g., exogenous) transgene(s) expressing immunomodulatory polypeptides that are controlled by endogenous adenovirus early promoters, thereby driving meaningfully higher expression levels than can be generally achieved in replication deficient viruses.
  • the higher expression levels from the recombinant transgenes in a replicating virus results in enhanced immunomodulatory effect(s) over the lower expression levels in replication deficient viruses.
  • a recombinant virus comprises a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is deleted.
  • a sufficient number of nucleotides in the range of ⁇ 305 to -141 are retained to maintain functional activity of the Ad packaging signal function and (near) optimal transcription of the E1a protein in tumor but not growth arrested normal cells.
  • At least one of Pea3 II, Pea3 III, Pea3 IV, and Pea3 V, or a functional portion thereof is deleted or modified (e.g., at least one nucleotide of the sequence is changed or an additional nucleotide is inserted into the sequence).
  • at least one of Pea3 II and Pea3 III, or a functional portion thereof is deleted or modified.
  • Pea3 II or a functional portion thereof, and Pea3 III or a functional portion thereof is deleted or modified.
  • at least one of Pea3 IV and Pea3 V, or a functional portion thereof is deleted or modified.
  • Pea3 I, or a functional portion thereof is retained.
  • the element is present in the recombinant adenoviral vector, preferably at the same location as a reference adenoviral vector. In one aspect, at least one E2F1 binding site, or a functional portion thereof, is retained.
  • the vector is d1309-6, TAV-255, d155, d1200, d1230, or d1200+230. In another aspect, the vector is TAV-255. In another aspect, the E1a deletions in d1309-6, TAV-255, d155, d1200, d1230, d1200+230, or other E1a modifications affecting Pea3 and/or E2F1 binding sites between -394 to -218, are paired with a non-d1309 based E3 deletion such as the E3 deletion found in pBHG10 (Microbix, Ad5 base pairs (bp) 28133-30818), d1327 (Ad5 bp 28593-30470) or a similar size E3 deletion such that >3 kb of exogenous DNA can be successfully packaged and expressed from a recombinant adenovirus with the E1a deletions listed above, in combination with a deletion between the start site of the E1b 19K
  • the vector is a d1309 vector having one or more mutations in reference to the wild type sequence of Ad5 (see, e.g., Chroboczek et al., Virology (1992) January; 186(1):280-5, herein incorporated by reference), including a disruption in the coding sequences for one or more of the 10.4K, 14.5K, and 14.7K proteins in the E3 region.
  • a recombinant virus selectively expresses at least one E1a isoform, e.g., E1a-12S or E1a-13S.
  • the sequence encoding the E1a isoform is operably linked to a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is deleted or modified.
  • a recombinant virus comprises a DNA sequence, e.g., a transgene, inserted into an E1b-19K insertion site.
  • the insertion site is located between the start site of E1b-19K and the start site of E1b 55K.
  • the insertion site comprises a deletion of 202 base pairs following the start site of E1b-19K.
  • a transgene also, “insert” may be a full natural sequence of the gene of interest or a fragment thereof. It may be modified too include a Kozak sequence, stop codon, or other regulatory elements.
  • a transgene may include one or more endonuclease restriction sites.
  • the transgene is operably linked to a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is modified or deleted.
  • the transgene may be located in the E1, E2, E3 and/or E4 regions of the adenovirus but its expression is controlled by E1a mediated activation of the endogenous upstream adenovirus promoter, generating high levels of transgene expression only during E1a mediated viral replication.
  • the transgene is located in the E3 region.
  • Exogenous transgenes can be inserted into one or more deleted regions of the adenovirus E3 region, generally such transgenes are inserted into the adenoviral vector as to have their expression controlled by the endogenous E3 promoter (Luo J et al, Clin Cancer Res 2008: 14 2450-2457). This results in high levels of transgene expression that are specifically expressed during periods of viral replication, as the E3 promoter only becomes transcriptionally active during these times.
  • expression from the introduced transgene is also limited to infected tumor cells and, generally, not normal cells.
  • the entire E3 region, or a substantial portion of the E3 region is deleted.
  • the transgene is located in the E4 region.
  • the E4 region of adenovirus there are regions in the E4 region of adenovirus that can be deleted without significant effect on viral growth characteristics (Gao, G P et al, J. Virol 1996: 70; 8934-8943).
  • at least ORF3 and/or ORF6 are retained in the adenoviral vector in which the remainder of the E4 region. It should be possible to insert a foreign transgene into one of these deletions in the E4 region and drive expression of this gene with the endogenous E4 promoter of adenovirus, restricting high level expression of this gene to conditions where viral replication is expected to occur.
  • the adenoviruses described herein can be engineered to express an immunomodulatory agent or immunomodulatory polypeptide, e.g., a polypeptide agonist of a co-stimulatory signal of an immune cell.
  • the polypeptide agonist is an agonist of a T effector cell and/or the polypeptide agonist functions as a polypeptide antagonist of an inhibitory signal of an immune cell such as a regulatory T cell.
  • an “immunomodulatory protein” or an “immunomodulatory polypeptide” includes any polypeptide or set of polypeptides capable of modulating (e.g., stimulating) the anti-tumor immune response induced by the adenovirus.
  • an “immunomodulatory polypeptide” includes a desired immunostimulatory activity.
  • An immunomodulatory polypeptide can include a set of polypeptides, linked or unlinked, that can form a multimer (e.g., a dimer) capable of modulating the anti-tumor immune response induced by adenovirus, e.g., IL-12 dimer formed from p40 and p35, with or without a linker.
  • Immunomodulatory polypeptides can be full length proteins as occur in nature or can be fusions, variants, or fragments thereof that retain at least about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10% of the immunomodulatory activity of the full-length protein.
  • an agonist refers to a molecule(s) that binds to another molecule and induces an increased biological reaction.
  • an agonist is a molecule that binds to a receptor on a cell and triggers or stimulates one or more signal transduction pathways.
  • an agonist can include an antibody or ligand that binds to a receptor on a cell and induces one or more signal transduction pathways.
  • the agonist facilitates the interaction of the native ligand with the native receptor.
  • the term “antagonist(s)” refers to a molecule(s) that inhibits the action of another molecule, optionally without provoking an independent biological response itself.
  • an antagonist is a molecule that binds to a receptor on a cell and blocks or dampens the biological activity of an agonist.
  • an antagonist can include an antibody or ligand that binds to a receptor on a cell and blocks or dampens binding of the native ligand to the cell, optionally without inducing one or more signal transduction pathways.
  • Another example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand.
  • a further example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand or blocks receptor internalization, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand.
  • the immunomodulatory polypeptide expressed by the adenovirus is a costimulatory ligand, e.g., GITR ligand (GITRL), OX40 ligand (OX40L), or CD40 ligand (CD40L).
  • GITRL GITR ligand
  • OX40L OX40 ligand
  • CD40L CD40 ligand
  • the immunomodulatory polypeptide expressed by the adenovirus is a pro-inflammatory cytokine, e.g., GMCSF, IL-7, IL-12, or an IL-15 hybrid (e.g., a hybrid of IL-15 and IL-15 receptor alpha).
  • a pro-inflammatory cytokine e.g., GMCSF, IL-7, IL-12, or an IL-15 hybrid (e.g., a hybrid of IL-15 and IL-15 receptor alpha).
  • IL-15 hybrid e.g., a hybrid of IL-15 and IL-15 receptor alpha
  • the immunomodulatory polypeptide expressed by the adenovirus is an inhibitor (e.g., a “receptor trap” or a “trap”) of an inhibitory cytokine, e.g., IL-10 or IL-27.
  • the immunomodulatory agent/inhibitor of an inhibitory cytokine is an antibody against, e.g., TGFB or IL-10R. Such inhibitory cytokines decrease T effector cell function.
  • inhibitory cytokine receptor traps results in a tumor-localized binding to and neutralization of the inhibitory cytokine, thereby reducing or preventing its inhibitory activity and increasing the anti-tumor activity of the pharmaceutical composition, and also increasing safety of the composition by decreasing or eliminating undesired effects of systemic administration of a blockage of an inhibitory cytokine.
  • the immunomodulatory polypeptide expressed by the adenovirus is an initiator of a localized immune response, e.g., a protein (e.g., secreted flagellin) that activates a toll-like receptor ligand such as TLR-5.
  • a protein e.g., secreted flagellin
  • TLR-5 a toll-like receptor ligand
  • the immunomodulatory polypeptide expressed by the adenovirus is an inhibitor (e.g., an antibody antagonist) of a co-inhibitory checkpoint molecule, e.g., CTLA4.
  • a co-inhibitory checkpoint molecule e.g., CTLA4.
  • Such inhibitory checkpoint molecules decrease T effector cell function.
  • Expression of one or more inhibitory checkpoint molecules at the surface of an activated T cell e.g., an activated T effector cell attenuates the functional activity of the T cell.
  • expression of an antagonist of the co-inhibitory checkpoint molecule in the tumor microenvironment results in blocking the co-inhibitory activity and increasing the anti-tumor activity of the pharmaceutical composition, and also increasing safety of the composition by decreasing or eliminating undesired effects of systemic administration of a blockage of a co-inhibitory checkpoint molecule.
  • the immunomodulatory polypeptide expressed by the adenovirus is a cluster of differentiation (CD) molecule or a ligand of a cluster of differentiation (CD) molecule such as CD27, e.g., CD70.
  • CD27 a cluster of differentiation
  • CD70 a ligand of a cluster of differentiation molecule
  • Expression of a CD27 ligand in the tumor microenvironment results in a tumor-localized NK-mediated tumor clearance and promotes the adaptive immune response against the tumor, thereby increasing the anti-tumor activity of the pharmaceutical composition.
  • CD molecules include CD1d, CD2, CD3, CD4, CD5, CD6, CD7, CD8a, CD8b, CD9, CD21, CD25, CD37, CD40, CD49b, CD53, CD57, CD69, CD80, CD81, CD82, CD86, CD99, CD103, CD134, CD152, CD154, CD165, CD244, CD267, CD272, CD273, CD274, CD278, CD305, CD314, CD357, and CD360, or the ligands thereof, or any modulator (e.g., a stimulator or an inhibitor) thereof.
  • modulator e.g., a stimulator or an inhibitor
  • the immunomodulatory polypeptide expressed by the adenovirus is selected from a polypeptide provided in Table 1.
  • the heterologous gene is a GITR ligand family gene, such as TNFSF18 (also known as GITRL) (See, e.g., Tone, M., Tone, Y., Adams, E., Yates, S. F., Frewin, M. R., Cobbold, S. P., & Waldmann, H. (2003).
  • TNFSF18 also known as GITRL
  • TNFSF18 See, e.g., Tone, M., Tone, Y., Adams, E., Yates, S. F., Frewin, M. R., Cobbold, S. P., & Waldmann, H. (2003).
  • Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proceedings of the National Academy of Sciences of the United States of America, 100(25), 15059-15064. doi:10.1073/pnas.2334901100).
  • the GITR/GITRL signaling pathway is associated with activation of immune cells Nocentini, G., Ronchetti, S., Petrillo, M. G., & Riccardi, C. (2012). Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. British Journal of Pharmacology, 165(7), 2089-99. doi:10.1111/j.1476-5381.2011.01753.x).
  • the GITRL protein has an immunomodulatory activity including inhibiting the suppressive activity of T regulatory cells and activation of T effector cells.
  • the intratumoral localization of effective amounts of GITRL protein results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • the costimulatory activity of GITRL protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation
  • the heterologous gene is an engineered IL-10 trap, such as IL-10 receptor fused to a human immunoglobulin Fc domain.
  • IL-10 trap such as IL-10 receptor fused to a human immunoglobulin Fc domain.
  • examples include IL10RA-Fc fusion protein or IL10RA-IL10RB-Fc fusion protein.
  • Cytokine traps multi-component, high-affinity blockers of cytokine action. Nature Medicine, 9(1), 47-52. doi:10.1038/nm811).
  • the IL-10 family is associated with inhibition of inflammatory response in immune cells through inhibition of the expression of proinflammatory cytokines and co-stimulatory molecules.
  • Expression of IL-10 by T regulatory cells suppresses the activity of T effector cells (Mosser D M, Zhang X. Immunol Rev. 2008 December; 226:205-18. Interleukin-10: new perspectives on an old cytokine).
  • the IL-10 trap protein has an immunomodulatory activity by inhibiting the anti-inflammatory activity of IL-10.
  • the intratumoral localization of effective amounts of IL-10 trap protein results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • IL-10 trap protein inhibition of IL-10's anti-inflammatory activity has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • the IL-10 Receptor Trap includes all or a portion of the extracellular domains of IL-10Ra and IL-10Ra.
  • the heterologous gene is an antibody (or domain or fragment thereof) that inhibits the function of CTLA4 (See, e.g., Leach D R, Krummel M F, Allison J P. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996 Mar. 22; 271(5256):1734-6.)
  • CTLA4 family is associated with inhibition of T cells through the interaction with ligands CD80 and CD86 (Krummel M F, Allison J P (1995). “CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation”. J. Exp. Med.
  • the anti-CTLA4 antibody has an immunomodulatory activity including blocking the inhibitory function of CTLA4 resulting in more efficient activation of T effector cells.
  • the intratumoral localization of effective amounts of anti-CTLA4 antibody results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • anti-CTLA4 inhibition of CTLA4's T cell inhibitory activity has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • the heterologous gene is a member of the Interleukin cytokine family such as IL-12.
  • the IL-12 cytokine family is associated with induction of IFN ⁇ and mediating T-cell dependent immunity.
  • IL-12 is an immunostimulatory cytokine with strong antiangiogenic effects.
  • IL-12 has immunomodulatory activity including cell proliferation, lymphocyte differentiation and NK cell activation.
  • the intratumoral localization of effective amounts of IL-12 results in the differentiation, proliferation, and maintenance of T helper 1 (Th1) responses that lead to IFN ⁇ and IL-2 production that in turn, promote T cell responses and macrophage activation.
  • Th1 T helper 1
  • the local expression of effective amounts of IL-12 from intratumoral injections may provide a safety benefit over systemic administration and side effects associated with high IL-12 serum levels.
  • the immunostimulatory activity and induction of cytotoxicity mediated by natural killer cells and T cells by IL-12 may have a synergistic effect with the tumor-directed cell-lytic and immune stimulating activity of our adenovirus providing a more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • the IL-12 polypeptide is a fusion of IL-12 subunits p35 and p40, linked by a 45 bp linker, including IL-12 ⁇ :p40 (NM_001303244) and IL-12 Alpha:p35 (NM_008351.1).
  • the heterologous gene is a cytokine such as Granulocyte-macrophage colony-stimulating factor (GM-CSF), also known as colony stimulating factor 2 (CSF2).
  • GM-CSF Granulocyte-macrophage colony-stimulating factor
  • CSF2 colony stimulating factor 2
  • Cytokines are secreted proteins or peptides that mediate and regulate immunity and inflammation.
  • GM-CSF has an immunomodulatory activity of functioning as an immune adjuvant and facilitates development of the immune system, acting as a growth factor for DCs and APCs.
  • the localized secretion of effective amounts of GM-CSF results in an increase in dendritic cell (DC) maturation and function as well as macrophage activity, recruiting immune cells to the inflammatory site of tumor treatment, resulting in more effective therapeutic treatment of human subjects suffering from cancer.
  • DC dendritic cell
  • GM-CSF has been demonstrated to be capable of induced long-lasting, specific anti-tumor immunity when combined with cancer vaccines, potentially providing a synergistic effect with the tumor-directed cell-lytic activity of our adenovirus.
  • an oncolytic herpes simplex virus armed with GM-CSF (T-VEC) showed durable response rates in advanced melanoma patients compared with GM-CSF protein alone.
  • the heterologous gene comes from a gram-negative bacterium in the Salmonellae family, such as the gene encoding flagellin.
  • Bacterial proteins, including flagellin are associated with the activation of the innate immune response, leading to production of proinflammatory cytokines and the up-regulation of costimulatory molecules.
  • flagellin is a TLR5 agonist, and binding of secreted flagellin to TLR5 stimulates production of TNF ⁇ , and induces infiltration of APC's and TIL's to the local tumor environment.
  • flagellin By acting as a strong adjuvant, flagellin is able to prime the immune system to elicit strong adaptive immune responses, resulting in enhanced and broadened immune response a more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • secreted Flagellin contains a murine IL-2 signal sequence (See NM_008366) and Salmonella Flagellin, GenBank: D13689.
  • the heterologous gene is an engineered TNF ⁇ ligand family gene, such as a non-cleavable (membrane-bound, transmembrane) form of TNF ⁇ (See, e.g., Li Q, Li L, Shi W, Jiang X, Xu Y, Gong F, Zhou M, Edwards C K 3rd, Li Z., Mechanism of action differences in the antitumor effects of transmembrane and secretory tumor necrosis factor-alpha in vitro and in vivo. Cancer Immunol Immunother. 2006: 55, 1470-9.).
  • a non-cleavable (membrane-bound, transmembrane) form of TNF ⁇ See, e.g., Li Q, Li L, Shi W, Jiang X, Xu Y, Gong F, Zhou M, Edwards C K 3rd, Li Z., Mechanism of action differences in the antitumor effects of transmembrane and secretory tumor necrosis factor-
  • TNF ⁇ belongs to a family of pro-inflammatory cytokines (Calcinotto A, Grioni M, Jachetti E, Curnis F, Mondino A, Policyani G, Corti A, Bellone M. Targeting TNF- ⁇ to neoangiogenic vessels enhances lymphocyte infiltration in tumors and increases the therapeutic potential of immunotherapy. J Immunol. 2012; 188: 2687-94.). Specifically, expression of TNF ⁇ in the tumor microenvironment is expected to increase the inflammatory milieu resulting in increased anti-tumor immune responses. Use of a non-cleavable TNF ⁇ results in a tethered form of TNF ⁇ which remains membrane-bound.
  • TNF ⁇ protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • local expression may provide a safety benefit over systemic administration of TNF ⁇ .
  • the immunomodulatory activity of the membrane-bound TNF ⁇ protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • the heterologous gene is an OX40L family gene, such as TNFSF4 (also called OX40L, CD252) (See, e.g., Dannull J, Nair S, Su Z, Boczkowski D, DeBeck C, Yang B, Gilboa E, Vieweg J. Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA encoding OX40 ligand. Blood. 2005, 105: 3206-13.).
  • the TNFSF is associated with activation of immune cells (Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev.
  • OX40L is a co-stimulatory ligand for TNFRSF4 (OX40, CD134) resulting in activation of T cells.
  • OX40L in the tumor microenvironment and binding to its cognate receptor (OX40) is expected to increase the activity (proliferation, cytokine release) of tumor infiltrating lymphocytes (TILs) resulting in antitumor activity.
  • TILs tumor infiltrating lymphocytes
  • the intratumoral localization of effective amounts of OX40L protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Local expression may provide a safety benefit over systemic administration of OX40L.
  • the immunomodulatory activity of the OX40L protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation IL-7.
  • the heterologous gene is an IL-7 cytokine family gene, such as IL-7 (See, e.g., Gao J, Zhao L, Wan Y Y, Zhu B., Mechanism of Action of IL-7 and Its Potential Applications and Limitations in Cancer Immunotherapy. Int J Mol Sci. 2015, 16: 10267-80.).
  • IL-7 belongs to a family of pro-inflammatory cytokines (Geiselhart L A, Humphries C A, Gregorio TA, Mou S, Subleski J, Komschlies KL.
  • IL-7 administration alters the CD4:CD8 ratio, increases T cell numbers, and increases T cell function in the absence of activation. J Immunol.
  • IL-7 in the tumor microenvironment is expected to increase the inflammatory milieu resulting in increased anti-tumor immune responses.
  • the intratumoral localization of effective amounts of IL-7 protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • the immunomodulatory activity of the IL-7 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • the heterologous gene is a member of the TNF superfamily, such as CD40L (also known as CD40LG or CD154) (See, e.g., Hassan G S, et al., 2015. “Role of CD154 in cancer pathogenesis and immunotherapy.” Cancer Treat Rev 4 1(5):431-40).
  • CD40L is the ligand for CD40 expressed on antigen presenting cells.
  • the CD40L protein has a costimulatory activity important for activation of T cell dependent immune responses (Sotomayor E M, et al., 1999. “Conversion of tumor-directed CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40.” Nat Med.
  • CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help. Nat Med. 5:548-553).
  • the intratumoral localization of effective amounts of CD40 protein results in activation of the immune system and lysis of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • the costimulatory immunomodulatory activity of CD40 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting in a local and systemic immune response against the tumor.
  • the heterologous gene is a cytokine family gene, such as Interleukin 15 (IL-15) (See, e.g., Di Sabatino A, et. al., 2011 “Role of IL-15 in immune-mediated and infectious diseases”. Cytokine Growth Factor Rev. 22 (1): 19-33; Steel J C, et al., 2012, “Interleukin-15 biology and its therapeutic implications in cancer”. Trends Pharmacol. Sci. 33 (1): 35-41).
  • IL-15 is a cytokine that regulates T cell and NK cell activation and proliferation. (Waldmann T A, et al., (1999).
  • IL-15 has an immunomodulatory activity by providing survival signals to maintain memory T cells in the absence of antigen.
  • IL-15 has also been shown to enhance the anti-tumor immunity of CD8+ T cells (See, Klebanoff C A, et al., “IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T Cells” Proc. Natl. Acad. Sci. U.S.A.
  • an IL-15 hybrid includes IL-15, P2A, and IL-15R ⁇ , IL-15 (NM_008357), 68 bp P2A, and IL-15R ⁇ (GenBank: BC132233.1).
  • the heterologous gene is a member of the TNF superfamily, such as CD70 (also known as TNFSF7 or CD27L).
  • CD70 also known as TNFSF7 or CD27L.
  • CD70 is expressed on activated T and B cells, as well as mature dendritic cells, and acts as a ligand for CD27.
  • CD70 plays a costimulatory role in promoting T cell expansion and differentiation (Keller A M, et al., 2008.
  • CD70 protein results in activation of the immune system and lysis of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer.
  • the immunomodulatory activity of CD70 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting in a local and systemic immune response against the tumor.
  • the agonist of a co-stimulatory signal of an immune cell expressed by the adenovirus is an agonist of a co-stimulatory receptor expressed by an immune cell.
  • co-stimulatory receptors include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB(CD137), CD40, CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA).
  • GITR glucocorticoid-induced tumor necrosis factor receptor
  • ICOS or CD278 Inducible T-cell co-stimulator
  • OX40 CD134
  • the agonist of a co-stimulatory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) or ligand that specifically binds to the co-stimulatory receptor.
  • the antibody is a monoclonal antibody.
  • the antibody is an sc-Fv.
  • the antibody is a bispecific antibody that binds to two receptors on an immune cell.
  • the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell.
  • the antibody is a human or humanized antibody.
  • the ligand or antibody is a chimeric protein.
  • the antagonist of an inhibitory signal of an immune cell is an antagonist of an inhibitory receptor expressed by an immune cell.
  • inhibitory receptors include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD160.
  • the antagonist of an inhibitory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) that specifically binds to the co-stimulatory receptor.
  • Antibodies include, but are not limited to, monoclonal antibodies, bispecific antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, single domain antibodies, camelid or camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked bispecific Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies), and epitope-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • an antibody is a human or humanized antibody.
  • an antibody is a monoclonal antibody or scFv.
  • an antibody is a human or humanized monoclonal antibody or scFv.
  • the antibody is a bispecific antibody.
  • the bispecific antibody specifically binds to a co-stimulatory receptor of an immune cell or an inhibitory receptor of an immune, and a receptor on a cancer cell. In some embodiments, the bispecific antibody specifically binds to two receptors immune cells, e.g., two co-stimulatory receptors on immune cells, two inhibitory receptors on immune cells, or one co-stimulatory receptor on immune cells and one inhibitory receptor on immune cells.
  • the recombinant AVs described herein may be engineered to express any agonist of a co-stimulatory signal and/or any antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage).
  • the agonist and/or antagonist is an agonist of a human co-stimulatory signal of an immune cell and/or antagonist of a human inhibitory signal of an immune cell.
  • the agonist of a co-stimulatory signal is an agonist of a co-stimulatory molecule (e.g., co-stimulatory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • a co-stimulatory molecule e.g., co-stimulatory receptor
  • immune cells such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • co-stimulatory molecules include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB (CD137), CD40, lymphotoxin alpha (LT alpha), LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes), CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA).
  • GITR glucocorticoid-induced tumor necrosis factor receptor
  • ICOS or CD278 Inducible T-cell co-stimulator
  • OX40 CD134
  • the agonist is an agonist of a human co-stimulatory receptor of an immune cell.
  • the agonist of a co-stimulatory receptor is not an agonist of ICOS.
  • the antagonist is an antagonist of an inhibitory molecule (e.g., inhibitory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages).
  • inhibitory molecules include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD 160, adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD 160.
  • the antagonist is an antagonist of a human inhibitory receptor of an immune cell.
  • the agonist of a co-stimulatory receptor is an antibody or antigen-binding fragment thereof that specifically binds to the co-stimulatory receptor.
  • co-stimulatory receptors include GITR, ICOS, OX40, CD27, CD28, 4-1BB, CD40, LT alpha, LIGHT, CD226, CRT AM, DR3, LTBR, TACI, BAFFR, and BCMA.
  • the antibody is a monoclonal antibody.
  • the antibody is an sc-Fv.
  • the antibody is a bispecific antibody that binds to two receptors on an immune cell.
  • the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell.
  • the antibody is a human or humanized antibody.
  • the agonist of a co-stimulatory receptor expressed by the adenovirus is a ligand of the co-stimulatory receptor.
  • the ligand is fragment of a native ligand.
  • native ligands include ICOSL, B7RP1, CD137L, OX40L, CD70, herpes virus entry mediator (HVEM), CD80, and CD86.
  • HVEM herpes virus entry mediator
  • the nucleotide sequences encoding native ligands as well as the amino acid sequences of native ligands are known in the art.
  • B7RP1 alsowise known as ICOSL; GenBank human: NM_015259.4, NP_056074.1 murine: NM_015790.3, NP_056605.1
  • CD137L GenBank human: NM_003811.3, NP_003802.1, murine: NM_009404.3, NP_033430.1
  • OX40L GenBank human: NM_003326.3, NP_003317.1, murine: NM_009452.2, NP_033478.1
  • CD70 GeneBank human: NM_001252.3, NP_001243.1, murine: NM_011617.2, AAD00274.1
  • CD80 GenBank human: NM_005191.3, NP_005182.1, murine: NM_009855.2, NP_033985.3
  • CD86 GenBank human: NM_005191.3, CAG46642.1, murine: NM_005191.3, CAG46642.1,
  • the ligand is a derivative (e.g., a fragment, domain, fusion, or other modification of a full-length polypeptide) a native ligand.
  • the ligand is a fusion protein comprising at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and a heterologous amino acid sequence.
  • the fusion protein comprises at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and the Fc portion of an immunoglobulin or a fragment thereof.
  • An example of a ligand fusion protein is a 4-IBB ligand fused to Fc portion of immunoglobulin (described by Meseck M et al., J Immunother. 2011 34: 175-82).
  • the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) or a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • native ligands for inhibitory receptors include PDL-1, PDL-2, B7-H3, B7-H4, HVEM, Gal9 and adenosine.
  • Specific examples of inhibitory receptors that bind to a native ligand include CTLA-4, PD-1, BTLA, KIR, LAG3, TIM3, and A2aR.
  • the antagonist of an inhibitory receptor expressed by the adenovirus is a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • the soluble receptor is a fragment of a native inhibitory receptor or a fragment of a derivative of a native inhibitory receptor that specifically binds to native ligand ⁇ e.g., the extracellular domain of a native inhibitory receptor or a derivative of an inhibitory receptor).
  • the soluble receptor is a fusion protein comprising at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor (e.g., the extracellular domain of the native inhibitory receptor or a derivative of the native inhibitory receptor), and a heterologous amino acid sequence.
  • the fusion protein comprises at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor, and the Fc portion of an immunoglobulin or a fragment thereof.
  • An example of a soluble receptor fusion protein is a LAG3-Ig fusion protein (described by Huard B et al, Eur J Immunol (1995) 25:2718-21).
  • the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s).
  • the antibody is a monoclonal antibody.
  • the antibody is an scFv.
  • the antibody is a human or humanized antibody.
  • a specific example of an antibody to inhibitory ligand is anti-PD-L1 antibody (Iwai Y, et al. PNAS 2002; 99: 12293-12297).
  • the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) or ligand that binds to the inhibitory receptor, but does not transduce an inhibitory signal(s).
  • inhibitory receptors include CTLA-4, PD1, BTLA, TIGIT, KIR, LAG3, TIM3, and A2aR.
  • the antibody is a monoclonal antibody.
  • the antibody is an scFv.
  • the antibody is a human or humanized antibody.
  • a specific example of an antibody to inhibitory receptor is anti-CTLA-4 antibody (Leach D R, et al. Science 1996; 271: 1734-1736).
  • Another example of an antibody to inhibitory receptor is anti-PD-1 antibody (Topalian S L, NEJM 2012; 28:3167-75).
  • a chimeric adenovirus described herein is engineered to produce an antagonist of CTLA-4, such as, e.g., ipilimumab or tremelimumab.
  • a chimeric adenovirus described herein is engineered to an antagonist of PD1, such as, e.g., MDX-1106 (BMS-936558), MK3475, CT-011, AMP-224, or MDX-1105.
  • a chimeric adenovirus described herein is engineered to express an antagonist of LAG3, such as, e.g., IMP321.
  • a chimeric adenovirus described herein is engineered to express an antibody (e.g., a monoclonal antibody or an antigen-binding fragment thereof, or scFv) that binds to B7-H3, such as, e.g., MGA271.
  • a chimeric adenovirus described herein is engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • adenovirus described herein is engineered to express anti-CD28 scFv, ICOSL, CD40L, OX40L, CD137L, GITRL, and/or CD70.
  • an agonist of a co-stimulatory signal of an immune cell expressed by the adenovirus induces (e.g., selectively) induces one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand.
  • an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%>, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands in the absence of the agonist.
  • an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not induce, or induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to
  • an agonist of a co-stimulatory signal of an immune cell activates or enhances (e.g., selectively activates or enhances) one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand.
  • an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%), or 75%) to 100% relative to the one or more signal transduction pathways induced by the binding of co-stimulatory receptor to one or more of its ligands in the absence of the agonist.
  • an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not activate or enhance, or activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5%
  • an antagonist of an inhibitory signal of an immune cell inhibits or reduces one or more of the signal transduction pathways induced by the binding of an inhibitory receptor to its ligand.
  • an antagonist of an inhibitory receptor inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands in the absence of the antagonist.
  • an antagonist of an inhibitory receptor (i) inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to the one particular ligand in the absence of the antagonist; and (ii) does not inhibit or reduce, or inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or
  • an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell induces, activates and/or enhances one or more immune activities, functions or responses.
  • the one or more immune activities, functions or responses can be in the form of, e.g., an antibody response (humoral response) or a cellular immune response, e.g., cytokine secretion (e.g., interferon-gamma), helper activity or cellular cytotoxicity.
  • expression of an activation marker on immune cells e.g., CD44, Granzyme, or Ki-67
  • expression of a co-stimulatory receptor on immune cells e.g., ICOS, CD28, OX40, or CD27
  • expression of a ligand for a co-stimulatory receptor e.g., B7HRP1, CD80, CD86, OX40L, or CD70
  • cytokine secretion infiltration of immune cells (e.g., T-lymphocytes, B lymphocytes and/or NK cells) to a tumor, antibody production, effector function, T cell activation, T cell differentiation, T cell proliferation, B cell differentiation, B cell proliferation, and/or NK cell proliferation is induced, activated and/or enhanced following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • myeloid-derived suppressor cell tumor infiltration and proliferation, Treg tumor infiltration, activation and proliferation, peripheral blood MDSC and Treg counts are inhibited following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • a chimeric adenovirus described herein is engineered to produce two or more immunomodulatory polypeptides.
  • the chimeric adenovirus produces a first immunomodulatory polypeptide and a second immunomodulatory polypeptide.
  • a first immunomodulatory polypeptide is a costimulatory ligand, a proinflammatory cytokine, an inhibitor of an inhibitory cytokine, an initiator of a localized immune response, an inhibitor of a co-inhibitory checkpoint molecule, or a ligand of a cluster of differentiation (CD) molecule
  • the second immunomodulatory polypeptide is a costimulatory ligand, a proinflammatory cytokine, an inhibitor of an inhibitory cytokine, an initiator of a localized immune response, an inhibitor of a co-inhibitory checkpoint molecule, or a ligand of a cluster of differentiation (CD) molecule.
  • a first immunomodulatory polypeptide is a costimulatory ligand
  • the second immunomodulatory polypeptide is a costimulatory ligand
  • a first immunomodulatory polypeptide is a costimulatory ligand and the second immunomodulatory polypeptide is a pro-inflammatory cytokine.
  • two or more immunomodulatory polypeptides are expressed from a single transcript.
  • an internal ribosome entry site (IRES) sequence is commonly used to drive expression of the second, third, fourth coding sequence, etc.
  • IRES internal ribosome entry site
  • the translational expression level of the second coding sequence is often significantly reduced (Furler et al. 2001. Gene Therapy 8:864-873).
  • the use of an IRES to control transcription of two or more coding sequences operably linked to the same promoter can result in lower level expression of the second, third, etc. coding sequence relative to the coding sequence adjacent the promoter.
  • an IRES sequence may be sufficiently long to impact complete packaging of the vector, e.g., the eCMV IRES has a length of 507 base pairs.
  • IRES Internal ribosome entry site
  • IRES immunoglobulin heavy-chain binding protein
  • VEGF vascular endothelial growth factor
  • FGF-2 fibroblast growth factor 2
  • IGFII insulin-like growth factor
  • EMCV encephelomyocarditis virus
  • IRES VEGF-associated vascular endothelial growth factor receptor 1 (Huez et al. 1998 . Mol. Cell. Biol. 18:6178-90). IRES have also been reported in different viruses such as cardiovirus, rhinovirus, aphthovirus, HCV, Friend murine leukemia virus (FrMLV) and Moloney murine leukemia virus (MoMLV).
  • IRES encompasses functional variations of IRES sequences as long as the variation is able to promote direct internal ribosome entry to the initiation codon of a cistron.
  • An IRES may be mammalian, viral or protozoan.
  • the IRES promotes direct internal ribosome entry to the initiation codon of a downstream cistron, leading to cap-independent translation.
  • the product of a downstream cistron can be expressed from a bicistronic (or multicistronic) mRNA, without requiring either cleavage of a polyprotein or generation of a monocistronic mRNA.
  • Internal ribosome entry sites are approximately 450 nucleotides in length and are characterized by moderate conservation of primary sequence and strong conservation of secondary structure.
  • the most significant primary sequence feature of the IRES is a pyrimidine-rich site whose start is located approximately 25 nucleotides upstream of the 3′ end of the IRES. See Jackson et al. (1990).
  • the cardio- and aphthovirus class for example, the encephalomyocarditis virus, Jang et al. 1990 . Gene Dev 4:1560-1572
  • the entero- and rhinovirus class for example, polioviruses, Borman et al. 1994 . EMBO J. 13:3149-3157
  • HAV hepatitis A virus
  • the ribosome entry site is an AUG triplet located at the 3′ end of the IRES, approximately 25 nucleotides downstream of a conserved oligopyrimidine tract.
  • Translation initiation can occur either at the ribosome entry site (cardioviruses) or at the next downstream AUG (entero/rhinovirus class). Initiation occurs at both sites in aphthoviruses.
  • HCV and pestiviruses such as bovine viral diarrhea virus (BVDV) or classical swine fever virus (CSFV) have 341 nt and 370 nt long 5′-UTR respectively. These 5′-UTR fragments form similar RNA secondary structures and can have moderately efficient IRES function (Tsukiyama-Kohara et al. 1992 . J. Virol. 66:1476-1483; Frolov et al. 1998 . RNA 4:1418-1435). Recent studies showed that both Friend-murine leukemia virus (MLV) 5′-UTR and rat retrotransposon virus-like 30S VL30) sequences contain IRES structure of retroviral origin (Torrent et al. 1996 . Hum.
  • MMV Friend-murine leukemia virus
  • IRES elements include, without limitation, immunoglobulin heavy-chain binding protein (BiP) (Macejak et al. 1991 . Nature 353:90-94), antennapedia mRNA of Drosophila (Oh et al. 1992.
  • BiP immunoglobulin heavy-chain binding protein
  • FGF-2 fibroblast growth factor-2
  • PDGF-B platelet-derived growth factor B
  • IRES element IRES element
  • IRES sequences include Picornavirus HAV (Glass et al. 1993. Virology 193:842-852); EMCV (Jang and Wimmer. 1990 . Gene Dev. 4:1560-1572); Poliovirus (Borman et al. 1994. EMBO J. 13:3149-3157); HCV (Tsukiyama-Kohara et al. 1992. J. Virol. 66:1476-1483); pestivirus BVDV (Frolov et al. 1998. RNA.
  • IRES may be prepared using standard recombinant and synthetic methods known in the art. For cloning convenience, restriction sites may be engineered into the ends of the IRES fragments to be used.
  • two immunomodulatory polypeptides are expressed from separate transcripts, i.e., a first transcript and a second transcript.
  • the two transcripts are encoded by a DNA insertion at the same location in the adenovirus, e.g., both inserted in E1b, E3, or E4.
  • the two transcripts are encoded by a DNA insertion at the different locations in the adenovirus, e.g., a first transcript DNA inserted in E1b and a second transcript DNA inserted in E3 or E4, or alternatively, a first transcript DNA inserted in E3 and a second transcript DNA inserted in E4.
  • Another aspect of the invention provides a virus for causing expression in a target cell of a plurality of recombinant immunomodulatory polypeptides or other protein(s) or polypeptides of interest, wherein the vector also includes a promoter operably linked to a first coding sequence for a first recombinant immunomodulatory polypeptide, a self-processing or other cleavage coding sequence, such as a 2A or 2A-like sequence or a protease recognition site, and a second coding sequence for a second recombinant immunomodulatory polypeptide, wherein the self-processing cleavage sequence or protease recognition site coding sequence is inserted between the first and the second coding sequences.
  • a promoter operably linked to a first coding sequence for a first recombinant immunomodulatory polypeptide
  • a self-processing or other cleavage coding sequence such as a 2A or 2A-like sequence or a protease recognition site
  • the viral vector comprises an expression vector as described above wherein the expression vector further comprises an additional proteolytic cleavage site between the first and second recombinant immunomodulatory polypeptides.
  • a preferred additional proteolytic cleavage site is a furin cleavage site with the consensus sequence RXR/K-R.
  • Oncolytic viruses were originally conceived as simply a means of targeted destruction of cancer cells. However, it is now thought that the most effective OV therapies will be those that combine tumor cell death with the stimulation of a host anti-tumor immune response. OVs engineered to express particular immunomodulatory cytokines in tumor cells will be able to specifically guide the immune system toward combating cancer cells. Combining the expression of these cytokines with the release of tumor-associated antigens (TAAs, i.e. tumor cell debris) upon viral lysis of cancer cells will allow for the development of cellular or antibody-mediated anti-tumor immune responses (Lichty et al., 2014, Nature Reviews Cancer, 14: 559-567).
  • TAAs tumor-associated antigens
  • a pharmaceutical composition of the invention comprises at least one of the vectors of the invention as described herein. Furthermore, the composition may comprise at least two, three or four different (i.e., expressing different transgenes) vectors of the invention.
  • a pharmaceutical composition may also comprise any other vectors, such as other adenoviral vectors, other therapeutically effective agents, any other agents such as pharmaceutically acceptable carriers, buffers, excipients, adjuvants, antiseptics, filling, stabilizing or thickening agents, and/or any components, e.g., such as components found in corresponding viral or pharmaceutical products.
  • the cancer is nasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer
  • the vector or pharmaceutical composition of the invention may be administered to any eukaryotic subject selected from a group consisting of animals and human beings, in a preferred embodiment of the invention, the subject is a human or a non-human animal.
  • An animal may be selected from a group consisting of pets, domestic animals and production animals.
  • the adenoviral vector(s) of the present invention may be administered to a subject, preferably suspended in a biologically compatible solution or pharmaceutically acceptable delivery vehicle.
  • a suitable vehicle includes sterile saline.
  • Other aqueous and non-aqueous isotonic sterile injection solutions and aqueous and non-aqueous sterile suspensions known to be pharmaceutically acceptable carriers and well known to those of skill in the art may be employed for this purpose.
  • the therapeutic of the present invention may also be administered directly to the tumor in the form of a liquid, gel or suspension introduced by intratumoral injection.
  • a study examining the treatment of mice bearing subcutaneous human pancreatic adenocarcinoma xenografts with recombinant Newcastle disease virus (rNDV) showed that intratumoral injection yielded better tumor regression than intravenous injection.
  • animals were injected intratumorally every other day for a total of 4 injections, each containing 5 ⁇ 10′ 50% Tissue Culture Infective Dose (TCID50) rNDV in 50 ⁇ l (Buijs et al., 2015, Viruses, 6: 2980-2998).
  • mice bearing subcutaneous bladder cancer xenografts with modified oncolytic adenovirus found that intratumoral injection significantly suppressed tumor growth.
  • animals were injected intratumorally twice at a 1-day interval with 5 ⁇ 10 8 infectious unit (IFU) viruses in 100 ⁇ l (Yang et al., 2015, Cell Death and Disease, e1760).
  • IFU infectious unit
  • HCC hepatocellular carcinoma
  • oncolytic adenoviral vectors of the invention may have therapeutic effects.
  • oncolytic adenoviral vectors or pharmaceutical compositions are administered several times during the treatment period.
  • Oncolytic adenoviral vectors or pharmaceutical compositions may be administered for example from 1 to 10 times in the first 2 weeks, 4 weeks, monthly or during the treatment period.
  • administration is done three to seven times in the first 2 weeks, then at 4 weeks and then monthly
  • administration is done four times in the first 2 weeks, then at 4 weeks and then monthly.
  • the length of the treatment period may vary, and for example may last from two to 12 months or more.
  • the therapeutic of the present invention is administered with an adjuvant.
  • Suitable adjuvants include aluminum salts (alum) such as aluminum hydroxide, aluminum phosphate, and aluminum sulfate, Incomplete Freund's Adjuvant (IFA), and monophosphoryl lipid A (MPL). These adjuvants are suitable for human administration, either alone or optionally all combinations thereof (Chang et al., “Adjuvant Activity of Incomplete Freund's Adjuvant,” Adv Drug Deliv Rev 32:173-186 (1998), which is hereby incorporated by reference in its entirety).
  • cytokines such as interleukins (IL-1, IL-2, and IL-12), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), di- and tri-palmitoyl-S-glyceryl cysteine (Pam2Cys and Pam3Cys, respectively), a TLR2 agonist, an anti-granulocyte macrophage colony-stimulating factor (GM-CSF) antibody, RR-XS15, Montanide®, and MALP-2.
  • cytokines such as interleukins (IL-1, IL-2, and IL-12
  • M-CSF macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • Pam2Cys and Pam3Cys di- and tri-palmitoyl-S-glyceryl cysteine
  • GM-CSF anti-granulocyte macrophage colony-stimulating factor
  • the adenoviral vector of the present invention can be administered orally, parenterally, for example, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes. They may be administered alone or with suitable pharmaceutical carriers, and can be in solid or liquid (e.g., aqueous) form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • the therapeutic of the present invention may be orally administered, for example, with an inert diluent, or with a suitable edible carrier, or they may be enclosed in hard or soft-shell capsules, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • the therapeutic may be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like.
  • Various other materials may be present as coatings or to modify the physical form of the dosage unit.
  • the therapeutic may also be administered parenterally.
  • Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • the therapeutic of the present invention may also be administered directly to the airways in the form of an aerosol.
  • the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • suitable propellants for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • suitable propellants for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • the materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • the therapeutic of this invention may be administered in sufficient amounts to transfect the desired cells and provide sufficient levels of integration and expression of the replicating virus to provide a therapeutic benefit without undue adverse effects or with medically acceptable physiological effects which can be determined by those skilled in the medical arts.
  • Dosages of the therapeutic will depend primarily on factors, such as the condition being treated, the age, weight, and health of the patient, and may thus vary among patients. The dosage will be adjusted to balance the therapeutic benefit against any viral toxicity or side effects.
  • the present invention also relates to a method of enhancing the delivery to and distribution within a tumor mass of therapeutic proteins expressed from viruses.
  • an adenovirus as described herein optionally in a pharmaceutical composition as described herein, can be injected into a tumor mass such that the virus infects and lyses one or more tumor cell. Combination Therapy.
  • the viral immunotherapy of the invention is effective alone, but combination of multiple adenoviral immunotherapies, or one or more adenoviral immunotherapies with any other therapies, such as traditional therapy, may be more effective than either one alone.
  • each agent of the combination therapy may work independently in the tumor tissue, the adenoviral vectors may sensitize cells to chemotherapy or radiotherapy and/or chemotherapeutic agents may enhance the level of virus replication or effect the receptor status of the target cells.
  • the agents of combination therapy may be administered simultaneously or sequentially.
  • the method or use further comprises administration of concurrent radiotherapy to a subject.
  • the method or use further comprises administration of concurrent chemotherapy to a subject.
  • concurrent refers to a therapy, which has been administered before, after or simultaneously with the gene therapy of the invention. The period for a concurrent therapy may vary from minutes to several weeks. In some embodiments, the concurrent therapy lasts for some hours.
  • Agents suitable for combination therapy include but are not limited to afatinib, all-trans retinoid acid, azacitidine, azathioprine, bleomycin, carboplatin, capecitabine, cisplatin, chlorambucil, cyclophosphamide, cytarabine, daunorubicin, docetaxel, doxifluridine, doxorubicin, epirubicin, epothilone, etoposide, fluorouracil, 5-fluorouracil, gemcitabine, hydroxyurea, idarubicin, imatinib, mechlorethamine, mercaptopurine, methotrexate, mitoxantrone, oxaliplatin, paclitaxel, pemetrexed, temozolomide, teniposide, tioguanine, valrubicin, vinblastine, vincristine, vindesine and vinorelbine.
  • the method or use further comprises administration of verapamil or another calcium channel blocker to a subject.
  • “Calcium channel blocker” refers to a class of drugs and natural substances which disrupt the conduction of calcium channels, and if may be selected from a group consisting of verapamil, dihydropyridines, gallopamil, diltiazem, mibefradil, bepridil, fluspirilene and fendiline.
  • the method or use further comprises administration of autophagy inducing agents to a subject.
  • Autophagy refers to a catabolic process involving the degradation of a cell's own components through the lysosomal machinery.
  • Autophagy inducing agents refer to agents capable of inducing autophagy and may be selected from a group consisting of, but not limited to, mTOR inhibitors (e.g., temsirolimus, sirolimus, everolimus, and ridaforolimus), P13K inhibitors (e.g., wortmannin, lithium, tamoxifen, chloroquine, bafilomycin, and temozolomide.
  • the method further comprises administration of temozolomide to a subject. Temozolomide may be either oral or intravenous temozolomide.
  • the method or use further comprises administration of chemotherapy or anti-CD20 therapy or other approaches for blocking of neutralizing antibodies.
  • Anti-CD20 therapy refers to agents capable of killing CD20 positive cells, and may be selected from a group consisting of rituximab and other anti-CD20 monoclonal antibodies.
  • Approaches for blocking of neutralizing antibodies refers to agents capable of inhibiting the generation of anti-viral antibodies that normally result from infection and may be selected from a group consisting of different chemotherapeutics, immunomodulatory substances, corticoids and other drugs.
  • These substances may be selected from a group consisting of, but not limited to, cyclophosphamide, ciclosporin, azathioprine, methylprednisolone, etoposide, CD40L, CTLA4, FK506 (tacrolimus), IL-12, IFN- ⁇ , interleukin 10, anti-CD8, anti-CD4 antibodies, hematopoietic stem cell transplantation (HSCT) and oral adenoviral proteins.
  • the oncolytic adenoviral vector of the invention induces virion-mediated oncolysis of tumor cells and activates human immune response against tumor cells.
  • the method or use further comprises administration of substances capable to downregulating regulatory T-cells in a subject in an amount to downregulate (e.g., by at least 10%, 20%, 50%, 70%, 90% or more) regulatory T-cells in the subject.
  • “Substances capable to downregulating regulatory T-cells” refers to agents that reduce the numbers of cells identified as T-suppressor or Regulatory T-cells. These cells have been identified as consisting one or many of the following immunophenotypic markers: CD4+, CD25+, FoxP3+, CD127- and GITR+.
  • Such agents reducing T-suppressor or Regulatory T-cells may be selected from a group consisting of anti-CD25 antibodies or chemotherapeutics.
  • the method or use further comprises administration of cyclophosphamide to a subject.
  • Cyclophosphamide is a common chemotherapeutic agent, which has also been used in some autoimmune disorders.
  • cyclophosphamide can be used to enhance viral replication and the effects of GM-CSF induced stimulation of NK and cytotoxic T-cells for enhanced immune response against the tumor. It can be used as intravenous bolus doses or low-dose oral metronomic administration.
  • the invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, reflecting approval by the agency of the manufacture, use or sale of the product for human administration.
  • the practice of the present invention employs conventional techniques of cell culture, molecular biology, microbiology, recombinant DNA manipulation, immunology science, which are within the skill of the art. Such techniques are explained fully in the literature. See, e.g. Cell Biology: a Laboratory Handbook: J. Cells (Ed). Academic Press. N.Y. (1996); Graham, F. L. and Prevec, L. Adenovirus-based expression vectors and recombinant vaccines. In: Vaccines: New Approaches to Immunological Problems. R. W. Ellis (ed) Butterworth. Pp 363-390; Grahan and Prevec Manipulation of adenovirus vectors. In: Methods in Molecular Biology, Vol.
  • Standard techniques are used for recombinant nucleic acid methods, polynucleotide synthesis, and microbial culture and transformation (e.g., electroporation, lipofection). Generally, enzymatic reactions and purification steps are performed according to the manufacturer's specifications. The techniques and procedures are generally performed according to conventional methods in the art and various general references (see generally, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd. edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which are provided throughout this document. The nomenclature used herein and the laboratory procedures in analytical chemistry, organic synthetic chemistry, and pharmaceutical formulation and delivery, and treatment of patients. Methods for the construction of adenoviral mutants are generally known in the art.
  • Adenoviruses and cell lines were generally obtained from American Type Culture Collection (ATCC), Manassas, Va.) unless otherwise noted. Cell lines used in the Examples below may include one or more of the cell lines listed in Table 2.
  • Viral stocks were propagated on HEK-293 cells and purified by standard methods such as column purification kits (Virapure, Millipore) or CsCl gradient centrifugation (as described in Tollefson, A., Hermiston, T. W., and Wold, W. S. M.; “Preparation and Titration of CsCl-banded Adenovirus Stock” in Adenovirus Methods and Protocols, Humana Press, 1999, pp 1-10, W. S. M. Wold, Ed.).
  • the method used to quantitate viral particles is based on simple OD 260/280 readings, e.g., using the method of Lehmberg et al. (1999) J. Chrom.
  • the A260 nm peak area of each sample is directly proportional to the number of viral particles in the sample.
  • the number of viral particles per ml in each test sample was calculated by multiplying the known number of viral particles per ml in the standard by the ratio of the A260 nm viral peak area of the sample to the A260 nm viral peak area of the standard.
  • One A260 unit contains approximately 1 ⁇ 10 12 viral particles.
  • Virus Infectious Units/ml (IU/ml) were determined by hexon staining of infected cells (e.g., using Adeno-XTM Rapid titer kit from Takara Bio USA, Inc. (TBUSA, formerly known as Clontech Laboratories, Inc.).
  • Initial screening of recombinant virus was based on isolated viral DNA rescued from transfected 293 cells where viral propagation based cytopathic effects (CPE) were observed.
  • Viral DNA was used as a template for PCR based detection of sequences flanking the site of the TAV-255 E1a enhancer deletion region. Wild-type sequence would produce a band of 350 bp, while DNA which had the E1a enhancer deletion would only generate a clearly distinguishable 300 bp band.
  • primers specific for internal sequences of the IL-12 transgene and for Ad hexon sequences were used to verify 400 bp and 3 kb PCR amplified bands respectively that would only be present in viral DNA generated by recombination between the two parental plasmids containing either the E1 region and transgene insert or the late Ad structural proteins derived solely from the pAdEasyTM plasmid.
  • Individual constructs were isolated by two rounds of plaque purification on A549 or 293 cells using standard methods (Tollefson, A., Hermiston, T. W., and Wold, W. S.
  • Tumor specific viral lysis was evaluated in both tumor and non-tumor cell lines of murine and human origin by infection of the cells in vitro with the viruses, followed by standard crystal violet for cell viability over time as instructed in the kit manuals.
  • DNA sequencing of the human Ad5-based recombinant adenovirus genomic DNAs was performed as follows. Viral DNA was purified from recombinant adenoviruses such as TAV-255 or other modifications of these constructs by standard column purification methods such as the Qia-Amp® blood DNA purification kit from Qiagen®. PCR primers were used to amplify and isolate regions covering the E1a modified regions and the regions containing the transgene insert and sent out for sequencing at a CRO. Isolated DNA was also analyzed by standard restriction digestion and SDS PAGE analysis for verification of appropriate sized bands of digested DNA. Sequence information was analyzed using the Vector NTI program (Informatix).
  • the base shuttle transfer vector includes pXC1 TAV 255 d19k (Zhang et al., Cancer Gene Ther. (2015) 22(1):17-22).
  • This plasmid has a deletion between bp-305 to -255 of the E1a enhancer region, removing two Pea3 and one E2F binding sites which restrict replication and oncolysis of an adenovirus with this deletion to infected tumor cells (Hedrun, F. H., Shantanu K., and Reid, T. (2011) Cancer Gene Therapy 18; 717-723.) It also contains sites allowing deletion of the E1b 19k region and exogenous transgene insert by a Sal I/Xho I digest.
  • cDNAs for each transgene of interest were synthesized (GeneArtTM) or isolated from commercial plasmid sources (GE Lifesciences or GeneCopoeiaTM) by PCR using primers which added on 5′ SalI and 3′ XhoI restriction sequences for insertion into the SalI/XhoI digested pXC1 TAV 255 plasmids.
  • the modified pXC1 TAV 255 gene insert plasmids were amplified in E. coli and purified using Qiagen® Maxi-prep plasmid kits.
  • the pXC1 TAV 255 gene insert-containing plasmids were co-transfected into HEK-293 cells (ATCC) with pBHG10 (Microbix) as described (Bett, A. J., Haddara, W., Prevec, L., and Graham, F. (1994) PNAS 91; 8802-8806), using the calcium phosphate transfection protocol from Molecular Cloning: A laboratory manual (Maniatis Vol. 3; 16.30-16.36) for 2-5 ⁇ g plasmid DNA (for both plasmids so the pXC1 will be in molar excess) per 60 mm dish of cells.
  • pBHG10 is provided as the adenoviral genome source, having substantial additional utility over vectors such as pJM17 (See, e.g., Hedrun et al, (2011)).
  • an E3 deletion and/or other modifications allow increased packaging capacity for exogenous genes in excess of pJM17 capacity.
  • the human IL-12 virus, TRZ627 was constructed using a modification of the pAdEasyTM Adenoviral Vector System (Agilent Technologies).
  • sequences from pXCI-TAV d19K plasmid (Hedjran F et al., Cancer Gene Therapy (2011) 18, 717-723) which included the 50-nucleotide deletion in the enhancer of E1A which restricts viral propagation to tumor vs. non-tumor cells, were subcloned into the pShuttleTM vector supplied in the pAdEasyTM kit to create the TAV-255 Shuttle E1 cloning plasmid.
  • the added sequences also included a modification introducing Sal I/Xho I cloning sites into the E1b 19k coding region which can be used to replace the E1b 19k ORF with an exogenous transgene insert whose expression would be driven by the Ad E1b promoter during viral replication.
  • Recombination between Pme I linearized pTAV-255 Shuttle E1 containing hIL-12 cloned in at the E1b 19k site and the pAdEasy vector took place in the recA proficient BJ5183 bacterial strain, which had been modified to already contain the pAdEasy plasmid.
  • DNA isolated from Kanamycin resistant plated colonies was screened for full-length viral DNA recombinants by restriction digest. Positive clones (TRZ-627, hIL-12) were subsequently digested with Pac I to free up the Ad ITRs and then transfected into 293 cells to amplify the virus.
  • Virus can be purified from the lysate by several methods, including Anion-exchange HPLC (Shabram, P. W., et al (1997) Human Gene Therapy 8; 453-465) or several commercially available kits based on affinity chromatography or size exclusion membranes or columns (e.g., Adeno-XTM Maxi Purification Kit, Clontech® (Takara), Adenovirus Purification Virakit®, Virapur®). Purified virus can also undergo clonal isolation by standard plaque purification methods, followed by re-amplification and purification of the plaque purified viral clone.
  • Anion-exchange HPLC Shabram, P. W., et al (1997) Human Gene Therapy 8; 453-465) or several commercially available kits based on affinity chromatography or size exclusion membranes or columns (e.g., Adeno-XTM Maxi Purification Kit, Clontech® (Takara), Adenovirus Purification Virakit®, Virapur®
  • Example 6 In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Encoding Immunomodulatory Polypeptides: Single Viruses with and without Antibody Treatment
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples were stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials were stored protected from light at ⁇ 80° C. On each day of dosing, one vial was thawed at room temperature for approximately 20 minutes. A single dose is 1 ⁇ 10 9 pfu.
  • mice Female Jackson 129S1 (129S1/Sv1mJ) mice were used in this study. They were 6-7 weeks old on Day 1 of the experiment. The animals were fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum.
  • the environment was controlled to a temperature range of 70° ⁇ 2° F. and a humidity range of 30-70%.
  • ADS-12 cells were grown in RPMI 1640 medium which was modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100 ⁇ Penicillin/Streptomycin/L-Glutamine (PSG).
  • FBS non-heat-inactivated Fetal Bovine Serum
  • PSG Penicillin/Streptomycin/L-Glutamine
  • the trypsin was inactivated by dilution with complete growth medium and any clumps of cells were separated by pipetting.
  • the cells were centrifuged at 200rcf for 8 minutes at 4° C., the supernatant was aspirated, and the pellet was re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) by pipetting.
  • DPBS cold Dulbecco's Phosphate Buffered Saline
  • An aliquot of the homogeneous cell suspension was diluted in a trypan blue solution and counted using a Luna automated cell counter. The cell suspension was centrifuged at 200rcf for 8 minutes at 4° C.
  • the supernatant was aspirated and the cell pellet was re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) to generate a final concentration of 1 ⁇ 10 7 trypan-excluding cells/ml.
  • DPBS cold Dulbecco's Phosphate Buffered Saline
  • the cell suspension was maintained on wet ice during implantation. Following implantation, an aliquot of the remaining cells was diluted with a trypan blue solution and counted to determine the post-implantation cell viability.
  • the cell viabilities of the suspensions used for implantation are listed in Table 3.
  • Test animals were implanted subcutaneously on both flanks (on the back between the spine and the hip), the right flank on Day 0 and the left flank on Day 8, with 1 ⁇ 10 6 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • mice were sorted into study groups based on caliper measurement estimation of tumor burden on Day 15 when the mean tumor burden for all animals on the right flank was approximately 82 mm 3 (range of group means, 75-90 mm 3 ). The mice were distributed to ensure that the mean tumor burden on the right flank for all groups was within 10% of the overall mean tumor burden for the study population.
  • the mean estimated right side tumor burden for all groups in the experiment on the first day of treatment was 82 mm3 and all of the groups in the experiment were well-matched (range of group means, 75-90 mm3). All animals weighed at least 13.3 g at the initiation of therapy. Mean group body weights at first treatment were also well-matched (range, 15.4-18.3 g).
  • a tumor burden of 500 mm3 was chosen for evaluation of efficacy by tumor growth delay for the right and left tumors.
  • the median Control Group ( FIG. 6A ) tumor burdens reached 500 mm3 on Day 47 for right tumors and Day 43 for left tumors.
  • the median tumor volume doubling times for the Control Group were 12.1 and 10.1 days for the right and left tumors, respectively.
  • mice with palpable left side tumors were put on study. Since in this experiment the implant was delayed 8 days, the left side tumors were not palpable at staging. These mice that had a left side tumor that remained a 0 throughout the study could have been triaged out of the study if implanted on the same day as the right side.
  • results of mouse inoculation and tumor growth are shown in FIG. 6 .
  • FIG. 6A is a graph showing the activity of various oncolytic viruses compared to the empty virus (“d19k”), 38 days after cell implantation (primary tumor) as a function of tumor growth inhibition. Black bars represent virus alone and hatched bars represent virus+anti-PD-L1 antibody.
  • FIG. 6B is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), each of five viruses alone (viruses carrying CTLA-4, IL-12, IL-7, CD70, and IL-10 transgenes) or combined with anti-PD-L1 antibody.
  • FIG. 6A is a graph showing the activity of various oncolytic viruses compared to the empty virus (“d19k”), 38 days after cell implantation (primary tumor) as a function of tumor growth inhibition. Black bars represent virus alone and hatched bars represent virus+
  • 6C shows similar data for three more viruses (viruses carrying OX40L, CD40L, and GM-CSF transgenes).
  • viruses carrying OX40L, CD40L, and GM-CSF transgenes.
  • the IL-12 oncolytic virus treatment alone or in combination with an anti-PD-L1 antibody, were the most effective at reducing tumor growth.
  • CD70, IL-7, and CTLA-4 viruses were also able to reduce tumor volume significantly when combined with the anti-PD-L1 antibody.
  • the following Figures demonstrate efficacy (or lack thereof) of various viruses with transgenes with or without PD-L1 on the primary tumor only; the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • Treatments are shown for the primary tumor (receiving the oncolytic virus injection) and are as follows: 6 D, virus buffer and control IgG only (left) and virus buffer and anti-PD-L1 antibody (right); 6 E, d19k (empty virus) and control IgG only (left) and d19k and anti-PD-L1 antibody (right); 6 F, CTLA-4 virus with control IgG (left) or anti-PD-L1 (right); 6 G, IL-12 virus with control IgG (left) or anti-PD-L1 antibody (right); 6 H, GM-CSF virus with control IgG (left) or anti-PD-L1 antibody (right); 6 I, IL-7 virus with control IgG (left) or anti-PD-L1 antibody (right); 6 J, CD40L virus with control IgG (left) or anti-PD-L1 antibody (right); 6 K, L10 trap virus with control IgG (left) or anti-PD-L1 antibody (right); and 6 L, OX40
  • Example 7 In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Encoding Immunomodulatory Polypeptides: Virus Mixing Study
  • the mouse tumor model in this Example uses syngeneic immunocompetent mice. Animals were injected with approximately 1,000,000 cells subcutaneously (s.c.) into the right hind flank of the mouse. At the same time, the mice were also injected with 1,000,000 cells s.c. into the left hind flank of the mouse to create a bilateral tumor model. When the right (primary) tumors reached 63-80 mm 3 in size and the left tumor was palpable, they were injected with 25 ⁇ l of virus buffer or 25 ⁇ l of virus at 4 ⁇ 10 9 pfu/ml (plaque forming units per ml) directly into the center of the primary tumor every fourth day for a total of three doses.
  • mice were also treated intraperitoneally with 250 ⁇ l of an anti-PD-L1 antibody at 2 mg/ml every fourth day for a total of three doses.
  • the antibody was administered 24 hours after administration of the virus.
  • a reduction in the size of the primary and distal (contralateral) tumor would be noted relative to the virus buffer control and additional controls such as the wild-type virus (not expressing a transgene) with or without administration of the anti-PD-L1 antibody.
  • a specific example of the treatment with oncolytic adenoviral vectors is described in more detail below for a panel of such vectors.
  • ADS-12 a murine KRAS-mutant lung adenocarcinoma cell line grown in its syngeneic mouse strain is a tumor model known to support adenoviral infection and replication and is useful in the evaluation of host immune responses to oncolytic human adenoviruses.
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples were stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials were stored protected from light at ⁇ 80° C. On each day of dosing, one vial was thawed at room temperature for approximately 20 minutes.
  • mice Female 12951 (129S1/SvImJ) mice from The Jackson Laboratory were used in this study. They were approximately 7-8 weeks old on Day 14 of the experiment. The animals were fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum. Animals were housed in static cages with Bed-O'CobsTM bedding inside Biobubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour. All treatments, body weight determinations, and tumor measurements were carried out in the bubble environment. The environment was controlled to a temperature range of 70° ⁇ 2° F. and a humidity range of 30-70%. All procedures carried out in this experiment were conducted in compliance with all the laws, regulations and guidelines of the National Institutes of Health (NIH) and with the approval of Molecular Imaging, Inc.'s Animal Care and Use Committee. Molecular Imaging, Inc. is an AAALAC accredited facility.
  • NASH National Institutes of Health
  • ADS-12 cells (murine KRAS-mutant lung adenocarcinoma) were grown in RPMI 1640 medium which is modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100 ⁇ Penicillin/Streptomycin/L-Glutamine (PSG).
  • FBS non-heat-inactivated Fetal Bovine Serum
  • PSG 100 ⁇ Penicillin/Streptomycin/L-Glutamine
  • the growth environment was maintained in an incubator with a 5% CO 2 atmosphere at 37° C. When expansion as complete, the cells are trypsinized using 0.25% trypsin/2.21 mM EDTA in HBSS solution. Following the cell viabilities of the suspensions used for implantation (two preps) are listed in the table below.
  • Test animals were implanted subcutaneously, on both flanks (on the back between the spine and the hip) on Day 0 with 1.00 ⁇ 10 6 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • mice All mice were sorted into study groups based on caliper measurement estimation of tumor burden on Day 14 when the mean tumor burden for all animals on the right flank is approximately 68 mm 3 (range of group means, 65-71 mm 3 ). The mice were distributed to ensure that the mean tumor burden for all groups was within 10% of the overall mean tumor burden for the study population.
  • the primary endpoints used to evaluate efficacy were: tumor growth delay, complete and partial tumor response, and the number of tumor-free survivors at the end of the study for both left and right tumors.
  • a complete response (CR) is defined as a decrease in tumor mass to an undetectable size ( ⁇ 63 mm 3 )
  • a partial response (PR) is defined as a smaller tumor mass at the last measurement compared to at the first treatment. PRs are exclusive of CRs.
  • results of mouse inoculation and tumor growth are shown in FIG. 7 .
  • the left-hand panel of each Figure represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor.
  • the thick line in each graph shows the average tumor volume in mm 3 .
  • the pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • Treatments shown are as follows: 7 A, virus buffer only; 7 B, empty virus only (TRZ000); 7 C, TRZ010 (IL-10trap)+empty virus; 7 D, TRZ011 (OX40 ligand)+empty virus; 7 E, TRZ009 (CD70)+empty virus; 7 F, TRZ007 (IL-7)+empty virus, 7 G, TRZ002 (IL-12)+empty virus; 7 H, TRZ004 (GM-CSF)+empty virus; 7 I, TRZ003 (flagellin)+empty virus; 7 J, TRZ002+TRZ010; 7 K, TRZ002+TRZ007; 7 L, TRZ007+TRZ010; 7 M, TRZ011+TRZ004; 7 N, TRZ009+TRZ003; 7 O, TRZ002+TRZ009; 7 P, TRZ007+TRZ009; 7 Q, TRZ007+TRZ004; 7 R, TR
  • FIG. 7G shows tumor volume in mice treated with an IL-12 adenovirus. 5 out of 8 mice in this Figure had a complete response, and one mouse had a partial response. In the contralateral tumor, however, no complete or partial responses were seen, indicating that there were no systemic immune effects from treatment with the IL-12 virus alone.
  • FIG. 7K shows mice treated with the same IL-12 adenovirus in combination with an IL-7 adenovirus.
  • mice 7 out of 8 had a complete response in the primary tumor (into which the virus was injected) and in the contralateral tumor two mice showed a complete response and one a partial response.
  • FIGS. 7V (IL-12 adenovirus+anti-PD-L1 antibody) and 7 AA (IL-12 adenovirus+CD70 adenovirus+anti-PD-L1 antibody) each group of 8 mice had 8 complete responses in the primary tumor and several in the contralateral tumor.
  • deletions in the adenovirus backbone are sufficiently large enough to allow packaging of viral DNA containing two exogenously added transgenes into the viral capsids, the two genes can be co-expressed by several methods from a single deletion site in adenovirus. Both added genes can be linked to each other by methods described below and have their expression controlled by an endogenous adenovirus promoter, not an exogenously added promoter, so that high expression will only occur during conditions of viral replication. Control of restricting viral replication to certain conditions such as after infection of tumor cells, is described elsewhere in this document.
  • Co-expression of two proteins from a single transcript can be achieved through the use of virus components such as internal ribosome entry site (IRES) elements (Renaud-Gabardos E et al, World J Exp Med 2015, 5: 11-20), insertion of self-cleaving 2A peptide sequences derived from viruses such as Foot and Mouth Disease virus (FMDV) (Garry A. Luke (2012), Translating 2A Research into Practice, Innovations in Biotechnology , Dr. Eddy C. Agbo (Ed.), ISBN: 978-953-51-0096-6, InTech), or by combining the sequences of the two transgenes into a single fusion protein.
  • An exemplary method would use one of the 2A sequences to direct more equal level of expression from both transgenes as opposed to lower expression levels typically seen from the second transgene when using IRES elements.
  • any of the transgenes may be inserted into the E1 or E3 region.
  • a dual transgene construct may have IL-12 inserted into the E1 region and IL-2 in the E3 region.
  • a dual transgene construct may have IL-2 inserted into the E1 region and IL-12 in the E3 region.
  • TRZ Number Brief Description
  • TRZ402 TAV-255-d19kE1-mIL-10T-E3-mIL-12
  • TRZ403 TAV-255-d19kE1-mIL-7-E3-mIL-12
  • TRZ404 TAV-255-d19kE1-mCD70-E3-IL-12
  • TRZ405 TAV-255-d19kE1-mOX40L-E3-mGM-CSF
  • TRZ406 TAV-255-d19kE1-mIL-10T-E3-mIL-7
  • TRZ407 TAV-255-d19kE1-mIL-12-E3-mIL-10T
  • TRZ408 TAV-255-d19k-E3-mIL-12
  • TRZ409 E1-mIL-12/E3-mIL-7
  • TRZ418 E1-trimeric mCD70/E3-mIL
  • the cDNA for each transgene can be inserted into separate deleted regions of adenovirus so that expression of each would be controlled separately by the endogenous upstream adenovirus promoter. No exogenous promoter would be added with the exogenous transgene sequence. Expression of the E1a proteins leads to the activation of the other adenovirus promoters and viral replication, so expression from each endogenous adenovirus promoter is linked to viral replication.
  • Exogenous transgenes inserted behind different adenovirus promoters such as the E1b promoter, the E3 promoter, and the E4 promoter, in place of deletions in these regions, leads to a construct where co-expression from each inserted transgene is limited to conditions of where viral replication occurs.
  • co-expression of both exogenous transgenes is restricted to tumor cells.
  • an ⁇ E1b19K site that includes a deletion of bp 1714-1916 (numbered according to hAd5 vector sequence), which increases packaging capacity by approximately 200 bp.
  • a deletion at the E3 site ⁇ E3 site as shown in FIG. 2 ); this deletion can be any deletion in the E3 region, generally at or about bp 27,900-30,800 bp (numbered according to hAd5 vector sequence), which increases packaging capacity up to an additional approximately 2400 bp, as compared to most wild type adenoviruses, which are typically limited to containing (i.e., packaging) approximately 1800 bp of exogenous sequences.
  • deletions in E3 open reading frames that are suitable for modification (e.g., truncation or deletion) without substantially decreasing viral propagation.
  • adenoviral vectors wherein the E3 12.5K coding region (27,852-28,175 bp) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 320 or greater than 320 bases are deleted from one or more truncation sites within the region) or entirely deleted.
  • adenoviral vectors wherein the E3 7.1K coding region (28,541-28,732) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or greater than 190 bases are deleted from one or more truncation sites within the region) or entirely deleted.
  • the 7.1K sequence is associated with inhibition of TRAIL apoptosis and associated with one or more RID proteins.
  • adenoviral vectors wherein the E3 gp19K (28,729-29211) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450 or greater than 450 bases are deleted from one or more truncation sites within the region) or entirely deleted.
  • the gp19K sequence is associated with inhibition of CTL killing.
  • E3 10.5 also called E3 11.6 (ADP)
  • ADP E3 11.6
  • 29, 485-29,766 adenoviral vectors wherein the E3 10.5 (also called E3 11.6 (ADP)) (29, 485-29,766) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, or greater than 250 bases are deleted from one or more truncation sites within the region) or entirely deleted.
  • the 10.5 sequence is associated with promotion of virus release.
  • adenoviral vectors wherein the E3 (RID ⁇ ) (29,778-29,969) and/or the E3 (RID ⁇ ) (30,057-30,455) is truncated (e.g., with respect to E3 (RID ⁇ ) at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or greater than 190 bases are deleted from one or more truncation sites within the region) or entirely deleted, or truncated (e.g., with respect to E3 (RID ⁇ ) at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 390, or greater than 390 bases are deleted from one or more truncation
  • adenoviral vectors wherein the E3 14.7K (30,488-30,834) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 340, or greater than 340 bases are deleted from one or more truncation sites within the region) or entirely deleted.
  • the 14.7K sequence is associated with inhibition of TNF, FasL, and TRAIL apoptosis.
  • a nucleic acid sequence encoding one or more immunomodulatory polypeptides is inserted into the adenoviral genome by truncation or deletion of a portion of the E4 region of the viral genome.
  • An E4 deletion and exogenous insertion can be utilized in combination with any other viral modification provided herein or otherwise known in the art, or alternatively, without any other viral modification.
  • Exemplary E4 regions useful as insertion sites include truncation or deletion of E4 ORF1 (35,136-35,522 bp) and/or E4 ORF2 (34,696-35,106 bp) (each numbered according to hAd5 vector sequence).
  • the expression of the one or more immunomodulatory polypeptides is controlled by the endogenous E4 promoter.
  • a schematic illustration of this embodiment is provided in FIG. 3 .
  • Adenoviral vectors are designed in which sequences in and around Pea3 sites I-V near the E1a Enhancer region of adenoviral vectors, in which sequences in and around Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics.
  • adenovirus vectors are engineered which have lower affinity Pea3 sites compared to wild-type adenoviral vectors.
  • Such vectors are designed to be efficient in cells in the tumor microenvironment where the concentration of transcription factors is high, but to be relatively inactive in normal (e.g., non-neoplastic) cells, thus reducing the possibility of side effects caused by damage to normal tissue cells.
  • FIG. 9A An illustration of the E1a area of wild-type and TAV-255 constructs is shown in FIG. 9A .
  • FIG. 9B A cartoon illustration of the proposed method of action of the E1a enhancer region mutants is shown in FIG. 9B .
  • Constructs NV1 to NV7 are based in part on the TAV-255 construct.
  • a vector is provided wherein, (in relation to the wild-type sequence) the sequence is removed between Pea3 IV and Pea3 III sites, a single mutation is made in Pea3 III, the sequence between Pea3 III and Pea3 II, and the Pea3 II site is mutated.
  • the sequence of the E1a enhancer region of the exemplary construct NV1 is set forth in SEQ ID NO:99.
  • a vector wherein, (in relation to the wild-type sequence) the sequence is removed between Pea3 IV and Pea3 III sites, both Pea3 III and Pea3 II are mutated, and Pea3 V flanking sites are mutated such that the resultant Pea3 V site has an affinity more similar to Pea3 III and enhancer 1.
  • the sequence of the E1a enhancer region of the exemplary construct NV2 is set forth in SEQ ID NO:100.
  • a vector wherein, (in relation to the wild-type sequence) the sequence between Pea3 V and Pea3 IV sites is replaced with the sequence between the Pea3 III and Pea3 II sites; the sequence between the Pea3 IV and Pea3 III is deleted; the sequence between the Pea3 III and Pea3 II is deleted; and the Pea3 III and Pea3 II sites are both mutated, as well as the residues 3 bp that are immediately 5′ of the Pea3 V site.
  • the sequence of the E1a enhancer region of the exemplary construct NV3 is set forth in SEQ ID NO:101.
  • a vector wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites (thus engineering a vector in which all Pea3 sites except Pea3 I are lower affinity).
  • the sequence of the E1a enhancer region of the exemplary construct NV4 is set forth in SEQ ID NO:102.
  • a vector is provided wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites, and a single point mutation is introduced in the Pea3 I site, rendering it a lower affinity Pea3 I site (thus engineering a vector in which all Pea3 sites are lower affinity).
  • the sequence of the E1a enhancer region of the exemplary construct NV5 is set forth in SEQ ID NO:103.
  • a vector is provided wherein, (in relation to the wild-type sequence), the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites, and the flanking regions of the Pea3 I site are altered, rendering it a lower affinity Pea3 I site (thus engineering a vector in which all Pea3 sites are lower affinity).
  • the sequence of the E1a enhancer region of the exemplary construct NV6 is set forth in SEQ ID NO:104.
  • a vector wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 I site are altered to produce a lower-affinity Pea3 I site, and the flanking sequences around the Pea3 II site is altered to mimic the lower affinity Pea3 IV site (thus engineering a vector in which all Pea3 sites except Pea3 III are lower affinity).
  • the sequence of the E1a enhancer region of the exemplary construct NV7 is set forth in SEQ ID NO:105.
  • Example 12 In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Comprising E1a Enhancer Region Alterations and Encoding Immunomodulatory Polypeptides: Single Viruses with and without Antibody Treatment
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples are stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials are stored protected from light at ⁇ 80° C. On each day of dosing, one vial is thawed at room temperature for approximately 20 minutes. A single dose is 1 ⁇ 10 9 pfu.
  • Viruses to be combined with anti-PD-L1 in this Example include NV1-NV7 (having E1a enhancer region sequences set forth in SEQ ID Nos:99-105).
  • mice Female Jackson 129S1 (12951/Sv1mJ) mice are used in this study. Mice are 6-7 weeks old on Day 1 of the experiment. The animals are fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum. Animals are housed in static cages with Bed-O'CobsTM bedding inside bioBubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour. All treatments, body weight determinations, and tumor measurements are carried out in the bubble environment. The environment is controlled to a temperature range of 70° ⁇ 2° F. and a humidity range of 30-70%.
  • ADS-12 cells are grown in RPMI 1640 medium which is modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100 ⁇ Penicillin/Streptomycin/L-Glutamine (PSG).
  • FBS non-heat-inactivated Fetal Bovine Serum
  • PSG Penicillin/Streptomycin/L-Glutamine
  • the trypsin is inactivated by dilution with complete growth medium and any clumps of cells are separated by pipetting.
  • the cells are centrifuged at 200rcf for 8 minutes at 4° C., the supernatant is aspirated, and the pellet is re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) by pipetting.
  • DPBS cold Dulbecco's Phosphate Buffered Saline
  • An aliquot of the homogeneous cell suspension is diluted in a trypan blue solution and counted using a Luna automated cell counter.
  • the cell suspension is centrifuged at 200rcf for 8 minutes at 4° C.
  • the supernatant is aspirated and the cell pellet is re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) to generate a final concentration of 1 ⁇ 10 7 trypan-excluding cells/ml.
  • DPBS cold Dulbecco's Phosphate Buffered Saline
  • the cell suspension is maintained on wet ice during implantation. Following implantation, an aliquot of the remaining cells is diluted with a trypan blue solution and counted to determine the post-implantation cell viability.
  • Test animals are implanted subcutaneously on both flanks (on the back between the spine and the hip), the right flank on Day 0 and the left flank on Day 8, with 1 ⁇ 10 6 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • mice are sorted into study groups based on caliper measurement estimation of tumor burden on Day 15 when the mean tumor burden for all animals on the right flank is approximately 82 mm 3 (range of group means, 75-90 mm 3 ). The mice are distributed to ensure that the mean tumor burden on the right flank for all groups is within 10% of the overall mean tumor burden for the study population.
  • the mean estimated right side tumor burden for all groups in the experiment on the first day of treatment is approximately 82 mm 3 and all of the groups in the experiment are well-matched (range of group means, 75-90 mm 3 ). All animals weigh at least 13.3 g at the initiation of therapy. Mean group body weights at first treatment are also well-matched (range, approximately 15.4-18.3 g).
  • a tumor burden of 500 mm 3 is chosen for evaluation of efficacy by tumor growth delay for the right and left tumors.
  • the median Control Group tumor burdens will reach 500 mm 3 on or about Day 47 for right tumors and on or about Day 43 for left tumors.
  • the median tumor volume doubling times for the Control Group will be approximately 12 and 10 days for the right and left tumors, respectively.
  • results of mouse inoculation and tumor growth will show that treatment of tumor bearing mice with oncolytic viruses comprising altered E1a regions and encoding various transgenes, with or without anti-PD-L1, show efficacy in reducing tumor volume.
  • Cancer immunotherapy is moving toward use of combinations to increase efficacy. Combining cancer immunotherapies can expand clinical benefits of existing approved monotherapies; however, systemically-administered combinations can produce excessive toxicity. Therefore, novel dual specificity oncolytic adenoviral vectors were developed having a transgene at both the E1 and the E3 regions in order to evaluate combinations of adenoviral-delivered immunomodulators to enhance systemic antitumor immunity.
  • Table 6 A summary of exemplary constructs contemplated for use by the methods disclosed herein (with additions to those disclosed in Table 5) is listed in Table 6.
  • a bilateral tumor model was prepared using ADS-12 tumor cells as described in the Examples above (e.g., Example 7). 1 ⁇ 10 6 ADS-12 tumor cells injected into primary ( ⁇ 2 days) and contralateral (0 days) flanks. At staging, mice are randomized based on contralateral tumors (88-150 mm 3 ) and primary tumors (88-250 mm 3 ). There are 8 mice/group, with tumors and body weight measured 3 times per week. An anti-PD-L1 (500 ⁇ g/dose) or an anti-PD-1 antibody (250 ⁇ g/dose) is administered i.p. to induce systemic T cell activation.
  • An anti-PD-L1 500 ⁇ g/dose
  • an anti-PD-1 antibody 250 ⁇ g/dose
  • the virus is injected intratumorally (i.t.) into primary tumors only; such injection leads to oncolysis, immune infiltration, and tumor shrinkage.
  • the contralateral tumor is not injected, and no oncolysis occurs. Rather, tumor shrinkage is solely due to antigen-specific activated tumor infiltrating lymphocytes.
  • FIG. 9A is a cartoon of the contrast between the single transgene constructs (top) used in the virus mixing examples, and the dual transgene vectors used to make the E1/E3 transgene adenoviruses.
  • FIG. 9B shows the results of mice injected i.t. with Empty virus+/ ⁇ anti-PD-L1 or with TRZ-409 (IL-12/IL-7 dual transgene)+/ ⁇ anti-PD-L1.
  • the tumor volume of the injected tumor is shown in the left panel, and the tumor volume of the contralateral tumor is shown in the right panel.
  • the left arrow indicates virus injection and the right arrow indicates anti-PD-L1 injection.
  • TRZ-409 injection reduced tumor volume significantly compared to empty virus.
  • the combination with anti-PD-L1 was slightly more efficacious in this study.
  • FIG. 9C shows the results of a second experiment using the inverse of TRZ-409, TRZ-403, in which the IL-7 transgene occupies the E1 region and the IL-12 gene occupies the E3 region (which has a stronger promoter than the E1).
  • TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1.
  • Plasma levels of IL-12 and IFN- ⁇ ( FIG. 9D ) for all mice injected with TRZ-403 show that expression of the transgenes is well tolerated.
  • mice having primary tumors injected with TRZ-403 (IL-7+IL-12), TRZ-403+anti-PD-L1, TRZ-403+control IgG, or TRZ-409 (IL-12+1L-7), and controls including untreated mice and mice injected with empty vector TRZ-d19K with anti-PD-L1, anti-PD-1, or a control IgG are shown in FIG. 9E , which illustrates the survival as a percentage of mice still on study.
  • mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days). Both TRZ-403 and TRZ-409 showed efficacy over the controls; all mice receiving control injections were deceased by day 56 of the study.
  • TRZ-403 11-7+IL-12
  • TRZ-403 showed the most efficacy in reducing tumor volume, followed by the mixture of viruses.
  • the single transgene IL-12 virus showed a small amount of efficacy by comparison.
  • TRZ-403 reduced the tumor volume, showing significant superiority over the mixture of viruses.

Landscapes

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

Abstract

Disclosed herein are compositions and methods for treating cancer in a subject. This involves administering an oncolytic virus containing a heterologous DNA sequence encoding one or more immunomodulatory and/or immunostimulatory polypeptide(s) of interest to the subject under conditions effective to enhance an anti-tumor immune response in the subject, and to treat cancer. It also relates to a method of enhancing the delivery to and distribution within a tumor mass of therapeutic viruses.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of co-pending U.S. application Ser. No. 15/834,690, filed Dec. 7, 2017, which claims the benefit of U.S. Provisional Application Nos. 62/572,206, filed Oct. 13, 2017; 62/440,646, filed Dec. 30, 2016; and 62/440,670, filed Dec. 30, 2016, each of which is incorporated herein by reference in its entirety.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 21, 2017, is named 42022US_CRF_sequencelisting.txt and is 266,186 bytes in size.
  • FIELD
  • The invention described herein relates generally to the fields of immunology, virology, molecular biology, and more specifically to oncolytic adenoviruses having therapeutic applications.
  • BACKGROUND
  • Cancer is a leading cause of death in the United States and elsewhere. Depending on the type of cancer, it is typically treated with surgery, chemotherapy, and/or radiation. These treatments often fail, and it is clear that new therapies are necessary, to be used alone or in combination with current standards of care.
  • Originally conceived as solely tumor-lysing therapeutics, viruses that can preferentially target tumor cells for destruction are being used experimentally as vectors for the delivery of immune-stimulating cargo. The propagation of a lasting anti-tumor host immune response in combination with the destruction of tumor cells is described in, e.g., Lichty et al., 2014, Nature Reviews Cancer, 14: 559-567.
  • Clinical trials employing adenovirus, reovirus, measles, herpes simplex, Newcastle disease virus and vaccinia as oncolytic viruses have suggested that these platforms may all be safe treatment approaches.
  • Adenoviruses are medium-sized (90-100 nm), non-enveloped icosahedral viruses, which have double stranded linear DNA of about 36 kilobase pairs in a protein capsid. The viral capsid has fiber structures that participate in attachment of the virus to the target cell. First, the knob domain of the fiber protein binds to the receptor of the target cell (e.g., CD46 or Coxsackie and adenovirus receptor (CAR)), secondly, the virus interacts with an integrin molecule and thirdly, the virus is endocytosed into the target cell. Next, the viral genome is transported from endosomes into the nucleus and the replication machinery of the target cell is utilized also for viral purposes.
  • The adenoviral genome has early (E1-E4), intermediate (IX and IVa2) and late genes (L1-L5), which are transcribed in sequential order. Early gene products affect defense mechanisms, cell cycle and cellular metabolism of the host cell. Intermediate and late genes encode structural viral proteins for production of new virions.
  • More than 60 different serotypes of adenoviruses have been found in humans. Serotypes are classified into six subgroups A-F and different serotypes are known to be associated with different conditions i.e. respiratory diseases, conjunctivitis and gastroenteritis. Adenovirus serotype 5 (Ad-5) is known to cause respiratory diseases and it is the most common serotype studied in the field of gene therapy. In the first Ad5 vectors E1 and/or E3 regions were deleted enabling insertion of foreign DNA to the vectors.
  • Furthermore, deletions of other regions as well as further mutations have provided extra properties to viral vectors. Indeed, various modifications of adenoviruses have been suggested for achieving efficient anti-tumor effects.
  • Adenoviral vectors mediate gene transfer at a high efficacy compared to other vector systems, and they are currently the most frequently used vectors for cancer gene therapy. A non-replicating p53 expressing adenoviral vector and a replication selective virus (H101) have received regulatory approval in China. Several attempts to achieve tumor-selective control through the insertion of tumor selective promoter elements upstream of the E1 or other adenovirus critical promoters have had variable levels of success, but ultimately were limited by “leaky” gene expression of viral proteins in non-tumor cells and by reduced ability to propagate and lyse tumor cells compared to wild-type virus infections.
  • SUMMARY
  • In a first aspect, a pharmaceutical composition is provided comprising an effective amount of a recombinant adenoviral vector comprising: a transgene insertion site located between the start site of adenoviral E1b-19K and the start site of adenoviral E1b-55K, wherein a first DNA sequence and a second DNA sequence are each inserted into the transgene insertion site; wherein the first DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, and wherein the second DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand; and wherein the adenoviral vector comprises a modified adenoviral E1a regulatory sequence wherein at least one Pea3 binding site, or a functional portion thereof, of the recombinant adenoviral vector is modified or deleted.
  • In one embodiment, the adenoviral vector comprises an IRES element or encodes a self-cleaving 2A peptide sequence between the first DNA sequence and the second DNA sequence. In another embodiment, the vector comprises a modified E3 region. In another embodiment, the vector comprises an intact E3 region. In another embodiment, the vector comprises a third DNA sequence inserted into the E3 region, wherein the third DNA sequence encodes a polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, or a human OX40 ligand. In one embodiment, the chimeric human IL-12 polypeptide comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide. In another embodiment, the chimeric human IL-12 polypeptide comprises a sequence as set forth in SEQ ID NO:46.
  • In one embodiment, the adenoviral vector comprises a nucleic acid sequence at least 95% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 85% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 75% identical in an E3 region to vector d1327.
  • In one embodiment, the pharmaceutical composition is formulated for systemic administration.
  • In another embodiment, the pharmaceutical composition is formulated for systemic administration.
  • In a second aspect is provided a pharmaceutical composition comprising an effective amount of a recombinant adenoviral vector comprising: a first transgene insertion site located between the start site of adenoviral E1b-19K and the start site of adenoviral E1b-55K; a second transgene insertion site located in adenoviral E3 region; a first DNA sequence, present in the first transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand; and a second DNA sequence, present in the second transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, wherein the adenoviral vector comprises a modified adenoviral E1a regulatory sequence. In one embodiment, the pharmaceutical composition further comprises a third DNA sequence inserted into a third transgene insertion site, encoding one or a plurality of polypeptides selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand.
  • In another embodiment, at least one of the first DNA sequence, the second DNA sequence, and the third DNA sequence independently comprises an IRES element and/or a self-cleaving 2A peptide. In one embodiment, at least one E1a regulatory sequence Pea3 binding site, or a functional portion thereof of the adenoviral vector, is modified or deleted. In another embodiment, a sequence between two Pea3 sites of the adenoviral vector is deleted. In one embodiment, the adenoviral vector comprises a modified E3 region. In another embodiment the adenoviral vector comprises a nucleic acid sequence at least 95% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 85% identical in an E3 region to vector d1327. In another embodiment, the adenoviral vector comprises a nucleic acid sequence at least 75% identical in an E3 region to vector d1327.
  • In one embodiment, the chimeric human IL-12 polypeptide comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide.
  • In another aspect is provided a method for treating a tumor in a human subject in need thereof, comprising administering to the human with the tumor a therapeutic amount of the pharmaceutical composition of the first aspect by systemic or intratumoral administration.
  • In another aspect is provided method for treating a tumor in a human subject in need thereof, comprising administering to the human with the tumor a therapeutic amount of the pharmaceutical composition of the second aspect by systemic or intratumoral administration.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a cartoon illustrating transcription factor binding regulation of conformational structure/activity of E1a enhancer region.
  • FIG. 2 is an illustration of a vector map demonstrating an adenoviral vector expressing two immunomodulatory polypeptides.
  • FIG. 3 is an illustration of a vector map demonstrating an adenoviral vector expressing at least one immunomodulatory polypeptide at an E4 site.
  • FIG. 4 is an illustration of the TRZ200 virus genome: an E1b 19k empty, E3-deleted virus.
  • FIG. 5 is an illustration of a vector map showing the plasmid comprising the TRZ6 hIL-12 plasmid.
  • FIG. 6A is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6A shows the activity of various oncolytic viruses compared to the empty virus (“d19k”), 38 days after cell implantation (primary tumor). Black bars represent virus alone and hatched bars represent virus+anti-PD-L1 antibody.
  • FIG. 6B is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6B is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), each of five viruses alone (CTLA-4, IL-12, IL-7, CD70, and IL-10) or combined with anti-PD-L1 antibody.
  • FIG. 6C is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6C is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), showing three viruses—OX40L, CD40L, and GM-CSF alone or combined with anti-PD-L1 antibody.
  • FIG. 6D is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6D shows virus buffer and control IgG only (left) and virus buffer and anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6E is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6E shows d19k (empty virus) and control IgG only (left) and d19k and anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6F is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6F shows CTLA-4 virus with control IgG (left) or anti-PD-L1 (right); the thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6G is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6G shows IL-12 virus with control IgG (left) or anti-PD-L1 antibody (right); the thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6H is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6H shows GM-C S F virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6I is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6I shows IL-7 virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6J is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6J shows CD40L virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6K is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6K shows L10 trap virus with control IgG (left) or anti-PD-L1 antibody (right); The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 6L is graph(s) showing results of treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising transgene(s), with or without anti-PD-L1. FIG. 6L shows OX40L virus with control IgG (left) or anti-PD-L1 antibody. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows).
  • FIG. 7A is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7A shows virus buffer only.
  • FIG. 7B is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7B shows empty virus only (TRZ000).
  • FIG. 7C is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7C shows TRZ010 (IL-10trap)+empty virus.
  • FIG. 7D is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7D shows TRZ011 (OX40 ligand)+empty virus.
  • FIG. 7E is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7E shows TRZ009 (CD70)+empty virus.
  • FIG. 7F is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7F shows TRZ007 (IL-7)+empty virus.
  • FIG. 7G is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7G shows TRZ002 (IL-12)+empty virus.
  • FIG. 7H is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7H shows TRZ004 (GM-CSF)+empty virus.
  • FIG. 7I is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7I shows TRZ003 (flagellin)+empty virus.
  • FIG. 7J is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7J shows TRZ002+TRZ010.
  • FIG. 7K is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7K shows TRZ002+TRZ007.
  • FIG. 7L is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7L shows TRZ007+TRZ010.
  • FIG. 7M is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7M shows TRZ011+TRZ004.
  • FIG. 7N is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7N shows TRZ009+TRZ003.
  • FIG. 7O is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7O shows TRZ002+TRZ009.
  • FIG. 7P is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7P shows TRZ007+TRZ009.
  • FIG. 7Q is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7Q shows TRZ007+TRZ004.
  • FIG. 7R is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7R shows TRZ002+TRZ011.
  • FIG. 7S is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7S shows TRZ010+TRZ004.
  • FIG. 7T is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7T shows TRZ002+TRZ004.
  • FIG. 7U is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7U shows virus buffer and anti-PD-L1.
  • FIG. 7V is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7V shows TRZ002+empty virus+anti-PD-L1.
  • FIG. 7W is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7W shows TRZ009+anti-PD-L1.
  • FIG. 7X is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7X shows TRZ007+empty virus+anti-PD-L1.
  • FIG. 7Y is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7Y shows TRZ002+TRZ007+anti-PD-L1.
  • FIG. 7Z is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7Z shows TRZ007+TRZ009+anti-PD-L1.
  • FIG. 8A is schematic representations of the rationale for modifications of the E1a Enhancer region of adenoviral vectors, in which Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics. FIG. 8A is an illustration of a wild-type adenoviral genome (top) and the TAV-255 adenoviral construct in which residues -305 to -255 are deleted (bottom).
  • FIG. 8B is schematic representations of the rationale for modifications of the E1a Enhancer region of adenoviral vectors, in which Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics. FIG. 8B is an illustration of the potential method of action of the TAV-255 construct, in which the deletion removes Pea3 sites II and III, which moves Pea3 sites IV and V closer to the promoter as distal control elements.
  • FIG. 9A is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. FIG. 9A is a cartoon of the contrast between the single transgene constructs (top) used in the virus mixing examples, and the dual transgene vectors used to make the E1/E3 transgene adenoviruses.
  • FIG. 9B is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. FIG. 9B shows the results of mice injected i.t. with Empty virus+/−anti-PD-L1 or with TRZ-409 (IL-12/IL-7 dual transgene)+/−anti-PD-L1. The tumor volume of the injected tumor is shown in the left panel, and the tumor volume of the contralateral tumor is shown in the right panel. For each pair of arrows, the left arrow indicates virus injection and the right arrow indicates anti-PD-L1 injection. As can be seen for both the primary and contralateral tumors, TRZ-409 injection reduced tumor volume significantly compared to empty virus. The combination with anti-PD-L1 was slightly more efficacious in this study.
  • FIG. 9C is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. FIG. 9C shows the results of a second experiment using the inverse of TRZ-409, TRZ-403, in which the IL-7 transgene occupies the E1 region and the IL-12 gene occupies the E3 region (which has a stronger promoter than the E1). As can be seen in the Figure, TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1.
  • FIG. 9D is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. As can be seen in the Figure, TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1. Plasma levels of IL-12 and IFN-γ (FIG. 9D) for all mice injected with TRZ-403 show that expression of the transgenes is well tolerated. The study was extended for all mice having tumors over 500 mm3.
  • FIG. 9E is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. As can be seen in the FIG. 9E, mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days). Mice having primary tumors injected with TRZ-403 (IL-7+IL-12), TRZ-403+anti-PD-L1, TRZ-403+control IgG, or TRZ-409 (IL-12+1L-7), and controls including untreated mice and mice injected with empty vector TRZ-d19K with anti-PD-L1, anti-PD-1, or a control IgG. As can be seen in the Figure, mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days). Both TRZ-403 and TRZ-409 showed efficacy over the controls; all mice receiving control injections were deceased by day 56 of the study.
  • FIG. 9F is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with dual transgene (expressed from E1 and E3) oncolytic viruses comprising transgene(s), with either anti-PD-L1 antibody or anti-PD-1 antibody treatment. FIG. 9F compares a mixture of two single transgene viruses with a dual transgene virus having the same transgenes. As can be seen in the primary tumor (left panel) TRZ-403 showed the most efficacy in reducing tumor volume, followed by the mixture of IL-7 and IL-12 single transgene viruses. The single transgene IL-12 virus showed a small amount of efficacy by comparison. In the contralateral tumor, however (right panel) only the dual transgene virus, TRZ-403, reduced the tumor volume, showing significant superiority over the mixture of viruses.
  • FIG. 10 is graph(s) showing treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising transgene(s), with or without anti-PD-L1 treatment. The left-hand panel of represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and antibody (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). FIG. 7AA shows TRZ002+TRZ009+anti-PD-L1.
  • DEFINITIONS
  • The term “replicating virus” is meant to include a virus that undergoes the process of intracellular viral multiplication, consisting of the synthesis of proteins, nucleic acids, and sometimes lipids, and their assembly into a new infectious particle.
  • As used herein, the term “adenovirus” refers to any of a group of DNA-containing viruses (small infectious agents) that cause conjunctivitis and upper respiratory tract infections in humans. Adenoviral vectors are described in Peng, Z., “Current Status of Gendicine in China: Recombinant Human Ad-p53 Agent for Treatment of Cancers,” Hum Gene Ther 16:1016-1027 (2005); No authors listed, “The End of the Beginning: Oncolytic Virotherapy Achieves Clinical Proof-of-concept,” Mol Ther 13:237-238 (2006); Vile et al., “The Oncolytic Virotherapy Treatment Platform for Cancer: Unique Biological and Biosafety Points to Consider,” Cancer Gene Ther 9:1062-1067 (2002); Harrison et al., “Wild-type Adenovirus Decreases Tumor Xenograft Growth, but Despite Viral Persistence Complete Tumor Responses are Rarely Achieved—Deletion of the Viral Elb-19-kD Gene Increases the Viral Oncolytic Effect,” Hum Gene Ther 12:1323-1332 (2001); Kim et al., “Clinical Research Results with d11520 (Onyx-015), a Replication-selective Adenovirus for the Treatment of Cancer: What Have We Learned?,” Gene Ther 8:89-98 (2001); and Thorne et al., “Oncolytic Virotherapy: Approaches to Tumor Targeting and Enhancing Antitumor Effects,” Semin Oncol 32:537-548 (2005), each of which is hereby incorporated by reference in their entirety. Adenoviral positions referenced herein are to positions in Adenovirus type 5 (GenBank 10 accession #M73260; the virus is available from the American Type Culture Collection, Rockville, Md., U.S.A., under accession number VR-5). It will be understood that corresponding positions can be identified in other adenovirus vectors by alignment using BLAST 2.0 under default settings (Altschul et al., J. Mol. Biol. 215:403-410 (1990)). Software for performing BLAST analyses is publicly available on the Web through the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov/).
  • Current research in the field of viral vectors is producing improved viral vectors with high-titer and high-efficiency of transduction in mammalian cells (see, e.g., U.S. Pat. No. 6,218,187 to Finer et al., which is hereby incorporated by reference in its entirety). Such vectors are suitable in the present invention, as is any viral vector that comprises a combination of desirable elements derived from one or more of the viral vectors described herein. It is not intended that the expression vector be limited to a particular viral vector.
  • Certain “control elements” or “regulatory sequences” can also be incorporated into the vector-construct. The term “control elements” refers collectively to promoter regions, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites (“IRES”), enhancers, and the like, which collectively provide for the replication, transcription, and translation of a coding sequence(s) in a recipient cell. Not all of these control elements need always be present so long as the selected coding sequence is capable of being replicated, transcribed, and translated in an appropriate host cell.
  • The term “promoter region” is used herein in its ordinary sense to refer to a nucleotide region comprising a DNA regulatory sequence, wherein the regulatory sequence is derived from a gene which is capable of binding RNA polymerase and initiating transcription of a downstream (3′-direction) coding sequence. Transcriptional control signals in eukaryotes comprise “promoter” and “enhancer” elements. Promoter and enhancer elements have been isolated from a variety of eukaryotic sources, including genes in yeast, insect, and mammalian cells, and viruses. Analogous control elements, i.e., promoters, are also found in prokaryotes. Such elements may vary in their strength and specificity. For example, promoters may be “constitutive” or “inducible.”
  • A constitutive promoter is a promoter that directs expression of a gene throughout the development and life of an organism. Examples of some constitutive promoters that are widely used for inducing expression of transgenes include the cytomegalovirus (CMV) early promoter, those derived from any of the several actin genes, which are known to be expressed in most cells types (U.S. Pat. No. 6,002,068 to Privalle et al., which is hereby incorporated by reference in its entirety), and the ubiquitin promoter, which is a gene product known to accumulate in many cell types.
  • To ensure efficient expression, 3′ polyadenylation regions can be present to provide for proper maturation of the mRNA transcripts. The 3′ polyadenylation region will preferably be from the adenovirus sequence downstream of the inserted transgene, but the native 3′-untranslated region of the immunomodulatory gene may be used, or an alternative polyadenylation signal from, for example, SV40, particularly including a splice site, which provides for more efficient expression, could also be used. Alternatively, the 3′-untranslated region derived from a gene highly expressed in a particular cell type could be fused with the immunomodulatory gene.
  • “Inhibitors,” “activators,” and “modulators” of expression or of activity are used to refer to inhibitory, activating, or modulating molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., ligands, agonists, antagonists, and their homologs and mimetics. The term “modulator” includes inhibitors and activators. Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or protease inhibitor activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein, e.g., antagonists. Activators are agents that, e.g., induce or activate the expression of a described target protein or bind to, stimulate, increase, open, activate, facilitate, enhance activation or protease inhibitor activity, sensitize or up regulate the activity of described target protein (or encoding polynucleotide), e.g., agonists. Modulators include naturally occurring and synthetic ligands, antagonists and agonists (e.g., small chemical molecules, antibodies and the like that function as either agonists or antagonists). Such assays for inhibitors and activators include, e.g., applying putative modulator compounds to cells expressing the described target protein and then determining the functional effects on the described target protein activity, as described above. Samples or assays comprising described target protein that are treated with a potential activator, inhibitor, or modulator are compared to control samples without the inhibitor, activator, or modulator to examine the extent of effect. Control samples (untreated with modulators) are assigned a relative activity value of 100%. Inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%. Activation of the described target protein is achieved when the activity value relative to the control is 110%, optionally 150%, optionally 200, 300%, 400%, 500%, or 1000-3000% or higher.
  • “Antibody” refers to a polypeptide comprising a framework region from an immunoglobulin gene or fragments thereof that specifically binds and recognizes an antigen. The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
  • Naturally occurring immunoglobulins have a common core structure in which two identical light chains (about 24 kD) and two identical heavy chains (about 55 or 70 kD) form a tetramer. The amino-terminal portion of each chain is known as the variable (V) region and can be distinguished from the more conserved constant (C) regions of the remainder of each chain. Within the variable region of the light chain is a C-terminal portion known as the J region. Within the variable region of the heavy chain, there is a D region in addition to the J region. Most of the amino acid sequence variation in immunoglobulins is confined to three separate locations in the V regions known as hypervariable regions or complementarity determining regions (CDRs) which are directly involved in antigen binding. Proceeding from the amino-terminus, these regions are designated CDR1, CDR2 and CDR3, respectively. The CDRs are held in place by more conserved framework regions (FRs). Proceeding from the amino-terminus, these regions are designated FR1, FR2, FR3, and FR4, respectively. The locations of CDR and FR regions and a numbering system have been defined by, e.g., Kabat et al. (Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, U.S. Government Printing Office (1991)).
  • An exemplary immunoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively.
  • Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′2 dimer into an Fab′ monomer. The Fab′ monomer is essentially Fab with part of the hinge region (see FUNDAMENTAL IMMUNOLOGY (Paul ed., 3d ed. 1993). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by using recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies, or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv) or those identified using phage display libraries (see, e.g., McCafferty et al., Nature 348:552-554 (1990)).
  • For preparation of monoclonal or polyclonal antibodies, any technique known in the art can be used (see, e.g., Kohler & Milstein, Nature 256:495-497 (1975); Kozbor et al., Immunology Today 4:72 (1983); Cole et al., pp. 77-96 in Monoclonal Antibodies and Cancer Therapy (1985)). “Monoclonal” antibodies refer to antibodies derived from a single clone. Techniques for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce antibodies to polypeptides of this invention. Also, transgenic mice, or other organisms such as other mammals, may be used to express humanized antibodies. Alternatively, phage display technology can be used to identify antibodies and heteromeric Fab fragments that specifically bind to selected antigens (see, e.g., McCafferty et al., Nature 348:552-554 (1990); Marks et al., Biotechnology 10:779-783 (1992)).
  • A “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • A “humanized” antibody is an antibody that retains the reactivity of a non-human antibody while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody with their human counterparts. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol., 44:65-92 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988); Padlan, Molec. Immun., 28:489-498 (1991); Padlan, Molec. Immun., 31(3):169-217 (1994).
  • As used herein, the terms “treat” and “treating” in the context of the administration of a therapy refers to a treatment/therapy from which a subject receives a beneficial effect, such as the reduction, decrease, attenuation, diminishment, stabilization, remission, suppression, inhibition or arrest of the development or progression of cancer, or a symptom thereof. In certain embodiments, the treatment/therapy that a subject receives results in at least one or more of the following effects: (i) the reduction or amelioration of the severity of cancer and/or a symptom associated therewith; (ii) the reduction in the duration of a symptom associated with cancer; (iii) the prevention in the recurrence of a symptom associated with cancer; (iv) the regression of cancer and/or a symptom associated therewith; (v) the reduction in hospitalization of a subject; (vi) the reduction in hospitalization length; (vii) the increase in the survival of a subject; (viii) the inhibition of the progression of cancer and/or a symptom associated therewith; (ix) the enhancement or improvement the therapeutic effect of another therapy; (x) a reduction or elimination in the cancer cell population; (xi) a reduction in the growth of a tumor or neoplasm; (xii) a decrease in tumor size; (xiii) a reduction in the formation of a tumor; (xiv) eradication, removal, or control of primary, regional and/or metastatic cancer; (xv) a decrease in the number or size of metastases; (xvi) a reduction in mortality; (xvii) an increase in cancer-free survival rate of patients; (xviii) an increase in relapse-free survival; (xix) an increase in the number of patients in remission; (xx) a decrease in hospitalization rate; (xxi) the size of the tumor is maintained and does not increase in size or increases the size of the tumor by less 5% or 10% after administration of a therapy as measured by conventional methods available to one of skill in the art, such as MRI, X-ray, and CAT Scan; (xxii) the prevention of the development or onset of cancer and/or a symptom associated therewith; (xxiii) an increase in the length of remission in patients; (xxiv) the reduction in the number of symptoms associated with cancer; (xxv) an increase in symptom-free survival of cancer patients; and/or (xxvi) limitation of or reduction in metastasis. In some embodiments, the treatment/therapy that a subject receives does not cure cancer, but prevents the progression or worsening of the disease. In certain embodiments, the treatment/therapy that a subject receives does not prevent the onset/development of cancer, but may prevent the onset of cancer symptoms.
  • As used herein, the term “in combination” in the context of the administration of (a) therapy(ies) to a subject, refers to the use of more than one therapy. The use of the term “in combination” does not restrict the order in which therapies are administered to a subject. A first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject.
  • The terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region, e.g., of the entire polypeptide sequences or specific region, if indicated), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be “substantially identical.”
  • For sequence comparison, one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • A “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of, e.g., a full-length sequence or from 20 to 600, about 50 to about 200, or about 100 to about 150 amino acids or nucleotides in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison are conducted by a BLAST 2.0 algorithm, which is described in Altschul et al. (1990) J Mol. Biol. 215:403-410. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a word length (W) of 11, an expectation (E) or 10, M=5, N=−4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a word length of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915) alignments (B) of 50, expectation (E) of 10, M=5, N=−4, and a comparison of both strands.
  • The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • DETAILED DESCRIPTION
  • Disclosed herein are compositions and methods of treating cancer in a subject. Provided are replicating viral vectors, specifically oncolytic adenoviral vectors, that contain one or more recombinant nucleic acid sequences encoding therapeutic polypeptides.
  • Oncolytic adenovirus (AV) expressing a transgene display highly oncolytic and immunogenic properties. Therefore, the recombinant adenoviral vectors provided herein have the potential to have broad activity against a primary tumor infected with adenovirus but also against tumors in the metastatic disease setting. In some embodiments, the metastatic tumors need not be directly injected with virus nor does the virus injected into the primary site travel to the metastatic setting (e.g. through the blood stream). However, due to expression of an immune stimulatory transgene from the adenovirus (for example, including but not limited to an E1b 19K deleted region in an exemplary vector such as TAV-255 (e.g., TAV-255 Δ19)), the immune system will be primed to fight cancer systemically in the entire body of the cancer patient as long as the metastatic tumor cells express the same tumor antigens as the primary tumor cells. Since metastases are derived from the primary tumors and genetically very similar to the primary tumor cells, metastatic tumor growth will be inhibited, in some embodiments, to the same extent as the primary tumor.
  • Provided are adenoviruses as described herein. In another aspect, a cell transformed with any one of the recombinant adenoviruses described herein is provided.
  • In another aspect, a method is provided of selectively expressing a peptide in a target cell comprises contacting the target cell with any one of the recombinant adenoviruses described herein. In one aspect, the recombinant adenovirus comprises a E1a regulatory sequence deletion mutant operably linked to a nucleotide sequence encoding a peptide, e.g., a peptide associated with viral replication or with cancer.
  • Also provided herein are methods of adenoviral therapy that utilize the oncolytic adenoviruses of the instant invention as adenoviral vectors that express one, two, or more recombinant immunomodulatory genes. The oncolytic adenovirus contains a heterologous gene that encodes a therapeutic protein, incorporated within the viral genome, such that the heterologous gene is expressed within an infected cell. A therapeutic protein, as used herein, refers to a protein that provides one or more therapeutic benefit when expressed in a given cell. In particular, the therapeutic benefit includes recruitment of the host immune system to the tumor.
  • Modified Regulatory Regions
  • E1a is the first protein produced by an adenovirus upon infection of a cell, activating other adenoviral promoters and facilitating infected cells to enter cell division. Rendering expression of this protein under tumor-selective control is an effective means of limiting expression of viral proteins and oncolysis to tumor cells.
  • Normal cells require mitogenic growth signals (GS) before they can move from a quiescent state into an active proliferative state. Tumor cells are able to generate many of their own growth signals or mimic normal growth signals, and transcription factors such as E2F1 and Pea3 are commonly overexpressed in tumor cells at levels that can cooperate in forming conformational structures optimal for driving E1a transcription during adenovirus infection and replication. (Hanahan D, Weinberg R A. The Hallmarks of Cancer. Cell 2000; 100: 57-70; de Launoit Y, Chotteau-Lelievre A, Beaudoin C, Coutte L, Netzer S, Brenner C et al., The PEA3 Group of ETS-Related Transcription Factors: Role in Breast Cancer Metastasis, Adv Exp Med Biol 2000; 480: 107-116; Bruder J T, Hearing P: Cooperative Binding of EF-1A to the E1A Enhancer Region Mediates Synergistic Effects on E1A Transcription During Adenovirus Infection. J Virol 1991; 65:5084-5087).
  • Small deletions selectively targeting the binding sites for E2F1 and Pea3 sites in the E1a enhancer region are an alternative and less disruptive method than complete replacement of the E1a enhancer region with a transcriptionally restricted promoter element. Cooperative binding and transcriptionally optimized conformation of the E1a enhancer region could still take place due to the over-abundance of transcription factors found in tumor cells, while in normal, non-dividing cells, the disruption of binding sites would further inhibit the ability to form optimized conformations in the limiting level of mitogenic growth signals.
  • Thus, disclosed herein are methods of engineering adenoviral vectors via the Pea3 binding sites. The five Pea3 transcription factor binding sites, also known as E1AF (or originally as EF-1A-enhancer binding factor to the E1a core motif) have differential effects on the production of E1a mRNA levels as demonstrated by specific deletions of individual and paired sites. The murine Pea3 sites are described herein as Pea3 sites I, II, III, IV, and V. The main binding sites for Pea3 are sites I and III, while sites II, IV, and V are slightly degenerate versions. Pea3 binds cooperatively between sites II and III, IV and V, and II and I, and this cooperative binding activates E1a transcription. (see, e.g., Hearing (J. Virol 65, 1991, Mol Cell Biol 9, 1989, and Nuc Acids Res 20, 1992). Pea3 itself is a dimer of both α
  • and β subunits, where the α subunit makes the primary DNA contact and the β subunit forms a heteromultimeric complex with the β subunit both in solution or on a dimeric binding site. During normal infection conditions, binding at sites I and III, followed by the cooperative binding at site II would cause conformational changes that serve to bring this protein/DNA complex closer to the activation transcription factor (ATF) binding site and TATA box for full activation of E1a mRNA expression. The two lower affinity sites, Pea3 IV and Pea3 5, do not appear to contribute much to activation under these normal circumstances, as they may be too far away or unoccupied. Deletion of site II causes the greatest reduction in transcriptional activity, pointing to the importance of the cooperative binding effect it has for sites III and I in activating transcription, presumably through a conformational change. Deletion of either site III or I had much less of a reduction, since presumably you could still have cooperative binding between site II and the remaining site III or I. Deletion of both sites I and III (but not II) reduces transcription to the levels seen with Pea3 site II deletion alone, and the combination of deleting both sites III and II, or sites II and I also results in similar (but not greater) levels of reduction.
  • Deletion of the region encompassing sites III and II removes this cooperative binding capacity derived from site II, but it also moves sites IV and V closer to site I. Although lower affinity, sites IV and V do show cooperative binding, and by moving them closer to site I, binding at these three sites may be able to mimic the conformation change normally occurring with binding of sites leading to transcriptional activation. Because sites IV and V are lower affinity sites, there may not be enough of this transcription factor around in normal cells to get full binding on the deleted construct for activation, but there are several publications reporting increased levels of E1AF/Pea3 with tumor progression and invasiveness (i.e., Horiuchi S. et. al., J Pathol 2003 August; 200(5): 568-76), indicating that tumor cells may have enough E1AF to bind the lower affinity binding sites IV and V, along with site I, and potentially lead to a conformational change needed to activate transcription of E1a in tumor but not normal cells with this deleted virus.
  • The present invention provides replicating adenoviruses. In some embodiments, the replicating vectors of the instant invention contain recombinant (e.g., exogenous) transgene(s) expressing immunomodulatory polypeptides that are controlled by endogenous adenovirus early promoters, thereby driving meaningfully higher expression levels than can be generally achieved in replication deficient viruses. In addition to the enhanced anti-tumor efficacy resulting from tumor-specific oncolysis from the replicating adenovirus, the higher expression levels from the recombinant transgenes in a replicating virus results in enhanced immunomodulatory effect(s) over the lower expression levels in replication deficient viruses.
  • In one aspect, a recombinant virus comprises a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is deleted. In one aspect, a sufficient number of nucleotides in the range of −305 to -141 are retained to maintain functional activity of the Ad packaging signal function and (near) optimal transcription of the E1a protein in tumor but not growth arrested normal cells.
  • Additional deletions or modifications of individual or combinations of Pea3 V, Pea3IV, Pea3III, Pea3II and/or E2F1 binding sites between base pairs-394 and -218 designed to inhibit binding of Pea3 and/or E2F1 to these sites and take part in cooperative binding induced conformational changes in the E1a enhancer region are also encompassed by this description.
  • In one aspect, at least one of Pea3 II, Pea3 III, Pea3 IV, and Pea3 V, or a functional portion thereof, is deleted or modified (e.g., at least one nucleotide of the sequence is changed or an additional nucleotide is inserted into the sequence). In another aspect, at least one of Pea3 II and Pea3 III, or a functional portion thereof, is deleted or modified. In one aspect, Pea3 II or a functional portion thereof, and Pea3 III or a functional portion thereof, is deleted or modified. In another aspect, at least one of Pea3 IV and Pea3 V, or a functional portion thereof, is deleted or modified. In another aspect, Pea3 I, or a functional portion thereof, is retained. By “retained” is meant that the element is present in the recombinant adenoviral vector, preferably at the same location as a reference adenoviral vector. In one aspect, at least one E2F1 binding site, or a functional portion thereof, is retained.
  • In one aspect, the vector is d1309-6, TAV-255, d155, d1200, d1230, or d1200+230. In another aspect, the vector is TAV-255. In another aspect, the E1a deletions in d1309-6, TAV-255, d155, d1200, d1230, d1200+230, or other E1a modifications affecting Pea3 and/or E2F1 binding sites between -394 to -218, are paired with a non-d1309 based E3 deletion such as the E3 deletion found in pBHG10 (Microbix, Ad5 base pairs (bp) 28133-30818), d1327 (Ad5 bp 28593-30470) or a similar size E3 deletion such that >3 kb of exogenous DNA can be successfully packaged and expressed from a recombinant adenovirus with the E1a deletions listed above, in combination with a deletion between the start site of the E1b 19K protein and the start site of the E1b 55K protein of approximately 202 base pairs. In one aspect, the vector is a d1309 vector having one or more mutations in reference to the wild type sequence of Ad5 (see, e.g., Chroboczek et al., Virology (1992) January; 186(1):280-5, herein incorporated by reference), including a disruption in the coding sequences for one or more of the 10.4K, 14.5K, and 14.7K proteins in the E3 region.
  • In one aspect, a recombinant virus selectively expresses at least one E1a isoform, e.g., E1a-12S or E1a-13S. In one aspect, the sequence encoding the E1a isoform is operably linked to a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is deleted or modified.
  • In one aspect, a recombinant virus comprises a DNA sequence, e.g., a transgene, inserted into an E1b-19K insertion site. In one aspect, the insertion site is located between the start site of E1b-19K and the start site of E1b 55K. In another aspect, the insertion site comprises a deletion of 202 base pairs following the start site of E1b-19K. A transgene (also, “insert”) may be a full natural sequence of the gene of interest or a fragment thereof. It may be modified too include a Kozak sequence, stop codon, or other regulatory elements. A transgene may include one or more endonuclease restriction sites.
  • In another aspect, expression the transgene is operably linked to a modified E1a regulatory sequence, wherein at least one Pea3 binding site, or a functional portion thereof, is modified or deleted. The transgene may be located in the E1, E2, E3 and/or E4 regions of the adenovirus but its expression is controlled by E1a mediated activation of the endogenous upstream adenovirus promoter, generating high levels of transgene expression only during E1a mediated viral replication.
  • In specific embodiments, the transgene is located in the E3 region. Exogenous transgenes can be inserted into one or more deleted regions of the adenovirus E3 region, generally such transgenes are inserted into the adenoviral vector as to have their expression controlled by the endogenous E3 promoter (Luo J et al, Clin Cancer Res 2008: 14 2450-2457). This results in high levels of transgene expression that are specifically expressed during periods of viral replication, as the E3 promoter only becomes transcriptionally active during these times. Using a modified adenovirus backbone, such as the E1a enhancer modification described herein, to limit viral replication to infected tumor cells, expression from the introduced transgene is also limited to infected tumor cells and, generally, not normal cells. In certain embodiments, the entire E3 region, or a substantial portion of the E3 region, is deleted.
  • In specific embodiments, the transgene is located in the E4 region. In a similar fashion to inserting exogenous transgenes into deleted regions in the E3 or E1 regions of adenovirus, there are regions in the E4 region of adenovirus that can be deleted without significant effect on viral growth characteristics (Gao, G P et al, J. Virol 1996: 70; 8934-8943). Generally, at least ORF3 and/or ORF6 are retained in the adenoviral vector in which the remainder of the E4 region. It should be possible to insert a foreign transgene into one of these deletions in the E4 region and drive expression of this gene with the endogenous E4 promoter of adenovirus, restricting high level expression of this gene to conditions where viral replication is expected to occur.
  • Tumor-Directed Recombinant Immunomodulatory Polypeptide Production.
  • The adenoviruses described herein can be engineered to express an immunomodulatory agent or immunomodulatory polypeptide, e.g., a polypeptide agonist of a co-stimulatory signal of an immune cell. In some embodiments, the polypeptide agonist is an agonist of a T effector cell and/or the polypeptide agonist functions as a polypeptide antagonist of an inhibitory signal of an immune cell such as a regulatory T cell. As provided herein, an “immunomodulatory protein” or an “immunomodulatory polypeptide” includes any polypeptide or set of polypeptides capable of modulating (e.g., stimulating) the anti-tumor immune response induced by the adenovirus. Generally, an “immunomodulatory polypeptide” includes a desired immunostimulatory activity. An immunomodulatory polypeptide can include a set of polypeptides, linked or unlinked, that can form a multimer (e.g., a dimer) capable of modulating the anti-tumor immune response induced by adenovirus, e.g., IL-12 dimer formed from p40 and p35, with or without a linker. Immunomodulatory polypeptides can be full length proteins as occur in nature or can be fusions, variants, or fragments thereof that retain at least about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10% of the immunomodulatory activity of the full-length protein.
  • As used herein, the term “agonist(s)” refers to a molecule(s) that binds to another molecule and induces an increased biological reaction. In a specific embodiment, an agonist is a molecule that binds to a receptor on a cell and triggers or stimulates one or more signal transduction pathways. For example, an agonist can include an antibody or ligand that binds to a receptor on a cell and induces one or more signal transduction pathways. In other embodiments, the agonist facilitates the interaction of the native ligand with the native receptor. As used herein, the term “antagonist(s)” refers to a molecule(s) that inhibits the action of another molecule, optionally without provoking an independent biological response itself. In a specific embodiment, an antagonist is a molecule that binds to a receptor on a cell and blocks or dampens the biological activity of an agonist. For example, an antagonist can include an antibody or ligand that binds to a receptor on a cell and blocks or dampens binding of the native ligand to the cell, optionally without inducing one or more signal transduction pathways. Another example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand. A further example of an antagonist includes an antibody or soluble receptor that competes with the native receptor on cells for binding to the native ligand or blocks receptor internalization, and thus, blocks or dampens one or more signal transduction pathways induced when the native receptor binds to the native ligand.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is a costimulatory ligand, e.g., GITR ligand (GITRL), OX40 ligand (OX40L), or CD40 ligand (CD40L). Expression of one or more costimulatory ligands in the tumor microenvironment and the specific binding to the cognate receptor results in an increase in activity of a tumor-infiltrating lymphocyte (TIL), such activity including TIL proliferation and cytokine release, thereby increasing the anti-tumor activity of the pharmaceutical composition.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is a pro-inflammatory cytokine, e.g., GMCSF, IL-7, IL-12, or an IL-15 hybrid (e.g., a hybrid of IL-15 and IL-15 receptor alpha). Expression of one or more pro-inflammatory cytokine in the tumor microenvironment results in a tumor-localized increase in the inflammatory milieu, thereby increasing the anti-tumor activity of the pharmaceutical composition, and also increasing safety of the composition by decreasing or eliminating undesired effects of systemic administration of a pro-inflammatory cytokine.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is an inhibitor (e.g., a “receptor trap” or a “trap”) of an inhibitory cytokine, e.g., IL-10 or IL-27. In other embodiments, the immunomodulatory agent/inhibitor of an inhibitory cytokine is an antibody against, e.g., TGFB or IL-10R. Such inhibitory cytokines decrease T effector cell function. Expression of one or more inhibitory cytokine receptor traps in the tumor microenvironment results in a tumor-localized binding to and neutralization of the inhibitory cytokine, thereby reducing or preventing its inhibitory activity and increasing the anti-tumor activity of the pharmaceutical composition, and also increasing safety of the composition by decreasing or eliminating undesired effects of systemic administration of a blockage of an inhibitory cytokine.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is an initiator of a localized immune response, e.g., a protein (e.g., secreted flagellin) that activates a toll-like receptor ligand such as TLR-5. Expression of one or more immune response initiators in the tumor microenvironment results in a tumor-localized immune response (e.g., infiltration of TILs and antigen presenting cells (APCs) and increasing the anti-tumor activity of the pharmaceutical composition.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is an inhibitor (e.g., an antibody antagonist) of a co-inhibitory checkpoint molecule, e.g., CTLA4. Such inhibitory checkpoint molecules decrease T effector cell function. Expression of one or more inhibitory checkpoint molecules at the surface of an activated T cell (e.g., an activated T effector cell) attenuates the functional activity of the T cell. Thus, expression of an antagonist of the co-inhibitory checkpoint molecule in the tumor microenvironment results in blocking the co-inhibitory activity and increasing the anti-tumor activity of the pharmaceutical composition, and also increasing safety of the composition by decreasing or eliminating undesired effects of systemic administration of a blockage of a co-inhibitory checkpoint molecule.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is a cluster of differentiation (CD) molecule or a ligand of a cluster of differentiation (CD) molecule such as CD27, e.g., CD70. Expression of a CD27 ligand in the tumor microenvironment results in a tumor-localized NK-mediated tumor clearance and promotes the adaptive immune response against the tumor, thereby increasing the anti-tumor activity of the pharmaceutical composition. Other suitable CD molecules include CD1d, CD2, CD3, CD4, CD5, CD6, CD7, CD8a, CD8b, CD9, CD21, CD25, CD37, CD40, CD49b, CD53, CD57, CD69, CD80, CD81, CD82, CD86, CD99, CD103, CD134, CD152, CD154, CD165, CD244, CD267, CD272, CD273, CD274, CD278, CD305, CD314, CD357, and CD360, or the ligands thereof, or any modulator (e.g., a stimulator or an inhibitor) thereof.
  • In specific embodiments, the immunomodulatory polypeptide expressed by the adenovirus is selected from a polypeptide provided in Table 1.
  • TABLE 1
    Exemplary Immunomodulatory polypeptides
    SEQ ID
    NO: Immunomodulatory Polypeptide Accession Number(s)
    1 GITR-Ligand (GITR-L; TNFSF18) NP_005083
    2 GITR-Ligand (GITR-L; TNFSF18) NM_183391
    3 CD28 ligand or agonist (CD80) EAW79565
    4 TNFα CAA78745
    5 Non-cleavable TNFα NM_013693
    6 GM-CSF NM_009969
    7 ICOS ligand or agonist NP_056074
    8 4-1BB ligand or agonist AAA53134 (human)
    9 OX40 ligand or agonist CAE11757
    10 OX40 ligand NM_009452
    11 CD40 ligand or agonist NP_000065
    12 CD40 ligand NM_011616
    13 CD27 ligand or agonist AAA36175
    14 CD70 ligand or agonist NM_011617
    15 Interleukin-2 (IL-2) AAB46883
    16 Interleukin-7 (IL-7) AAH47698
    17 Interleukin-7 (IL-7) NM_00837
    18 Interleukin-12 (IL-12) alpha subunit AAD16432
    19 Interleukin-12 (IL-12) beta subunit NP_005526
    20 Interleukin-12 fusion polypeptide N/A
    21 IL-15 CAA62616
    22 IL-15 hybrid N/A
    23 IL-10R TRAP (IL-10 antagonist) NP_001549
    [alpha]
    24 IL-10R Trap N/A
    25 IL-27R TRAP (IL-27 antagonist) NP_004834
    26 IL-13 AAH96140
    27 IL-17 AAC50341
    28 IL-33 Isoform a NP_001300974
    29 IL-33 Isoform a NP_001300973
    30 IL-33 Isoform b NP_001186569
    31 IL-33 Isoform c NP_001186570
    32 IL-33 Isoform d NP_001300975;
    NP_001300976
    33 IL-33 Isoform e NP_001300977
    34 IFN-gamma AAB59534
    35 secreted flagellin N/A
    36 anti-CTLA4 (CTLA-4 antagonist N/A
    antibody)
    38 Interleukin-2 (IL-2) (murine) NM_008366.3
    39 Xc11 (murine) GenBank: BC062249.1
    42 4-1BBL (TNFSF9) (murine) GenBank: BC138767.1
    55 IL-15Rα N/A
    58 hαCTLA4 N/a com1-19
    Anti-PD-1 (PD-1 antagonist
    antibody)
    Anti-PD-L1 (PD-L1 antagonist
    antibody)
    Anti-TIGIT (TIGIT antagonist
    antibody)
    Anti-TGFβ (TGFβ antagonist
    antibody)
    Anti-IL6R (IL-6 receptor antibody)
  • Additional detail about the above-discussed agonists that can be expressed from an adenovirus vector as described herein is provided below:
  • GITR Ligand.
  • In one embodiment, the heterologous gene is a GITR ligand family gene, such as TNFSF18 (also known as GITRL) (See, e.g., Tone, M., Tone, Y., Adams, E., Yates, S. F., Frewin, M. R., Cobbold, S. P., & Waldmann, H. (2003). Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proceedings of the National Academy of Sciences of the United States of America, 100(25), 15059-15064. doi:10.1073/pnas.2334901100). The GITR/GITRL signaling pathway is associated with activation of immune cells Nocentini, G., Ronchetti, S., Petrillo, M. G., & Riccardi, C. (2012). Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. British Journal of Pharmacology, 165(7), 2089-99. doi:10.1111/j.1476-5381.2011.01753.x). Specifically, the GITRL protein has an immunomodulatory activity including inhibiting the suppressive activity of T regulatory cells and activation of T effector cells. The intratumoral localization of effective amounts of GITRL protein results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, the costimulatory activity of GITRL protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation
  • IL-10 TRAP.
  • In one embodiment, the heterologous gene is an engineered IL-10 trap, such as IL-10 receptor fused to a human immunoglobulin Fc domain. Examples include IL10RA-Fc fusion protein or IL10RA-IL10RB-Fc fusion protein. (See, e.g., Economides, A. N., Carpenter, L. R., Rudge, J. S., Wong, V., Koehler-Stec, E. M., Hartnett, C., . . . Stahl, N. (2003). Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nature Medicine, 9(1), 47-52. doi:10.1038/nm811). The IL-10 family is associated with inhibition of inflammatory response in immune cells through inhibition of the expression of proinflammatory cytokines and co-stimulatory molecules. Expression of IL-10 by T regulatory cells suppresses the activity of T effector cells (Mosser D M, Zhang X. Immunol Rev. 2008 December; 226:205-18. Interleukin-10: new perspectives on an old cytokine). Specifically, the IL-10 trap protein has an immunomodulatory activity by inhibiting the anti-inflammatory activity of IL-10. The intratumoral localization of effective amounts of IL-10 trap protein results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, IL-10 trap protein inhibition of IL-10's anti-inflammatory activity has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation. In one embodiment, the IL-10 Receptor Trap includes all or a portion of the extracellular domains of IL-10Ra and IL-10Ra.
  • ANTI-CTLA4.
  • In one embodiment, the heterologous gene is an antibody (or domain or fragment thereof) that inhibits the function of CTLA4 (See, e.g., Leach D R, Krummel M F, Allison J P. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996 Mar. 22; 271(5256):1734-6.) The CTLA4 family is associated with inhibition of T cells through the interaction with ligands CD80 and CD86 (Krummel M F, Allison J P (1995). “CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation”. J. Exp. Med. 182 (2): 459-65.) Specifically, the anti-CTLA4 antibody has an immunomodulatory activity including blocking the inhibitory function of CTLA4 resulting in more efficient activation of T effector cells. The intratumoral localization of effective amounts of anti-CTLA4 antibody results in stimulation of the immune system and inhibition of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, anti-CTLA4 inhibition of CTLA4's T cell inhibitory activity has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • IL-12:
  • In one embodiment, the heterologous gene is a member of the Interleukin cytokine family such as IL-12. The IL-12 cytokine family is associated with induction of IFNγ and mediating T-cell dependent immunity. Specifically, IL-12 is an immunostimulatory cytokine with strong antiangiogenic effects. IL-12 has immunomodulatory activity including cell proliferation, lymphocyte differentiation and NK cell activation. The intratumoral localization of effective amounts of IL-12 results in the differentiation, proliferation, and maintenance of T helper 1 (Th1) responses that lead to IFNγ and IL-2 production that in turn, promote T cell responses and macrophage activation. The local expression of effective amounts of IL-12 from intratumoral injections may provide a safety benefit over systemic administration and side effects associated with high IL-12 serum levels. Moreover, the immunostimulatory activity and induction of cytotoxicity mediated by natural killer cells and T cells by IL-12 may have a synergistic effect with the tumor-directed cell-lytic and immune stimulating activity of our adenovirus providing a more effective viral-based therapeutic treatment of human subjects suffering from cancer. In one embodiment, the IL-12 polypeptide is a fusion of IL-12 subunits p35 and p40, linked by a 45 bp linker, including IL-12 β:p40 (NM_001303244) and IL-12 Alpha:p35 (NM_008351.1).
  • GM-CSF:
  • In one embodiment, the heterologous gene is a cytokine such as Granulocyte-macrophage colony-stimulating factor (GM-CSF), also known as colony stimulating factor 2 (CSF2). Cytokines are secreted proteins or peptides that mediate and regulate immunity and inflammation. Specifically, GM-CSF has an immunomodulatory activity of functioning as an immune adjuvant and facilitates development of the immune system, acting as a growth factor for DCs and APCs. The localized secretion of effective amounts of GM-CSF results in an increase in dendritic cell (DC) maturation and function as well as macrophage activity, recruiting immune cells to the inflammatory site of tumor treatment, resulting in more effective therapeutic treatment of human subjects suffering from cancer. Moreover, GM-CSF has been demonstrated to be capable of induced long-lasting, specific anti-tumor immunity when combined with cancer vaccines, potentially providing a synergistic effect with the tumor-directed cell-lytic activity of our adenovirus. In a recent phase III clinical trial, an oncolytic herpes simplex virus armed with GM-CSF (T-VEC) showed durable response rates in advanced melanoma patients compared with GM-CSF protein alone.
  • Secreted Flagellin:
  • In one embodiment, the heterologous gene comes from a gram-negative bacterium in the Salmonellae family, such as the gene encoding flagellin. Bacterial proteins, including flagellin, are associated with the activation of the innate immune response, leading to production of proinflammatory cytokines and the up-regulation of costimulatory molecules. Specifically, flagellin is a TLR5 agonist, and binding of secreted flagellin to TLR5 stimulates production of TNFα, and induces infiltration of APC's and TIL's to the local tumor environment. By acting as a strong adjuvant, flagellin is able to prime the immune system to elicit strong adaptive immune responses, resulting in enhanced and broadened immune response a more effective viral-based therapeutic treatment of human subjects suffering from cancer. In some embodiments, secreted Flagellin contains a murine IL-2 signal sequence (See NM_008366) and Salmonella Flagellin, GenBank: D13689.
  • TNFA AND NON-CLEAVABLE TNFA.
  • In one embodiment, the heterologous gene is an engineered TNFα ligand family gene, such as a non-cleavable (membrane-bound, transmembrane) form of TNFα (See, e.g., Li Q, Li L, Shi W, Jiang X, Xu Y, Gong F, Zhou M, Edwards C K 3rd, Li Z., Mechanism of action differences in the antitumor effects of transmembrane and secretory tumor necrosis factor-alpha in vitro and in vivo. Cancer Immunol Immunother. 2006: 55, 1470-9.). TNFα belongs to a family of pro-inflammatory cytokines (Calcinotto A, Grioni M, Jachetti E, Curnis F, Mondino A, Parmiani G, Corti A, Bellone M. Targeting TNF-α to neoangiogenic vessels enhances lymphocyte infiltration in tumors and increases the therapeutic potential of immunotherapy. J Immunol. 2012; 188: 2687-94.). Specifically, expression of TNFα in the tumor microenvironment is expected to increase the inflammatory milieu resulting in increased anti-tumor immune responses. Use of a non-cleavable TNFα results in a tethered form of TNFα which remains membrane-bound. The intratumoral localization of effective amounts of TNFα protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. In addition, local expression may provide a safety benefit over systemic administration of TNFα. Moreover, the immunomodulatory activity of the membrane-bound TNFα protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • OX40L.
  • In one embodiment, the heterologous gene is an OX40L family gene, such as TNFSF4 (also called OX40L, CD252) (See, e.g., Dannull J, Nair S, Su Z, Boczkowski D, DeBeck C, Yang B, Gilboa E, Vieweg J. Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA encoding OX40 ligand. Blood. 2005, 105: 3206-13.). The TNFSF is associated with activation of immune cells (Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev. 2009; 229:173-91.). Specifically, OX40L is a co-stimulatory ligand for TNFRSF4 (OX40, CD134) resulting in activation of T cells. Expression of OX40L in the tumor microenvironment and binding to its cognate receptor (OX40) is expected to increase the activity (proliferation, cytokine release) of tumor infiltrating lymphocytes (TILs) resulting in antitumor activity. The intratumoral localization of effective amounts of OX40L protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Local expression may provide a safety benefit over systemic administration of OX40L. Moreover, the immunomodulatory activity of the OX40L protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation IL-7.
  • In one embodiment, the heterologous gene is an IL-7 cytokine family gene, such as IL-7 (See, e.g., Gao J, Zhao L, Wan Y Y, Zhu B., Mechanism of Action of IL-7 and Its Potential Applications and Limitations in Cancer Immunotherapy. Int J Mol Sci. 2015, 16: 10267-80.). IL-7 belongs to a family of pro-inflammatory cytokines (Geiselhart L A, Humphries C A, Gregorio TA, Mou S, Subleski J, Komschlies KL. IL-7 administration alters the CD4:CD8 ratio, increases T cell numbers, and increases T cell function in the absence of activation. J Immunol. 2001; 166: 3019-27.). Specifically, expression of IL-7 in the tumor microenvironment is expected to increase the inflammatory milieu resulting in increased anti-tumor immune responses. The intratumoral localization of effective amounts of IL-7 protein results in stimulation of the immune system and inhibition of growth of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, the immunomodulatory activity of the IL-7 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting from activation and/or recruitment of the immune system to the tumor and enhanced antigen presentation.
  • CD40L
  • In one embodiment, the heterologous gene is a member of the TNF superfamily, such as CD40L (also known as CD40LG or CD154) (See, e.g., Hassan G S, et al., 2015. “Role of CD154 in cancer pathogenesis and immunotherapy.” Cancer Treat Rev 4 1(5):431-40). The CD40L is the ligand for CD40 expressed on antigen presenting cells. Specifically, the CD40L protein has a costimulatory activity important for activation of T cell dependent immune responses (Sotomayor E M, et al., 1999. “Conversion of tumor-directed CD4+ T-cell tolerance to T-cell priming through in vivo ligation of CD40.” Nat Med. 5:780-787; French R R, et al., 1999. “CD40 antibody evokes a cytotoxic T-cell response that eradicates lymphoma and bypasses T-cell help. Nat Med. 5:548-553). The intratumoral localization of effective amounts of CD40 protein results in activation of the immune system and lysis of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, the costimulatory immunomodulatory activity of CD40 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting in a local and systemic immune response against the tumor.
  • IL-15 AND IL-15 HYBRID
  • In one embodiment, the heterologous gene is a cytokine family gene, such as Interleukin 15 (IL-15) (See, e.g., Di Sabatino A, et. al., 2011 “Role of IL-15 in immune-mediated and infectious diseases”. Cytokine Growth Factor Rev. 22 (1): 19-33; Steel J C, et al., 2012, “Interleukin-15 biology and its therapeutic implications in cancer”. Trends Pharmacol. Sci. 33 (1): 35-41). IL-15 is a cytokine that regulates T cell and NK cell activation and proliferation. (Waldmann T A, et al., (1999). “The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK cell differentiation and host response to intracellular pathogens”. Annu. Rev. Immunol. 17: 19-49). Specifically, the IL-15 protein has an immunomodulatory activity by providing survival signals to maintain memory T cells in the absence of antigen. IL-15 has also been shown to enhance the anti-tumor immunity of CD8+ T cells (See, Klebanoff C A, et al., “IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T Cells” Proc. Natl. Acad. Sci. U.S.A. 101 (7): 1969-74; and Teague R M, et al., “Interleukin-15 rescues tolerant CD8+ T cells for use in adoptive immunotherapy of established tumors” Nat. Med. 12 (3): 335-41). Expression of an IL-15 hybrid molecule (IL-15 linked to the IL-15 Receptor alpha, see Tosic et al., PLos ONE 9(10): e109801 (2014)) leads to stabilization and increased bioactivity (Bergamaschi et al., 2008. “Intracellular Interaction of Interleukin-15 with Its Receptor a during Production Leads to Mutual Stabilization and Increased Bioactivity”, JBC, 283(7):4189-99; Bergamaschi et al., 2013. “Circulating IL-15 exists as heterodimeric complex with soluble IL-15Rα in human and mouse serum”, Blood 120(1):e1). The intratumoral localization of effective amounts of IL-15 protein results in activation of the immune system and lysis of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, the survival signals provided by IL-15 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting in a local and systemic immune response against the tumor. In some embodiments, an IL-15 hybrid includes IL-15, P2A, and IL-15Rα, IL-15 (NM_008357), 68 bp P2A, and IL-15Rα (GenBank: BC132233.1).
  • CD70
  • In one embodiment, the heterologous gene is a member of the TNF superfamily, such as CD70 (also known as TNFSF7 or CD27L). (See, e.g., Denoeud J and Moser M., 2011. “Role of CD27/CD70 pathway of activation in immunity and tolerance.” J Leukoc Biol. 89(2):195-203). CD70 is expressed on activated T and B cells, as well as mature dendritic cells, and acts as a ligand for CD27. CD70 plays a costimulatory role in promoting T cell expansion and differentiation (Keller A M, et al., 2008. “Expression of costimulatory ligand CD70 on steady-state dendritic cells breaks CD8+ T cell tolerance and permits effective immunity,” Immunity 29(6):934-46; Bonehill A, et al., 2008. “Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA,” Mol Ther. 16(6):1170-80). In addition, CD70 expression in the tumor microenvironment will increase NK-mediated tumor clearance and promote an adaptive immune response against the tumor (Kelly J M, et al., 2002. “Induction of tumor-directed T cell memory by NK cell-mediated tumor rejection,” Nat Immunol. 3(1):83-90). The intratumoral localization of effective amounts of CD70 protein results in activation of the immune system and lysis of the tumor, resulting in more effective viral-based therapeutic treatment of human subjects suffering from cancer. Moreover, the immunomodulatory activity of CD70 protein has a synergistic effect with the tumor-directed cell-lytic activity of the adenovirus, resulting in a local and systemic immune response against the tumor.
  • In a specific embodiment, the agonist of a co-stimulatory signal of an immune cell expressed by the adenovirus is an agonist of a co-stimulatory receptor expressed by an immune cell. Specific examples of co-stimulatory receptors that can be expressed by the adenovirus include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB(CD137), CD40, CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA). In a specific embodiment, the agonist of a co-stimulatory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) or ligand that specifically binds to the co-stimulatory receptor. In one embodiment, the antibody is a monoclonal antibody. In another embodiment, the antibody is an sc-Fv. In a specific embodiment, the antibody is a bispecific antibody that binds to two receptors on an immune cell. In one embodiment, the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell. In specific embodiments, the antibody is a human or humanized antibody. In certain embodiments, the ligand or antibody is a chimeric protein.
  • In a specific embodiment, the antagonist of an inhibitory signal of an immune cell is an antagonist of an inhibitory receptor expressed by an immune cell. Specific examples of inhibitory receptors include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD160. In a specific embodiment, the antagonist of an inhibitory receptor expressed by an immune cell is an antibody (or an antigen-binding fragment thereof) that specifically binds to the co-stimulatory receptor.
  • Antibodies include, but are not limited to, monoclonal antibodies, bispecific antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, single domain antibodies, camelid or camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked bispecific Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies), and epitope-binding fragments of any of the above. In particular, antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. In a specific embodiment, an antibody is a human or humanized antibody. In another specific embodiment, an antibody is a monoclonal antibody or scFv. In certain embodiments, an antibody is a human or humanized monoclonal antibody or scFv. In other specific embodiments, the antibody is a bispecific antibody. In certain embodiments, the bispecific antibody specifically binds to a co-stimulatory receptor of an immune cell or an inhibitory receptor of an immune, and a receptor on a cancer cell. In some embodiments, the bispecific antibody specifically binds to two receptors immune cells, e.g., two co-stimulatory receptors on immune cells, two inhibitory receptors on immune cells, or one co-stimulatory receptor on immune cells and one inhibitory receptor on immune cells.
  • The recombinant AVs described herein may be engineered to express any agonist of a co-stimulatory signal and/or any antagonist of an inhibitory signal of an immune cell, such as, e.g., a T-lymphocyte, NK cell or antigen-presenting cell (e.g., a dendritic cell or macrophage). In specific embodiments, the agonist and/or antagonist is an agonist of a human co-stimulatory signal of an immune cell and/or antagonist of a human inhibitory signal of an immune cell. In certain embodiments, the agonist of a co-stimulatory signal is an agonist of a co-stimulatory molecule (e.g., co-stimulatory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages). Specific examples of co-stimulatory molecules include glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB (CD137), CD40, lymphotoxin alpha (LT alpha), LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes), CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA). In specific embodiments, the agonist is an agonist of a human co-stimulatory receptor of an immune cell. In certain embodiments, the agonist of a co-stimulatory receptor is not an agonist of ICOS. In some embodiments, the antagonist is an antagonist of an inhibitory molecule (e.g., inhibitory receptor) found on immune cells, such as, e.g., T-lymphocytes (e.g., CD4+ or CD8+T-lymphocytes), NK cells and/or antigen-presenting cells (e.g., dendritic cells or macrophages). Specific examples of inhibitory molecules include cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD 160, adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD 160. In specific embodiments, the antagonist is an antagonist of a human inhibitory receptor of an immune cell.
  • In a specific embodiment, the agonist of a co-stimulatory receptor is an antibody or antigen-binding fragment thereof that specifically binds to the co-stimulatory receptor. Specific examples of co-stimulatory receptors include GITR, ICOS, OX40, CD27, CD28, 4-1BB, CD40, LT alpha, LIGHT, CD226, CRT AM, DR3, LTBR, TACI, BAFFR, and BCMA. In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an sc-Fv. In a specific embodiment, the antibody is a bispecific antibody that binds to two receptors on an immune cell. In other embodiments, the bispecific antibody binds to a receptor on an immune cell and to another receptor on a cancer cell. In specific embodiments, the antibody is a human or humanized antibody.
  • In another embodiment, the agonist of a co-stimulatory receptor expressed by the adenovirus is a ligand of the co-stimulatory receptor. In certain embodiments, the ligand is fragment of a native ligand. Specific examples of native ligands include ICOSL, B7RP1, CD137L, OX40L, CD70, herpes virus entry mediator (HVEM), CD80, and CD86. The nucleotide sequences encoding native ligands as well as the amino acid sequences of native ligands are known in the art. For example, the nucleotide and amino acid sequences of B7RP1 (otherwise known as ICOSL; GenBank human: NM_015259.4, NP_056074.1 murine: NM_015790.3, NP_056605.1), CD137L(GenBank human: NM_003811.3, NP_003802.1, murine: NM_009404.3, NP_033430.1), OX40L(GenBank human: NM_003326.3, NP_003317.1, murine: NM_009452.2, NP_033478.1), CD70(GenBank human: NM_001252.3, NP_001243.1, murine: NM_011617.2, AAD00274.1), CD80(GenBank human: NM_005191.3, NP_005182.1, murine: NM_009855.2, NP_033985.3), and CD86 (GenBank human: NM_005191.3, CAG46642.1, murine: NM_019388.3, NP_062261.3) can be found in GenBank. In other embodiments, the ligand is a derivative (e.g., a fragment, domain, fusion, or other modification of a full-length polypeptide) a native ligand. In some embodiments, the ligand is a fusion protein comprising at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and a heterologous amino acid sequence. In specific embodiments, the fusion protein comprises at least a portion of the native ligand or a derivative of the native ligand that specifically binds to the co-stimulatory receptor, and the Fc portion of an immunoglobulin or a fragment thereof. An example of a ligand fusion protein is a 4-IBB ligand fused to Fc portion of immunoglobulin (described by Meseck M et al., J Immunother. 2011 34: 175-82).
  • In another embodiment, the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) or a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). Specific examples of native ligands for inhibitory receptors include PDL-1, PDL-2, B7-H3, B7-H4, HVEM, Gal9 and adenosine. Specific examples of inhibitory receptors that bind to a native ligand include CTLA-4, PD-1, BTLA, KIR, LAG3, TIM3, and A2aR.
  • In specific embodiments, the antagonist of an inhibitory receptor expressed by the adenovirus is a soluble receptor that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). In certain embodiments, the soluble receptor is a fragment of a native inhibitory receptor or a fragment of a derivative of a native inhibitory receptor that specifically binds to native ligand {e.g., the extracellular domain of a native inhibitory receptor or a derivative of an inhibitory receptor). In some embodiments, the soluble receptor is a fusion protein comprising at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor (e.g., the extracellular domain of the native inhibitory receptor or a derivative of the native inhibitory receptor), and a heterologous amino acid sequence. In specific embodiments, the fusion protein comprises at least a portion of the native inhibitory receptor or a derivative of the native inhibitory receptor, and the Fc portion of an immunoglobulin or a fragment thereof. An example of a soluble receptor fusion protein is a LAG3-Ig fusion protein (described by Huard B et al, Eur J Immunol (1995) 25:2718-21).
  • In specific embodiments, the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) that specifically binds to the native ligand for the inhibitory receptor and blocks the native ligand from binding to the inhibitory receptor and transducing an inhibitory signal(s). In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an scFv. In particular embodiments, the antibody is a human or humanized antibody. A specific example of an antibody to inhibitory ligand is anti-PD-L1 antibody (Iwai Y, et al. PNAS 2002; 99: 12293-12297).
  • In another embodiment, the antagonist of an inhibitory receptor expressed by the adenovirus is an antibody (or an antigen-binding fragment) or ligand that binds to the inhibitory receptor, but does not transduce an inhibitory signal(s). Specific examples of inhibitory receptors include CTLA-4, PD1, BTLA, TIGIT, KIR, LAG3, TIM3, and A2aR. In certain specific embodiments, the antibody is a monoclonal antibody. In other specific embodiments, the antibody is an scFv. In particular embodiments, the antibody is a human or humanized antibody. A specific example of an antibody to inhibitory receptor is anti-CTLA-4 antibody (Leach D R, et al. Science 1996; 271: 1734-1736). Another example of an antibody to inhibitory receptor is anti-PD-1 antibody (Topalian S L, NEJM 2012; 28:3167-75).
  • In certain embodiments, a chimeric adenovirus described herein is engineered to produce an antagonist of CTLA-4, such as, e.g., ipilimumab or tremelimumab. In certain embodiments, a chimeric adenovirus described herein is engineered to an antagonist of PD1, such as, e.g., MDX-1106 (BMS-936558), MK3475, CT-011, AMP-224, or MDX-1105. In certain embodiments, a chimeric adenovirus described herein is engineered to express an antagonist of LAG3, such as, e.g., IMP321. In certain embodiments, a chimeric adenovirus described herein is engineered to express an antibody (e.g., a monoclonal antibody or an antigen-binding fragment thereof, or scFv) that binds to B7-H3, such as, e.g., MGA271. In specific embodiments, a chimeric adenovirus described herein is engineered to express an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. In specific embodiments, adenovirus described herein is engineered to express anti-CD28 scFv, ICOSL, CD40L, OX40L, CD137L, GITRL, and/or CD70.
  • In certain embodiments, an agonist of a co-stimulatory signal of an immune cell expressed by the adenovirus induces (e.g., selectively) induces one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand. In specific embodiments, an agonist of a co-stimulatory receptor induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%>, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands in the absence of the agonist. In specific embodiments, an agonist of a co-stimulatory receptor: (i) induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not induce, or induces one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to such one or more other ligands in the absence of the agonist.
  • In certain embodiments, an agonist of a co-stimulatory signal of an immune cell activates or enhances (e.g., selectively activates or enhances) one or more of the signal transduction pathways induced by the binding of a co-stimulatory receptor to its ligand. In specific embodiments, an agonist of a co-stimulatory receptor activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%), or 75%) to 100% relative to the one or more signal transduction pathways induced by the binding of co-stimulatory receptor to one or more of its ligands in the absence of the agonist. In specific embodiments, an agonist of a co-stimulatory receptor: (i) an agonist of a co-stimulatory signal activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to the particular ligand in the absence of the agonist; and (ii) does not activate or enhance, or activates or enhances one or more of the signal transduction pathways induced by the binding of the co-stimulatory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of the co-stimulatory receptor to such one or more other ligands in the absence of the agonist.
  • In some embodiments, an antagonist of an inhibitory signal of an immune cell (e.g., selectively) inhibits or reduces one or more of the signal transduction pathways induced by the binding of an inhibitory receptor to its ligand. In specific embodiments, an antagonist of an inhibitory receptor inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to one or more of its ligands in the absence of the antagonist. In specific embodiments, an antagonist of an inhibitory receptor: (i) inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one particular ligand by at least 25%, 30%, 40%, 50%, 60%, 75%, 80%, 85%, 90%, 95%, 98% or 99%, or in the range of between 25% to 50%, 25% to 75%, 50% to 75%, 50% to 95%, 75% to 95%, or 75% to 100% relative to the one or more signal transduction pathways induced by the binding of the inhibitory receptor to the one particular ligand in the absence of the antagonist; and (ii) does not inhibit or reduce, or inhibits or reduces one or more of the signal transduction pathways induced by the binding of the inhibitory receptor to one or more other ligands by less than 20%, 15%, 10%, 5%, or 2%, or in the range of between 2% to 5%, 2% to 10%, 5% to 10%, 5% to 15%, 5% to 20%, 10% to 15%, or 15% to 20% relative to the one or more signal transduction pathways induced by the binding of inhibitory receptor to such one or more other ligands in the absence of the antagonist.
  • In specific embodiments, an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell induces, activates and/or enhances one or more immune activities, functions or responses. The one or more immune activities, functions or responses can be in the form of, e.g., an antibody response (humoral response) or a cellular immune response, e.g., cytokine secretion (e.g., interferon-gamma), helper activity or cellular cytotoxicity. In one embodiment, expression of an activation marker on immune cells (e.g., CD44, Granzyme, or Ki-67), expression of a co-stimulatory receptor on immune cells (e.g., ICOS, CD28, OX40, or CD27), expression of a ligand for a co-stimulatory receptor (e.g., B7HRP1, CD80, CD86, OX40L, or CD70), cytokine secretion, infiltration of immune cells (e.g., T-lymphocytes, B lymphocytes and/or NK cells) to a tumor, antibody production, effector function, T cell activation, T cell differentiation, T cell proliferation, B cell differentiation, B cell proliferation, and/or NK cell proliferation is induced, activated and/or enhanced following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell. In another embodiment, myeloid-derived suppressor cell (MDSC) tumor infiltration and proliferation, Treg tumor infiltration, activation and proliferation, peripheral blood MDSC and Treg counts are inhibited following contact with an agonist of a co-stimulatory signal of an immune cell and/or an antagonist of an inhibitory signal of an immune cell.
  • In certain embodiments, a chimeric adenovirus described herein is engineered to produce two or more immunomodulatory polypeptides. In some embodiments, the chimeric adenovirus produces a first immunomodulatory polypeptide and a second immunomodulatory polypeptide.
  • For example, a first immunomodulatory polypeptide is a costimulatory ligand, a proinflammatory cytokine, an inhibitor of an inhibitory cytokine, an initiator of a localized immune response, an inhibitor of a co-inhibitory checkpoint molecule, or a ligand of a cluster of differentiation (CD) molecule, and the second immunomodulatory polypeptide is a costimulatory ligand, a proinflammatory cytokine, an inhibitor of an inhibitory cytokine, an initiator of a localized immune response, an inhibitor of a co-inhibitory checkpoint molecule, or a ligand of a cluster of differentiation (CD) molecule.
  • For example, a first immunomodulatory polypeptide is a costimulatory ligand, and the second immunomodulatory polypeptide is a costimulatory ligand.
  • For another example, a first immunomodulatory polypeptide is a costimulatory ligand and the second immunomodulatory polypeptide is a pro-inflammatory cytokine.
  • In some embodiments, two or more immunomodulatory polypeptides are expressed from a single transcript. To express two or more proteins from a single transcript determined by a viral or non-viral vector, an internal ribosome entry site (IRES) sequence is commonly used to drive expression of the second, third, fourth coding sequence, etc. When two coding sequences are linked via an IRES, the translational expression level of the second coding sequence is often significantly reduced (Furler et al. 2001. Gene Therapy 8:864-873). In fact, the use of an IRES to control transcription of two or more coding sequences operably linked to the same promoter can result in lower level expression of the second, third, etc. coding sequence relative to the coding sequence adjacent the promoter. In addition, an IRES sequence may be sufficiently long to impact complete packaging of the vector, e.g., the eCMV IRES has a length of 507 base pairs.
  • Internal ribosome entry site (IRES) elements were first discovered in picornavirus mRNAs (Jackson et al. 1990. Trends Biochem. Sci. 15:477-83) and Jackson and Kaminski, RNA (1995) 1:985-1000). Examples of IRES generally employed by those of skill in the art include those referenced in Table I and Appendix A, as well as those described in U.S. Pat. No. 6,692,736. Examples of “IRES” known in the art include, but are not limited to IRES obtainable from picornavirus (Jackson et al., 1990) and IRES obtainable from viral or cellular mRNA sources, such as for example, immunoglobulin heavy-chain binding protein (BiP), the vascular endothelial growth factor (VEGF) (Huez et al. 1998, Mol. Cell. Biol. 18:6178-6190), the fibroblast growth factor 2 (FGF-2), and insulin-like growth factor (IGFII), the translational initiation factor eIF4G and yeast transcription factors TFIID and HAP4, the encephelomyocarditis virus (EMCV) which is commercially available from Novagen (Duke et al. 1992. J. Virol 66:1602-9) and the VEGF IRES (Huez et al. 1998. Mol. Cell. Biol. 18:6178-90). IRES have also been reported in different viruses such as cardiovirus, rhinovirus, aphthovirus, HCV, Friend murine leukemia virus (FrMLV) and Moloney murine leukemia virus (MoMLV). As used herein, “IRES” encompasses functional variations of IRES sequences as long as the variation is able to promote direct internal ribosome entry to the initiation codon of a cistron. An IRES may be mammalian, viral or protozoan. The IRES promotes direct internal ribosome entry to the initiation codon of a downstream cistron, leading to cap-independent translation. Thus, the product of a downstream cistron can be expressed from a bicistronic (or multicistronic) mRNA, without requiring either cleavage of a polyprotein or generation of a monocistronic mRNA. Internal ribosome entry sites are approximately 450 nucleotides in length and are characterized by moderate conservation of primary sequence and strong conservation of secondary structure. The most significant primary sequence feature of the IRES is a pyrimidine-rich site whose start is located approximately 25 nucleotides upstream of the 3′ end of the IRES. See Jackson et al. (1990). Three major classes of picornavirus IRES have been identified and characterized: the cardio- and aphthovirus class (for example, the encephalomyocarditis virus, Jang et al. 1990. Gene Dev 4:1560-1572); the entero- and rhinovirus class (for example, polioviruses, Borman et al. 1994. EMBO J. 13:3149-3157); and the hepatitis A virus (HAV) class, Glass et al. 1993. Virol 193:842-852). For the first two classes, two general principles apply. First, most of the 450-nucleotide sequence of the IRES functions to maintain particular secondary and tertiary structures conducive to ribosome binding and translational initiation. Second, the ribosome entry site is an AUG triplet located at the 3′ end of the IRES, approximately 25 nucleotides downstream of a conserved oligopyrimidine tract. Translation initiation can occur either at the ribosome entry site (cardioviruses) or at the next downstream AUG (entero/rhinovirus class). Initiation occurs at both sites in aphthoviruses. HCV and pestiviruses such as bovine viral diarrhea virus (BVDV) or classical swine fever virus (CSFV) have 341 nt and 370 nt long 5′-UTR respectively. These 5′-UTR fragments form similar RNA secondary structures and can have moderately efficient IRES function (Tsukiyama-Kohara et al. 1992. J. Virol. 66:1476-1483; Frolov et al. 1998. RNA 4:1418-1435). Recent studies showed that both Friend-murine leukemia virus (MLV) 5′-UTR and rat retrotransposon virus-like 30S VL30) sequences contain IRES structure of retroviral origin (Torrent et al. 1996. Hum. Gene Ther 7:603-612). In eukaryotic cells, translation is normally initiated by the ribosome scanning from the capped mRNA 5′ end, under the control of initiation factors. However, several cellular mRNAs have been found to have IRES structure to mediate the cap-independent translation (van der Velde, et al. 1999. Int Biochem Cell Biol. 31:87-106). Examples of IRES elements include, without limitation, immunoglobulin heavy-chain binding protein (BiP) (Macejak et al. 1991. Nature 353:90-94), antennapedia mRNA of Drosophila (Oh et al. 1992. Gene and Dev 6:1643-1653), fibroblast growth factor-2 (FGF-2) (Vagner et al. 1995. Mol. Cell. Biol. 15:35-44), platelet-derived growth factor B (PDGF-B) (Bernstein et al. 1997. J. Biol. Chem. 272:9356-9362), insulin-like growth factor II (Teerink et al. (1995) Biochim. Biophys. Acta 1264:403-408), and the translation initiation factor eIF4G (Gan et al. 1996. J. Biol. Chem. 271:623-626). Recently, vascular endothelial growth factor (VEGF) was also found to have IRES element (Stein et al. 1998. Mol. Cell. Biol. 18:3112-3119; Huez et al. 1998. Mol. Cell. Biol. 18:6178-6190). Further examples of IRES sequences include Picornavirus HAV (Glass et al. 1993. Virology 193:842-852); EMCV (Jang and Wimmer. 1990. Gene Dev. 4:1560-1572); Poliovirus (Borman et al. 1994. EMBO J. 13:3149-3157); HCV (Tsukiyama-Kohara et al. 1992. J. Virol. 66:1476-1483); pestivirus BVDV (Frolov et al. 1998. RNA. 4:1418-1435); Leishmania LRV-1 (Maga et al. 1995. Mol. Cell. Biol. 15:4884-4889); Retroviruses: MoMLV (Torrent et al. 1996. Hum. Gene Ther. 7:603-612). VL30, Harvey murine sarcoma virus, REV (Lopez-Lastra et al. 1997. Hum. Gene Ther. 8:1855-1865). IRES may be prepared using standard recombinant and synthetic methods known in the art. For cloning convenience, restriction sites may be engineered into the ends of the IRES fragments to be used.
  • In some embodiments, two immunomodulatory polypeptides are expressed from separate transcripts, i.e., a first transcript and a second transcript. In some embodiments, the two transcripts are encoded by a DNA insertion at the same location in the adenovirus, e.g., both inserted in E1b, E3, or E4. In some embodiments, the two transcripts are encoded by a DNA insertion at the different locations in the adenovirus, e.g., a first transcript DNA inserted in E1b and a second transcript DNA inserted in E3 or E4, or alternatively, a first transcript DNA inserted in E3 and a second transcript DNA inserted in E4.
  • Another aspect of the invention provides a virus for causing expression in a target cell of a plurality of recombinant immunomodulatory polypeptides or other protein(s) or polypeptides of interest, wherein the vector also includes a promoter operably linked to a first coding sequence for a first recombinant immunomodulatory polypeptide, a self-processing or other cleavage coding sequence, such as a 2A or 2A-like sequence or a protease recognition site, and a second coding sequence for a second recombinant immunomodulatory polypeptide, wherein the self-processing cleavage sequence or protease recognition site coding sequence is inserted between the first and the second coding sequences. In a related embodiment, the viral vector comprises an expression vector as described above wherein the expression vector further comprises an additional proteolytic cleavage site between the first and second recombinant immunomodulatory polypeptides. A preferred additional proteolytic cleavage site is a furin cleavage site with the consensus sequence RXR/K-R.
  • Interaction of Oncolytic Activity and Immunomodulatory Activity.
  • Oncolytic viruses (OVs) were originally conceived as simply a means of targeted destruction of cancer cells. However, it is now thought that the most effective OV therapies will be those that combine tumor cell death with the stimulation of a host anti-tumor immune response. OVs engineered to express particular immunomodulatory cytokines in tumor cells will be able to specifically guide the immune system toward combating cancer cells. Combining the expression of these cytokines with the release of tumor-associated antigens (TAAs, i.e. tumor cell debris) upon viral lysis of cancer cells will allow for the development of cellular or antibody-mediated anti-tumor immune responses (Lichty et al., 2014, Nature Reviews Cancer, 14: 559-567).
  • Pharmaceutical Compositions.
  • A pharmaceutical composition of the invention comprises at least one of the vectors of the invention as described herein. Furthermore, the composition may comprise at least two, three or four different (i.e., expressing different transgenes) vectors of the invention. In addition to the vector, a pharmaceutical composition may also comprise any other vectors, such as other adenoviral vectors, other therapeutically effective agents, any other agents such as pharmaceutically acceptable carriers, buffers, excipients, adjuvants, antiseptics, filling, stabilizing or thickening agents, and/or any components, e.g., such as components found in corresponding viral or pharmaceutical products.
  • The vector(s) described herein can be administered (e.g., in a pharmaceutical composition) to any human or animal, including but not limited to a human or non-human animal having or diagnosed with cancer. According to one embodiment, the cancer is nasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T-cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger-Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter cancer, brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondrosarcoma, Ewing's sarcoma, cancer of unknown primary site, carcinoid, carcinoid of gastrointestinal tract, fibrosarcoma, breast cancer, Paget's disease, cervical cancer, colorectal cancer, rectal cancer, esophagus cancer, gall bladder cancer, head cancer, eye cancer, neck cancer, kidney cancer, Wilms' tumor, liver cancer, Kaposi's sarcoma, prostate cancer, lung cancer, testicular cancer, Hodgkin's disease, non-Hodgkin's lymphoma, oral cancer, skin cancer, mesothelioma, multiple myeloma, ovarian cancer, endocrine pancreatic cancer, glucagonoma, pancreatic cancer, parathyroid cancer, penis cancer, pituitary cancer, soft tissue sarcoma, retinoblastoma, small intestine cancer, stomach cancer, thymus cancer, thyroid cancer, trophoblastic cancer, hydatidiform mole, uterine cancer, endometrial cancer, vagina cancer, vulva cancer, acoustic neuroma, mycosis fungoides, insulinoma, carcinoid syndrome, somatostatinoma, gum cancer, heart cancer, lip cancer, meninges cancer, mouth cancer, nerve cancer, palate cancer, parotid gland cancer, peritoneum cancer, pharynx cancer, pleural cancer, salivary gland cancer, tongue cancer, tonsil cancer. The vector or pharmaceutical composition of the invention may be administered to any eukaryotic subject selected from a group consisting of animals and human beings, in a preferred embodiment of the invention, the subject is a human or a non-human animal. An animal may be selected from a group consisting of pets, domestic animals and production animals.
  • The adenoviral vector(s) of the present invention may be administered to a subject, preferably suspended in a biologically compatible solution or pharmaceutically acceptable delivery vehicle. A suitable vehicle includes sterile saline. Other aqueous and non-aqueous isotonic sterile injection solutions and aqueous and non-aqueous sterile suspensions known to be pharmaceutically acceptable carriers and well known to those of skill in the art may be employed for this purpose.
  • The therapeutic of the present invention may also be administered directly to the tumor in the form of a liquid, gel or suspension introduced by intratumoral injection. A study examining the treatment of mice bearing subcutaneous human pancreatic adenocarcinoma xenografts with recombinant Newcastle disease virus (rNDV) showed that intratumoral injection yielded better tumor regression than intravenous injection. In this study, animals were injected intratumorally every other day for a total of 4 injections, each containing 5×10′ 50% Tissue Culture Infective Dose (TCID50) rNDV in 50 μl (Buijs et al., 2015, Viruses, 6: 2980-2998). Similarly, another study examining the treatment of mice bearing subcutaneous bladder cancer xenografts with modified oncolytic adenovirus found that intratumoral injection significantly suppressed tumor growth. In this study, animals were injected intratumorally twice at a 1-day interval with 5×108 infectious unit (IFU) viruses in 100 μl (Yang et al., 2015, Cell Death and Disease, e1760). Shown to be well-tolerated in Phase 1 trials, Pexa-Vec (JX-594), an oncolytic and immunotherapeutic vaccinia virus has been examined as an intratumorally-administered treatment for patients with advanced hepatocellular carcinoma (HCC). In this study design, patients were to be injected intratumorally 3 times every 2 weeks at one of 2 dose levels: 1×108 plaque forming units (pfu), or 1×109 pfu (Walther and Stein, 2015, Methods in Molecular Biology, 1317: 343-357).
  • A single administration of oncolytic adenoviral vectors of the invention may have therapeutic effects. However, in some embodiments of the invention, oncolytic adenoviral vectors or pharmaceutical compositions are administered several times during the treatment period. Oncolytic adenoviral vectors or pharmaceutical compositions may be administered for example from 1 to 10 times in the first 2 weeks, 4 weeks, monthly or during the treatment period. In one embodiment of the invention, administration is done three to seven times in the first 2 weeks, then at 4 weeks and then monthly, in a specific embodiment of the invention, administration is done four times in the first 2 weeks, then at 4 weeks and then monthly. The length of the treatment period may vary, and for example may last from two to 12 months or more.
  • To improve the efficacy of the present invention, in some embodiments, the therapeutic of the present invention is administered with an adjuvant. Suitable adjuvants include aluminum salts (alum) such as aluminum hydroxide, aluminum phosphate, and aluminum sulfate, Incomplete Freund's Adjuvant (IFA), and monophosphoryl lipid A (MPL). These adjuvants are suitable for human administration, either alone or optionally all combinations thereof (Chang et al., “Adjuvant Activity of Incomplete Freund's Adjuvant,” Adv Drug Deliv Rev 32:173-186 (1998), which is hereby incorporated by reference in its entirety). Other adjuvants include cytokines, such as interleukins (IL-1, IL-2, and IL-12), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), di- and tri-palmitoyl-S-glyceryl cysteine (Pam2Cys and Pam3Cys, respectively), a TLR2 agonist, an anti-granulocyte macrophage colony-stimulating factor (GM-CSF) antibody, RR-XS15, Montanide®, and MALP-2.
  • The adenoviral vector of the present invention can be administered orally, parenterally, for example, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes. They may be administered alone or with suitable pharmaceutical carriers, and can be in solid or liquid (e.g., aqueous) form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • The therapeutic of the present invention may be orally administered, for example, with an inert diluent, or with a suitable edible carrier, or they may be enclosed in hard or soft-shell capsules, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the therapeutic may be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like. Various other materials may be present as coatings or to modify the physical form of the dosage unit.
  • The therapeutic may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • The therapeutic of the present invention may also be administered directly to the airways in the form of an aerosol. For use as aerosols, the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • The therapeutic of this invention may be administered in sufficient amounts to transfect the desired cells and provide sufficient levels of integration and expression of the replicating virus to provide a therapeutic benefit without undue adverse effects or with medically acceptable physiological effects which can be determined by those skilled in the medical arts.
  • Dosages of the therapeutic will depend primarily on factors, such as the condition being treated, the age, weight, and health of the patient, and may thus vary among patients. The dosage will be adjusted to balance the therapeutic benefit against any viral toxicity or side effects.
  • The present invention also relates to a method of enhancing the delivery to and distribution within a tumor mass of therapeutic proteins expressed from viruses. For example, an adenovirus as described herein, optionally in a pharmaceutical composition as described herein, can be injected into a tumor mass such that the virus infects and lyses one or more tumor cell. Combination Therapy.
  • The viral immunotherapy of the invention is effective alone, but combination of multiple adenoviral immunotherapies, or one or more adenoviral immunotherapies with any other therapies, such as traditional therapy, may be more effective than either one alone. For example, each agent of the combination therapy may work independently in the tumor tissue, the adenoviral vectors may sensitize cells to chemotherapy or radiotherapy and/or chemotherapeutic agents may enhance the level of virus replication or effect the receptor status of the target cells. The agents of combination therapy may be administered simultaneously or sequentially.
  • In a preferred embodiment of the invention, the method or use further comprises administration of concurrent radiotherapy to a subject. In another preferred embodiment of the invention, the method or use further comprises administration of concurrent chemotherapy to a subject. As used herein “concurrent” refers to a therapy, which has been administered before, after or simultaneously with the gene therapy of the invention. The period for a concurrent therapy may vary from minutes to several weeks. In some embodiments, the concurrent therapy lasts for some hours.
  • Agents suitable for combination therapy include but are not limited to afatinib, all-trans retinoid acid, azacitidine, azathioprine, bleomycin, carboplatin, capecitabine, cisplatin, chlorambucil, cyclophosphamide, cytarabine, daunorubicin, docetaxel, doxifluridine, doxorubicin, epirubicin, epothilone, etoposide, fluorouracil, 5-fluorouracil, gemcitabine, hydroxyurea, idarubicin, imatinib, mechlorethamine, mercaptopurine, methotrexate, mitoxantrone, oxaliplatin, paclitaxel, pemetrexed, temozolomide, teniposide, tioguanine, valrubicin, vinblastine, vincristine, vindesine and vinorelbine.
  • In some embodiments, the method or use further comprises administration of verapamil or another calcium channel blocker to a subject. “Calcium channel blocker” refers to a class of drugs and natural substances which disrupt the conduction of calcium channels, and if may be selected from a group consisting of verapamil, dihydropyridines, gallopamil, diltiazem, mibefradil, bepridil, fluspirilene and fendiline.
  • In some embodiments, the method or use further comprises administration of autophagy inducing agents to a subject. Autophagy refers to a catabolic process involving the degradation of a cell's own components through the lysosomal machinery. “Autophagy inducing agents” refer to agents capable of inducing autophagy and may be selected from a group consisting of, but not limited to, mTOR inhibitors (e.g., temsirolimus, sirolimus, everolimus, and ridaforolimus), P13K inhibitors (e.g., wortmannin, lithium, tamoxifen, chloroquine, bafilomycin, and temozolomide. In a specific embodiment of the invention, the method further comprises administration of temozolomide to a subject. Temozolomide may be either oral or intravenous temozolomide.
  • In some embodiments, the method or use further comprises administration of chemotherapy or anti-CD20 therapy or other approaches for blocking of neutralizing antibodies. “Anti-CD20 therapy” refers to agents capable of killing CD20 positive cells, and may be selected from a group consisting of rituximab and other anti-CD20 monoclonal antibodies. “Approaches for blocking of neutralizing antibodies” refers to agents capable of inhibiting the generation of anti-viral antibodies that normally result from infection and may be selected from a group consisting of different chemotherapeutics, immunomodulatory substances, corticoids and other drugs. These substances may be selected from a group consisting of, but not limited to, cyclophosphamide, ciclosporin, azathioprine, methylprednisolone, etoposide, CD40L, CTLA4, FK506 (tacrolimus), IL-12, IFN-γ, interleukin 10, anti-CD8, anti-CD4 antibodies, hematopoietic stem cell transplantation (HSCT) and oral adenoviral proteins.
  • In some embodiments, the oncolytic adenoviral vector of the invention induces virion-mediated oncolysis of tumor cells and activates human immune response against tumor cells. In some embodiments, the method or use further comprises administration of substances capable to downregulating regulatory T-cells in a subject in an amount to downregulate (e.g., by at least 10%, 20%, 50%, 70%, 90% or more) regulatory T-cells in the subject. “Substances capable to downregulating regulatory T-cells” refers to agents that reduce the numbers of cells identified as T-suppressor or Regulatory T-cells. These cells have been identified as consisting one or many of the following immunophenotypic markers: CD4+, CD25+, FoxP3+, CD127- and GITR+. Such agents reducing T-suppressor or Regulatory T-cells may be selected from a group consisting of anti-CD25 antibodies or chemotherapeutics.
  • In some embodiments, the method or use further comprises administration of cyclophosphamide to a subject. Cyclophosphamide is a common chemotherapeutic agent, which has also been used in some autoimmune disorders. In the present invention, cyclophosphamide can be used to enhance viral replication and the effects of GM-CSF induced stimulation of NK and cytotoxic T-cells for enhanced immune response against the tumor. It can be used as intravenous bolus doses or low-dose oral metronomic administration.
  • Kits
  • The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, reflecting approval by the agency of the manufacture, use or sale of the product for human administration.
  • EXAMPLES
  • The present invention is further described by the following examples, which are illustrative of specific embodiments of the invention, and various uses thereof. These exemplifications, which illustrating certain specific aspects of the invention, do not portray the limitations or circumscribe the scope of the disclosed invention.
  • Unless otherwise indicated, the practice of the present invention employs conventional techniques of cell culture, molecular biology, microbiology, recombinant DNA manipulation, immunology science, which are within the skill of the art. Such techniques are explained fully in the literature. See, e.g. Cell Biology: a Laboratory Handbook: J. Cells (Ed). Academic Press. N.Y. (1996); Graham, F. L. and Prevec, L. Adenovirus-based expression vectors and recombinant vaccines. In: Vaccines: New Approaches to Immunological Problems. R. W. Ellis (ed) Butterworth. Pp 363-390; Grahan and Prevec Manipulation of adenovirus vectors. In: Methods in Molecular Biology, Vol. 7: Gene Transfer and Expression Techniques. E. J. Murray and J. M. Walker (eds) Humana Press Inc., Clifton, N.J. pp 109-128, 1991; Sambrook et al. (1989), Molecular Cloning, A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press; Sambrook et al. (1989), and Ausubel et al. (1995), Short Protocols in Molecular Biology, John Wiley and Sons.
  • Example 1. Virology and Recombinant Nucleic Acids
  • Standard techniques are used for recombinant nucleic acid methods, polynucleotide synthesis, and microbial culture and transformation (e.g., electroporation, lipofection). Generally, enzymatic reactions and purification steps are performed according to the manufacturer's specifications. The techniques and procedures are generally performed according to conventional methods in the art and various general references (see generally, Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd. edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which are provided throughout this document. The nomenclature used herein and the laboratory procedures in analytical chemistry, organic synthetic chemistry, and pharmaceutical formulation and delivery, and treatment of patients. Methods for the construction of adenoviral mutants are generally known in the art. See Bett, A. J. et al, PNAS 1994 vol: 91, pages 8802-8806, Mittal, S. K., Virus Res., 1993, vol: 28, pages 67-90; and Hermiston, T. et al., Methods in Molecular Medicine: Adenovirus Methods and Protocols, W. S. M. Wold, ed, Humana Press, 1999. Further, the adenovirus 5 genome is registered as GenBank 10 accession #M73260, and the virus is available from the American Type Culture Collection, Rockville, Md., U.S.A., under accession number VR-5.
  • Viruses and Cell Lines.
  • Adenoviruses and cell lines were generally obtained from American Type Culture Collection (ATCC), Manassas, Va.) unless otherwise noted. Cell lines used in the Examples below may include one or more of the cell lines listed in Table 2.
  • TABLE 2
    Viruses and Cell Lines
    Cell Line Cell/Tissue Type Catalog #
    HEK-293 Normal human embryonic kidney ATCC ® CRL-1573 ™
    MRC-5 Normal human lung ATCC ® CCL-171 ™
    HMEC-1 Normal human endothelium ATCC ® CRL3243 ™
    MCF 10A Normal human mammary epithelial ATCC ® CRL-10317 ™
    NHLF Normal human lung fibroblasts Lonza CC-2512
    HUVEC Normal human umbilical ATCC ® CRL-1730 ™
    endothelium
    HCC827 lung adenocarcinoma ATCC ® CRL-2868 ™
    A549 human lung carcinoma ATCC ® CCL-185 ™
    ADS-12 a derivative from a murine
    KRAS mutant lung
    adenocarcinoma cell line (LKR-
    13)
    NCI-H1734 NSCLC adenocarcinoma ATCC ® CRL-5891 ™
    NCI-H2110 NSCLC metastatic carcinoma ATCC ® CRL-5924 ™
    HT-1080 Connective tissue fibrosarcoma ATCC ® CCL121 ™
    PC-3 Prostate adenocarcinoma ATCC ® CRL1435 ™,
    SNU-449 Hepatocellular carcinoma ATCC ® CRL-2234 ™
    HepG2 Hepatocellular carcinoma ATCC ® HB-8065 ™
    MDA-MB-231 Breast adenocarcinoma ATCC ® HTB-26 ™
    PANC-1 Pancreatic carcinoma ATCC ® CRL-1469 ™
    SW780 Bladder carcinoma ATCC ® CRL-2169 ™
    FaDu Head and neck squamous cell ATCC ® HTB43 ™
    carcinoma
    DLD-1 Colorectal adenocarcinoma ATCC ® CCL-221 ™
    CT26 Murine colon carcinoma ATCC ® CRL-2638 ™
    U-87 brain glioblastoma ATCC ® HTB14 ™
    MBT-2 Murine bladder cancer described, e.g., in Takahashi et
    al., J Urol, 166(6), 2506-2511
    B16F10 Murine skin melanoma ATCC ® CRL-6475 ™
    TAV-255 D19 Adenovirus described in, e.g., Zhang et al.,
    Cancer Gene Ther. (2015)
    22(1): 17-22.
    Ad5 Adenoid 75 strain ATCC ® VR-5 ™
  • Viral Purification and Quantitation
  • Viral stocks were propagated on HEK-293 cells and purified by standard methods such as column purification kits (Virapure, Millipore) or CsCl gradient centrifugation (as described in Tollefson, A., Hermiston, T. W., and Wold, W. S. M.; “Preparation and Titration of CsCl-banded Adenovirus Stock” in Adenovirus Methods and Protocols, Humana Press, 1999, pp 1-10, W. S. M. Wold, Ed.). The method used to quantitate viral particles is based on simple OD 260/280 readings, e.g., using the method of Lehmberg et al. (1999) J. Chrom. B, 732:411-423. In the viral concentration range used, the A260 nm peak area of each sample is directly proportional to the number of viral particles in the sample. The number of viral particles per ml in each test sample was calculated by multiplying the known number of viral particles per ml in the standard by the ratio of the A260 nm viral peak area of the sample to the A260 nm viral peak area of the standard. One A260 unit contains approximately 1×1012 viral particles. Virus Infectious Units/ml (IU/ml) were determined by hexon staining of infected cells (e.g., using Adeno-X™ Rapid titer kit from Takara Bio USA, Inc. (TBUSA, formerly known as Clontech Laboratories, Inc.).
  • Example 2. Bio Selection
  • Initial screening of recombinant virus was based on isolated viral DNA rescued from transfected 293 cells where viral propagation based cytopathic effects (CPE) were observed. Viral DNA was used as a template for PCR based detection of sequences flanking the site of the TAV-255 E1a enhancer deletion region. Wild-type sequence would produce a band of 350 bp, while DNA which had the E1a enhancer deletion would only generate a clearly distinguishable 300 bp band. In addition, primers specific for internal sequences of the IL-12 transgene and for Ad hexon sequences were used to verify 400 bp and 3 kb PCR amplified bands respectively that would only be present in viral DNA generated by recombination between the two parental plasmids containing either the E1 region and transgene insert or the late Ad structural proteins derived solely from the pAdEasy™ plasmid. Individual constructs were isolated by two rounds of plaque purification on A549 or 293 cells using standard methods (Tollefson, A., Hermiston, T. W., and Wold, W. S. M.; “Preparation and Titration of CsCl-banded Adenovirus Stock” in Adenovirus Methods and Protocols, Humana Press, 1999, pp 1-10, W. S. M. Wold, Ed). Dilutions of adenoviral lysates were used to infect A549 or 293 cells in a standard plaque assay. Well-individuated plaques were harvested, and the same plaque assay method was used to generate a second round of individual plaques from these harvests. Well isolated plaques from the second round of plaque purification were deemed pure, infected cultures are prepared using these purified plaques, and the oncolytic potency and selectivity of these culture supernatants was determined.
  • Example 3. Cytolytic Assay
  • Tumor specific viral lysis was evaluated in both tumor and non-tumor cell lines of murine and human origin by infection of the cells in vitro with the viruses, followed by standard crystal violet for cell viability over time as instructed in the kit manuals.
  • Example 4. DNA Sequencing
  • DNA sequencing of the human Ad5-based recombinant adenovirus genomic DNAs was performed as follows. Viral DNA was purified from recombinant adenoviruses such as TAV-255 or other modifications of these constructs by standard column purification methods such as the Qia-Amp® blood DNA purification kit from Qiagen®. PCR primers were used to amplify and isolate regions covering the E1a modified regions and the regions containing the transgene insert and sent out for sequencing at a CRO. Isolated DNA was also analyzed by standard restriction digestion and SDS PAGE analysis for verification of appropriate sized bands of digested DNA. Sequence information was analyzed using the Vector NTI program (Informatix).
  • Example 5. Construction of Recombinant Viruses
  • The base shuttle transfer vector includes pXC1 TAV 255 d19k (Zhang et al., Cancer Gene Ther. (2015) 22(1):17-22). This plasmid has a deletion between bp-305 to -255 of the E1a enhancer region, removing two Pea3 and one E2F binding sites which restrict replication and oncolysis of an adenovirus with this deletion to infected tumor cells (Hedrun, F. H., Shantanu K., and Reid, T. (2011) Cancer Gene Therapy 18; 717-723.) It also contains sites allowing deletion of the E1b 19k region and exogenous transgene insert by a Sal I/Xho I digest. Without the digest, the majority of the E1b 19k region is intact, but non-functional. cDNAs for each transgene of interest were synthesized (GeneArt™) or isolated from commercial plasmid sources (GE Lifesciences or GeneCopoeia™) by PCR using primers which added on 5′ SalI and 3′ XhoI restriction sequences for insertion into the SalI/XhoI digested pXC1 TAV 255 plasmids. The modified pXC1 TAV 255 gene insert plasmids were amplified in E. coli and purified using Qiagen® Maxi-prep plasmid kits.
  • To obtain recombinant adenoviruses, the pXC1 TAV 255 gene insert-containing plasmids were co-transfected into HEK-293 cells (ATCC) with pBHG10 (Microbix) as described (Bett, A. J., Haddara, W., Prevec, L., and Graham, F. (1994) PNAS 91; 8802-8806), using the calcium phosphate transfection protocol from Molecular Cloning: A laboratory manual (Maniatis Vol. 3; 16.30-16.36) for 2-5 μg plasmid DNA (for both plasmids so the pXC1 will be in molar excess) per 60 mm dish of cells. Recombination between homologous adenovirus sequences from each plasmid generates a full length, replication competent adenovirus containing the E1 modifications described earlier and a specific transgene inserted into the E1b 19k deletion site and the E3 deletion supplied by the pBHG10 plasmid. Optionally, the pBHG10 is provided as the adenoviral genome source, having substantial additional utility over vectors such as pJM17 (See, e.g., Hedrun et al, (2011)). Preferentially, an E3 deletion and/or other modifications allow increased packaging capacity for exogenous genes in excess of pJM17 capacity.
  • In another embodiment, the human IL-12 virus, TRZ627, was constructed using a modification of the pAdEasy™ Adenoviral Vector System (Agilent Technologies). First, sequences from pXCI-TAV d19K plasmid (Hedjran F et al., Cancer Gene Therapy (2011) 18, 717-723) which included the 50-nucleotide deletion in the enhancer of E1A which restricts viral propagation to tumor vs. non-tumor cells, were subcloned into the pShuttle™ vector supplied in the pAdEasy™ kit to create the TAV-255 Shuttle E1 cloning plasmid. Sequences between the first Pac I site (6) and the single Mfe I site (807) in pShuttle were replaced by pXC1 TAV d19K sequences from the beginning of the 5′ ITR sequence (21) to the Mfe I site (3874), corresponding to the same Mfe I site in pShuttle. A Pac I cloning site was added by PCR onto the 5′ end of this fragment, which brought in Ad E1a and E1b sequence not found in the original pShuttle plasmid. The TAV-255 deletion of 50 bp in the E1a enhancer region removes the Pea3 III, Pea3 II, and an E2F transcription factor binding sites and corresponds to human wild-type Ad5 sequence of bases 194-244. The added sequences also included a modification introducing Sal I/Xho I cloning sites into the E1b 19k coding region which can be used to replace the E1b 19k ORF with an exogenous transgene insert whose expression would be driven by the Ad E1b promoter during viral replication. Recombination between Pme I linearized pTAV-255 Shuttle E1 containing hIL-12 cloned in at the E1b 19k site and the pAdEasy vector took place in the recA proficient BJ5183 bacterial strain, which had been modified to already contain the pAdEasy plasmid. DNA isolated from Kanamycin resistant plated colonies was screened for full-length viral DNA recombinants by restriction digest. Positive clones (TRZ-627, hIL-12) were subsequently digested with Pac I to free up the Ad ITRs and then transfected into 293 cells to amplify the virus.
  • Infected HEK-293 cells were collected when visible sign of cytopathic effects due to viral replication were observed up to 2 weeks post-infection and resuspended in their media and lysed by 3 rounds of freeze/thaw. Virus can be purified from the lysate by several methods, including Anion-exchange HPLC (Shabram, P. W., et al (1997) Human Gene Therapy 8; 453-465) or several commercially available kits based on affinity chromatography or size exclusion membranes or columns (e.g., Adeno-X™ Maxi Purification Kit, Clontech® (Takara), Adenovirus Purification Virakit®, Virapur®). Purified virus can also undergo clonal isolation by standard plaque purification methods, followed by re-amplification and purification of the plaque purified viral clone.
  • Example 6. In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Encoding Immunomodulatory Polypeptides: Single Viruses with and without Antibody Treatment
  • Materials and Methods
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples were stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials were stored protected from light at −80° C. On each day of dosing, one vial was thawed at room temperature for approximately 20 minutes. A single dose is 1×109 pfu.
  • The mouse tumor model in this Example uses syngeneic immunocompetent mice. Female Jackson 129S1 (129S1/Sv1mJ) mice were used in this study. They were 6-7 weeks old on Day 1 of the experiment. The animals were fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum.
  • Animals were housed in static cages with Bed-O'Cobs™ bedding inside bioBubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour.
  • All treatments, body weight determinations, and tumor measurements were carried out in the bubble environment. The environment was controlled to a temperature range of 70°±2° F. and a humidity range of 30-70%.
  • Cell Preparation
  • ADS-12 cells were grown in RPMI 1640 medium which was modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100× Penicillin/Streptomycin/L-Glutamine (PSG). The growth environment was maintained in an incubator with a 5% CO2 atmosphere at 37° C. When expansion was complete, the cells (passage 7) were trypsinized using 0.25% trypsin/2.21 mM EDTA in HBSS solution. Following cell detachment, the trypsin was inactivated by dilution with complete growth medium and any clumps of cells were separated by pipetting. The cells were centrifuged at 200rcf for 8 minutes at 4° C., the supernatant was aspirated, and the pellet was re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) by pipetting. An aliquot of the homogeneous cell suspension was diluted in a trypan blue solution and counted using a Luna automated cell counter. The cell suspension was centrifuged at 200rcf for 8 minutes at 4° C. The supernatant was aspirated and the cell pellet was re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) to generate a final concentration of 1×107 trypan-excluding cells/ml. The cell suspension was maintained on wet ice during implantation. Following implantation, an aliquot of the remaining cells was diluted with a trypan blue solution and counted to determine the post-implantation cell viability. The cell viabilities of the suspensions used for implantation (two preps) are listed in Table 3.
  • TABLE 3
    Implantation Cell Viability
    Pre-Implant Viability (%) Post-Implant Viability (%)
    Cell Prep 1 95 89
    Cell Prep 2 95 89
  • Test animals were implanted subcutaneously on both flanks (on the back between the spine and the hip), the right flank on Day 0 and the left flank on Day 8, with 1×106 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • All mice were sorted into study groups based on caliper measurement estimation of tumor burden on Day 15 when the mean tumor burden for all animals on the right flank was approximately 82 mm3 (range of group means, 75-90 mm3). The mice were distributed to ensure that the mean tumor burden on the right flank for all groups was within 10% of the overall mean tumor burden for the study population.
  • Results
  • The mean estimated right side tumor burden for all groups in the experiment on the first day of treatment was 82 mm3 and all of the groups in the experiment were well-matched (range of group means, 75-90 mm3). All animals weighed at least 13.3 g at the initiation of therapy. Mean group body weights at first treatment were also well-matched (range, 15.4-18.3 g). A tumor burden of 500 mm3 was chosen for evaluation of efficacy by tumor growth delay for the right and left tumors. The median Control Group (FIG. 6A) tumor burdens reached 500 mm3 on Day 47 for right tumors and Day 43 for left tumors. The median tumor volume doubling times for the Control Group were 12.1 and 10.1 days for the right and left tumors, respectively. Control animals experienced a 7.3% mean weight gain during the treatment regimen. There were no spontaneous regressions in the Control Group for either right or left side tumors, however, 50% of the tumors on the left side never reached the palpation limit. In some embodiments, mice with palpable left side tumors were put on study. Since in this experiment the implant was delayed 8 days, the left side tumors were not palpable at staging. These mice that had a left side tumor that remained a 0 throughout the study could have been triaged out of the study if implanted on the same day as the right side.
  • Results of mouse inoculation and tumor growth are shown in FIG. 6. The series of graphs shows treatment of tumor bearing mice (n=8 in each group) with oncolytic viruses comprising various transgenes, with or without anti-PD-L1.
  • FIG. 6A is a graph showing the activity of various oncolytic viruses compared to the empty virus (“d19k”), 38 days after cell implantation (primary tumor) as a function of tumor growth inhibition. Black bars represent virus alone and hatched bars represent virus+anti-PD-L1 antibody. FIG. 6B is a graph comparing the average tumor size (primary tumor) over time of tumors injected with virus buffer (black circles), empty vector (blue solid squares), anti-PD-L1 alone (antibody given on days 16, 20, 24, and 28, right-hand arrow in each pair of arrows), each of five viruses alone (viruses carrying CTLA-4, IL-12, IL-7, CD70, and IL-10 transgenes) or combined with anti-PD-L1 antibody. FIG. 6C shows similar data for three more viruses (viruses carrying OX40L, CD40L, and GM-CSF transgenes). As shown in the Figures, the IL-12 oncolytic virus treatment, alone or in combination with an anti-PD-L1 antibody, were the most effective at reducing tumor growth. CD70, IL-7, and CTLA-4 viruses were also able to reduce tumor volume significantly when combined with the anti-PD-L1 antibody.
  • The following Figures demonstrate efficacy (or lack thereof) of various viruses with transgenes with or without PD-L1 on the primary tumor only; the thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows). Treatments are shown for the primary tumor (receiving the oncolytic virus injection) and are as follows: 6D, virus buffer and control IgG only (left) and virus buffer and anti-PD-L1 antibody (right); 6E, d19k (empty virus) and control IgG only (left) and d19k and anti-PD-L1 antibody (right); 6F, CTLA-4 virus with control IgG (left) or anti-PD-L1 (right); 6G, IL-12 virus with control IgG (left) or anti-PD-L1 antibody (right); 6H, GM-CSF virus with control IgG (left) or anti-PD-L1 antibody (right); 6I, IL-7 virus with control IgG (left) or anti-PD-L1 antibody (right); 6J, CD40L virus with control IgG (left) or anti-PD-L1 antibody (right); 6K, L10 trap virus with control IgG (left) or anti-PD-L1 antibody (right); and 6L, OX40L virus with control IgG (left) or anti-PD-L1 antibody.
  • As shown in the Figures, combinations with the IL-12 adenovirus (FIG. 6G), showed the greatest ability to reduce tumor volume. As in FIG. 6A, the IL-7 (FIG. 6I) and CTLA-4 (FIG. 6F) oncolytic viruses also showed activity when combined with anti-PD-L1 antibody. Treatment with the oncolytic virus encoding the CD40 ligand also showed some activity when combined with the anti-PD-L1 antibody (FIG. 6J).
  • Example 7. In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Encoding Immunomodulatory Polypeptides: Virus Mixing Study
  • The mouse tumor model in this Example uses syngeneic immunocompetent mice. Animals were injected with approximately 1,000,000 cells subcutaneously (s.c.) into the right hind flank of the mouse. At the same time, the mice were also injected with 1,000,000 cells s.c. into the left hind flank of the mouse to create a bilateral tumor model. When the right (primary) tumors reached 63-80 mm3 in size and the left tumor was palpable, they were injected with 25 μl of virus buffer or 25 μl of virus at 4×109 pfu/ml (plaque forming units per ml) directly into the center of the primary tumor every fourth day for a total of three doses. The mice were also treated intraperitoneally with 250 μl of an anti-PD-L1 antibody at 2 mg/ml every fourth day for a total of three doses. The antibody was administered 24 hours after administration of the virus. A reduction in the size of the primary and distal (contralateral) tumor would be noted relative to the virus buffer control and additional controls such as the wild-type virus (not expressing a transgene) with or without administration of the anti-PD-L1 antibody. A specific example of the treatment with oncolytic adenoviral vectors is described in more detail below for a panel of such vectors.
  • Evaluation of the primary tumor (right flank) was used to determine the direct effect of the oncolytic viruses whereas evaluation of the contralateral tumor (left flank) was used to see the systemic effects of the oncolytic viruses. ADS-12 (a murine KRAS-mutant lung adenocarcinoma cell line) grown in its syngeneic mouse strain is a tumor model known to support adenoviral infection and replication and is useful in the evaluation of host immune responses to oncolytic human adenoviruses.
  • Materials and Methods
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples were stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials were stored protected from light at −80° C. On each day of dosing, one vial was thawed at room temperature for approximately 20 minutes.
  • Animal Studies
  • Female 12951 (129S1/SvImJ) mice from The Jackson Laboratory were used in this study. They were approximately 7-8 weeks old on Day 14 of the experiment. The animals were fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum. Animals were housed in static cages with Bed-O'Cobs™ bedding inside Biobubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour. All treatments, body weight determinations, and tumor measurements were carried out in the bubble environment. The environment was controlled to a temperature range of 70°±2° F. and a humidity range of 30-70%. All procedures carried out in this experiment were conducted in compliance with all the laws, regulations and guidelines of the National Institutes of Health (NIH) and with the approval of Molecular Imaging, Inc.'s Animal Care and Use Committee. Molecular Imaging, Inc. is an AAALAC accredited facility.
  • Cell Preparation
  • ADS-12 cells (murine KRAS-mutant lung adenocarcinoma) were grown in RPMI 1640 medium which is modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100×Penicillin/Streptomycin/L-Glutamine (PSG). The growth environment was maintained in an incubator with a 5% CO2 atmosphere at 37° C. When expansion as complete, the cells are trypsinized using 0.25% trypsin/2.21 mM EDTA in HBSS solution. Following the cell viabilities of the suspensions used for implantation (two preps) are listed in the table below.
  • TABLE 4
    Implantation Cell Viability
    Pre-Implant Viability (%) Post-Implant Viability (%)
    Cell Prep 1 94 92
    Cell Prep 2 97 92
  • Test animals were implanted subcutaneously, on both flanks (on the back between the spine and the hip) on Day 0 with 1.00×106 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • Treatment
  • All mice were sorted into study groups based on caliper measurement estimation of tumor burden on Day 14 when the mean tumor burden for all animals on the right flank is approximately 68 mm3 (range of group means, 65-71 mm3). The mice were distributed to ensure that the mean tumor burden for all groups was within 10% of the overall mean tumor burden for the study population.
  • Measurement and Endpoints
  • Tumor burden (mm3) was estimated from caliper measurements by the formula for the volume of a prolate ellipsoid assuming unit density as: Tumor burden (mm3)=(L× W2)/2, where L and W are the respective orthogonal tumor length and width measurements (mm). All groups were compared to the virus buffer control group.
  • The primary endpoints used to evaluate efficacy were: tumor growth delay, complete and partial tumor response, and the number of tumor-free survivors at the end of the study for both left and right tumors. A complete response (CR) is defined as a decrease in tumor mass to an undetectable size (<63 mm3), and a partial response (PR) is defined as a smaller tumor mass at the last measurement compared to at the first treatment. PRs are exclusive of CRs.
  • All animals were observed for clinical signs at least once daily. Animals were weighed on each day of treatment. Individual body weights were recorded three times weekly. Animals with combined tumor burdens in excess of 2000 mm3 were euthanized, as were those found in obvious distress or in a moribund condition. Treatment-related weight loss in excess of 20% is generally considered unacceptably toxic. In this Example, a dosage level was determined to be tolerated if treatment-related weight loss (during and two weeks after treatment) was <20% and mortality during this period in the absence of potentially lethal tumor burdens was <10%.
  • Results
  • Results of mouse inoculation and tumor growth are shown in FIG. 7. The series of graphs shows treatment of tumor bearing mice (n=8 in each group) with single and combinations of oncolytic viruses comprising various transgenes, with or without anti-PD-L1. The left-hand panel of each Figure represents the primary tumor into which the virus was injected; the right-hand panel represents the contralateral tumor. The thick line in each graph shows the average tumor volume in mm3. The pairs of arrows on the x-axis represent day of treatment with virus (intratumoral, left arrows) and the anti-PD-L1 antibody, if used (intraperitoneal, right arrows). A summary of the responses of the 8 mice is shown in a table at the bottom of each graph, wherein CR=Complete Response (tumor volume=0) and PR=Partial Response (tumor volume on last day of measurements is smaller than tumor volume on the first day of measurements). Treatments shown are as follows: 7A, virus buffer only; 7B, empty virus only (TRZ000); 7C, TRZ010 (IL-10trap)+empty virus; 7D, TRZ011 (OX40 ligand)+empty virus; 7E, TRZ009 (CD70)+empty virus; 7F, TRZ007 (IL-7)+empty virus, 7G, TRZ002 (IL-12)+empty virus; 7H, TRZ004 (GM-CSF)+empty virus; 7I, TRZ003 (flagellin)+empty virus; 7J, TRZ002+TRZ010; 7K, TRZ002+TRZ007; 7L, TRZ007+TRZ010; 7M, TRZ011+TRZ004; 7N, TRZ009+TRZ003; 7O, TRZ002+TRZ009; 7P, TRZ007+TRZ009; 7Q, TRZ007+TRZ004; 7R, TRZ002+TRZ011; 7S, TRZ010+TRZ004; 7T, TRZ002+TRZ004; 7U, virus buffer and anti-PD-L1; 7V, TRZ002+empty virus+anti-PD-L1; 7W, TRZ009+anti-PD-L1; 7X, TRZ007+empty virus+anti-PD-L1; 7Y, TRZ002+TRZ007+anti-PD-L1; 7Z, TRZ007+TRZ009+anti-PD-L1; 7AA, TRZ002+TRZ009+anti-PD-L1. All viruses were administered at 1×108 pfu/dose for each virus, resulting in 2×108 pfu total in virus combinations.
  • As can be seen in the Figure, certain combinations of virus transgenes were extremely successful in lysing tumor cells, including a number of examples of complete response. For example, FIG. 7G shows tumor volume in mice treated with an IL-12 adenovirus. 5 out of 8 mice in this Figure had a complete response, and one mouse had a partial response. In the contralateral tumor, however, no complete or partial responses were seen, indicating that there were no systemic immune effects from treatment with the IL-12 virus alone. A better result is seen in FIG. 7K, which shows mice treated with the same IL-12 adenovirus in combination with an IL-7 adenovirus. In this group of mice, 7 out of 8 had a complete response in the primary tumor (into which the virus was injected) and in the contralateral tumor two mice showed a complete response and one a partial response. In FIGS. 7V (IL-12 adenovirus+anti-PD-L1 antibody) and 7AA (IL-12 adenovirus+CD70 adenovirus+anti-PD-L1 antibody), each group of 8 mice had 8 complete responses in the primary tumor and several in the contralateral tumor. These data demonstrate that certain combination therapies comprising oncolytic adenoviruses can be useful in treating primary and metastatic tumors.
  • Example 8. Adenoviral Vectors Expressing Multiple Immunomodulatory Polypeptides
  • If deletions in the adenovirus backbone are sufficiently large enough to allow packaging of viral DNA containing two exogenously added transgenes into the viral capsids, the two genes can be co-expressed by several methods from a single deletion site in adenovirus. Both added genes can be linked to each other by methods described below and have their expression controlled by an endogenous adenovirus promoter, not an exogenously added promoter, so that high expression will only occur during conditions of viral replication. Control of restricting viral replication to certain conditions such as after infection of tumor cells, is described elsewhere in this document. Co-expression of two proteins from a single transcript can be achieved through the use of virus components such as internal ribosome entry site (IRES) elements (Renaud-Gabardos E et al, World J Exp Med 2015, 5: 11-20), insertion of self-cleaving 2A peptide sequences derived from viruses such as Foot and Mouth Disease virus (FMDV) (Garry A. Luke (2012), Translating 2A Research into Practice, Innovations in Biotechnology, Dr. Eddy C. Agbo (Ed.), ISBN: 978-953-51-0096-6, InTech), or by combining the sequences of the two transgenes into a single fusion protein. An exemplary method would use one of the 2A sequences to direct more equal level of expression from both transgenes as opposed to lower expression levels typically seen from the second transgene when using IRES elements.
  • Descriptions of exemplary dual transgene constructs shown in Table 5 in this Example and Table 6 in Example 13. As can be seen in the table, any of the transgenes may be inserted into the E1 or E3 region. For example, in one embodiment a dual transgene construct may have IL-12 inserted into the E1 region and IL-2 in the E3 region. In another embodiment, a dual transgene construct may have IL-2 inserted into the E1 region and IL-12 in the E3 region. Use of the dual transgene constructs in therapeutic oncolytic adenoviruses is described in Examples 9 and 13.
  • TABLE 5
    Exemplary dual transgene constructs.
    TRZ Number Brief Description
    TRZ402* TAV-255-d19kE1-mIL-10T-E3-mIL-12
    TRZ403* TAV-255-d19kE1-mIL-7-E3-mIL-12
    TRZ404* TAV-255-d19kE1-mCD70-E3-IL-12
    TRZ405 TAV-255-d19kE1-mOX40L-E3-mGM-CSF
    TRZ406 TAV-255-d19kE1-mIL-10T-E3-mIL-7
    TRZ407 TAV-255-d19kE1-mIL-12-E3-mIL-10T
    TRZ408 TAV-255-d19k-E3-mIL-12
    TRZ409 E1-mIL-12/E3-mIL-7
    TRZ413 E1-mIL-7/E3-mIL-12-P2A-ADP
    TRZ418* E1-trimeric mCD70/E3-mIL-12
    TRZ421* E1-mIL-2/E3-mIL-12
    TRZ501 TAV-255-d19K Dual mIL-10T-P2A-mIL-12, E3 deleted
    TRZ510 E1-mIL-7-P2A-mIL-12
    TRZ512 E1-mIL-12-P2A-mIL-7
    m = murine;
    P2A = cleavage site
    *See also Table 6
  • Example 9. Adenoviral Vectors Expressing Multiple Immunomodulatory Polypeptides
  • To maintain co-expression from two single gene inserts at different sites, the cDNA for each transgene can be inserted into separate deleted regions of adenovirus so that expression of each would be controlled separately by the endogenous upstream adenovirus promoter. No exogenous promoter would be added with the exogenous transgene sequence. Expression of the E1a proteins leads to the activation of the other adenovirus promoters and viral replication, so expression from each endogenous adenovirus promoter is linked to viral replication. Exogenous transgenes inserted behind different adenovirus promoters, such as the E1b promoter, the E3 promoter, and the E4 promoter, in place of deletions in these regions, leads to a construct where co-expression from each inserted transgene is limited to conditions of where viral replication occurs. Combined with modifications in the E1a enhancer region as described previously in this document to restrict viral replication to tumor cells, co-expression of both exogenous transgenes is restricted to tumor cells.
  • As shown schematically in FIG. 2, provided is an ΔE1b19K site that includes a deletion of bp 1714-1916 (numbered according to hAd5 vector sequence), which increases packaging capacity by approximately 200 bp. Also provided is a deletion at the E3 site (ΔE3 site as shown in FIG. 2); this deletion can be any deletion in the E3 region, generally at or about bp 27,900-30,800 bp (numbered according to hAd5 vector sequence), which increases packaging capacity up to an additional approximately 2400 bp, as compared to most wild type adenoviruses, which are typically limited to containing (i.e., packaging) approximately 1800 bp of exogenous sequences.
  • Provided herein are deletions in E3 open reading frames that are suitable for modification (e.g., truncation or deletion) without substantially decreasing viral propagation. By way of non-limiting example, provided are adenoviral vectors wherein the E3 12.5K coding region (27,852-28,175 bp) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 320 or greater than 320 bases are deleted from one or more truncation sites within the region) or entirely deleted. Also provided are adenoviral vectors wherein the E3 7.1K coding region (28,541-28,732) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or greater than 190 bases are deleted from one or more truncation sites within the region) or entirely deleted. The 7.1K sequence is associated with inhibition of TRAIL apoptosis and associated with one or more RID proteins. Also provided are adenoviral vectors wherein the E3 gp19K (28,729-29211) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450 or greater than 450 bases are deleted from one or more truncation sites within the region) or entirely deleted. The gp19K sequence is associated with inhibition of CTL killing. Also provided are adenoviral vectors wherein the E3 10.5 (also called E3 11.6 (ADP)) (29, 485-29,766) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, or greater than 250 bases are deleted from one or more truncation sites within the region) or entirely deleted. The 10.5 sequence is associated with promotion of virus release. Also provided are adenoviral vectors wherein the E3 (RIDα) (29,778-29,969) and/or the E3 (RIDβ) (30,057-30,455) is truncated (e.g., with respect to E3 (RIDα) at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or greater than 190 bases are deleted from one or more truncation sites within the region) or entirely deleted, or truncated (e.g., with respect to E3 (RIDβ) at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 390, or greater than 390 bases are deleted from one or more truncation sites within the region) or entirely deleted. These RID sequences are associated with inhibition of TNF, FasL, and TRAIL apoptosis and degrade EGFR. Any combination of the above-referenced deletions is also provided.
  • Also provided are adenoviral vectors wherein the E3 14.7K (30,488-30,834) is truncated (e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 340, or greater than 340 bases are deleted from one or more truncation sites within the region) or entirely deleted. The 14.7K sequence is associated with inhibition of TNF, FasL, and TRAIL apoptosis.
  • Example 10. Adenoviral Vectors Expressing an Immunomodulatory Polypeptide from the E4 Region
  • A nucleic acid sequence encoding one or more immunomodulatory polypeptides is inserted into the adenoviral genome by truncation or deletion of a portion of the E4 region of the viral genome. An E4 deletion and exogenous insertion can be utilized in combination with any other viral modification provided herein or otherwise known in the art, or alternatively, without any other viral modification. Exemplary E4 regions useful as insertion sites include truncation or deletion of E4 ORF1 (35,136-35,522 bp) and/or E4 ORF2 (34,696-35,106 bp) (each numbered according to hAd5 vector sequence). The expression of the one or more immunomodulatory polypeptides is controlled by the endogenous E4 promoter. A schematic illustration of this embodiment is provided in FIG. 3.
  • Example 11. Adenoviral Vectors Comprising Altered Configuration of Pea3 Sites
  • Adenoviral vectors are designed in which sequences in and around Pea3 sites I-V near the E1a Enhancer region of adenoviral vectors, in which sequences in and around Pea3 sites are altered, moved, or deleted in order to produce vectors with a variety of expression characteristics. In one embodiment, adenovirus vectors are engineered which have lower affinity Pea3 sites compared to wild-type adenoviral vectors. Such vectors are designed to be efficient in cells in the tumor microenvironment where the concentration of transcription factors is high, but to be relatively inactive in normal (e.g., non-neoplastic) cells, thus reducing the possibility of side effects caused by damage to normal tissue cells. An illustration of the E1a area of wild-type and TAV-255 constructs is shown in FIG. 9A. A cartoon illustration of the proposed method of action of the E1a enhancer region mutants is shown in FIG. 9B. Constructs NV1 to NV7 are based in part on the TAV-255 construct.
  • In one embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the sequence is removed between Pea3 IV and Pea3 III sites, a single mutation is made in Pea3 III, the sequence between Pea3 III and Pea3 II, and the Pea3 II site is mutated. The sequence of the E1a enhancer region of the exemplary construct NV1 is set forth in SEQ ID NO:99.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the sequence is removed between Pea3 IV and Pea3 III sites, both Pea3 III and Pea3 II are mutated, and Pea3 V flanking sites are mutated such that the resultant Pea3 V site has an affinity more similar to Pea3 III and enhancer 1. The sequence of the E1a enhancer region of the exemplary construct NV2 is set forth in SEQ ID NO:100.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the sequence between Pea3 V and Pea3 IV sites is replaced with the sequence between the Pea3 III and Pea3 II sites; the sequence between the Pea3 IV and Pea3 III is deleted; the sequence between the Pea3 III and Pea3 II is deleted; and the Pea3 III and Pea3 II sites are both mutated, as well as the residues 3 bp that are immediately 5′ of the Pea3 V site. The sequence of the E1a enhancer region of the exemplary construct NV3 is set forth in SEQ ID NO:101.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites (thus engineering a vector in which all Pea3 sites except Pea3 I are lower affinity). The sequence of the E1a enhancer region of the exemplary construct NV4 is set forth in SEQ ID NO:102.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites, and a single point mutation is introduced in the Pea3 I site, rendering it a lower affinity Pea3 I site (thus engineering a vector in which all Pea3 sites are lower affinity). The sequence of the E1a enhancer region of the exemplary construct NV5 is set forth in SEQ ID NO:103.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence), the flanking sequences around the Pea3 III and Pea3 II sites are altered to mimic the lower affinity Pea3 V and Pea3 IV sites, and the flanking regions of the Pea3 I site are altered, rendering it a lower affinity Pea3 I site (thus engineering a vector in which all Pea3 sites are lower affinity). The sequence of the E1a enhancer region of the exemplary construct NV6 is set forth in SEQ ID NO:104.
  • In another embodiment, a vector is provided wherein, (in relation to the wild-type sequence) the flanking sequences around the Pea3 I site are altered to produce a lower-affinity Pea3 I site, and the flanking sequences around the Pea3 II site is altered to mimic the lower affinity Pea3 IV site (thus engineering a vector in which all Pea3 sites except Pea3 III are lower affinity). The sequence of the E1a enhancer region of the exemplary construct NV7 is set forth in SEQ ID NO:105.
  • Example 12. In Vivo Demonstration of Reduction in Tumor Volume with Oncolytic Adenoviral Vectors Comprising E1a Enhancer Region Alterations and Encoding Immunomodulatory Polypeptides: Single Viruses with and without Antibody Treatment Materials and Methods
  • An anti-mPD-L1 antibody is, e.g., from BioXCell®, Catalog# BE0101 (Rat IgG2b). This antibody was used in the below experiments.
  • Virus samples are stored in 25 mM NaCl, 10 mM Tris Tris(hydroxymethyl)aminomethane), and 5% glycerol with a pH value of 8.0. Vials are stored protected from light at −80° C. On each day of dosing, one vial is thawed at room temperature for approximately 20 minutes. A single dose is 1×109 pfu. Viruses to be combined with anti-PD-L1 in this Example include NV1-NV7 (having E1a enhancer region sequences set forth in SEQ ID Nos:99-105).
  • The mouse tumor model in this Example uses syngeneic immunocompetent mice. Female Jackson 129S1 (12951/Sv1mJ) mice are used in this study. Mice are 6-7 weeks old on Day 1 of the experiment. The animals are fed irradiated Harlan 2918.15 Rodent Diet and water ad libitum. Animals are housed in static cages with Bed-O'Cobs™ bedding inside bioBubble® Clean Rooms that provide H.E.P.A filtered air into the bubble environment at 100 complete air changes per hour. All treatments, body weight determinations, and tumor measurements are carried out in the bubble environment. The environment is controlled to a temperature range of 70°±2° F. and a humidity range of 30-70%.
  • Cell Preparation
  • ADS-12 cells are grown in RPMI 1640 medium which is modified with 1% 100 mM Na pyruvate, 1% 200 mM L-glutamine, 1% 1M HEPES buffer, 1% of a 45% glucose solution and supplemented with 10% non-heat-inactivated Fetal Bovine Serum (FBS) and 1% 100× Penicillin/Streptomycin/L-Glutamine (PSG). The growth environment is maintained in an incubator with a 5% CO2 atmosphere at 37° C. When expansion is complete, the cells (passage 7) are trypsinized using 0.25% trypsin/2.21 mM EDTA in HBSS solution. Following cell detachment, the trypsin is inactivated by dilution with complete growth medium and any clumps of cells are separated by pipetting. The cells are centrifuged at 200rcf for 8 minutes at 4° C., the supernatant is aspirated, and the pellet is re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) by pipetting. An aliquot of the homogeneous cell suspension is diluted in a trypan blue solution and counted using a Luna automated cell counter. The cell suspension is centrifuged at 200rcf for 8 minutes at 4° C. The supernatant is aspirated and the cell pellet is re-suspended in cold Dulbecco's Phosphate Buffered Saline (DPBS) to generate a final concentration of 1×107 trypan-excluding cells/ml. The cell suspension is maintained on wet ice during implantation. Following implantation, an aliquot of the remaining cells is diluted with a trypan blue solution and counted to determine the post-implantation cell viability.
  • Test animals are implanted subcutaneously on both flanks (on the back between the spine and the hip), the right flank on Day 0 and the left flank on Day 8, with 1×106 cells in 0.1 ml of serum-free medium using a 28-gauge insulin syringe with a fixed needle.
  • All mice are sorted into study groups based on caliper measurement estimation of tumor burden on Day 15 when the mean tumor burden for all animals on the right flank is approximately 82 mm3 (range of group means, 75-90 mm3). The mice are distributed to ensure that the mean tumor burden on the right flank for all groups is within 10% of the overall mean tumor burden for the study population.
  • Results
  • The mean estimated right side tumor burden for all groups in the experiment on the first day of treatment is approximately 82 mm3 and all of the groups in the experiment are well-matched (range of group means, 75-90 mm3). All animals weigh at least 13.3 g at the initiation of therapy. Mean group body weights at first treatment are also well-matched (range, approximately 15.4-18.3 g). A tumor burden of 500 mm3 is chosen for evaluation of efficacy by tumor growth delay for the right and left tumors. The median Control Group tumor burdens will reach 500 mm3 on or about Day 47 for right tumors and on or about Day 43 for left tumors. The median tumor volume doubling times for the Control Group will be approximately 12 and 10 days for the right and left tumors, respectively.
  • Results of mouse inoculation and tumor growth will show that treatment of tumor bearing mice with oncolytic viruses comprising altered E1a regions and encoding various transgenes, with or without anti-PD-L1, show efficacy in reducing tumor volume.
  • Example 13. In Vivo Efficacy of Dual-Specificity Oncolytic Adenoviral Vectors in a Bilateral Tumor Model
  • Cancer immunotherapy is moving toward use of combinations to increase efficacy. Combining cancer immunotherapies can expand clinical benefits of existing approved monotherapies; however, systemically-administered combinations can produce excessive toxicity. Therefore, novel dual specificity oncolytic adenoviral vectors were developed having a transgene at both the E1 and the E3 regions in order to evaluate combinations of adenoviral-delivered immunomodulators to enhance systemic antitumor immunity. A summary of exemplary constructs contemplated for use by the methods disclosed herein (with additions to those disclosed in Table 5) is listed in Table 6.
  • TABLE 6
    Exemplary E1/E3 Dual Transgene Constructs and Single
    Transgene Comparators (*)
    Name E1 E3
    TRZ-001* CTLA-4 Del
    TRZ-009* CD70 intact
    TRZ-018* CD70 trimer Del
    TRZ-021* Empty IL-2
    TRZ-032* IL-2 Empty (has 12.5K protein)
    TRZ-401 CTLA4 IL-12
    TRZ-402 IL-10 Trap IL-12
    TRZ-403 IL-7 IL-12 (has 12.5K protein)
    TRZ-404 CD70 IL-12
    TRZ-408* Empty IL-12
    TRZ-409 IL-12 IL-7
    TRZ-411 OX40L IL-12
    TRZ-412 IL-2 IL-12
    TRZ-414 CD40L IL-12
    TRZ-415 IL-12 CTLA4
    TRZ-416 OX40L (trimer) IL-12
    TRZ-417 CD40L (trimer) IL-12
    TRZ-418 CD70 (trimer) IL-12
    TRZ-421 IL-12 IL-2
    TRZ-510 IL-7-P2A-IL-12 TRZ-202-like (no 12.5)
    TRZ-TBD IL-7-P2A-IL-12 IL-2
  • A bilateral tumor model was prepared using ADS-12 tumor cells as described in the Examples above (e.g., Example 7). 1×106 ADS-12 tumor cells injected into primary (−2 days) and contralateral (0 days) flanks. At staging, mice are randomized based on contralateral tumors (88-150 mm3) and primary tumors (88-250 mm3). There are 8 mice/group, with tumors and body weight measured 3 times per week. An anti-PD-L1 (500 μg/dose) or an anti-PD-1 antibody (250 μg/dose) is administered i.p. to induce systemic T cell activation.
  • In this model, the virus is injected intratumorally (i.t.) into primary tumors only; such injection leads to oncolysis, immune infiltration, and tumor shrinkage. The contralateral tumor is not injected, and no oncolysis occurs. Rather, tumor shrinkage is solely due to antigen-specific activated tumor infiltrating lymphocytes.
  • Results are shown in FIG. 9. FIG. 9A is a cartoon of the contrast between the single transgene constructs (top) used in the virus mixing examples, and the dual transgene vectors used to make the E1/E3 transgene adenoviruses. FIG. 9B shows the results of mice injected i.t. with Empty virus+/−anti-PD-L1 or with TRZ-409 (IL-12/IL-7 dual transgene)+/−anti-PD-L1. The tumor volume of the injected tumor is shown in the left panel, and the tumor volume of the contralateral tumor is shown in the right panel. For each pair of arrows, the left arrow indicates virus injection and the right arrow indicates anti-PD-L1 injection. As can be seen for both the primary and contralateral tumors, TRZ-409 injection reduced tumor volume significantly compared to empty virus. The combination with anti-PD-L1 was slightly more efficacious in this study.
  • FIG. 9C shows the results of a second experiment using the inverse of TRZ-409, TRZ-403, in which the IL-7 transgene occupies the E1 region and the IL-12 gene occupies the E3 region (which has a stronger promoter than the E1). As can be seen in the Figure, TRZ-403 strongly inhibits primary and distant tumor growth, with or without anti-PD-L1. Plasma levels of IL-12 and IFN-γ (FIG. 9D) for all mice injected with TRZ-403 show that expression of the transgenes is well tolerated.
  • The study was extended for all mice having tumors over 500 mm3. Mice having primary tumors injected with TRZ-403 (IL-7+IL-12), TRZ-403+anti-PD-L1, TRZ-403+control IgG, or TRZ-409 (IL-12+1L-7), and controls including untreated mice and mice injected with empty vector TRZ-d19K with anti-PD-L1, anti-PD-1, or a control IgG are shown in FIG. 9E, which illustrates the survival as a percentage of mice still on study. As can be seen in the Figure, mice injected with TRZ-403 had a much higher percentage of survival over TRZ-409 at the time end of the study (70 days). Both TRZ-403 and TRZ-409 showed efficacy over the controls; all mice receiving control injections were deceased by day 56 of the study.
  • Next, a direct comparison was made between the dual transgene virus TRZ-403 (11-7+IL-12) and a mixture of IL-7 and IL-12 single transgene viruses. As can be seen in FIG. 9F, in the primary tumor (left panel) TRZ-403 showed the most efficacy in reducing tumor volume, followed by the mixture of viruses. The single transgene IL-12 virus showed a small amount of efficacy by comparison. In the contralateral tumor, however (right panel) only the dual transgene virus, TRZ-403, reduced the tumor volume, showing significant superiority over the mixture of viruses.
  • Endnotes
  • Although the invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.
  • All publications, databases, GenBank sequences, patents, and patent applications cited in this specification are herein incorporated by reference as if each specifically and individually indicated to be incorporated by reference.
  • APPENDIX A
    SEQUENCE REFERENCE
    Accession
    SEQ Number(s)
    ID or
    NO: Molecule description Sequence
    1 hGITR- NP_005083  MTLHPSPITCEFLFSTALISPKMCLSHLENMPL
    Ligand SHSRTQGAQRSSWKLWLFCSIVMLLFLCSFS
    (GITR-L; WLIFIFLQLETAKEPCMAKFGPLPSKWQMASS
    TNFSF18) EPPCVNKVSDWKLEILQNGLYLIY
    GQVAPNANYNDVAPFEVRLYKNKDMIQTLT
    NKSKIQNVGGTYELHVGDTIDLIFNSEHQVLK
    NNTYWGIILLANPQFIS
    2 mGITR- NM_183391 MEEMPLRESSPQRAERCKKSWLLCIVALLLM
    Ligand LLCSLGTLIYTSLKPTAIESCMVKFELSSSKWH
    (GITR-L; MTSPKPHCVNTTSDGKLKILQSGTYLIYGQVI
    TNFSF18) PVDKKYIKDNAPFVVQIYKKNDVLQTLMNDF
    QILPIGGVYELHAGDNIYLKFNSKDHIQKTNT
    YWGIILMPDLPFIS
    3 hCD28 ligand EAW79565 MGHTRRQGTSPSKCPYLNFFQLLVLAGLSHF
    or agonist CSGVIHVTKEVKEVATLSCGHNVSVEELAQT
    (CD80) RIYWQKEKKMVLTMMSGDMNIWPEYKNRTI
    FDITNNLSIVILALRPSDEGTYECVVLK
    YEKDAFKREHLAEVTLSVKADFPTPSISDFEIP
    TSNIRRIICSTSGGFPEPHLSWLENGEELNAINT
    TVSQDPETELYAVSSKLDFNMTTNHSFMCLIK
    YGHLRVNQTFNWNTTKQEHFP
    DNLLPSWAITLISVNGIFVICCLTYCFAPRCRE
    RRRNERLRRESVRPV
    4 hTNFα CAA78745 MSTESMIRDVELAEEALPKKTGGPQGSRRCLF
    LSLFSFLIVAGATTLFCLLHFGVIGPQREEFPR
    DLSLISPLAQAVRSSSRTPSDKPVAHVVANPQ
    AEGQLQWLNRRANALLANGVELR
    DNQLVVPSEGLYLIYSQVLFKGQGCPSTHVLL
    THTISRIAVSYQTKVNLLSAIKSPCQRETPEGA
    EAKPWYEPIYLGGVFQLEKGDRLSAEINRPDY
    LDFAESGQVYFGIIAL
    5 Non-cleavable NM_013693 MIETYSQPSPRSVATGLPASMKIFMYLLTVFLI
    mTNFα TQMIGSVLFAVYLHRRLDKVEEEVNLHEDFV
    FIKKLKRCNKGEGSLSLLNCEEMRRQFEDLV
    KDITLNKEEKKEDEDPVAHVVANHQVEEQLE
    WLSQRANALLANGMDLKDNQLVVPADGLY
    LVYSQVLFKGQGCPDYVLLTHTVSRFAISYQE
    KVNLLSAVKSPCPKDTPEGAELKPWYEPIYLG
    GVFQLEKGDQLSAEVNLPKYLDFAESGQVYF
    GIIAL
    6 mGM-CSF NM_009969 MWLQNLLFLGIVVYSLSAPTRSPITVTRPWKH
    VEAIKEALNLLDDMPVTLNEEVEVVSNEFSFK
    KLTCVQTRLKIFEQGLRGNFTKLKGALNMTA
    SYYQTYCPPTPETDCETQVTTYADFIDSLKTF
    LTDIPFECKKPGQK
    7 hICOS ligand NP_056074  MRLGSPGLLFLLFSSLRADTQEKEVRAMVGS
    or agonist DVELSCACPEGSRFDLNDVYVYWQTSESKTV
    VTYHIPQNSSLENVDSRYRNRALMSPAGMLR
    GDFSLRLFNVTPQDEQKFHCLVLSQSL
    GFQEVLSVEVTLHVAANFSVPVVSAPHSPSQ
    DELTFTCTSINGYPRPNVYWINKTDNSLLDQA
    LQNDTVFLNMRGLYDVVSVLRIARTPSVNIG
    CCIENVLLQQNLTVGSQTGNDIGERD
    KITENPVSTGEKNAATWSILAVLCLLVVVAV
    AIGWVCRDRCLQHSYAGAWAVSPETELTGH
    V
    8 h4-1BB AAA53134 MEYASDASLDPEAPWPPAPRARACRVLPWA
    ligand or LVAGLLLLLLLAAACAVFLACPWAVSGARAS
    agonist PGSAASPRLREGPELSPDDPAGLLDLRQGMFA
    QLVAQNVLLIDGPLSWYSDPGLAGVSL
    TGGLSYKEDTKELVVAKAGVYYVFFQLELRR
    VVAGEGSGSVSLALHLQPLRSAAGAAALALT
    VDLPPASSEARNSAFGFQGRLLHLSAGQRLG
    VHLHTEARARHAWQLTQGATVLGLFRV
    TPEIPAGLPSPRSE
    9 hOX40 ligand CAE11757 MCVGARRLGRGPCAALLLLGLGLSTVTGLHC
    or agonist VGDTYPSNDRCCHECRPGNGMVSRCSRSQNT
    VCRPCGPGFYNDVVSSKPCKPCTWCNLRSGS
    ERKQLCTATQDTVCRCRAGTQPLDSYK
    PGVDCAPCPPGHFSPGDNQACKPWTNCTLAG
    KHTLQPASNSSDAICEDRDPPATQPQETQGPP
    ARPITVQPTEAWPRTSQGPSTRPVEVPGGRAV
    AAILGLGLVLGLLGPLAILLALYLL
    RRDQRLPPDAHKPPGGGSFRTPIQEEQADAHS
    TLAKI
    10 mOX40 NM_009452 MEGEGVQPLDENLENGSRPRFKWKKTLRLV
    ligand VSGIKGAGMLLCFIYVCLQLSSSPAKDPPIQRL
    RGAVTRCEDGQLFISSYKNEYQTMEVQNNSV
    VIKCDGLYIIYLKGSFFQEVKIDLHFREDHNPI
    SIPMLNDGRRIVFTVVASLAFKDKVYLTVNAP
    DTLCEHLQINDGELIVVQLTPGYCAPEGSYHS
    TVNQVPL
    11 hCD40 ligand NP_000065 MIETYNQTSPRSAATGLPISMKIFMYLLTVFLI
    or agonist TQMIGSALFAVYLHRRLDKIEDERNLHEDFVF
    MKTIQRCNTGERSLSLLNCEEIKSQFEGFVKDI
    MLNKEETKKENSFEMQKGDQNP
    QIAAHVISEASSKTTSVLQWAEKGYYTMSNN
    LVTLENGKQLTVKRQGLYYIYAQVTFCSNRE
    ASSQAPFIASLCLKSPGRFERILLRAANTHSSA
    KPCGQQSIHLGGVFELQPGASVFVN
    VTDPSQVSHGTGFTSFGLLKL
    12 mCD40 NM_011616 MIETYSQPSPRSVATGLPASMKIFMYLLTVFLI
    ligand TQMIGSVLFAVYLHRRLDKVEEEVNLHEDFV
    FIKKLKRCNKGEGSLSLLNCEEMRRQFEDLV
    KDITLNKEEKKENSFEMQRGDEDPQIAAHVV
    SEANSNAASVLQWAKKGYYTMKSNLVMLE
    NGKQLTVKREGLYYVYTQVTFCSNREPSSQR
    PFIVGLWLKPSSGSERILLKAANTHSSSQLCEQ
    QSVHLGGVFELQAGASVFVNVTEASQVIHRV
    GFSSFGLLKL
    13 hCD27 ligand  AAA36175 MPEEGSGCSVRRRPYGCVLRAALVPLVAGLV
    or agonist ICLVVCIQRFAQAQQQLPLESLGWDVAELQL
    NHTGPQQDPRLYWQGGPALGRSFLHGPELDK
    GQLRIHRDGIYMVHIQVTLAICSSTTA
    SRHHPTTLAVGICSPASRSISLLRLSFHQGCTIV
    SQRLTPLARGDTLCTNLTGTLLPSRNTDETFF
    GVQWVRP
    14 mCD70 NM_011617 MPEEGRPCPWVRWSGTAFQRQWPWLLLVVF
    ligand or ITVFCCWFHCSGLLSKQQQRLLEHPEPHTAEL
    agonist QLNLTVPRKDPTLRWGAGPALGRSFTHGPEL
    EEGHLRIHQDGLYRLHIQVTLANCSSPGSTLQ
    HRATLAVGICSPAAHGISLLRGRFGQDCTVAL
    QRLTYLVHGDVLCTNLTLPLLPSRNADETFFG
    VQWICP
    15 hInterleukin-2 AAB46883 MYRMQLLSCIALSLALVTNSAPTSSSTKKTQL
    (IL-2) QLEHLLLDLQMILNGINNYKNPKLTRMLTFK
    FYMPKKATELKHLQCLEEELKPLEEVLNLAQ
    SKNFHLRPRDLISNINVIVLELKGSE
    TTFMCEYADETATIVEFLNRWITFCQSIISTLT
    16 hInterleukin-7 AAH47698 MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKD
    (IL-7) GKQYESVLMVSIDQLLDSMKEIGSNCLNNEF
    NFFKRHICDANKEGMFLFRAARKLRQFLKMN
    STGDFDLHLLKVSEGTTILLNCTGQ
    VKGRKPAALGEAQPTKSLEENKSLKEQKKLN
    DLCFLKRLLQEIKTCWNKILMGTKEH
    17 murine MM_008371.5 MFHVSFRYIFGIPPLILVLLPVTSSECHIKDKEG
    Interleukin-7 KAYESVLMISIDELDKMTGTDSNCPNNEPNFF
    (IL-7) RKHVCDDTKEAAFLNRAARKLKQFLKMNISE
    EFNVHLLTVSQGTQTLVNCTSKEEKNVKEQK
    KNDACFLKRLLREIKTCWNKILKGSI
    18 Interleukin-12 AAD16432 MWPPGSASQPPPSPAAATGLHPAARPVSLQC
    (IL-12) alpha RLSMCPARSLLLVATLVLLDHLSLARNLPVA
    subunit TPDPGMFPCLHHSQNLLRAVSNMLQKARQTL
    EFYPCTSEEIDHEDITKDKTSTVEACL
    PLELTKNESCLNSRETSFITNGSCLASRKTSFM
    MALCLSSIYEDLKMYQVEFKTMNAKLLMDP
    KRQIFLDQNMLAVIDELMQALNFNSETVPQK
    SSLEEPDFYKTKIKLCILLHAFRIRAVTIDRVM
    SYLNAS
    19 hInterleukin- NP_005526 MEPLVTWVVPLLFLFLLSRQGAACRTSECCF
    12 (IL-12) QDPPYPDADSGSASGPRDLRCYRISSDRYECS
    receptor WQYEGPTAGVSHFLRCCLSSGRCCYFAAGSA
    subunit beta-1 TRLQFSDQAGVSVLYTVTLWVESWAR
    isoform NQTEKSPEVTLQLYNSVKYEPPLGDIKVSKLA
    precursor GQLRMEWETPDNQVGAEVQFRHRTPSSPWK
    LGDCGPQDDDTESCLCPLEMNVAQEFQLRRR
    QLGSQGSSWSKWSSPVCVPPENPPQPQ
    VRFSVEQLGQDGRRRLTLKEQPTQLELPEGC
    QGLAPGTEVTYRLQLHMLSCPCKAKATRTLH
    LGKMPYLSGAAYNVAVISSNQFGPGLNQTW
    HIPADTHTEPVALNISVGTNGTTMYWPA
    RAQSMTYCIEWQPVGQDGGLATCSLTAPQDP
    DPAGMATYSWSRESGAMGQEKCYYITIFASA
    HPEKLTLWSTVLSTYHFGGNASAAGTPHHVS
    VKNHSLDSVSVDWAPSLLSTCPGVLKE
    YVVRCRDEDSKQVSEHPVQPTETQVTLSGLR
    AGVAYTVQVRADTAWLRGVWSQPQRFSIEV
    QVSDWLIFFASLGSFLSILLVGVLGYLGLNRA
    ARHLCPPLPTPCASSAIEFPGGKETWQ
    WINPVDFQEEASLQEALVVEMSWDKGERTEP
    LEKTELPEGAPELALDTELSLEDGDRCKAKM
    20 murine N/A MCPQKLTISWFAIVLLVSPLMAMWELEKDVY
    Interleukin-12 VVEVDWTPDAPGETVNLTCDTPEEDDITWTS
    fusion DQRHGVIGSGKTLTITVKEFLDAGQYTCHKG
    polypeptide GETLSHSHLLLHKKENGIWSTEILKNFKNKTF
    LKCEAPNYSGRFTCSWLVQRNMDLKFNIKSS
    SSSPDSRAVTCGMASLSAEKVTLDQRDYEKY
    SVSCQEDVTCPTAEETLPIELALEARQQNKYE
    NYSTSFFIRDIIKPDPPKNLQMKPLKNSQVEVS
    WEYPDSWSTPHSYFSLKFFVRIQRKKEKMKE
    TEEGCNQKGAFLVEKTSTEVQCKGGNVCVQ
    AQDRYYNSSCSKWACVPCRVRSGGGGSGGG
    GSGGGGSRVIPVSGPARCLSQSRNLLKTTDD
    MVKTAREKLKHYSCTAEDIDHEDITRDQTST
    LKTCLPLELHKNESCLATRETSSTTRGSCLPPQ
    KTSLMMTLCLGSIYEDLKMYQTEFQAINAAL
    QNHNHQQIILDKGMLVAIDELMQSLNHNGET
    LRQKPPVGEADPYRVKMKLCILLHAFSTRVV
    TINRVMGYLSSA
    21 hIL-15 CAA62616 MRISKPHLRSISIQCYLCLLLNSHFLTEAGIHV
    FILGCFSAGLPKTEANWVNVISDLKKIEDLIQS
    MHIDATLYTESDVHPSCKVTAMKCFLLELQV
    ISLESGDASIHDTVENLIILANNSLSSNGNVTES
    GCKECEELEEKNIKEFLQSFVHIVQMFINTS
    22 mIL-15 N/A MKILKPYMRNTSISCYLCFLLNSHFLTEAGIH
    hybrid VFILGCVSVGLPKTEANWIDVRYDLEKIESLIQ
    SIHIDTTLYTDSDFHPSCKVTAMNCFLLELQVI
    LHEYSNMTLNETVRNVLYLANSTLSSNKNVA
    ESGCKECEELEEKTFTEFLQSFIRIVQMFINTS
    GSGATNFSLLKQAGDVEENPGPGTTCPPPVSI
    EHADIRVKNYSVNSRERYVCNSGFKRKAGTS
    TLIECVINKNTNVAHWTTPSLKCIRDPSLAHY
    SPVPTVVTPKVTSQPESPSPSAKEPEAFSPKSD
    TAMTTETAIMPGSRLTPSQTTSAGTTGTGSHK
    SSRAPSLAATMTLEPTASTSLRITEISPHSSKM
    TK
    23 hIL-10 NP_001549 MLPCLVVLLAALLSLRLGSDAHGTELPSPPSV
    receptor WFEAEFFHHILHWTPIPNQSESTCYEVALLRY
    subunit alpha GIESWNSISNCSQTLSYDLTAVTLDLYHSNGY
    precursor RARVRAVDGSRHSNWTVTNTRFSV
    DEVTLTVGSVNLEIHNGFILGKIQLPRPKMAP
    ANDTYESIFSHFREYEIAIRKVPGNFTFTHKKV
    KHENFSLLTSGEVGEFCVQVKPSVASRSNKG
    MWSKEECISLTRQYFTVTNVIIFF
    AFVLLLSGALAYCLALQLYVRRRKKLPSVLL
    FKKPSPFIFISQRPSPETQDTIHPLDEEAFLKVS
    PELKNLDLHGSTDSGFGSTKPSLQTEEPQFLLP
    DPHPQADRTLGNREPPVLGDSC
    SSGSSNSTDSGICLQEPSLSPSTGPTWEQQVGS
    NSRGQDDSGIDLVQNSEGRAGDTQGGSALGH
    HSPPEPEVPGEEDPAAVAFQGYLRQTRCAEE
    KATKTGCLEEESPLTDGLGPKFGRC
    LVDEAGLHPPALAKGYLKQDPLEMTLASSGA
    PTGQWNQPTEEWSLLALSSCSDLGISDWSFA
    HDLAPLGCVAAPGGLLGSFNSDLVTLPLISSL
    QSSE
    24 mIL-10R N/A MLSRLLPFLVTISSLSLEFIAYGTELPSPSYVW
    Trap FEARFFQHILHWKPIPNQSESTYYEVALKQYG
    NSTWNDIHICRKAQALSCDLTTFTLDLYHRSY
    GYRARVRAVDNSQYSNWTTTETRFTVDEVIL
    TVDSVTLKAMDGIIYGTIHPPRPTITPAGDEYE
    QVFKDLRVYKISIRKFSELKNATKRVKQETFT
    LTVPIGVRKFCVKVLPRLESRINKAEWSEEQC
    LLITTEQYFTVTNLSHPASSTKVDKKIVPRDC
    GCKPCICTVPEVSSVFIFPPKPKDVLTITLTPKV
    TCVVVDISKDDPEVQFSWFVDDVEVHTAQTQ
    PREEQFNSTFRSVSELPIMHQDWLNGKEFKCR
    VNSAAFPAPIEKTISKTKGRPKAPQVYTIPPPK
    EQMAKDKVSLTCMITDFFPEDITVEWQWNG
    QPAENYKNTQPIMDTDGSYFVYSKLNVQKSN
    WEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK
    25 hIL-27 NP_004834 MRGGRGAPFWLWPLPKLALLPLLWVLFQRT
    Receptor RPQGSAGPLQCYGVGPLGDLNCSWEPLGDLG
    subunit alpha APSELHLQSQKYRSNKTQTVAVAAGRSWVAI
    precursor PREQLTMSDKLLVWGTKAGQPLWPPVFV
    NLETQMKPNAPRLGPDVDFSEDDPLEATVHW
    APPTWPSHKVLICQFHYRRCQEAAWTLLEPE
    LKTIPLTPVEIQDLELATGYKVYGRCRMEKEE
    DLWGEWSPILSFQTPPSAPKDVWVSG
    NLCGTPGGEEPLLLWKAPGPCVQVSYKVWF
    WVGGRELSPEGITCCCSLIPSGAEWARVSAVN
    ATSWEPLTNLSLVCLDSASAPRSVAVSSIAGS
    TELLVTWQPGPGEPLEHVVDWARDGD
    PLEKLNWVRLPPGNLSALLPGNFTVGVPYRIT
    VTAVSASGLASASSVWGFREELAPLVGPTLW
    RLQDAPPGTPAIAWGEVPRHQLRGHLTHYTL
    CAQSGTSPSVCMNVSGNTQSVTLPDL
    PWGPCELWVTASTIAGQGPPGPILRLHLPDNT
    LRWKVLPGILFLWGLFLLGCGLSLATSGRCY
    HLRHKVLPRWVWEKVPDPANSSSGQPHMEQ
    VPEAQPLGDLPILEVEEMEPPPVMESS
    QPAQATAPLDSGYEKHFLPTPEELGLLGPPRP
    QVLA
    26 hIL-13 AAH96140 MVWSINLTAGMYCAALESLINVSGCSAIEKT
    QRMLSGFCPHKVSAGQFSSLHVRDTKIEVAQ
    FVKDLLLHLKKLFREGRFN
    27 hIL-17 AAC50341 MTPGKTSLVSLLLLLSLEAIVKAGITIPRNPGC
    PNSEDKNFPRTVMVNLNIHNRNTNTNPKRSS
    DYYNRSTSPWNLHRNEDPERYPSVIWEAKCR
    HLGCINADGNVDYHMNSVPIQQEIL
    VLRREPPHCPNSFRLEKILVSVGCTCVTPIVHH
    VA
    28 hIL-33 NP_001300974 MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform a  KSQQKAKEVCPMYFMKLRSGLMIKKEACYF
    RRETTKRPSLKTGRKHKRHLVLAACQQQSTV
    ECFAFGISGVQKYTRALHDSSITGISPIT
    EYLASLSTYNDQSITFALEDESYEIYVEDLKK
    DEKKDKVLLSYYESQHPSNESGDGVDGKML
    MVTLSPTKDFWLHANNKEHSVELHKCEKPLP
    DQAFFVLHNMHSNCVSFECKTDPGVFI
    GVKDNHLALIKVDSSENLCTENILFKLSET
    29 hIL-33 NP_001300973 MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform a KSQQKAKEVCPMYFMKLRSGLMIKKEACYF
    RRETTKRPSLKTGRKHKRHLVLAACQQQSTV
    ECFAFGISGVQKYTRALHDSSITGISPIT
    EYLASLSTYNDQSITFALEDESYEIYVEDLKK
    DEKKDKVLLSYYESQHPSNESGDGVDGKML
    MVTLSPTKDFWLHANNKEHSVELHKCEKPLP
    DQAFFVLHNMHSNCVSFECKTDPGVFI
    GVKDNHLALIKVDSSENLCTENILFKLSET
    30 hIL-33 NP_001186569 MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform b KSQQKAKEVCPMYFMKLRSGLMIKKEACYF
    RRETTKRPSLKTGRKHKRHLVLAACQQQSTV
    ECFAFGISGVQKYTRALHDSSITDKVLLS
    YYESQHPSNESGDGVDGKMLMVTLSPTKDF
    WLHANNKEHSVELHKCEKPLPDQAFFVLHN
    MHSNCVSFECKTDPGVFIGVKDNHLALIKVD
    SSENLCTENILFKLSET
    31 hIL-33 NP_001186570 MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform c NKVLLSYYESQHPSNESGDGVDGKMLMVTL
    SPTKDFWLHANNKEHSVELHKCEKPLPDQAF
    FVLHNMHSNCVSFECKTDPGVFIGVKDNH
    LALIKVDSSENLCTENILFKLSET
    32 hIL-33 NP 001300975; MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform d NP_001300976 KSQQKAKEVCPMYFMKLRSGLMIKKEACYF
    RRETTKRPSLKTGRKHKRHLVLAACQQQSTV
    ECFAFGISGVQKYTRALHDSSITEYLASL
    STYNDQSITFALEDESYEIYVEDLKKDEKKDK
    VLLSYYESQHPSNESGDGVDGKMLMVTLSPT
    KDFWLHANNKEHSVELHKCEKPLPDQAFFVL
    HNMHSNCVSFECKTDPGVFIGVKDNH
    LALIKVDSSENLCTENILFKLSET
    33 hIL-33 NP_001300977 MKPKMKYSTNKISTAKWKNTASKALCFKLG
    Isoform e KSQQKAKEVCPMYFMKLRSGLMIKKEACYF
    RRETTKRPSLKTGISPITEYLASLSTYNDQSITF
    ALEDESYEIYVEDLKKDEKKDKVLLS
    YYESQHPSNESGDGVDGKMLMVTLSPTKDF
    WLHANNKEHSVELHKCEKPLPDQAFFVLHN
    MHSNCVSFECKTDPGVFIGVKDNHLALIKVD
    SSENLCTENILFKLSET
    34 hIFN-γ AAB59534 MKYTSYILAFQLCIVLGSLGCYCQDPYVKEA
    ENLKKYFNAGHSDVADNGTLFLGILKNWKEE
    SDRKIMQSQIVSFYFKLFKNFKDDQSIQKSVE
    TIKEDMNVKFFNSNKKKRDDFEKLTN
    YSVTDLNVQRKAIHELIQVMAELSPAAKTGK
    RKRSQMLFRGRRASQ
    35 Flagellin p06179 MAQVINTNSLSLLTQNNLNKSQSALGTAIERL
    SSGLRINSAKDDAAGQAIANRFTANIKGLTQA
    SRNANDGISIAQTTEGALNEINNNLQRVRELA
    VQSANSTNSQSDLDSIQAEITQRLNEIDRVSG
    QTQFNGVKVLAQDNTLTIQVGANDGETIDID
    LKQINSQTLGLDTLNVQQKYKVSDTAATVTG
    YADTTIALDNSTFKASATGLGGTDQKIDGDL
    KFDDTTGKYYAKVTVTGGTGKDGYYEVSVD
    KTNGEVTLAGGATSPLTGGLPATATEDVKNV
    QVANADLTEAKAALTAAGVTGTASVVKMSY
    TDNNGKTIDGGLAVKVGDDYYSATQNKDGSI
    SINTTKYTADDGTSKTALNKLGGADGKTEVV
    SIGGKTYAASKAEGHNFKAQPDLAEAAATTT
    ENPLQKIDAALAQVDTLRSDLGAVQNRFNSA
    ITNLGNTVNNLTSARSRIEDSDYATEVSNMSR
    AQILQQAGTSVLAQANQVPQNVLSLLR
    36 anti-CTLA4 N/A MDMRVPAQLLGLLLLWLRGARCDIVMTQTT
    (CTLA-4 LSLPVSLGDQASISCRSSQSIVHSNGNTYLEW
    antagonist YLQKPGQSPKLLIYKVSNRFSGVPDRFSGSGS
    antibody) GTDFTLKISRVEAEDLGVYYCFQGSHVPYTFG
    GGTKLEIKRADAAPTVSGSGGGSGGGSGGGS
    EAKLQESGPVLVKPGASVKMSCKASGYTFTD
    YYMNWVKQSHGKSLEWIGVINPYNGDTSYN
    QKFKGKATLTVDKSSSTAYMELNSLTSEDSA
    VYYCARYYGSWFAYWGQGTLITVSTEPRGPT
    IKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMIS
    LSPIVTCVVVDVSEDDPDVQISWFVNNVEVH
    TAQTQTHREDYNSTLRVVSALPIQHQDWMSG
    KEFKCKVNNKDLPAPIERTISKPKGSVRAPQV
    YVLPPPEEEMTKKQVTLTCMVTDFMPEDIYV
    EWTNNGKTELNYKNTEPVLDSDGSYFMYSK
    LRVEKKNWVERNSYSCSVVHEGLHNHHTTK
    SFSRTPGK
    37 murine IL-2 DNA TATCACCCTTGCTAATCACTCCTCACAGTGA
    (NM_008366.3) CCTCAAGTCCTGCAGGCATGTACAGCATGC
    AGCTCGCATCCTGTGTCACATTGACACTTGT
    GCTCCTTGTCAACAGCGCACCCACTTCAAG
    CTCCACTTCAAGCTCTACAGCGGAAGCACA
    GCAGCAGCAGCAGCAGCAGCAGCAGCAGC
    AGCAGCACCTGGAGCAGCTGTTGATGGACC
    TACAGGAGCTCCTGAGCAGGATGGAGAATT
    ACAGGAACCTGAAACTCCCCAGGATGCTCA
    CCTTCAAATTTTACTTGCCCAAGCAGGCCA
    CAGAATTGAAAGATCTTCAGTGCCTAGAAG
    ATGAACTTGGACCTCTGCGGCATGTTCTGG
    ATTTGACTCAAAGCAAAAGCTTTCAATTGG
    AAGATGCTGAGAATTTCATCAGCAATATCA
    GAGTAACTGTTGTAAAACTAAAGGGCTCTG
    ACAACACATTTGAGTGCCAATTCGATGATG
    AGTCAGCAACTGTGGTGGACTTTCTGAGGA
    GATGGATAGCCTTCTGTCAAAGCATCATCT
    CAACAAGCCCTCAATAACTATGTACCTCCT
    GCTTACAACACATAAGGCTCTCTATTTATTT
    AAATATTTAACTTTAATTTATTTTTGGATGT
    ATTGTTTACTATCTTTTGTAACTACTAGTCT
    TCAGATGATAAATATGGATCTTTAAAGATT
    CTTTTTGTAAGCCCCAAGGGCTCAAAAATG
    TTTTAAACTATTTATCTGAAATTATTTATTA
    TATTGAATTGTTAAATATCATGTGTAGGTA
    GACTCATTAATAAAAGTATTTAGATGATTC
    AAATATAAATAAGCTCAGATGTCTGTCATT
    TTTAGGACAGCACAAAGTAAGCGCTAAAAT
    AACTTCTCAGTTATTCCTGTGAACTCTATGT
    TAATCAGTGTTTTCAAGAAATAAAGCTCTC
    CTCTAAAAAAAAAAAAAAA
    38 murine IL-2 amino acid MYSMQLASCVTLTLVLLVNSAPTSSSTSSSTA
    CAA25909 EAQQQQQQQQQQQQHLEQLLMDLQELLSRM
    ENYRNLKLPRMLTFKFYLPKQATELKDLQCL
    EDELGPLRHVLDLTQSKSFQLEDAENFISNIRV
    TVVKLKGSDNTFECQFDDESATVVDFLRRWI
    AFCQSIISTSPQ
    39 murine Xc11 DNA AGCCCAGCAAGACCTCAGCCATGAGACTTC
    (GenBank:  TCCTCCTGACTTTCCTGGGAGTCTGCTGCCT
    BC062249.1) CACCCCATGGGTTGTGGAAGGTGTGGGGAC
    TGAAGTCCTAGAAGAGAGTAGCTGTGTGAA
    CTTACAAACCCAGCGGCTGCCAGTTCAAAA
    AATCAAGACCTATATCATCTGGGAGGGGGC
    CATGAGAGCTGTAATTTTTGTCACCAAACG
    AGGACTAAAAATTTGTGCTGATCCAGAAGC
    CAAATGGGTGAAAGCAGCGATCAAGACTGT
    GGATGGCAGGGCCAGTACCAGAAAGAACA
    TGGCTGAAACTGTTCCCACAGGAGCCCAGA
    GGTCCACCAGCACAGCGATAACCCTGACTG
    GGTAACAGCCTCCAGGACAATGTTTCCTCA
    CTCGTTAAGCAGCTCATCTCAGTTCCCAAA
    CCCATTGCACAAATACTTATTTTTATTTTTA
    ACGACATTCACATTCATTTCAAATGTTATAA
    GTAATAAATATTTATTATTGATGAAAAAAA
    AAAAAAAAAAAAA
    40 murine Xc11 amino acid MRLLLLTFLGVCCLTPWVVEGVGTEVLEESS
    (GenBank: CVNLQTQRLPVQKIKTYIIWEGAMRAVIFVTK
    BC062249.1) RGLKICADPEAKWVKAAIKTVDGRASTRKN
    MAETVPTGAQRSTSTAITLTG
    41 Murine 4- (DNA) GAGACGTGCACTGACCGACCGTGGTAATGG
    1BBL GenBank: ACCAGCACACACTTGATGTGGAGGATACCG
    (TNFSF9) BC138767.1 CGGATGCCAGACATCCAGCAGGTACTTCGT
    GCCCCTCGGATGCGGCGCTCCTCAGAGATA
    CCGGGCTCCTCGCGGACGCTGCGCTCCTCT
    CAGATACTGTGCGCCCCACAAATGCCGCGC
    TCCCCACGGATGCTGCCTACCCTGCGGTTA
    ATGTTCGGGATCGCGAGGCCGCGTGGCCGC
    CTGCACTGAACTTCTGTTCCCGCCACCCAA
    AGCTCTATGGCCTAGTCGCTTTGGTTTTGCT
    GCTTCTGATCGCCGCCTGTGTTCCTATCTTC
    ACCCGCACCGAGCCTCGGCCAGCGCTCACA
    ATCACCACCTCGCCCAACCTGGGTACCCGA
    GAGAATAATGCAGACCAGGTCACCCCTGTT
    TCCCACATTGGCTGCCCCAACACTACACAA
    CAGGGCTCTCCTGTGTTCGCCAAGCTACTG
    GCTAAAAACCAAGCATCGTTGTGCAATACA
    ACTCTGAACTGGCACAGCCAAGATGGAGCT
    GGGAGCTCATACCTATCTCAAGGTCTGAGG
    TACGAAGAAGACAAAAAGGAGTTGGTGGT
    AGACAGTCCCGGGCTCTACTACGTATTTTTG
    GAACTGAAGCTCAGTCCAACATTCACAAAC
    ACAGGCCACAAGGTGCAGGGCTGGGTCTCT
    CTTGTTTTGCAAGCAAAGCCTCAGGTAGAT
    GACTTTGACAACTTGGCCCTGACAGTGGAA
    CTGTTCCCTTGCTCCATGGAGAACAAGTTA
    GTGGACCGTTCCTGGAGTCAACTGTTGCTC
    CTGAAGGCTGGCCACCGCCTCAGTGTGGGT
    CTGAGGGCTTATCTGCATGGAGCCCAGGAT
    GCATACAGAGACTGGGAGCTGTCTTATCCC
    AACACCACCAGCTTTGGACTCTTTCTTGTGA
    AACCCGACAACCCATGGGAATGAGAACTAT
    CCTTCTTGTGACTCCTAGTTGCTAAGTCCTC
    AAGCTGCTATGTTTTATGGGGTCTGAGCAG
    GGGT
    42 Murine 4- (protein)  MDQHTLDVEDTADARHPAGTSCPSDAALLR
    1BBL GenBank: DTGLLADAALLSDTVRPTNAALPTDAAYPAV
    (TNFSF9) BC138767.1 NVRDREAAWPPALNFCSRHPKLYGLVALVLL
    LLIAACVPIFTRTEPRPALTITTSPNLGTRENNA
    DQVTPVSHIGCPNTTQQGSPVFAKLLAKNQA
    SLCNTTLNWHSQDGAGSSYLSQGLRYEEDKK
    ELVVDSPGLYYVFLELKLSPTFTNTGHKVQG
    WVSLVLQAKPQVDDFDNLALTVELFPCSMEN
    KLVDRSWSQLLLLKAGHRLSVGLRAYLHGA
    QDAYRDWELSYPNTTSFGLFLVKPDNPWE
    43 Mouse 4- with human  MDMRVPAQLLGLLLLWLRGARCRMKQIEDK
    1BBL IGKV1-39 IEEILSKIYHIENEIARIKKLIGERGGGSGGGSG
    trimeric kappa LC GGSRTEPRPALTITTSPNLGTRENNADQVTPV
    version signal SHIGCPNTTQQGSPVFAKLLAKNQASLCNTTL
    sequence, NWHSQDGAGSSYLSQGLRYEEDKKELVVDS
    trimerization PGLYYVFLELKLSPTFTNTGHKVQGWVSLVL
    domain QAKPQVDDFDNLALTVELFPCSMENKLVDRS
    from yeast, WSQLLLLKAGHRLSVGLRAYLHGAQDAYRD
    and Linker WELSYPNTTSFGLFLVKPDNPWE**
    1, followed
    by 4-1BBL
    (TNFSF9,
    Acc#
    P41274)
    residues
    104-309
    (minus ECD
    and
    transmembrane 
    domain)
    44 murine 4- residues RTEPRPALTITTSPNLGTRENNADQVTPVSHIG
    1BBL 104-209 CPNTTQQGSPVFAKLLAKNQASLCNTTLNWH
    (TNFSF9, (minus ECD  SQDGAGSSYLSQGLRYEEDKKELVVDSPGLY
    p41274) and YVFLELKLSPTFTNTGHKVQGWVSLVLQAKP
    transmem- QVDDFDNLALTVELFPCSMENKLVDRSWSQL
    brane LLLKAGHRLSVGLRAYLHGAQDAYRDWELS
    domain) YPNTTSFGLFLVKPDNPWE**
    45 Linker 1 15-mer GGGGSGGGGSGGGGS
    46 TRZ202 AA MCHQQLVISWFSLVFLASPLVAIWELKKDVY
    (hIL-12 Sequence VVELDWYPDAPGEMVVLTCDTPEEDGITWTL
    Insert) DQSSEVLGSGKTLTIQVKEFGDAGQYTCHKG
    GEVLSHSLLLLHKKEDGIWSTDILKDQKEPKN
    KTFLRCEAKNYSGRFTCWWLTTISTDLTFSVK
    SSRGSSDPQGVTCGAATLSAERVRGDNKEYE
    YSVECQEDSACPAAEESLPIEVMVDAVHKLK
    YENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQV
    EVSWEYPDTWSTPHSYFSLTFCVQVQGKSKR
    EKKDRVFTDKTSATVICRKNASISVRAQDRY
    YSSSWSEWASVPCSGGGGSGGGGSGGGGSR
    NLPVATPDPGMFPCLHHSQNLLRAVSNMLQK
    ARQTLEFYPCTSEEIDHEDITKDKTSTVEACLP
    LELTKNESCLNSRETSFITNGSCLASRKTSFM
    MALCLSSIYEDLKMYQVEFKTMNAKLLMDP
    KRQIFLDQNMLAVIDELMQALNFNSETVPQK
    SSLEEPDFYKTKIKLCILLHAFRIRAVTIDRVM
    SYLNAS**
    47 hIL-12 p40β AF180563 MCHQQLVISWFSLVFLASPLVAIWELKKDVY
    VVELDWYPDAPGEMVVLTCDTPEEDGITWTL
    DQSSEVLGSGKTLTIQVKEFGDAGQYTCHKG
    GEVLSHSLLLLHKKEDGIWSTDILKDQKEPKN
    KTFLRCEAKNYSGRFTCWWLTTISTDLTFSVK
    SSRGSSDPQGVTCGAATLSAERVRGDNKEYE
    YSVECQEDSACPAAEESLPIEVMVDAVHKLK
    YENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQV
    EVSWEYPDTWSTPHSYFSLTFCVQVQGKSKR
    EKKDRVFTDKTSATVICRKNASISVRAQDRY
    YSSSWSEWASVPCS
    48 hIL-12 p35α AA RNLPVATPDPGMFPCLHHSQNLLRAVSNMLQ
    Sequence of KARQTLEFYPCTSEEIDHEDITKDKTSTVEAC
    Accession LPLELTKNESCLNSRETSFITNGSCLASRKTSF
    #AF101062.1 MMALCLSSIYEDLKMYQVEFKTMNAKLLMD
    (native PKRQIFLDQNMLAVIDELMQALNFNSETVPQ
    start codon  KSSLEEPDFYKTKIKLCILLHAFRIRAVTIDRV
    and signal MSYLNAS**
    sequence
    removed)
    SEQ (TRZ015) AA GTCGACGCCACCMDMRVPAQLLGLLLLWL
    ID hGITRL Sequence, RGARCRMKQIEDKIEEILSKIYHIENEIARIKKL
    NOS insert, including IGERGGGSGGGSGGGSETAKEPCMAKFGPLP
    49 Trimeric Kozak SKWQMASSEPPCVNKVSDWKLEILQNGLYLI
    and version sequences, YGQVAPNANYNDVAPFEVRLYKNKDMIQTL
    107 RE sites, TNKSKIQNVGGTYELHVGDTIDLIFNSEHQVL
    and stop KNNTYWGIILLANPQFIS** CTCGAG
    codons
    50 hIGKV1-39 AA MDMRVPAQLLGLLLLWLRGARC
    kappa LC Sequence
    signal
    sequence
    51 Trimerization AA RMKQIEDKIEEILSKIYHIENEIARIKKLIGER
    domain Sequence
    (yeast)
    52 GITRL minus AA ETAKEPCMAKFGPLPSKWQMASSEPPCVNKV
    ECD and Sequence SDWKLEILQNGLYLIYGQVAPNANYNDVAPF
    transmembrane EVRLYKNKDMIQTLTNKSKIQNVGGTYELHV
    domains GDTIDLIFNSEHQVLKNNTYWGIILLANPQFIS**
    53 Linker 2 12-mer GGGSGGGSGGGS
    54 TRZ006 : hIL- hIL-15Rα- MAPRRARGCRTLGLPALLLLLLLRPPATRGIT
    15 hybrid Linker-hIL- CPPPMSVEHADIWVKSYSLYSRERYICNSGFK
    15 RKAGTSSLTECVLNKATNVAHWTTPSLKCIR
    DPALVHQRPAPPSTVTTASGGSGGGGSGGGS
    GGGGSNWVNVISDLKKIEDLIQSMHIDATLYT
    ESDVHPSCKVTAMKCFLLELQVISLESGDASI
    HDTVENLIILANNSLSSNGNVTESGCKECEEL
    EEKNIKEFLQSFVHIVQMFINTS**
    55 hIL-15Rα AA MAPRRARGCRTLGLPALLLLLLLRPPATRG
    endogenous Sequence
    signal peptide
    56 Sushi domain AA MSVEHADIWVKSYSLYSRERYICNSGFKRKA
    from IL-15Rα Sequence GTSSLTECVLNKATNVAHWTTPSLKCIRDPAL
    VHQRPAPPSTVTTA
    57 mature hIL-15 AA NWVNVISDLKKIEDLIQSMHIDATLYTESDVH
    Sequence PSCKVTAMKCFLLELQVISLESGDASIHDTVE
    NLIILANNSLSSNGNVTESGCKECEELEEKNIK
    EFLQSFVHIVQMFINTS
    58 TRZ201: Orientation MDMRVPAQLLGLLLLWLRGARCEIVLTQSPA
    hαCTLA4 2 ANT2054 TLSLSPGERATLSCSASSSISYMHWFQQRPGQ
    scFv-IgG1 SPRRWIYDTSKLASGVPARFSGSGSGTDYTLT
    Fc ISSLEPEDFATYYCHQRTSYPLTFGQGTKLEIK
    GGGGSGGGGSGGGGSQVQLVQSGAELKKPG
    ASVKVSCKASGYTFTSYWINWIRQAPGQGLE
    WIGRIAPGSGTTYYNEVFKGRVTITVDKSTST
    AYMELSSLRSEDTAVYFCARGDYGSYWGQG
    TLVTVSSDKTHTCPPCPAPELLGGPSVFLFPPK
    PKDTLMISRTPEVTCVVVDVSHEDPEVKFNW
    YVDGVEVHNAKTKPREEQYNSTYRVVSVLT
    VLHQDWLNGKEYKCKVSNKALPAPIEKTISK
    AKGQPREPQVYTLPPSREEMTKNQVSLTCLV
    KGFYPSDIAVEWESNGQPENNYKTTPPVLDS
    DGSFFLYSKLTVDKSRWQQGNVFSCSVMHE
    ALHNHYTQKSLSLSPGK**
    59 Kappa AA EIVLTQSPATLSLSPGERATLSCSASSSISYMH
    variable Sequence WFQQRPGQSPRRWIYDTSKLASGVPARFSGS
    GSGTDYTLTISSLEPEDFATYYCHQRTSYPLTF
    GQGTKLEIK
    60 Heavy AA QVQLVQSGAELKKPGASVKVSCKASGYTFTS
    variable Sequence YWINWIRQAPGQGLEWIGRIAPGSGTTYYNE
    VFKGRVTITVDKSTSTAYMELSSLRSEDTAVY
    FCARGDYGSYWGQGTLVTVSS
    61 human Fc AA DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI
    IgG1 Sequence SRTPEVTCVVVDVSHEDPEVKFNWYVDGVE
    VHNAKTKPREEQYNSTYRVVSVLTVLHQDW
    LNGKEYKCKVSNKALPAPIEKTISKAKGQPRE
    PQVYTLPPSREEMTKNQVSLTCLVKGFYPSDI
    AVEWESNGQPENNYKTTPPVLDSDGSFFLYS
    KLTVDKSRWQQGNVFSCSVMHEALHNHYTQ
    KSLSLSPGK**
    62 TRZ106 hIL-15 MAPRRARGCRTLGLPALLLLLLLRPPATRGIT
    hybrid (hIL- CPPPMSVEHADIWVKSYSLYSRERYICNSGFK
    15Rα- RKAGTSSLTECVLNKATNVAHWTTPSLKCIR
    Linker 2- DPALVHQRPAPPSTVTTASGGSGGGGSGGGS
    hIL-15) GGGGSNWVNVISDLKKIEDLIQSMHIDATLYT
    (hIL-15Rα ESDVHPSCKVTAMKCFLLELQVISLESGDASI
    endogenous HDTVENLIILANNSLSSNGNVTESGCKECEEL
    signal EEKNIKEFLQSFVHIVQMFINTS**
    peptide,
    Sushi
    domain
    from IL-
    15Rα,
    linker,
    mature hIL-
    15)
    63 hIL-15Ra MAPRRARGCRTLGLPALLLLLLLRPPATRG
    endogenous
    signal peptide
    64 mature hIL-15 NWVNVISDLKKIEDLIQSMHIDATLYTESDVH
    PSCKVTAMKCFLLELQVISLESGDASIHDTVE
    NLIILANNSLSSNGNVTESGCKECEELEEKNIK
    EFLQSFVHIVQMFINTS
    65 TRZ108: full length MIETYNQTSPRSAATGLPISMKIFMYLLTVFLI
    hCD40 monomeric TQMIGSALFAVYLHRRLDKIEDERNLHEDFVF
    version, MKTIQRCNTGERSLSLLNCEEIKSQFEGFVKDI
    membrane MLNKEETKKENSFEMQKGDQNPQIAAHVISE
    bound ASSKTTSVLQWAEKGYYTMSNNLVTLENGK
    QLTVKRQGLYYIYAQVTFCSNREASSQAPFIA
    SLCLKSPGRFERILLRAANTHSSAKPCGQQSIH
    LGGVFELQPGASVFVNVTDPSQVSHGTGFTSF
    GLLKL**
    66 TRZ111: full length MERVQPLEENVGNAARPRFERNKLLLVASVI
    hOX40L monomeric QGLGLLLCFTYICLHFSALQVSHRYPRIQSIKV
    variant 1 version, QFTEYKKEKGFILTSQKEDEIMKVQNNSVIIN
    membrane CDGFYLISLKGYFSQEVNISLHYQKDEEPLFQ
    bound LKKVRSVNSLMVASLTYKDKVYLNVTTDNT
    SLDDFHVNGGELILIHQNPGEFCVL**
    67 TRZ114: hIL- Extracellular MLPCLVVLLAALLSLRLGSDAHGTELPSPPSV
    10RTrap domain of WFEAEFFHHILHWTPIPNQSESTCYEVALLRY
    Version2 hIL-10Rα GIESWNSISNCSQTLSYDLTAVTLDLYHSNGY
    with RARVRAVDGSRHSNWTVTNTRFSVDEVTLT
    endogenous  VGSVNLEIHNGFILGKIQLPRPKMAPANDTYE
    signal SIFSHFREYEIAIRKVPGNFTFTHKKVKHENFS
    sequence, LLTSGEVGEFCVQVKPSVASRSNKGMWSKEE
    without stop CISLTRQYFTVTNDKTHTCPPCPAPELLGGPS
    and human VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDP
    Fc IgG1 EVKFNWYVDGVEVHNAKTKPREEQYNSTYR
    VVSVLTVLHQDWLNGKEYKCKVSNKALPAPI
    EKTISKAKGQPREPQVYTLPPSREEMTKNQVS
    LTCLVKGFYPSDIAVEWESNGQPENNYKTTPP
    VLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV
    MHEALHNHYTQKSLSLSPGK**
    68 Extracellular HGTELPSPPSVWFEAEFFHHILHWTPIPNQSES
    domain of TCYEVALLRYGIESWNSISNCSQTLSYDLTAV
    hIL-10Rα TLDLYHSNGYRARVRAVDGSRHSNWTVTNT
    RFSVDEVTLTVGSVNLEIHNGFILGKIQLPRPK
    MAPANDTYESIFSHFREYEIAIRKVPGNFTFTH
    KKVKHENFSLLTSGEVGEFCVQVKPSVASRS
    NKGMWSKEECISLTRQYFTVTN
    69 endogenous MLPCLVVLLAALLSLRLGSDA
    signal
    sequence in
    TRZ114
    70 human Fc DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI
    IgG1 SRTPEVTCVVVDVSHEDPEVKFNWYVDGVE
    VHNAKTKPREEQYNSTYRVVSVLTVLHQDW
    LNGKEYKCKVSNKALPAPIEKTISKAKGQPRE
    PQVYTLPPSREEMTKNQVSLTCLVKGFYPSDI
    AVEWESNGQPENNYKTTPPVLDSDGSFFLYS
    KLTVDKSRWQQGNVFSCSVMHEALHNHYTQ
    KSLSLSPGK**
    71 TRZ116: Trimeric MDMRVPAQLLGLLLLWLRGARCRMKQIEDK
    hOX40L version IEEILSKIYHIENEIARIKKLIGERGGGSGGGSG
    w/human GGSETAKEPCMAKFGPLPSKWQMASSEPPCV
    IGKV1-39 NKVSDWKLEILQNGLYLIYGQVAPNANYND
    kappa LC VAPFEVRLYKNKDMIQTLTNKSKIQNVGGTY
    signal ELHVGDTIDLIFNSEHQVLKNNTYWGIILLAN
    sequence, PQFIS**
    trimerization
    domain
    from yeast,
    and Linker
    1, followed
    by OX4OL
    minus ECD
    and
    transmem-
    brane
    domains
    72 hOX4OL QVSHRYPRIQSIKVQFTEYKKEKGFILTSQKE
    minus ECD DEIMKVQNNSVIINCDGFYLISLKGYFSQEVNI
    and SLHYQKDEEPLFQLKKVRSVNSLMVASLTYK
    transmembrane DKVYLNVTTDNTSLDDFHVNGGELILIHQNP
    domains GEFCVL**
    73 TRZ117: Trimeric MDMRVPAQLLGLLLLWLRGARCRMKQIEDK
    hCD4OL version IEEILSKIYHIENEIARIKKLIGERGGGSGGGSG
    w/human GGSMQKGDQNPQIAAHVISEASSKTTSVLQW
    IGKV1-39 AEKGYYTMSNNLVTLENGKQLTVKRQGLYY
    kappa LC IYAQVTFCSNREASSQAPFIASLCLKSPGREER
    signal ILLRAANTHSSAKPCGQQSIHLGGVFELQPGA
    sequence, SVFVNVTDPSQVSHGTGFTSFGLLKL**
    trimerization
    domain
    from yeast,
    and Linker
    1, followed
    by CD40L
    minus ECD
    and
    transmem-
    brane
    domains
    76 hCD4OL MQKGDQNPQIAAHVISEASSKTTSVLQWAEK
    minus ECD GYYTMSNNLVTLENGKQLTVKRQGLYYIYA
    and QVTFCSNREASSQAPFIASLCLKSPGRFERILL
    transmem- RAANTHSSAKPCGQQSIHLGGVFELQPGASVF
    brane VNVTDPSQVSHGTGFTSFGLLKL**
    domains
    77 TRZ307: hIL- MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKD
    7 variant 1 GKQYESVLMVSIDQLLDSMKEIGSNCLNNEF
    NFFKRHICDANKEGMFLFRAARKLRQFLKMN
    STGDFDLHLLKVSEGTTILLNCTGQVKGRKPA
    ALGEAQPTKSLEENKSLKEQKKLNDLCFLKR
    LLQEIKTCWNKILMGTKEH**
    106 hIL-7 MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKD
    GKQYESVLMVSIDQLLDSMKEIGSNCLNNEF
    NFFKRHICDANKEGMFLFRAARKLRQFLKMN
    STGDFDLHLLKVSEGTTILLNCTGQVKGRKPA
    ALGEAQPTKSLEENKSLKEQKKLNDLCFLKR
    LLQEIKTCWNKILMGTKEH**
    78 TRZ304: MWLQSLLLLGTVACSISAPARSPSPSTQPWEH
    hGM-CSF VNAIQEARRLLNLSRDTAAEMNETVEVISEM
    FDLQEPTCLQTRLELYKQGLRGSLTKLKGPLT
    MMASHYKQHCPPTPETSCATQIITFESFKENL
    KDFLLVIPFDCWEPVQE**
    79 hGM-CSF MWLQSLLLLGTVACSISAPARSPSPSTQPWEH
    VNAIQEARRLLNLSRDTAAEMNETVEVISEM
    FDLQEPTCLQTRLELYKQGLRGSLTKLKGPLT
    MMASHYKQHCPPTPETSCATQIITFESFKENL
    KDFLLVIPFDCWEPVQE**
    80 TRZ309: (residue in MPEEGSGCSVRRRPYGCVLRAALVPLVAGLV
    hCD70 bold to be  ICLVVCIQRFAQAQQQLPLESLGWDVAELQL
    removed) NHTGPQQDPRLYWQGGPALGRSFLHGPELDK
    GQLRIHRDGIYMVHIQVTLAICSSTTASRHHP
    TTLAVGICSPASRSISLLRLSFHQGCTIASQRLT
    PLARGDTLCTNLTGTLLPSRNTDETFFGVQW
    VRP**
    81 hCD70 (residue in MPEEGSGCSVRRRPYGCVLRAALVPLVAGLV
    bold to be  ICLVVCIQRFAQAQQQLPLESLGWDVAELQL
    modified to NHTGPQQDPRLYWQGGPALGRSFLHGPELDK
    avoid GQLRIHRDGIYMVHIQVTLAICSSTTASRHHP
    introducing TTLAVGICSPASRSISLLRLSFHQGCTIASQRLT
    new Xho I PLARGDTLCTNLTGTLLPSRNTDETFFGVQW
    site) VRP**
    82 E3 14.7k Region ATGACTGACACCCTAGATCTAGAAATGGA
    overlapping CGGAATTATTACAGAGCAGCGCCTGCTAGA
    with E3 AAGACGCAGGGCAGCGGCCGAGCAACAGC
    14.5k ORF GCATGAATCAAGAGCTCCAAGACATGGTTA
    in bold ACTTGCACCAGTGCAAAAGGGGTATCTTTT
    GTCTGGTAAAGCAGGCCAAAGTCACCTACG
    ACAGTAATACCACCGGACACCGCCTTAGCT
    ACAAGTTGCCAACCAAGCGTCAGAAATTGG
    TGGTCATGGTGGGAGAAAAGCCCATTACCA
    TAACTCAGCACTCGGTAGAAACCGAAGGCT
    GCATTCACTCACCTTGTCAAGGACCTGAGG
    ATCTCTGCACCCTTATTAAGACCCTGTGCGG
    TCTCAAAGATCTTATTCCCTTTAACTAA
    83 E3 14.5k Region ATGAAATTTACTGTGACTTTTCTGCTGATTA
    overlapping TTTGCACCCTATCTGCGTTTTGTTCCCCGAC
    with E3 CTCCAAGCCTCAAAGACATATATCATGCAG
    14.7k in ATTCACTCGTATATGGAATATTCCAAGTTGC
    bold TACAATGAAAAAAGCGATCTTTCCGAAGCC
    TGGTTATATGCAATCATCTCTGTTATGGTGT
    TCTGCAGTACCATCTTAGCCCTAGCTATATA
    TCCCTACCTTGACATTGGCTGGAACGCAAT
    AGATGCCATGAACCACCCAACTTTCCCCGC
    GCCCGCTATGCTTCCACTGCAACAAGTTGTT
    GCCGGCGGCTTTGTCCCAGCCAATCAGCCT
    CGCCCACCTTCTCCCACCCCCACTGAAATC
    AGCTACTTTAATCTAACAGGAGGAGATGAC
    TGA
    84 E3 10.4K ATGATTCCTCGAGTTTTTATATTACTGACCC
    TTGTTGCGCTTTTTTTGTGCGTGCTCCACAT
    TGGCTGCGGTTTCTCACATCGAAGTAGACT
    GCATTCCAGCCTTCACAGTCTATTTGCTTTA
    CGGATTTGTCACCCTCACGCTCATCTGCAGC
    CTCATCACTGTGGTCATCGCCTTTATCCAGT
    GCATTGA
    85 E3 10.5k ATGACCAACACAACCAACGCGGCCGCCGCT
    (ADP) ACCGGACTTACATCTACCACAAATACACCC
    CAAGTTTCTGCCTTTGTCAATAACTGGGATA
    ACTTGGGCATGTGGTGGTTCTCCATAGCGC
    TTATGTTTGTATGCCTTATTATTATGTGGCT
    CATCTGCTGCCTAAAGCGCAAACGCGCCCG
    ACCACCCATCTATAGTCCCATCATTGTGCTA
    CACCCAAACAATGATGGAATCCATAGATTG
    GACGGACTGAAACACATGTTCTTTTCTCTTA
    CAGTATGA
    86 TRZ000 Region ATGATTAGGTACATAATCCTAGGTTTACTC
    intact E3 overlapping ACCCTTGCGTCAGCCCACGGTACCACCCAA
    gp19k ORF with E3 AAGGTGGATTTTAAGGAGCCAGCCTGTAAT
    gp19k ORF GTTACATTCGCAGCTGAAGCTAATGAGTGC
    in bold ACCACTCTTATAAAATGCACCACAGAACAT
    GAAAAGCTGCTTATTCGCCACAAAAACAAA
    ATTGGCAAGTATGCTGTTTATGCTATTTGGC
    AGCCAGGTGACACTACAGAGTATAATGTTA
    CAGTTTTCCAGGGTAAAAGTCATAAAACTT
    TTATGTATACTTTTCCATTTTATGAAATGTG
    CGACATTACCATGTACATGAGCAAACAGTA
    TAAGTTGTGGCCCCCACAAAATTGTGTGGA
    AAACACTGGCACTTTCTGCTGCACTGCTAT
    GCTAATTACAGTGCTCGCTTTGGTCTGTACC
    CTACTCTATATTAAATACAAAAGCAGACGC
    AGCTTTATTGAGGAAAAGAAAATGCCTTAA
    87 TRZ000 Region ATGAACAATTCAAGCAACTCTACGGGCTAT
    intact E3 7.1K overlapping TCTAATTCAGGTTTCTCTAGAATCGGGGTTG
    with E3 GGGTTATTCTCTGTCTTGTGATTCTCTTTATT
    gp19k ORF  CTTATACTAACGCTTCTCTGCCTAAGGCTCG
    in bold CCGCCTGCTGTGTGCACATTTGCATTTATTG
    TCAGCTTTTTAAACGCTGGGGTCGCCACCC
    AAGATGA
    88 TRZ200 ATGTTAAGTGGAGAGGCAGAGCAACTGCGC
    intact E3 CTGAAACACCTGGTCCACTGTCGCCGCCAC
    12.5k AAGTGCTTTGCCCGCGACTCCGGTGAGTTTT
    GCTACTTTGAATTGCCCGAGGATCATATCG
    AGGGCCCGGCGCACGGCGTCCGGCTTACCG
    CCCAGGGAGAGCTTGCCCGTAGCCTGATTC
    GGGAGTTTACCCAGCGCCCCCTGCTAGTTG
    AGCGGGACAGGGGACCCTGTGTTCTCACTG
    TGATTTGCAACTGTCCTAACCCTGGATTACA
    TCAAGATCTTTGTTGCCATCTCTGTGCTGAG
    TATAATAAATACAGAAATTAA
    89 TRZ000 CACCCTAGATCTAGAAATGGACGGAATTAT
    intact E3 TACAGAGCAGCGCCTGCTAGAAAGACGCA
    14.7k ORF GGGCAGCGGCCGAGCAACAGCGCATGAAT
    CAAGAGCTCCAAGACATGGTTAACTTGCAC
    CAGTGCAAAAGGGGTATCTTTTGTCTGGTA
    AAGCAGGCCAAAGTCACCTACGACAGTAAT
    ACCACCGGACACCGCCTTAGCTACAAGTTG
    CCAACCAAGCGTCAGAAATTGGTGGTCATG
    GTGGGAGAAAAGCCCATTACCATAACTCAG
    CACTCGGTAGAAACCGAAGGCTGCATTCAC
    TCACCTTGTCAAGGACCTGAGGATCTCTGC
    ACCCTTATTAAGACCCTGTGCGGTCTCAAA
    GATCTTATTCCCTTTAACTAA
    90 TRZ200 E3 AGACGCCCAGGCCGAAGTTCAGATGACTAA
    region (not CTCAGGGGCGCAGCTTGCGGGCGGCTTTCG
    full E3-this is TCACAGGGTGCGGTCGCCCGGGCAGGGTAT
    deleted AACTCACCTGACAATCAGAGGGCGAGGTAT
    version) TCAGCTCAACGACGAGTCGGTGAGCTCCTC
    GCTTGGTCTCCGTCCGGACGGGACATTTCA
    GATCGGCGGCGCCGGCCGCTCTTCATTCAC
    GCCTCGTCAGGCAATCCTAACTCTGCAGAC
    CTCGTCCTCTGAGCCGCGCTCTGGAGGCAT
    TGGAACTCTGCAATTTATTGAGGAGTTTGT
    GCCATCGGTCTACTTTAACCCCTTCTCGGGA
    CCTCCCGGCCACTATCCGGATCAATTTATTC
    CTAACTTTGACGCGGTAAAGGACTCGGCGG
    ACGGCTACGACTGAATGTTAAGTGGAGAGG
    CAGAGCAACTGCGCCTGAAACACCTGGTCC
    ACTGTCGCCGCCACAAGTGCTTTGCCCGCG
    ACTCCGGTGAGTTTTGCTACTTTGAATTGCC
    CGAGGATCATATCGAGGGCCCGGCGCACGG
    CGTCCGGCTTACCGCCCAGGGAGAGCTTGC
    CCGTAGCCTGATTCGGGAGTTTACCCAGCG
    CCCCCTGCTAGTTGAGCGGGACAGGGGACC
    CTGTGTTCTCACTGTGATTTGCAACTGTCCT
    AACCCTGGATTACATCAAGATCCTCTAGTT
    AATGTCAGGTCGCCTAAGTCGATTAACTAG
    AGTACCCGGGGATCTTATTCCCTTTAACTAA
    91 TRZ200 GATCTTATTCCCTTTAACTAA
    deleted E3
    14.7k partial
    ORF
    92 TRZ200 ATGTTAAGTGGAGAGGCAGAGCAACTGCGC
    deleted E3 CTGAAACACCTGGTCCACTGTCGCCGCCAC
    12.5K partial AAGTGCTTTGCCCGCGACTCCGGTGAGTTTT
    ORF GCTACTTTGAATTGCCCGAGGATCATATCG
    AGGGCCCGGCGCACGGCGTCCGGCTTACCG
    CCCAGGGAGAGCTTGCCCGTAGCCTGATTC
    GGGAGTTTACCCAGCGCCCCCTGCTAGTTG
    AGCGGGACAGGGGACCCTGTGTTCTCACTG
    TGATTTGCAACTGTCCTAACCCTGGATTACA
    TCAAGAT
    93 E3 Promoter AGACGCCCAGGCCGAAGTTCAGATGACTA
    (E3 deleted or
    E3 intact)
    94 Full TRZ200 TTTTGGATTGAAGCCAATATGATAATGAGG
    virus GGGTGGAGTTTGTGACGTGGCGCGGGGCGT
    sequence GGGAACGGGGCGGGTGACGTAGTAGTGTG
    beginning at GCGGAAGTGTGATGTTGCAAGTGTGGCGGA
    5′ ITR ACACATGTAAGCGACGGATGTGGCAAAAGT
    through 3′ GACGTTTTTGGTGTGCGCCGGTGTTTTGGGC
    ITR GTAACCGAGTAAGATTTGGCCATTTTCGCG
    GGAAAACTGAATAAGAGGAAGTGAAATCT
    GAATAATTTTGTGTTACTCATAGCGCGTAAT
    ATTTGTCTAGGGCCGCGGGGACTTTGACCG
    TTTACGTGGAGACTCGCCCAGGTGTTTTTCT
    CAGGTGTTTTCCGCGTTCCGGGTCAAAGTT
    GGCGTTTTATTATTATAGTCAGCTGACGTGT
    AGTGTATTTATACCCGGTGAGTTCCTCAAG
    AGGCCACTCTTGAGTGCCAGCGAGTAGAGT
    TTTCTCCTCCGAGCCGCTCCGACACCGGGA
    CTGAAAATGAGACATATTATCTGCCACGGA
    GGTGTTATTACCGAAGAAATGGCCGCCAGT
    CTTTTGGACCAGCTGATCGAAGAGGTACTG
    GCTGATAATCTTCCACCTCCTAGCCATTTTG
    AACCACCTACCCTTCACGAACTGTATGATTT
    AGACGTGACGGCCCCCGAAGATCCCAACGA
    GGAGGCGGTTTCGCAGATTTTTCCCGACTCT
    GTAATGTTGGCGGTGCAGGAAGGGATTGAC
    TTACTCACTTTTCCGCCGGCGCCCGGTTCTC
    CGGAGCCGCCTCACCTTTCCCGGCAGCCCG
    AGCAGCCGGAGCAGAGAGCCTTGGGTCCG
    GTTTCTATGCCAAACCTTGTACCGGAGGTG
    ATCGATCTTACCTGCCACGAGGCTGGCTTTC
    CACCCAGTGACGACGAGGATGAAGAGGGT
    GAGGAGTTTGTGTTAGATTATGTGGAGCAC
    CCCGGGCACGGTTGCAGGTCTTGTCATTAT
    CACCGGAGGAATACGGGGGACCCAGATATT
    ATGTGTTCGCTTTGCTATATGAGGACCTGTG
    GCATGTTTGTCTACAGTAAGTGAAAATTAT
    GGGCAGTGGGTGATAGAGTGGTGGGTTTGG
    TGTGGTAATTTTTTTTTTAATTTTTACAGTTT
    TGTGGTTTAAAGAATTTTGTATTGTGATTTT
    TTTAAAAGGTCCTGTGTCTGAACCTGAGCC
    TGAGCCCGAGCCAGAACCGGAGCCTGCAA
    GACCTACCCGCCGTCCTAAAATGGCGCCTG
    CTATCCTGAGACGCCCGACATCACCTGTGT
    CTAGAGAATGCAATAGTAGTACGGATAGCT
    GTGACTCCGGTCCTTCTAACACACCTCCTGA
    GATACACCCGGTGGTCCCGCTGTGCCCCAT
    TAAACCAGTTGCCGTGAGAGTTGGTGGGCG
    TCGCCAGGCTGTGGAATGTATCGAGGACTT
    GCTTAACGAGCCTGGGCAACCTTTGGACTT
    GAGCTGTAAACGCCCCAGGCCATAAGGTGT
    AAACCTGTGATTGCGTGTGTGGTTAACGCC
    TTTGTTTGCTGAATGAGTTGATGTAAGTTTA
    ATAAAGGGTGAGATAATGTTTAACTTGCAT
    GGCGTGTTAAATGGGGCGGGGCTTAAAGGG
    TATATAATGCGCCGTGGGCTAATCTTGGTT
    ACATCTGACCTCGTCGACGCTTGGGAGTGT
    TTGGAAGATTTTTCTGCTGTGCGTAACTTGC
    TGGAACAGAGCTCTAACAGTACCTCTTGGT
    TTTGGAGGTTTCTGTGGGGCTCATCCCAGG
    CAAAGTTAGTCTGCAGAATTAAGGAGGATT
    ACAAGTGGGAATTTGAAGAGCTTTTGAAAT
    CCTGTGGTGAGCTGTTTGATTCTTTGAACTC
    GAGTCACCAGGCGCTTTTCCAAGAGAAGGT
    CATCAAGACTTTGGATTTTTCCACACCGGG
    GCGCGCTGCGGCTGCTGTTGCTTTTTTGAGT
    TTTATAAAGGATAAATGGAGCGAAGAAACC
    CATCTGAGCGGGGGGTACCTGCTGGATTTT
    CTGGCCATGCATCTGTGGAGAGCGGTTGTG
    AGACACAAGAATCGCCTGCTACTGTTGTCT
    TCCGTCCGCCCGGCGATAATACCGACGGAG
    GAGCAGCAGCAGCAGCAGGAGGAAGCCAG
    GCGGCGGCGGCAGGAGCAGAGCCCATGGA
    ACCCGAGAGCCGGCCTGGACCCTCGGGAAT
    GAATGTTGTACAGGTGGCTGAACTGTATCC
    AGAACTGAGACGCATTTTGACAATTACAGA
    GGATGGGCAGGGGCTAAAGGGGGTAAAGA
    GGGAGCGGGGGGCTTGTGAGGCTACAGAG
    GAGGCTAGGAATCTAGCTTTTAGCTTAATG
    ACCAGACACCGTCCTGAGTGTATTACTTTTC
    AACAGATCAAGGATAATTGCGCTAATGAGC
    TTGATCTGCTGGCGCAGAAGTATTCCATAG
    AGCAGCTGACCACTTACTGGCTGCAGCCAG
    GGGATGATTTTGAGGAGGCTATTAGGGTAT
    ATGCAAAGGTGGCACTTAGGCCAGATTGCA
    AGTACAAGATCAGCAAACTTGTAAATATCA
    GGAATTGTTGCTACATTTCTGGGAACGGGG
    CCGAGGTGGAGATAGATACGGAGGATAGG
    GTGGCCTTTAGATGTAGCATGATAAATATG
    TGGCCGGGGGTGCTTGGCATGGACGGGGTG
    GTTATTATGAATGTAAGGTTTACTGGCCCC
    AATTTTAGCGGTACGGTTTTCCTGGCCAATA
    CCAACCTTATCCTACACGGTGTAAGCTTCTA
    TGGGTTTAACAATACCTGTGTGGAAGCCTG
    GACCGATGTAAGGGTTCGGGGCTGTGCCTT
    TTACTGCTGCTGGAAGGGGGTGGTGTGTCG
    CCCCAAAAGCAGGGCTTCAATTAAGAAATG
    CCTCTTTGAAAGGTGTACCTTGGGTATCCTG
    TCTGAGGGTAACTCCAGGGTGCGCCACAAT
    GTGGCCTCCGACTGTGGTTGCTTCATGCTAG
    TGAAAAGCGTGGCTGTGATTAAGCATAACA
    TGGTATGTGGCAACTGCGAGGACAGGGCCT
    CTCAGATGCTGACCTGCTCGGACGGCAACT
    GTCACCTGCTGAAGACCATTCACGTAGCCA
    GCCACTCTCGCAAGGCCTGGCCAGTGTTTG
    AGCATAACATACTGACCCGCTGTTCCTTGC
    ATTTGGGTAACAGGAGGGGGGTGTTCCTAC
    CTTACCAATGCAATTTGAGTCACACTAAGA
    TATTGCTTGAGCCCGAGAGCATGTCCAAGG
    TGAACCTGAACGGGGTGTTTGACATGACCA
    TGAAGATCTGGAAGGTGCTGAGGTACGATG
    AGACCCGCACCAGGTGCAGACCCTGCGAGT
    GTGGCGGTAAACATATTAGGAACCAGCCTG
    TGATGCTGGATGTGACCGAGGAGCTGAGGC
    CCGATCACTTGGTGCTGGCCTGCACCCGCG
    CTGAGTTTGGCTCTAGCGATGAAGATACAG
    ATTGAGGTACTGAAATGTGTGGGCGTGGCT
    TAAGGGTGGGAAAGAATATATAAGGTGGG
    GGTCTTATGTAGTTTTGTATCTGTTTTGCAG
    CAGCCGCCGCCGCCATGAGCACCAACTCGT
    TTGATGGAAGCATTGTGAGCTCATATTTGA
    CAACGCGCATGCCCCCATGGGCCGGGGTGC
    GTCAGAATGTGATGGGCTCCAGCATTGATG
    GTCGCCCCGTCCTGCCCGCAAACTCTACTA
    CCTTGACCTACGAGACCGTGTCTGGAACGC
    CGTTGGAGACTGCAGCCTCCGCCGCCGCTT
    CAGCCGCTGCAGCCACCGCCCGCGGGATTG
    TGACTGACTTTGCTTTCCTGAGCCCGCTTGC
    AAGCAGTGCAGCTTCCCGTTCATCCGCCCG
    CGATGACAAGTTGACGGCTCTTTTGGCACA
    ATTGGATTCTTTGACCCGGGAACTTAATGTC
    GTTTCTCAGCAGCTGTTGGATCTGCGCCAG
    CAGGTTTCTGCCCTGAAGGCTTCCTCCCCTC
    CCAATGCGGTTTAAAACATAAATAAAAAAC
    CAGACTCTGTTTGGATTTGGATCAAGCAAG
    TGTCTTGCTGTCTTTATTTAGGGGTTTTGCG
    CGCGCGGTAGGCCCGGGACCAGCGGTCTCG
    GTCGTTGAGGGTCCTGTGTATTTTTTCCAGG
    ACGTGGTAAAGGTGACTCTGGATGTTCAGA
    TACATGGGCATAAGCCCGTCTCTGGGGTGG
    AGGTAGCACCACTGCAGAGCTTCATGCTGC
    GGGGTGGTGTTGTAGATGATCCAGTCGTAG
    CAGGAGCGCTGGGCGTGGTGCCTAAAAATG
    TCTTTCAGTAGCAAGCTGATTGCCAGGGGC
    AGGCCCTTGGTGTAAGTGTTTACAAAGCGG
    TTAAGCTGGGATGGGTGCATACGTGGGGAT
    ATGAGATGCATCTTGGACTGTATTTTTAGGT
    TGGCTATGTTCCCAGCCATATCCCTCCGGG
    GATTCATGTTGTGCAGAACCACCAGCACAG
    TGTATCCGGTGCACTTGGGAAATTTGTCAT
    GTAGCTTAGAAGGAAATGCGTGGAAGAACT
    TGGAGACGCCCTTGTGACCTCCAAGATTTT
    CCATGCATTCGTCCATAATGATGGCAATGG
    GCCCACGGGCGGCGGCCTGGGCGAAGATAT
    TTCTGGGATCACTAACGTCATAGTTGTGTTC
    CAGGATGAGATCGTCATAGGCCATTTTTAC
    AAAGCGCGGGCGGAGGGTGCCAGACTGCG
    GTATAATGGTTCCATCCGGCCCAGGGGCGT
    AGTTACCCTCACAGATTTGCATTTCCCACGC
    TTTGAGTTCAGATGGGGGGATCATGTCTAC
    CTGCGGGGCGATGAAGAAAACGGTTTCCGG
    GGTAGGGGAGATCAGCTGGGAAGAAAGCA
    GGTTCCTGAGCAGCTGCGACTTACCGCAGC
    CGGTGGGCCCGTAAATCACACCTATTACCG
    GCTGCAACTGGTAGTTAAGAGAGCTGCAGC
    TGCCGTCATCCCTGAGCAGGGGGGCCACTT
    CGTTAAGCATGTCCCTGACTCGCATGTTTTC
    CCTGACCAAATCCGCCAGAAGGCGCTCGCC
    GCCCAGCGATAGCAGTTCTTGCAAGGAAGC
    AAAGTTTTTCAACGGTTTGAGACCGTCCGC
    CGTAGGCATGCTTTTGAGCGTTTGACCAAG
    CAGTTCCAGGCGGTCCCACAGCTCGGTCAC
    CTGCTCTACGGCATCTCGATCCAGCATATCT
    CCTCGTTTCGCGGGTTGGGGCGGCTTTCGCT
    GTACGGCAGTAGTCGGTGCTCGTCCAGACG
    GGCCAGGGTCATGTCTTTCCACGGGCGCAG
    GGTCCTCGTCAGCGTAGTCTGGGTCACGGT
    GAAGGGGTGCGCTCCGGGCTGCGCGCTGGC
    CAGGGTGCGCTTGAGGCTGGTCCTGCTGGT
    GCTGAAGCGCTGCCGGTCTTCGCCCTGCGC
    GTCGGCCAGGTAGCATTTGACCATGGTGTC
    ATAGTCCAGCCCCTCCGCGGCGTGGCCCTT
    GGCGCGCAGCTTGCCCTTGGAGGAGGCGCC
    GCACGAGGGGCAGTGCAGACTTTTGAGGGC
    GTAGAGCTTGGGCGCGAGAAATACCGATTC
    CGGGGAGTAGGCATCCGCGCCGCAGGCCCC
    GCAGACGGTCTCGCATTCCACGAGCCAGGT
    GAGCTCTGGCCGTTCGGGGTCAAAAACCAG
    GTTTCCCCCATGCTTTTTGATGCGTTTCTTA
    CCTCTGGTTTCCATGAGCCGGTGTCCACGCT
    CGGTGACGAAAAGGCTGTCCGTGTCCCCGT
    ATACAGACTTGAGAGGCCTGTCCTCGAGCG
    GTGTTCCGCGGTCCTCCTCGTATAGAAACTC
    GGACCACTCTGAGACAAAGGCTCGCGTCCA
    GGCCAGCACGAAGGAGGCTAAGTGGGAGG
    GGTAGCGGTCGTTGTCCACTAGGGGGTCCA
    CTCGCTCCAGGGTGTGAAGACACATGTCGC
    CCTCTTCGGCATCAAGGAAGGTGATTGGTT
    TGTAGGTGTAGGCCACGTGACCGGGTGTTC
    CTGAAGGGGGGCTATAAAAGGGGGTGGGG
    GCGCGTTCGTCCTCACTCTCTTCCGCATCGC
    TGTCTGCGAGGGCCAGCTGTTGGGGTGAGT
    ACTCCCTCTGAAAAGCGGGCATGACTTCTG
    CGCTAAGATTGTCAGTTTCCAAAAACGAGG
    AGGATTTGATATTCACCTGGCCCGCGGTGA
    TGCCTTTGAGGGTGGCCGCATCCATCTGGT
    CAGAAAAGACAATCTTTTTGTTGTCAAGCT
    TGGTGGCAAACGACCCGTAGAGGGCGTTGG
    ACAGCAACTTGGCGATGGAGCGCAGGGTTT
    GGTTTTTGTCGCGATCGGCGCGCTCCTTGGC
    CGCGATGTTTAGCTGCACGTATTCGCGCGC
    AACGCACCGCCATTCGGGAAAGACGGTGGT
    GCGCTCGTCGGGCACCAGGTGCACGCGCCA
    ACCGCGGTTGTGCAGGGTGACAAGGTCAAC
    GCTGGTGGCTACCTCTCCGCGTAGGCGCTC
    GTTGGTCCAGCAGAGGCGGCCGCCCTTGCG
    CGAGCAGAATGGCGGTAGGGGGTCTAGCTG
    CGTCTCGTCCGGGGGGTCTGCGTCCACGGT
    AAAGACCCCGGGCAGCAGGCGCGCGTCGA
    AGTAGTCTATCTTGCATCCTTGCAAGTCTAG
    CGCCTGCTGCCATGCGCGGGCGGCAAGCGC
    GCGCTCGTATGGGTTGAGTGGGGGACCCCA
    TGGCATGGGGTGGGTGAGCGCGGAGGCGT
    ACATGCCGCAAATGTCGTAAACGTAGAGGG
    GCTCTCTGAGTATTCCAAGATATGTAGGGT
    AGCATCTTCCACCGCGGATGCTGGCGCGCA
    CGTAATCGTATAGTTCGTGCGAGGGAGCGA
    GGAGGTCGGGACCGAGGTTGCTACGGGCG
    GGCTGCTCTGCTCGGAAGACTATCTGCCTG
    AAGATGGCATGTGAGTTGGATGATATGGTT
    GGACGCTGGAAGACGTTGAAGCTGGCGTCT
    GTGAGACCTACCGCGTCACGCACGAAGGAG
    GCGTAGGAGTCGCGCAGCTTGTTGACCAGC
    TCGGCGGTGACCTGCACGTCTAGGGCGCAG
    TAGTCCAGGGTTTCCTTGATGATGTCATACT
    TATCCTGTCCCTTTTTTTTCCACAGCTCGCG
    GTTGAGGACAAACTCTTCGCGGTCTTTCCA
    GTACTCTTGGATCGGAAACCCGTCGGCCTC
    CGAACGGTAAGAGCCTAGCATGTAGAACTG
    GTTGACGGCCTGGTAGGCGCAGCATCCCTT
    TTCTACGGGTAGCGCGTATGCCTGCGCGGC
    CTTCCGGAGCGAGGTGTGGGTGAGCGCAAA
    GGTGTCCCTGACCATGACTTTGAGGTACTG
    GTATTTGAAGTCAGTGTCGTCGCATCCGCC
    CTGCTCCCAGAGCAAAAAGTCCGTGCGCTT
    TTTGGAACGCGGATTTGGCAGGGCGAAGGT
    GACATCGTTGAAGAGTATCTTTCCCGCGCG
    AGGCATAAAGTTGCGTGTGATGCGGAAGGG
    TCCCGGCACCTCGGAACGGTTGTTAATTAC
    CTGGGCGGCGAGCACGATCTCGTCAAAGCC
    GTTGATGTTGTGGCCCACAATGTAAAGTTC
    CAAGAAGCGCGGGATGCCCTTGATGGAAG
    GCAATTTTTTAAGTTCCTCGTAGGTGAGCTC
    TTCAGGGGAGCTGAGCCCGTGCTCTGAAAG
    GGCCCAGTCTGCAAGATGAGGGTTGGAAGC
    GACGAATGAGCTCCACAGGTCACGGGCCAT
    TAGCATTTGCAGGTGGTCGCGAAAGGTCCT
    AAACTGGCGACCTATGGCCATTTTTTCTGG
    GGTGATGCAGTAGAAGGTAAGCGGGTCTTG
    TTCCCAGCGGTCCCATCCAAGGTTCGCGGC
    TAGGTCTCGCGCGGCAGTCACTAGAGGCTC
    ATCTCCGCCGAACTTCATGACCAGCATGAA
    GGGCACGAGCTGCTTCCCAAAGGCCCCCAT
    CCAAGTATAGGTCTCTACATCGTAGGTGAC
    AAAGAGACGCTCGGTGCGAGGATGCGAGC
    CGATCGGGAAGAACTGGATCTCCCGCCACC
    AATTGGAGGAGTGGCTATTGATGTGGTGAA
    AGTAGAAGTCCCTGCGACGGGCCGAACACT
    CGTGCTGGCTTTTGTAAAAACGTGCGCAGT
    ACTGGCAGCGGTGCACGGGCTGTACATCCT
    GCACGAGGTTGACCTGACGACCGCGCACAA
    GGAAGCAGAGTGGGAATTTGAGCCCCTCGC
    CTGGCGGGTTTGGCTGGTGGTCTTCTACTTC
    GGCTGCTTGTCCTTGACCGTCTGGCTGCTCG
    AGGGGAGTTACGGTGGATCGGACCACCACG
    CCGCGCGAGCCCAAAGTCCAGATGTCCGCG
    CGCGGCGGTCGGAGCTTGATGACAACATCG
    CGCAGATGGGAGCTGTCCATGGTCTGGAGC
    TCCCGCGGCGTCAGGTCAGGCGGGAGCTCC
    TGCAGGTTTACCTCGCATAGACGGGTCAGG
    GCGCGGGCTAGATCCAGGTGATACCTAATT
    TCCAGGGGCTGGTTGGTGGCGGCGTCGATG
    GCTTGCAAGAGGCCGCATCCCCGCGGCGCG
    ACTACGGTACCGCGCGGCGGGCGGTGGGCC
    GCGGGGGTGTCCTTGGATGATGCATCTAAA
    AGCGGTGACGCGGGCGAGCCCCCGGAGGT
    AGGGGGGGCTCCGGACCCGCCGGGAGAGG
    GGGCAGGGGCACGTCGGCGCCGCGCGCGG
    GCAGGAGCTGGTGCTGCGCGCGTAGGTTGC
    TGGCGAACGCGACGACGCGGCGGTTGATCT
    CCTGAATCTGGCGCCTCTGCGTGAAGACGA
    CGGGCCCGGTGAGCTTGAACCTGAAAGAGA
    GTTCGACAGAATCAATTTCGGTGTCGTTGA
    CGGCGGCCTGGCGCAAAATCTCCTGCACGT
    CTCCTGAGTTGTCTTGATAGGCGATCTCGGC
    CATGAACTGCTCGATCTCTTCCTCCTGGAGA
    TCTCCGCGTCCGGCTCGCTCCACGGTGGCG
    GCGAGGTCGTTGGAAATGCGGGCCATGAGC
    TGCGAGAAGGCGTTGAGGCCTCCCTCGTTC
    CAGACGCGGCTGTAGACCACGCCCCCTTCG
    GCATCGCGGGCGCGCATGACCACCTGCGCG
    AGATTGAGCTCCACGTGCCGGGCGAAGACG
    GCGTAGTTTCGCAGGCGCTGAAAGAGGTAG
    TTGAGGGTGGTGGCGGTGTGTTCTGCCACG
    AAGAAGTACATAACCCAGCGTCGCAACGTG
    GATTCGTTGATATCCCCCAAGGCCTCAAGG
    CGCTCCATGGCCTCGTAGAAGTCCACGGCG
    AAGTTGAAAAACTGGGAGTTGCGCGCCGAC
    ACGGTTAACTCCTCCTCCAGAAGACGGATG
    AGCTCGGCGACAGTGTCGCGCACCTCGCGC
    TCAAAGGCTACAGGGGCCTCTTCTTCTTCTT
    CAATCTCCTCTTCCATAAGGGCCTCCCCTTC
    TTCTTCTTCTGGCGGCGGTGGGGGAGGGGG
    GACACGGCGGCGACGACGGCGCACCGGGA
    GGCGGTCGACAAAGCGCTCGATCATCTCCC
    CGCGGCGACGGCGCATGGTCTCGGTGACGG
    CGCGGCCGTTCTCGCGGGGGCGCAGTTGGA
    AGACGCCGCCCGTCATGTCCCGGTTATGGG
    TTGGCGGGGGGCTGCCATGCGGCAGGGATA
    CGGCGCTAACGATGCATCTCAACAATTGTT
    GTGTAGGTACTCCGCCGCCGAGGGACCTGA
    GCGAGTCCGCATCGACCGGATCGGAAAACC
    TCTCGAGAAAGGCGTCTAACCAGTCACAGT
    CGCAAGGTAGGCTGAGCACCGTGGCGGGC
    GGCAGCGGGCGGCGGTCGGGGTTGTTTCTG
    GCGGAGGTGCTGCTGATGATGTAATTAAAG
    TAGGCGGTCTTGAGACGGCGGATGGTCGAC
    AGAAGCACCATGTCCTTGGGTCCGGCCTGC
    TGAATGCGCAGGCGGTCGGCCATGCCCCAG
    GCTTCGTTTTGACATCGGCGCAGGTCTTTGT
    AGTAGTCTTGCATGAGCCTTTCTACCGGCA
    CTTCTTCTTCTCCTTCCTCTTGTCCTGCATCT
    CTTGCATCTATCGCTGCGGCGGCGGCGGAG
    TTTGGCCGTAGGTGGCGCCCTCTTCCTCCCA
    TGCGTGTGACCCCGAAGCCCCTCATCGGCT
    GAAGCAGGGCTAGGTCGGCGACAACGCGC
    TCGGCTAATATGGCCTGCTGCACCTGCGTG
    AGGGTAGACTGGAAGTCATCCATGTCCACA
    AAGCGGTGGTATGCGCCCGTGTTGATGGTG
    TAAGTGCAGTTGGCCATAACGGACCAGTTA
    ACGGTCTGGTGACCCGGCTGCGAGAGCTCG
    GTGTACCTGAGACGCGAGTAAGCCCTCGAG
    TCAAATACGTAGTCGTTGCAAGTCCGCACC
    AGGTACTGGTATCCCACCAAAAAGTGCGGC
    GGCGGCTGGCGGTAGAGGGGCCAGCGTAG
    GGTGGCCGGGGCTCCGGGGGCGAGATCTTC
    CAACATAAGGCGATGATATCCGTAGATGTA
    CCTGGACATCCAGGTGATGCCGGCGGCGGT
    GGTGGAGGCGCGCGGAAAGTCGCGGACGC
    GGTTCCAGATGTTGCGCAGCGGCAAAAAGT
    GCTCCATGGTCGGGACGCTCTGGCCGGTCA
    GGCGCGCGCAATCGTTGACGCTCTAGCGTG
    CAAAAGGAGAGCCTGTAAGCGGGCACTCTT
    CCGTGGTCTGGTGGATAAATTCGCAAGGGT
    ATCATGGCGGACGACCGGGGTTCGAGCCCC
    GTATCCGGCCGTCCGCCGTGATCCATGCGG
    TTACCGCCCGCGTGTCGAACCCAGGTGTGC
    GACGTCAGACAACGGGGGAGTGCTCCTTTT
    GGCTTCCTTCCAGGCGCGGCGGCTGCTGCG
    CTAGCTTTTTTGGCCACTGGCCGCGCGCAG
    CGTAAGCGGTTAGGCTGGAAAGCGAAAGC
    ATTAAGTGGCTCGCTCCCTGTAGCCGGAGG
    GTTATTTTCCAAGGGTTGAGTCGCGGGACC
    CCCGGTTCGAGTCTCGGACCGGCCGGACTG
    CGGCGAACGGGGGTTTGCCTCCCCGTCATG
    CAAGACCCCGCTTGCAAATTCCTCCGGAAA
    CAGGGACGAGCCCCTTTTTTGCTTTTCCCAG
    ATGCATCCGGTGCTGCGGCAGATGCGCCCC
    CCTCCTCAGCAGCGGCAAGAGCAAGAGCA
    GCGGCAGACATGCAGGGCACCCTCCCCTCC
    TCCTACCGCGTCAGGAGGGGCGACATCCGC
    GGTTGACGCGGCAGCAGATGGTGATTACGA
    ACCCCCGCGGCGCCGGGCCCGGCACTACCT
    GGACTTGGAGGAGGGCGAGGGCCTGGCGC
    GGCTAGGAGCGCCCTCTCCTGAGCGGCACC
    CAAGGGTGCAGCTGAAGCGTGATACGCGTG
    AGGCGTACGTGCCGCGGCAGAACCTGTTTC
    GCGACCGCGAGGGAGAGGAGCCCGAGGAG
    ATGCGGGATCGAAAGTTCCACGCAGGGCGC
    GAGCTGCGGCATGGCCTGAATCGCGAGCGG
    TTGCTGCGCGAGGAGGACTTTGAGCCCGAC
    GCGCGAACCGGGATTAGTCCCGCGCGCGCA
    CACGTGGCGGCCGCCGACCTGGTAACCGCA
    TACGAGCAGACGGTGAACCAGGAGATTAA
    CTTTCAAAAAAGCTTTAACAACCACGTGCG
    TACGCTTGTGGCGCGCGAGGAGGTGGCTAT
    AGGACTGATGCATCTGTGGGACTTTGTAAG
    CGCGCTGGAGCAAAACCCAAATAGCAAGC
    CGCTCATGGCGCAGCTGTTCCTTATAGTGC
    AGCACAGCAGGGACAACGAGGCATTCAGG
    GATGCGCTGCTAAACATAGTAGAGCCCGAG
    GGCCGCTGGCTGCTCGATTTGATAAACATC
    CTGCAGAGCATAGTGGTGCAGGAGCGCAGC
    TTGAGCCTGGCTGACAAGGTGGCCGCCATC
    AACTATTCCATGCTTAGCCTGGGCAAGTTTT
    ACGCCCGCAAGATATACCATACCCCTTACG
    TTCCCATAGACAAGGAGGTAAAGATCGAGG
    GGTTCTACATGCGCATGGCGCTGAAGGTGC
    TTACCTTGAGCGACGACCTGGGCGTTTATC
    GCAACGAGCGCATCCACAAGGCCGTGAGC
    GTGAGCCGGCGGCGCGAGCTCAGCGACCGC
    GAGCTGATGCACAGCCTGCAAAGGGCCCTG
    GCTGGCACGGGCAGCGGCGATAGAGAGGC
    CGAGTCCTACTTTGACGCGGGCGCTGACCT
    GCGCTGGGCCCCAAGCCGACGCGCCCTGGA
    GGCAGCTGGGGCCGGACCTGGGCTGGCGGT
    GGCACCCGCGCGCGCTGGCAACGTCGGCGG
    CGTGGAGGAATATGACGAGGACGATGAGT
    ACGAGCCAGAGGACGGCGAGTACTAAGCG
    GTGATGTTTCTGATCAGATGATGCAAGACG
    CAACGGACCCGGCGGTGCGGGCGGCGCTGC
    AGAGCCAGCCGTCCGGCCTTAACTCCACGG
    ACGACTGGCGCCAGGTCATGGACCGCATCA
    TGTCGCTGACTGCGCGCAATCCTGACGCGT
    TCCGGCAGCAGCCGCAGGCCAACCGGCTCT
    CCGCAATTCTGGAAGCGGTGGTCCCGGCGC
    GCGCAAACCCCACGCACGAGAAGGTGCTG
    GCGATCGTAAACGCGCTGGCCGAAAACAG
    GGCCATCCGGCCCGACGAGGCCGGCCTGGT
    CTACGACGCGCTGCTTCAGCGCGTGGCTCG
    TTACAACAGCGGCAACGTGCAGACCAACCT
    GGACCGGCTGGTGGGGGATGTGCGCGAGG
    CCGTGGCGCAGCGTGAGCGCGCGCAGCAGC
    AGGGCAACCTGGGCTCCATGGTTGCACTAA
    ACGCCTTCCTGAGTACACAGCCCGCCAACG
    TGCCGCGGGGACAGGAGGACTACACCAACT
    TTGTGAGCGCACTGCGGCTAATGGTGACTG
    AGACACCGCAAAGTGAGGTGTACCAGTCTG
    GGCCAGACTATTTTTTCCAGACCAGTAGAC
    AAGGCCTGCAGACCGTAAACCTGAGCCAGG
    CTTTCAAAAACTTGCAGGGGCTGTGGGGGG
    TGCGGGCTCCCACAGGCGACCGCGCGACCG
    TGTCTAGCTTGCTGACGCCCAACTCGCGCCT
    GTTGCTGCTGCTAATAGCGCCCTTCACGGA
    CAGTGGCAGCGTGTCCCGGGACACATACCT
    AGGTCACTTGCTGACACTGTACCGCGAGGC
    CATAGGTCAGGCGCATGTGGACGAGCATAC
    TTTCCAGGAGATTACAAGTGTCAGCCGCGC
    GCTGGGGCAGGAGGACACGGGCAGCCTGG
    AGGCAACCCTAAACTACCTGCTGACCAACC
    GGCGGCAGAAGATCCCCTCGTTGCACAGTT
    TAAACAGCGAGGAGGAGCGCATTTTGCGCT
    ACGTGCAGCAGAGCGTGAGCCTTAACCTGA
    TGCGCGACGGGGTAACGCCCAGCGTGGCGC
    TGGACATGACCGCGCGCAACATGGAACCGG
    GCATGTATGCCTCAAACCGGCCGTTTATCA
    ACCGCCTAATGGACTACTTGCATCGCGCGG
    CCGCCGTGAACCCCGAGTATTTCACCAATG
    CCATCTTGAACCCGCACTGGCTACCGCCCC
    CTGGTTTCTACACCGGGGGATTCGAGGTGC
    CCGAGGGTAACGATGGATTCCTCTGGGACG
    ACATAGACGACAGCGTGTTTTCCCCGCAAC
    CGCAGACCCTGCTAGAGTTGCAACAGCGCG
    AGCAGGCAGAGGCGGCGCTGCGAAAGGAA
    AGCTTCCGCAGGCCAAGCAGCTTGTCCGAT
    CTAGGCGCTGCGGCCCCGCGGTCAGATGCT
    AGTAGCCCATTTCCAAGCTTGATAGGGTCT
    CTTACCAGCACTCGCACCACCCGCCCGCGC
    CTGCTGGGCGAGGAGGAGTACCTAAACAAC
    TCGCTGCTGCAGCCGCAGCGCGAAAAAAAC
    CTGCCTCCGGCATTTCCCAACAACGGGATA
    GAGAGCCTAGTGGACAAGATGAGTAGATG
    GAAGACGTACGCGCAGGAGCACAGGGACG
    TGCCAGGCCCGCGCCCGCCCACCCGTCGTC
    AAAGGCACGACCGTCAGCGGGGTCTGGTGT
    GGGAGGACGATGACTCGGCAGACGACAGC
    AGCGTCCTGGATTTGGGAGGGAGTGGCAAC
    CCGTTTGCGCACCTTCGCCCCAGGCTGGGG
    AGAATGTTTTAAAAAAAAAAAAGCATGATG
    CAAAATAAAAAACTCACCAAGGCCATGGC
    ACCGAGCGTTGGTTTTCTTGTATTCCCCTTA
    GTATGCGGCGCGCGGCGATGTATGAGGAAG
    GTCCTCCTCCCTCCTACGAGAGTGTGGTGA
    GCGCGGCGCCAGTGGCGGCGGCGCTGGGTT
    CTCCCTTCGATGCTCCCCTGGACCCGCCGTT
    TGTGCCTCCGCGGTACCTGCGGCCTACCGG
    GGGGAGAAACAGCATCCGTTACTCTGAGTT
    GGCACCCCTATTCGACACCACCCGTGTGTA
    CCTGGTGGACAACAAGTCAACGGATGTGGC
    ATCCCTGAACTACCAGAACGACCACAGCAA
    CTTTCTGACCACGGTCATTCAAAACAATGA
    CTACAGCCCGGGGGAGGCAAGCACACAGA
    CCATCAATCTTGACGACCGGTCGCACTGGG
    GCGGCGACCTGAAAACCATCCTGCATACCA
    ACATGCCAAATGTGAACGAGTTCATGTTTA
    CCAATAAGTTTAAGGCGCGGGTGATGGTGT
    CGCGCTTGCCTACTAAGGACAATCAGGTGG
    AGCTGAAATACGAGTGGGTGGAGTTCACGC
    TGCCCGAGGGCAACTACTCCGAGACCATGA
    CCATAGACCTTATGAACAACGCGATCGTGG
    AGCACTACTTGAAAGTGGGCAGACAGAAC
    GGGGTTCTGGAAAGCGACATCGGGGTAAA
    GTTTGACACCCGCAACTTCAGACTGGGGTT
    TGACCCCGTCACTGGTCTTGTCATGCCTGGG
    GTATATACAAACGAAGCCTTCCATCCAGAC
    ATCATTTTGCTGCCAGGATGCGGGGTGGAC
    TTCACCCACAGCCGCCTGAGCAACTTGTTG
    GGCATCCGCAAGCGGCAACCCTTCCAGGAG
    GGCTTTAGGATCACCTACGATGATCTGGAG
    GGTGGTAACATTCCCGCACTGTTGGATGTG
    GACGCCTACCAGGCGAGCTTGAAAGATGAC
    ACCGAACAGGGCGGGGGTGGCGCAGGCGG
    CAGCAACAGCAGTGGCAGCGGCGCGGAAG
    AGAACTCCAACGCGGCAGCCGCGGCAATGC
    AGCCGGTGGAGGACATGAACGATCATGCCA
    TTCGCGGCGACACCTTTGCCACACGGGCTG
    AGGAGAAGCGCGCTGAGGCCGAAGCAGCG
    GCCGAAGCTGCCGCCCCCGCTGCGCAACCC
    GAGGTCGAGAAGCCTCAGAAGAAACCGGT
    GATCAAACCCCTGACAGAGGACAGCAAGA
    AACGCAGTTACAACCTAATAAGCAATGACA
    GCACCTTCACCCAGTACCGCAGCTGGTACC
    TTGCATACAACTACGGCGACCCTCAGACCG
    GAATCCGCTCATGGACCCTGCTTTGCACTCC
    TGACGTAACCTGCGGCTCGGAGCAGGTCTA
    CTGGTCGTTGCCAGACATGATGCAAGACCC
    CGTGACCTTCCGCTCCACGCGCCAGATCAG
    CAACTTTCCGGTGGTGGGCGCCGAGCTGTT
    GCCCGTGCACTCCAAGAGCTTCTACAACGA
    CCAGGCCGTCTACTCCCAACTCATCCGCCA
    GTTTACCTCTCTGACCCACGTGTTCAATCGC
    TTTCCCGAGAACCAGATTTTGGCGCGCCCG
    CCAGCCCCCACCATCACCACCGTCAGTGAA
    AACGTTCCTGCTCTCACAGATCACGGGACG
    CTACCGCTGCGCAACAGCATCGGAGGAGTC
    CAGCGAGTGACCATTACTGACGCCAGACGC
    CGCACCTGCCCCTACGTTTACAAGGCCCTG
    GGCATAGTCTCGCCGCGCGTCCTATCGAGC
    CGCACTTTTTGAGCAAGCATGTCCATCCTTA
    TATCGCCCAGCAATAACACAGGCTGGGGCC
    TGCGCTTCCCAAGCAAGATGTTTGGCGGGG
    CCAAGAAGCGCTCCGACCAACACCCAGTGC
    GCGTGCGCGGGCACTACCGCGCGCCCTGGG
    GCGCGCACAAACGCGGCCGCACTGGGCGC
    ACCACCGTCGATGACGCCATCGACGCGGTG
    GTGGAGGAGGCGCGCAACTACACGCCCAC
    GCCGCCACCAGTGTCCACAGTGGACGCGGC
    CATTCAGACCGTGGTGCGCGGAGCCCGGCG
    CTATGCTAAAATGAAGAGACGGCGGAGGC
    GCGTAGCACGTCGCCACCGCCGCCGACCCG
    GCACTGCCGCCCAACGCGCGGCGGCGGCCC
    TGCTTAACCGCGCACGTCGCACCGGCCGAC
    GGGCGGCCATGCGGGCCGCTCGAAGGCTGG
    CCGCGGGTATTGTCACTGTGCCCCCCAGGT
    CCAGGCGACGAGCGGCCGCCGCAGCAGCC
    GCGGCCATTAGTGCTATGACTCAGGGTCGC
    AGGGGCAACGTGTATTGGGTGCGCGACTCG
    GTTAGCGGCCTGCGCGTGCCCGTGCGCACC
    CGCCCCCCGCGCAACTAGATTGCAAGAAAA
    AACTACTTAGACTCGTACTGTTGTATGTATC
    CAGCGGCGGCGGCGCGCAACGAAGCTATGT
    CCAAGCGCAAAATCAAAGAAGAGATGCTC
    CAGGTCATCGCGCCGGAGATCTATGGCCCC
    CCGAAGAAGGAAGAGCAGGATTACAAGCC
    CCGAAAGCTAAAGCGGGTCAAAAAGAAAA
    AGAAAGATGATGATGATGAACTTGACGACG
    AGGTGGAACTGCTGCACGCTACCGCGCCCA
    GGCGACGGGTACAGTGGAAAGGTCGACGC
    GTAAAACGTGTTTTGCGACCCGGCACCACC
    GTAGTCTTTACGCCCGGTGAGCGCTCCACC
    CGCACCTACAAGCGCGTGTATGATGAGGTG
    TACGGCGACGAGGACCTGCTTGAGCAGGCC
    AACGAGCGCCTCGGGGAGTTTGCCTACGGA
    AAGCGGCATAAGGACATGCTGGCGTTGCCG
    CTGGACGAGGGCAACCCAACACCTAGCCTA
    AAGCCCGTAACACTGCAGCAGGTGCTGCCC
    GCGCTTGCACCGTCCGAAGAAAAGCGCGGC
    CTAAAGCGCGAGTCTGGTGACTTGGCACCC
    ACCGTGCAGCTGATGGTACCCAAGCGCCAG
    CGACTGGAAGATGTCTTGGAAAAAATGACC
    GTGGAACCTGGGCTGGAGCCCGAGGTCCGC
    GTGCGGCCAATCAAGCAGGTGGCGCCGGG
    ACTGGGCGTGCAGACCGTGGACGTTCAGAT
    ACCCACTACCAGTAGCACCAGTATTGCCAC
    CGCCACAGAGGGCATGGAGACACAAACGT
    CCCCGGTTGCCTCAGCGGTGGCGGATGCCG
    CGGTGCAGGCGGTCGCTGCGGCCGCGTCCA
    AGACCTCTACGGAGGTGCAAACGGACCCGT
    GGATGTTTCGCGTTTCAGCCCCCCGGCGCC
    CGCGCCGTTCGAGGAAGTACGGCGCCGCCA
    GCGCGCTACTGCCCGAATATGCCCTACATC
    CTTCCATTGCGCCTACCCCCGGCTATCGTGG
    CTACACCTACCGCCCCAGAAGACGAGCAAC
    TACCCGACGCCGAACCACCACTGGAACCCG
    CCGCCGCCGTCGCCGTCGCCAGCCCGTGCT
    GGCCCCGATTTCCGTGCGCAGGGTGGCTCG
    CGAAGGAGGCAGGACCCTGGTGCTGCCAAC
    AGCGCGCTACCACCCCAGCATCGTTTAAAA
    GCCGGTCTTTGTGGTTCTTGCAGATATGGCC
    CTCACCTGCCGCCTCCGTTTCCCGGTGCCGG
    GATTCCGAGGAAGAATGCACCGTAGGAGG
    GGCATGGCCGGCCACGGCCTGACGGGCGGC
    ATGCGTCGTGCGCACCACCGGCGGCGGCGC
    GCGTCGCACCGTCGCATGCGCGGCGGTATC
    CTGCCCCTCCTTATTCCACTGATCGCCGCGG
    CGATTGGCGCCGTGCCCGGAATTGCATCCG
    TGGCCTTGCAGGCGCAGAGACACTGATTAA
    AAACAAGTTGCATGTGGAAAAATCAAAATA
    AAAAGTCTGGACTCTCACGCTCGCTTGGTC
    CTGTAACTATTTTGTAGAATGGAAGACATC
    AACTTTGCGTCTCTGGCCCCGCGACACGGC
    TCGCGCCCGTTCATGGGAAACTGGCAAGAT
    ATCGGCACCAGCAATATGAGCGGTGGCGCC
    TTCAGCTGGGGCTCGCTGTGGAGCGGCATT
    AAAAATTTCGGTTCCACCGTTAAGAACTAT
    GGCAGCAAGGCCTGGAACAGCAGCACAGG
    CCAGATGCTGAGGGATAAGTTGAAAGAGC
    AAAATTTCCAACAAAAGGTGGTAGATGGCC
    TGGCCTCTGGCATTAGCGGGGTGGTGGACC
    TGGCCAACCAGGCAGTGCAAAATAAGATTA
    ACAGTAAGCTTGATCCCCGCCCTCCCGTAG
    AGGAGCCTCCACCGGCCGTGGAGACAGTGT
    CTCCAGAGGGGCGTGGCGAAAAGCGTCCGC
    GCCCCGACAGGGAAGAAACTCTGGTGACGC
    AAATAGACGAGCCTCCCTCGTACGAGGAGG
    CACTAAAGCAAGGCCTGCCCACCACCCGTC
    CCATCGCGCCCATGGCTACCGGAGTGCTGG
    GCCAGCACACACCCGTAACGCTGGACCTGC
    CTCCCCCCGCCGACACCCAGCAGAAACCTG
    TGCTGCCAGGCCCGACCGCCGTTGTTGTAA
    CCCGTCCTAGCCGCGCGTCCCTGCGCCGCG
    CCGCCAGCGGTCCGCGATCGTTGCGGCCCG
    TAGCCAGTGGCAACTGGCAAAGCACACTGA
    ACAGCATCGTGGGTCTGGGGGTGCAATCCC
    TGAAGCGCCGACGATGCTTCTGATAGCTAA
    CGTGTCGTATGTGTGTCATGTATGCGTCCAT
    GTCGCCGCCAGAGGAGCTGCTGAGCCGCCG
    CGCGCCCGCTTTCCAAGATGGCTACCCCTTC
    GATGATGCCGCAGTGGTCTTACATGCACAT
    CTCGGGCCAGGACGCCTCGGAGTACCTGAG
    CCCCGGGCTGGTGCAGTTTGCCCGCGCCAC
    CGAGACGTACTTCAGCCTGAATAACAAGTT
    TAGAAACCCCACGGTGGCGCCTACGCACGA
    CGTGACCACAGACCGGTCCCAGCGTTTGAC
    GCTGCGGTTCATCCCTGTGGACCGTGAGGA
    TACTGCGTACTCGTACAAGGCGCGGTTCAC
    CCTAGCTGTGGGTGATAACCGTGTGCTGGA
    CATGGCTTCCACGTACTTTGACATCCGCGG
    CGTGCTGGACAGGGGCCCTACTTTTAAGCC
    CTACTCTGGCACTGCCTACAACGCCCTGGC
    TCCCAAGGGTGCCCCAAATCCTTGCGAATG
    GGATGAAGCTGCTACTGCTCTTGAAATAAA
    CCTAGAAGAAGAGGACGATGACAACGAAG
    ACGAAGTAGACGAGCAAGCTGAGCAGCAA
    AAAACTCACGTATTTGGGCAGGCGCCTTAT
    TCTGGTATAAATATTACAAAGGAGGGTATT
    CAAATAGGTGTCGAAGGTCAAACACCTAAA
    TATGCCGATAAAACATTTCAACCTGAACCT
    CAAATAGGAGAATCTCAGTGGTACGAAACA
    GAAATTAATCATGCAGCTGGGAGAGTCCTA
    AAAAAGACTACCCCAATGAAACCATGTTAC
    GGTTCATATGCAAAACCCACAAATGAAAAT
    GGAGGGCAAGGCATTCTTGTAAAGCAACAA
    AATGGAAAGCTAGAAAGTCAAGTGGAAAT
    GCAATTTTTCTCAACTACTGAGGCAGCCGC
    AGGCAATGGTGATAACTTGACTCCTAAAGT
    GGTATTGTACAGTGAAGATGTAGATATAGA
    AACCCCAGACACTCATATTTCTTACATGCCC
    ACTATTAAGGAAGGTAACTCACGAGAACTA
    ATGGGCCAACAATCTATGCCCAACAGGCCT
    AATTACATTGCTTTTAGGGACAATTTTATTG
    GTCTAATGTATTACAACAGCACGGGTAATA
    TGGGTGTTCTGGCGGGCCAAGCATCGCAGT
    TGAATGCTGTTGTAGATTTGCAAGACAGAA
    ACACAGAGCTTTCATACCAGCTTTTGCTTGA
    TTCCATTGGTGATAGAACCAGGTACTTTTCT
    ATGTGGAATCAGGCTGTTGACAGCTATGAT
    CCAGATGTTAGAATTATTGAAAATCATGGA
    ACTGAAGATGAACTTCCAAATTACTGCTTT
    CCACTGGGAGGTGTGATTAATACAGAGACT
    CTTACCAAGGTAAAACCTAAAACAGGTCAG
    GAAAATGGATGGGAAAAAGATGCTACAGA
    ATTTTCAGATAAAAATGAAATAAGAGTTGG
    AAATAATTTTGCCATGGAAATCAATCTAAA
    TGCCAACCTGTGGAGAAATTTCCTGTACTC
    CAACATAGCGCTGTATTTGCCCGACAAGCT
    AAAGTACAGTCCTTCCAACGTAAAAATTTC
    TGATAACCCAAACACCTACGACTACATGAA
    CAAGCGAGTGGTGGCTCCCGGGCTAGTGGA
    CTGCTACATTAACCTTGGAGCACGCTGGTC
    CCTTGACTATATGGACAACGTCAACCCATT
    TAACCACCACCGCAATGCTGGCCTGCGCTA
    CCGCTCAATGTTGCTGGGCAATGGTCGCTA
    TGTGCCCTTCCACATCCAGGTGCCTCAGAA
    GTTCTTTGCCATTAAAAACCTCCTTCTCCTG
    CCGGGCTCATACACCTACGAGTGGAACTTC
    AGGAAGGATGTTAACATGGTTCTGCAGAGC
    TCCCTAGGAAATGACCTAAGGGTTGACGGA
    GCCAGCATTAAGTTTGATAGCATTTGCCTTT
    ACGCCACCTTCTTCCCCATGGCCCACAACA
    CCGCCTCCACGCTTGAGGCCATGCTTAGAA
    ACGACACCAACGACCAGTCCTTTAACGACT
    ATCTCTCCGCCGCCAACATGCTCTACCCTAT
    ACCCGCCAACGCTACCAACGTGCCCATATC
    CATCCCCTCCCGCAACTGGGCGGCTTTCCG
    CGGCTGGGCCTTCACGCGCCTTAAGACTAA
    GGAAACCCCATCACTGGGCTCGGGCTACGA
    CCCTTATTACACCTACTCTGGCTCTATACCC
    TACCTAGATGGAACCTTTTACCTCAACCAC
    ACCTTTAAGAAGGTGGCCATTACCTTTGAC
    TCTTCTGTCAGCTGGCCTGGCAATGACCGC
    CTGCTTACCCCCAACGAGTTTGAAATTAAG
    CGCTCAGTTGACGGGGAGGGTTACAACGTT
    GCCCAGTGTAACATGACCAAAGACTGGTTC
    CTGGTACAAATGCTAGCTAACTATAACATT
    GGCTACCAGGGCTTCTATATCCCAGAGAGC
    TACAAGGACCGCATGTACTCCTTCTTTAGA
    AACTTCCAGCCCATGAGCCGTCAGGTGGTG
    GATGATACTAAATACAAGGACTACCAACAG
    GTGGGCATCCTACACCAACACAACAACTCT
    GGATTTGTTGGCTACCTTGCCCCCACCATGC
    GCGAAGGACAGGCCTACCCTGCTAACTTCC
    CCTATCCGCTTATAGGCAAGACCGCAGTTG
    ACAGCATTACCCAGAAAAAGTTTCTTTGCG
    ATCGCACCCTTTGGCGCATCCCATTCTCCAG
    TAACTTTATGTCCATGGGCGCACTCACAGA
    CCTGGGCCAAAACCTTCTCTACGCCAACTC
    CGCCCACGCGCTAGACATGACTTTTGAGGT
    GGATCCCATGGACGAGCCCACCCTTCTTTA
    TGTTTTGTTTGAAGTCTTTGACGTGGTCCGT
    GTGCACCAGCCGCACCGCGGCGTCATCGAA
    ACCGTGTACCTGCGCACGCCCTTCTCGGCC
    GGCAACGCCACAACATAAAGAAGCAAGCA
    ACATCAACAACAGCTGCCGCCATGGGCTCC
    AGTGAGCAGGAACTGAAAGCCATTGTCAAA
    GATCTTGGTTGTGGGCCATATTTTTTGGGCA
    CCTATGACAAGCGCTTTCCAGGCTTTGTTTC
    TCCACACAAGCTCGCCTGCGCCATAGTCAA
    TACGGCCGGTCGCGAGACTGGGGGCGTACA
    CTGGATGGCCTTTGCCTGGAACCCGCACTC
    AAAAACATGCTACCTCTTTGAGCCCTTTGG
    CTTTTCTGACCAGCGACTCAAGCAGGTTTA
    CCAGTTTGAGTACGAGTCACTCCTGCGCCG
    TAGCGCCATTGCTTCTTCCCCCGACCGCTGT
    ATAACGCTGGAAAAGTCCACCCAAAGCGTA
    CAGGGGCCCAACTCGGCCGCCTGTGGACTA
    TTCTGCTGCATGTTTCTCCACGCCTTTGCCA
    ACTGGCCCCAAACTCCCATGGATCACAACC
    CCACCATGAACCTTATTACCGGGGTACCCA
    ACTCCATGCTCAACAGTCCCCAGGTACAGC
    CCACCCTGCGTCGCAACCAGGAACAGCTCT
    ACAGCTTCCTGGAGCGCCACTCGCCCTACT
    TCCGCAGCCACAGTGCGCAGATTAGGAGCG
    CCACTTCTTTTTGTCACTTGAAAAACATGTA
    AAAATAATGTACTAGAGACACTTTCAATAA
    AGGCAAATGCTTTTATTTGTACACTCTCGGG
    TGATTATTTACCCCCACCCTTGCCGTCTGCG
    CCGTTTAAAAATCAAAGGGGTTCTGCCGCG
    CATCGCTATGCGCCACTGGCAGGGACACGT
    TGCGATACTGGTGTTTAGTGCTCCACTTAAA
    CTCAGGCACAACCATCCGCGGCAGCTCGGT
    GAAGTTTTCACTCCACAGGCTGCGCACCAT
    CACCAACGCGTTTAGCAGGTCGGGCGCCGA
    TATCTTGAAGTCGCAGTTGGGGCCTCCGCC
    CTGCGCGCGCGAGTTGCGATACACAGGGTT
    GCAGCACTGGAACACTATCAGCGCCGGGTG
    GTGCACGCTGGCCAGCACGCTCTTGTCGGA
    GATCAGATCCGCGTCCAGGTCCTCCGCGTT
    GCTCAGGGCGAACGGAGTCAACTTTGGTAG
    CTGCCTTCCCAAAAAGGGCGCGTGCCCAGG
    CTTTGAGTTGCACTCGCACCGTAGTGGCAT
    CAAAAGGTGACCGTGCCCGGTCTGGGCGTT
    AGGATACAGCGCCTGCATAAAAGCCTTGAT
    CTGCTTAAAAGCCACCTGAGCCTTTGCGCC
    TTCAGAGAAGAACATGCCGCAAGACTTGCC
    GGAAAACTGATTGGCCGGACAGGCCGCGTC
    GTGCACGCAGCACCTTGCGTCGGTGTTGGA
    GATCTGCACCACATTTCGGCCCCACCGGTT
    CTTCACGATCTTGGCCTTGCTAGACTGCTCC
    TTCAGCGCGCGCTGCCCGTTTTCGCTCGTCA
    CATCCATTTCAATCACGTGCTCCTTATTTAT
    CATAATGCTTCCGTGTAGACACTTAAGCTC
    GCCTTCGATCTCAGCGCAGCGGTGCAGCCA
    CAACGCGCAGCCCGTGGGCTCGTGATGCTT
    GTAGGTCACCTCTGCAAACGACTGCAGGTA
    CGCCTGCAGGAATCGCCCCATCATCGTCAC
    AAAGGTCTTGTTGCTGGTGAAGGTCAGCTG
    CAACCCGCGGTGCTCCTCGTTCAGCCAGGT
    CTTGCATACGGCCGCCAGAGCTTCCACTTG
    GTCAGGCAGTAGTTTGAAGTTCGCCTTTAG
    ATCGTTATCCACGTGGTACTTGTCCATCAGC
    GCGCGCGCAGCCTCCATGCCCTTCTCCCAC
    GCAGACACGATCGGCACACTCAGCGGGTTC
    ATCACCGTAATTTCACTTTCCGCTTCGCTGG
    GCTCTTCCTCTTCCTCTTGCGTCCGCATACC
    ACGCGCCACTGGGTCGTCTTCATTCAGCCG
    CCGCACTGTGCGCTTACCTCCTTTGCCATGC
    TTGATTAGCACCGGTGGGTTGCTGAAACCC
    ACCATTTGTAGCGCCACATCTTCTCTTTCTT
    CCTCGCTGTCCACGATTACCTCTGGTGATGG
    CGGGCGCTCGGGCTTGGGAGAAGGGCGCTT
    CTTTTTCTTCTTGGGCGCAATGGCCAAATCC
    GCCGCCGAGGTCGATGGCCGCGGGCTGGGT
    GTGCGCGGCACCAGCGCGTCTTGTGATGAG
    TCTTCCTCGTCCTCGGACTCGATACGCCGCC
    TCATCCGCTTTTTTGGGGGCGCCCGGGGAG
    GCGGCGGCGACGGGGACGGGGACGACACG
    TCCTCCATGGTTGGGGGACGTCGCGCCGCA
    CCGCGTCCGCGCTCGGGGGTGGTTTCGCGC
    TGCTCCTCTTCCCGACTGGCCATTTCCTTCT
    CCTATAGGCAGAAAAAGATCATGGAGTCAG
    TCGAGAAGAAGGACAGCCTAACCGCCCCCT
    CTGAGTTCGCCACCACCGCCTCCACCGATG
    CCGCCAACGCGCCTACCACCTTCCCCGTCG
    AGGCACCCCCGCTTGAGGAGGAGGAAGTG
    ATTATCGAGCAGGACCCAGGTTTTGTAAGC
    GAAGACGACGAGGACCGCTCAGTACCAAC
    AGAGGATAAAAAGCAAGACCAGGACAACG
    CAGAGGCAAACGAGGAACAAGTCGGGCGG
    GGGGACGAAAGGCATGGCGACTACCTAGA
    TGTGGGAGACGACGTGCTGTTGAAGCATCT
    GCAGCGCCAGTGCGCCATTATCTGCGACGC
    GTTGCAAGAGCGCAGCGATGTGCCCCTCGC
    CATAGCGGATGTCAGCCTTGCCTACGAACG
    CCACCTATTCTCACCGCGCGTACCCCCCAA
    ACGCCAAGAAAACGGCACATGCGAGCCCA
    ACCCGCGCCTCAACTTCTACCCCGTATTTGC
    CGTGCCAGAGGTGCTTGCCACCTATCACAT
    CTTTTTCCAAAACTGCAAGATACCCCTATCC
    TGCCGTGCCAACCGCAGCCGAGCGGACAAG
    CAGCTGGCCTTGCGGCAGGGCGCTGTCATA
    CCTGATATCGCCTCGCTCAACGAAGTGCCA
    AAAATCTTTGAGGGTCTTGGACGCGACGAG
    AAGCGCGCGGCAAACGCTCTGCAACAGGA
    AAACAGCGAAAATGAAAGTCACTCTGGAGT
    GTTGGTGGAACTCGAGGGTGACAACGCGCG
    CCTAGCCGTACTAAAACGCAGCATCGAGGT
    CACCCACTTTGCCTACCCGGCACTTAACCTA
    CCCCCCAAGGTCATGAGCACAGTCATGAGT
    GAGCTGATCGTGCGCCGTGCGCAGCCCCTG
    GAGAGGGATGCAAATTTGCAAGAACAAAC
    AGAGGAGGGCCTACCCGCAGTTGGCGACG
    AGCAGCTAGCGCGCTGGCTTCAAACGCGCG
    AGCCTGCCGACTTGGAGGAGCGACGCAAAC
    TAATGATGGCCGCAGTGCTCGTTACCGTGG
    AGCTTGAGTGCATGCAGCGGTTCTTTGCTG
    ACCCGGAGATGCAGCGCAAGCTAGAGGAA
    ACATTGCACTACACCTTTCGACAGGGCTAC
    GTACGCCAGGCCTGCAAGATCTCCAACGTG
    GAGCTCTGCAACCTGGTCTCCTACCTTGGA
    ATTTTGCACGAAAACCGCCTTGGGCAAAAC
    GTGCTTCATTCCACGCTCAAGGGCGAGGCG
    CGCCGCGACTACGTCCGCGACTGCGTTTAC
    TTATTTCTATGCTACACCTGGCAGACGGCC
    ATGGGCGTTTGGCAGCAGTGCTTGGAGGAG
    TGCAACCTCAAGGAGCTGCAGAAACTGCTA
    AAGCAAAACTTGAAGGACCTATGGACGGCC
    TTCAACGAGCGCTCCGTGGCCGCGCACCTG
    GCGGACATCATTTTCCCCGAACGCCTGCTT
    AAAACCCTGCAACAGGGTCTGCCAGACTTC
    ACCAGTCAAAGCATGTTGCAGAACTTTAGG
    AACTTTATCCTAGAGCGCTCAGGAATCTTG
    CCCGCCACCTGCTGTGCACTTCCTAGCGACT
    TTGTGCCCATTAAGTACCGCGAATGCCCTC
    CGCCGCTTTGGGGCCACTGCTACCTTCTGCA
    GCTAGCCAACTACCTTGCCTACCACTCTGA
    CATAATGGAAGACGTGAGCGGTGACGGTCT
    ACTGGAGTGTCACTGTCGCTGCAACCTATG
    CACCCCGCACCGCTCCCTGGTTTGCAATTCG
    CAGCTGCTTAACGAAAGTCAAATTATCGGT
    ACCTTTGAGCTGCAGGGTCCCTCGCCTGAC
    GAAAAGTCCGCGGCTCCGGGGTTGAAACTC
    ACTCCGGGGCTGTGGACGTCGGCTTACCTT
    CGCAAATTTGTACCTGAGGACTACCACGCC
    CACGAGATTAGGTTCTACGAAGACCAATCC
    CGCCCGCCTAATGCGGAGCTTACCGCCTGC
    GTCATTACCCAGGGCCACATTCTTGGCCAA
    TTGCAAGCCATCAACAAAGCCCGCCAAGAG
    TTTCTGCTACGAAAGGGACGGGGGGTTTAC
    TTGGACCCCCAGTCCGGCGAGGAGCTCAAC
    CCAATCCCCCCGCCGCCGCAGCCCTATCAG
    CAGCAGCCGCGGGCCCTTGCTTCCCAGGAT
    GGCACCCAAAAAGAAGCTGCAGCTGCCGCC
    GCCACCCACGGACGAGGAGGAATACTGGG
    ACAGTCAGGCAGAGGAGGTTTTGGACGAG
    GAGGAGGAGGACATGATGGAAGACTGGGA
    GAGCCTAGACGAGGAAGCTTCCGAGGTCGA
    AGAGGTGTCAGACGAAACACCGTCACCCTC
    GGTCGCATTCCCCTCGCCGGCGCCCCAGAA
    ATCGGCAACCGGTTCCAGCATGGCTACAAC
    CTCCGCTCCTCAGGCGCCGCCGGCACTGCC
    CGTTCGCCGACCCAACCGTAGATGGGACAC
    CACTGGAACCAGGGCCGGTAAGTCCAAGCA
    GCCGCCGCCGTTAGCCCAAGAGCAACAACA
    GCGCCAAGGCTACCGCTCATGGCGCGGGCA
    CAAGAACGCCATAGTTGCTTGCTTGCAAGA
    CTGTGGGGGCAACATCTCCTTCGCCCGCCG
    CTTTCTTCTCTACCATCACGGCGTGGCCTTC
    CCCCGTAACATCCTGCATTACTACCGTCATC
    TCTACAGCCCATACTGCACCGGCGGCAGCG
    GCAGCAACAGCAGCGGCCACACAGAAGCA
    AAGGCGACCGGATAGCAAGACTCTGACAA
    AGCCCAAGAAATCCACAGCGGCGGCAGCA
    GCAGGAGGAGGAGCGCTGCGTCTGGCGCCC
    AACGAACCCGTATCGACCCGCGAGCTTAGA
    AACAGGATTTTTCCCACTCTGTATGCTATAT
    TTCAACAGAGCAGGGGCCAAGAACAAGAG
    CTGAAAATAAAAAACAGGTCTCTGCGATCC
    CTCACCCGCAGCTGCCTGTATCACAAAAGC
    GAAGATCAGCTTCGGCGCACGCTGGAAGAC
    GCGGAGGCTCTCTTCAGTAAATACTGCGCG
    CTGACTCTTAAGGACTAGTTTCGCGCCCTTT
    CTCAAATTTAAGCGCGAAAACTACGTCATC
    TCCAGCGGCCACACCCGGCGCCAGCACCTG
    TTGTCAGCGCCATTATGAGCAAGGAAATTC
    CCACGCCCTACATGTGGAGTTACCAGCCAC
    AAATGGGACTTGCGGCTGGAGCTGCCCAAG
    ACTACTCAACCCGAATAAACTACATGAGCG
    CGGGACCCCACATGATATCCCGGGTCAACG
    GAATACGCGCCCACCGAAACCGAATTCTCC
    TGGAACAGGCGGCTATTACCACCACACCTC
    GTAATAACCTTAATCCCCGTAGTTGGCCCG
    CTGCCCTGGTGTACCAGGAAAGTCCCGCTC
    CCACCACTGTGGTACTTCCCAGAGACGCCC
    AGGCCGAAGTTCAGATGACTAACTCAGGGG
    CGCAGCTTGCGGGCGGCTTTCGTCACAGGG
    TGCGGTCGCCCGGGCAGGGTATAACTCACC
    TGACAATCAGAGGGCGAGGTATTCAGCTCA
    ACGACGAGTCGGTGAGCTCCTCGCTTGGTC
    TCCGTCCGGACGGGACATTTCAGATCGGCG
    GCGCCGGCCGCTCTTCATTCACGCCTCGTCA
    GGCAATCCTAACTCTGCAGACCTCGTCCTCT
    GAGCCGCGCTCTGGAGGCATTGGAACTCTG
    CAATTTATTGAGGAGTTTGTGCCATCGGTCT
    ACTTTAACCCCTTCTCGGGACCTCCCGGCCA
    CTATCCGGATCAATTTATTCCTAACTTTGAC
    GCGGTAAAGGACTCGGCGGACGGCTACGA
    CTGAATGTTAAGTGGAGAGGCAGAGCAACT
    GCGCCTGAAACACCTGGTCCACTGTCGCCG
    CCACAAGTGCTTTGCCCGCGACTCCGGTGA
    GTTTTGCTACTTTGAATTGCCCGAGGATCAT
    ATCGAGGGCCCGGCGCACGGCGTCCGGCTT
    ACCGCCCAGGGAGAGCTTGCCCGTAGCCTG
    ATTCGGGAGTTTACCCAGCGCCCCCTGCTA
    GTTGAGCGGGACAGGGGACCCTGTGTTCTC
    ACTGTGATTTGCAACTGTCCTAACCCTGGAT
    TACATCAAGATCCTCTAGTTAATGTCAGGT
    CGCCTAAGTCGATTAACTAGAGTACCCGGG
    GATCTTATTCCCTTTAACTAATAAAAAAAA
    ATAATAAAGCATCACTTACTTAAAATCAGT
    TAGCAAATTTCTGTCCAGTTTATTCAGCAGC
    ACCTCCTTGCCCTCCTCCCAGCTCTGGTATT
    GCAGCTTCCTCCTGGCTGCAAACTTTCTCCA
    CAATCTAAATGGAATGTCAGTTTCCTCCTGT
    TCCTGTCCATCCGCACCCACTATCTTCATGT
    TGTTGCAGATGAAGCGCGCAAGACCGTCTG
    AAGATACCTTCAACCCCGTGTATCCATATG
    ACACGGAAACCGGTCCTCCAACTGTGCCTT
    TTCTTACTCCTCCCTTTGTATCCCCCAATGG
    GTTTCAAGAGAGTCCCCCTGGGGTACTCTC
    TTTGCGCCTATCCGAACCTCTAGTTACCTCC
    AATGGCATGCTTGCGCTCAAAATGGGCAAC
    GGCCTCTCTCTGGACGAGGCCGGCAACCTT
    ACCTCCCAAAATGTAACCACTGTGAGCCCA
    CCTCTCAAAAAAACCAAGTCAAACATAAAC
    CTGGAAATATCTGCACCCCTCACAGTTACC
    TCAGAAGCCCTAACTGTGGCTGCCGCCGCA
    CCTCTAATGGTCGCGGGCAACACACTCACC
    ATGCAATCACAGGCCCCGCTAACCGTGCAC
    GACTCCAAACTTAGCATTGCCACCCAAGGA
    CCCCTCACAGTGTCAGAAGGAAAGCTAGCC
    CTGCAAACATCAGGCCCCCTCACCACCACC
    GATAGCAGTACCCTTACTATCACTGCCTCA
    CCCCCTCTAACTACTGCCACTGGTAGCTTGG
    GCATTGACTTGAAAGAGCCCATTTATACAC
    AAAATGGAAAACTAGGACTAAAGTACGGG
    GCTCCTTTGCATGTAACAGACGACCTAAAC
    ACTTTGACCGTAGCAACTGGTCCAGGTGTG
    ACTATTAATAATACTTCCTTGCAAACTAAA
    GTTACTGGAGCCTTGGGTTTTGATTCACAA
    GGCAATATGCAACTTAATGTAGCAGGAGGA
    CTAAGGATTGATTCTCAAAACAGACGCCTT
    ATACTTGATGTTAGTTATCCGTTTGATGCTC
    AAAACCAACTAAATCTAAGACTAGGACAG
    GGCCCTCTTTTTATAAACTCAGCCCACAACT
    TGGATATTAACTACAACAAAGGCCTTTACT
    TGTTTACAGCTTCAAACAATTCCAAAAAGC
    TTGAGGTTAACCTAAGCACTGCCAAGGGGT
    TGATGTTTGACGCTACAGCCATAGCCATTA
    ATGCAGGAGATGGGCTTGAATTTGGTTCAC
    CTAATGCACCAAACACAAATCCCCTCAAAA
    CAAAAATTGGCCATGGCCTAGAATTTGATT
    CAAACAAGGCTATGGTTCCTAAACTAGGAA
    CTGGCCTTAGTTTTGACAGCACAGGTGCCA
    TTACAGTAGGAAACAAAAATAATGATAAGC
    TAACTTTGTGGACCACACCAGCTCCATCTCC
    TAACTGTAGACTAAATGCAGAGAAAGATGC
    TAAACTCACTTTGGTCTTAACAAAATGTGG
    CAGTCAAATACTTGCTACAGTTTCAGTTTTG
    GCTGTTAAAGGCAGTTTGGCTCCAATATCT
    GGAACAGTTCAAAGTGCTCATCTTATTATA
    AGATTTGACGAAAATGGAGTGCTACTAAAC
    AATTCCTTCCTGGACCCAGAATATTGGAAC
    TTTAGAAATGGAGATCTTACTGAAGGCACA
    GCCTATACAAACGCTGTTGGATTTATGCCT
    AACCTATCAGCTTATCCAAAATCTCACGGT
    AAAACTGCCAAAAGTAACATTGTCAGTCAA
    GTTTACTTAAACGGAGACAAAACTAAACCT
    GTAACACTAACCATTACACTAAACGGTACA
    CAGGAAACAGGAGACACAACTCCAAGTGC
    ATACTCTATGTCATTTTCATGGGACTGGTCT
    GGCCACAACTACATTAATGAAATATTTGCC
    ACATCCTCTTACACTTTTTCATACATTGCCC
    AAGAATAAAGAATCGTTTGTGTTATGTTTC
    AACGTGTTTATTTTTCAATTGCAGAAAATTT
    CAAGTCATTTTTCATTCAGTAGTATAGCCCC
    ACCACCACATAGCTTATACAGATCACCGTA
    CCTTAATCAAACTCACAGAACCCTAGTATT
    CAACCTGCCACCTCCCTCCCAACACACAGA
    GTACACAGTCCTTTCTCCCCGGCTGGCCTTA
    AAAAGCATCATATCATGGGTAACAGACATA
    TTCTTAGGTGTTATATTCCACACGGTTTCCT
    GTCGAGCCAAACGCTCATCAGTGATATTAA
    TAAACTCCCCGGGCAGCTCACTTAAGTTCA
    TGTCGCTGTCCAGCTGCTGAGCCACAGGCT
    GCTGTCCAACTTGCGGTTGCTTAACGGGCG
    GCGAAGGAGAAGTCCACGCCTACATGGGG
    GTAGAGTCATAATCGTGCATCAGGATAGGG
    CGGTGGTGCTGCAGCAGCGCGCGAATAAAC
    TGCTGCCGCCGCCGCTCCGTCCTGCAGGAA
    TACAACATGGCAGTGGTCTCCTCAGCGATG
    ATTCGCACCGCCCGCAGCATAAGGCGCCTT
    GTCCTCCGGGCACAGCAGCGCACCCTGATC
    TCACTTAAATCAGCACAGTAACTGCAGCAC
    AGCACCACAATATTGTTCAAAATCCCACAG
    TGCAAGGCGCTGTATCCAAAGCTCATGGCG
    GGGACCACAGAACCCACGTGGCCATCATAC
    CACAAGCGCAGGTAGATTAAGTGGCGACCC
    CTCATAAACACGCTGGACATAAACATTACC
    TCTTTTGGCATGTTGTAATTCACCACCTCCC
    GGTACCATATAAACCTCTGATTAAACATGG
    CGCCATCCACCACCATCCTAAACCAGCTGG
    CCAAAACCTGCCCGCCGGCTATACACTGCA
    GGGAACCGGGACTGGAACAATGACAGTGG
    AGAGCCCAGGACTCGTAACCATGGATCATC
    ATGCTCGTCATGATATCAATGTTGGCACAA
    CACAGGCACACGTGCATACACTTCCTCAGG
    ATTACAAGCTCCTCCCGCGTTAGAACCATA
    TCCCAGGGAACAACCCATTCCTGAATCAGC
    GTAAATCCCACACTGCAGGGAAGACCTCGC
    ACGTAACTCACGTTGTGCATTGTCAAAGTG
    TTACATTCGGGCAGCAGCGGATGATCCTCC
    AGTATGGTAGCGCGGGTTTCTGTCTCAAAA
    GGAGGTAGACGATCCCTACTGTACGGAGTG
    CGCCGAGACAACCGAGATCGTGTTGGTCGT
    AGTGTCATGCCAAATGGAACGCCGGACGTA
    GTCATATTTCCTGAAGCAAAACCAGGTGCG
    GGCGTGACAAACAGATCTGCGTCTCCGGTC
    TCGCCGCTTAGATCGCTCTGTGTAGTAGTTG
    TAGTATATCCACTCTCTCAAAGCATCCAGG
    CGCCCCCTGGCTTCGGGTTCTATGTAAACTC
    CTTCATGCGCCGCTGCCCTGATAACATCCA
    CCACCGCAGAATAAGCCACACCCAGCCAAC
    CTACACATTCGTTCTGCGAGTCACACACGG
    GAGGAGCGGGAAGAGCTGGAAGAACCATG
    TTTTTTTTTTTATTCCAAAAGATTATCCAAA
    ACCTCAAAATGAAGATCTATTAAGTGAACG
    CGCTCCCCTCCGGTGGCGTGGTCAAACTCT
    ACAGCCAAAGAACAGATAATGGCATTTGTA
    AGATGTTGCACAATGGCTTCCAAAAGGCAA
    ACGGCCCTCACGTCCAAGTGGACGTAAAGG
    CTAAACCCTTCAGGGTGAATCTCCTCTATA
    AACATTCCAGCACCTTCAACCATGCCCAAA
    TAATTCTCATCTCGCCACCTTCTCAATATAT
    CTCTAAGCAAATCCCGAATATTAAGTCCGG
    CCATTGTAAAAATCTGCTCCAGAGCGCCCT
    CCACCTTCAGCCTCAAGCAGCGAATCATGA
    TTGCAAAAATTCAGGTTCCTCACAGACCTG
    TATAAGATTCAAAAGCGGAACATTAACAAA
    AATACCGCGATCCCGTAGGTCCCTTCGCAG
    GGCCAGCTGAACATAATCGTGCAGGTCTGC
    ACGGACCAGCGCGGCCACTTCCCCGCCAGG
    AACCATGACAAAAGAACCCACACTGATTAT
    GACACGCATACTCGGAGCTATGCTAACCAG
    CGTAGCCCCGATGTAAGCTTGTTGCATGGG
    CGGCGATATAAAATGCAAGGTGCTGCTCAA
    AAAATCAGGCAAAGCCTCGCGCAAAAAAG
    AAAGCACATCGTAGTCATGCTCATGCAGAT
    AAAGGCAGGTAAGCTCCGGAACCACCACA
    GAAAAAGACACCATTTTTCTCTCAAACATG
    TCTGCGGGTTTCTGCATAAACACAAAATAA
    AATAACAAAAAAACATTTAAACATTAGAAG
    CCTGTCTTACAACAGGAAAAACAACCCTTA
    TAAGCATAAGACGGACTACGGCCATGCCGG
    CGTGACCGTAAAAAAACTGGTCACCGTGAT
    TAAAAAGCACCACCGACAGCTCCTCGGTCA
    TGTCCGGAGTCATAATGTAAGACTCGGTAA
    ACACATCAGGTTGATTCACATCGGTCAGTG
    CTAAAAAGCGACCGAAATAGCCCGGGGGA
    ATACATACCCGCAGGCGTAGAGACAACATT
    ACAGCCCCCATAGGAGGTATAACAAAATTA
    ATAGGAGAGAAAAACACATAAACACCTGA
    AAAACCCTCCTGCCTAGGCAAAATAGCACC
    CTCCCGCTCCAGAACAACATACAGCGCTTC
    CACAGCGGCAGCCATAACAGTCAGCCTTAC
    CAGTAAAAAAGAAAACCTATTAAAAAAAC
    ACCACTCGACACGGCACCAGCTCAATCAGT
    CACAGTGTAAAAAAGGGCCAAGTGCAGAG
    CGAGTATATATAGGACTAAAAAATGACGTA
    ACGGTTAAAGTCCACAAAAAACACCCAGA
    AAACCGCACGCGAACCTACGCCCAGAAAC
    GAAAGCCAAAAAACCCACAACTTCCTCAAA
    TCGTCACTTCCGTTTTCCCACGTTACGTCAC
    TTCCCATTTTAAGAAAACTACAATTCCCAA
    CACATACAAGTTACTCCGCCCTAAAACCTA
    CGTCACCCGCCCCGTTCCCACGCCCCGCGC
    CACGTCACAAACTCCACCCCCTCATTATCAT
    ATTGGCTTCAATCCAAAATAAGGTATATTA
    TTGATGATG
    95 Full (human IL- TTTTGGATTGAAGCCAATATGATAATGAGG
    nucleotide 12 insert GGGTGGAGTTTGTGACGTGGCGCGGGGCGT
    sequence of  sequence in GGGAACGGGGCGGGTGACGTAGTAGTGTG
    TRZ627 hIL-  grey GCGGAAGTGTGATGTTGCAAGTGTGGCGGA
    12 virus highlight- ACACATGTAAGCGACGGATGTGGCAAAAGT
    (beginning at codon GACGTTTTTGGTGTGCGCCGGTGTTTTGGGC
    Ad5 5′ end optimized) GTAACCGAGTAAGATTTGGCCATTTTCGCG
    ITR) GGAAAACTGAATAAGAGGAAGTGAAATCT
    GAATAATTTTGTGTTACTCATAGCGCGTAAT
    ATTTGTCTAGGGCCGCGGGGACTTTGACCG
    TTTACGTGGAGACTCGCCCAGGTGTTTTTCT
    CAGGTGTTTTCCGCGTTCCGGGTCAAAGTT
    GGCGTTTTATTATTATAGTCAGCTGACGTGT
    AGTGTATTTATACCCGGTGAGTTCCTCAAG
    AGGCCACTCTTGAGTGCCAGCGAGTAGAGT
    TTTCTCCTCCGAGCCGCTCCGACACCGGGA
    CTGAAAATGAGACATATTATCTGCCACGGA
    GGTGTTATTACCGAAGAAATGGCCGCCAGT
    CTTTTGGACCAGCTGATCGAAGAGGTACTG
    GCTGATAATCTTCCACCTCCTAGCCATTTTG
    AACCACCTACCCTTCACGAACTGTATGATTT
    AGACGTGACGGCCCCCGAAGATCCCAACGA
    GGAGGCGGTTTCGCAGATTTTTCCCGACTCT
    GTAATGTTGGCGGTGCAGGAAGGGATTGAC
    TTACTCACTTTTCCGCCGGCGCCCGGTTCTC
    CGGAGCCGCCTCACCTTTCCCGGCAGCCCG
    AGCAGCCGGAGCAGAGAGCCTTGGGTCCG
    GTTTCTATGCCAAACCTTGTACCGGAGGTG
    ATCGATCTTACCTGCCACGAGGCTGGCTTTC
    CACCCAGTGACGACGAGGATGAAGAGGGT
    GAGGAGTTTGTGTTAGATTATGTGGAGCAC
    CCCGGGCACGGTTGCAGGTCTTGTCATTAT
    CACCGGAGGAATACGGGGGACCCAGATATT
    ATGTGTTCGCTTTGCTATATGAGGACCTGTG
    GCATGTTTGTCTACAGTAAGTGAAAATTAT
    GGGCAGTGGGTGATAGAGTGGTGGGTTTGG
    TGTGGTAATTTTTTTTTTAATTTTTACAGTTT
    TGTGGTTTAAAGAATTTTGTATTGTGATTTT
    TTTAAAAGGTCCTGTGTCTGAACCTGAGCC
    TGAGCCCGAGCCAGAACCGGAGCCTGCAA
    GACCTACCCGCCGTCCTAAAATGGCGCCTG
    CTATCCTGAGACGCCCGACATCACCTGTGT
    CTAGAGAATGCAATAGTAGTACGGATAGCT
    GTGACTCCGGTCCTTCTAACACACCTCCTGA
    GATACACCCGGTGGTCCCGCTGTGCCCCAT
    TAAACCAGTTGCCGTGAGAGTTGGTGGGCG
    TCGCCAGGCTGTGGAATGTATCGAGGACTT
    GCTTAACGAGCCTGGGCAACCTTTGGACTT
    GAGCTGTAAACGCCCCAGGCCATAAGGTGT
    AAACCTGTGATTGCGTGTGTGGTTAACGCC
    TTTGTTTGCTGAATGAGTTGATGTAAGTTTA
    ATAAAGGGTGAGATAATGTTTAACTTGCAT
    GGCGTGTTAAATGGGGCGGGGCTTAAAGGG
    TATATAATGCGCCGTGGGCTAATCTTGGTT
    Figure US20190142967A1-20190516-C00001
    Figure US20190142967A1-20190516-C00002
    Figure US20190142967A1-20190516-C00003
    GAAGGTCATCAAGACTTTGGATTTTTCCAC
    ACCGGGGCGCGCTGCGGCTGCTGTTGCTTT
    TTTGAGTTTTATAAAGGATAAATGGAGCGA
    AGAAACCCATCTGAGCGGGGGGTACCTGCT
    GGATTTTCTGGCCATGCATCTGTGGAGAGC
    GGTTGTGAGACACAAGAATCGCCTGCTACT
    GTTGTCTTCCGTCCGCCCGGCGATAATACC
    GACGGAGGAGCAGCAGCAGCAGCAGGAGG
    AAGCCAGGCGGCGGCGGCAGGAGCAGAGC
    CCATGGAACCCGAGAGCCGGCCTGGACCCT
    CGGGAATGAATGTTGTACAGGTGGCTGAAC
    TGTATCCAGAACTGAGACGCATTTTGACAA
    TTACAGAGGATGGGCAGGGGCTAAAGGGG
    GTAAAGAGGGAGCGGGGGGCTTGTGAGGC
    TACAGAGGAGGCTAGGAATCTAGCTTTTAG
    CTTAATGACCAGACACCGTCCTGAGTGTAT
    TACTTTTCAACAGATCAAGGATAATTGCGC
    TAATGAGCTTGATCTGCTGGCGCAGAAGTA
    TTCCATAGAGCAGCTGACCACTTACTGGCT
    GCAGCCAGGGGATGATTTTGAGGAGGCTAT
    TAGGGTATATGCAAAGGTGGCACTTAGGCC
    AGATTGCAAGTACAAGATCAGCAAACTTGT
    AAATATCAGGAATTGTTGCTACATTTCTGG
    GAACGGGGCCGAGGTGGAGATAGATACGG
    AGGATAGGGTGGCCTTTAGATGTAGCATGA
    TAAATATGTGGCCGGGGGTGCTTGGCATGG
    ACGGGGTGGTTATTATGAATGTAAGGTTTA
    CTGGCCCCAATTTTAGCGGTACGGTTTTCCT
    GGCCAATACCAACCTTATCCTACACGGTGT
    AAGCTTCTATGGGTTTAACAATACCTGTGT
    GGAAGCCTGGACCGATGTAAGGGTTCGGGG
    CTGTGCCTTTTACTGCTGCTGGAAGGGGGT
    GGTGTGTCGCCCCAAAAGCAGGGCTTCAAT
    TAAGAAATGCCTCTTTGAAAGGTGTACCTT
    GGGTATCCTGTCTGAGGGTAACTCCAGGGT
    GCGCCACAATGTGGCCTCCGACTGTGGTTG
    CTTCATGCTAGTGAAAAGCGTGGCTGTGAT
    TAAGCATAACATGGTATGTGGCAACTGCGA
    GGACAGGGCCTCTCAGATGCTGACCTGCTC
    GGACGGCAACTGTCACCTGCTGAAGACCAT
    TCACGTAGCCAGCCACTCTCGCAAGGCCTG
    GCCAGTGTTTGAGCATAACATACTGACCCG
    CTGTTCCTTGCATTTGGGTAACAGGAGGGG
    GGTGTTCCTACCTTACCAATGCAATTTGAGT
    CACACTAAGATATTGCTTGAGCCCGAGAGC
    ATGTCCAAGGTGAACCTGAACGGGGTGTTT
    GACATGACCATGAAGATCTGGAAGGTGCTG
    AGGTACGATGAGACCCGCACCAGGTGCAG
    ACCCTGCGAGTGTGGCGGTAAACATATTAG
    GAACCAGCCTGTGATGCTGGATGTGACCGA
    GGAGCTGAGGCCCGATCACTTGGTGCTGGC
    CTGCACCCGCGCTGAGTTTGGCTCTAGCGA
    TGAAGATACAGATTGAGGTACTGAAATGTG
    TGGGCGTGGCTTAAGGGTGGGAAAGAATAT
    ATAAGGTGGGGGTCTTATGTAGTTTTGTATC
    TGTTTTGCAGCAGCCGCCGCCGCCATGAGC
    ACCAACTCGTTTGATGGAAGCATTGTGAGC
    TCATATTTGACAACGCGCATGCCCCCATGG
    GCCGGGGTGCGTCAGAATGTGATGGGCTCC
    AGCATTGATGGTCGCCCCGTCCTGCCCGCA
    AACTCTACTACCTTGACCTACGAGACCGTG
    TCTGGAACGCCGTTGGAGACTGCAGCCTCC
    GCCGCCGCTTCAGCCGCTGCAGCCACCGCC
    CGCGGGATTGTGACTGACTTTGCTTTCCTGA
    GCCCGCTTGCAAGCAGTGCAGCTTCCCGTT
    CATCCGCCCGCGATGACAAGTTGACGGCTC
    TTTTGGCACAATTGGATTCTTTGACCCGGGA
    ACTTAATGTCGTTTCTCAGCAGCTGTTGGAT
    CTGCGCCAGCAGGTTTCTGCCCTGAAGGCT
    TCCTCCCCTCCCAATGCGGTTTAAAACATA
    AATAAAAAACCAGACTCTGTTTGGATTTGG
    ATCAAGCAAGTGTCTTGCTGTCTTTATTTAG
    GGGTTTTGCGCGCGCGGTAGGCCCGGGACC
    AGCGGTCTCGGTCGTTGAGGGTCCTGTGTA
    TTTTTTCCAGGACGTGGTAAAGGTGACTCT
    GGATGTTCAGATACATGGGCATAAGCCCGT
    CTCTGGGGTGGAGGTAGCACCACTGCAGAG
    CTTCATGCTGCGGGGTGGTGTTGTAGATGA
    TCCAGTCGTAGCAGGAGCGCTGGGCGTGGT
    GCCTAAAAATGTCTTTCAGTAGCAAGCTGA
    TTGCCAGGGGCAGGCCCTTGGTGTAAGTGT
    TTACAAAGCGGTTAAGCTGGGATGGGTGCA
    TACGTGGGGATATGAGATGCATCTTGGACT
    GTATTTTTAGGTTGGCTATGTTCCCAGCCAT
    ATCCCTCCGGGGATTCATGTTGTGCAGAAC
    CACCAGCACAGTGTATCCGGTGCACTTGGG
    AAATTTGTCATGTAGCTTAGAAGGAAATGC
    GTGGAAGAACTTGGAGACGCCCTTGTGACC
    TCCAAGATTTTCCATGCATTCGTCCATAATG
    ATGGCAATGGGCCCACGGGCGGCGGCCTGG
    GCGAAGATATTTCTGGGATCACTAACGTCA
    TAGTTGTGTTCCAGGATGAGATCGTCATAG
    GCCATTTTTACAAAGCGCGGGCGGAGGGTG
    CCAGACTGCGGTATAATGGTTCCATCCGGC
    CCAGGGGCGTAGTTACCCTCACAGATTTGC
    ATTTCCCACGCTTTGAGTTCAGATGGGGGG
    ATCATGTCTACCTGCGGGGCGATGAAGAAA
    ACGGTTTCCGGGGTAGGGGAGATCAGCTGG
    GAAGAAAGCAGGTTCCTGAGCAGCTGCGAC
    TTACCGCAGCCGGTGGGCCCGTAAATCACA
    CCTATTACCGGCTGCAACTGGTAGTTAAGA
    GAGCTGCAGCTGCCGTCATCCCTGAGCAGG
    GGGGCCACTTCGTTAAGCATGTCCCTGACT
    CGCATGTTTTCCCTGACCAAATCCGCCAGA
    AGGCGCTCGCCGCCCAGCGATAGCAGTTCT
    TGCAAGGAAGCAAAGTTTTTCAACGGTTTG
    AGACCGTCCGCCGTAGGCATGCTTTTGAGC
    GTTTGACCAAGCAGTTCCAGGCGGTCCCAC
    AGCTCGGTCACCTGCTCTACGGCATCTCGA
    TCCAGCATATCTCCTCGTTTCGCGGGTTGGG
    GCGGCTTTCGCTGTACGGCAGTAGTCGGTG
    CTCGTCCAGACGGGCCAGGGTCATGTCTTT
    CCACGGGCGCAGGGTCCTCGTCAGCGTAGT
    CTGGGTCACGGTGAAGGGGTGCGCTCCGGG
    CTGCGCGCTGGCCAGGGTGCGCTTGAGGCT
    GGTCCTGCTGGTGCTGAAGCGCTGCCGGTC
    TTCGCCCTGCGCGTCGGCCAGGTAGCATTT
    GACCATGGTGTCATAGTCCAGCCCCTCCGC
    GGCGTGGCCCTTGGCGCGCAGCTTGCCCTT
    GGAGGAGGCGCCGCACGAGGGGCAGTGCA
    GACTTTTGAGGGCGTAGAGCTTGGGCGCGA
    GAAATACCGATTCCGGGGAGTAGGCATCCG
    CGCCGCAGGCCCCGCAGACGGTCTCGCATT
    CCACGAGCCAGGTGAGCTCTGGCCGTTCGG
    GGTCAAAAACCAGGTTTCCCCCATGCTTTTT
    GATGCGTTTCTTACCTCTGGTTTCCATGAGC
    CGGTGTCCACGCTCGGTGACGAAAAGGCTG
    TCCGTGTCCCCGTATACAGACTTGAGAGGC
    CTGTCCTCGAGCGGTGTTCCGCGGTCCTCCT
    CGTATAGAAACTCGGACCACTCTGAGACAA
    AGGCTCGCGTCCAGGCCAGCACGAAGGAG
    GCTAAGTGGGAGGGGTAGCGGTCGTTGTCC
    ACTAGGGGGTCCACTCGCTCCAGGGTGTGA
    AGACACATGTCGCCCTCTTCGGCATCAAGG
    AAGGTGATTGGTTTGTAGGTGTAGGCCACG
    TGACCGGGTGTTCCTGAAGGGGGGCTATAA
    AAGGGGGTGGGGGCGCGTTCGTCCTCACTC
    TCTTCCGCATCGCTGTCTGCGAGGGCCAGC
    TGTTGGGGTGAGTACTCCCTCTGAAAAGCG
    GGCATGACTTCTGCGCTAAGATTGTCAGTTT
    CCAAAAACGAGGAGGATTTGATATTCACCT
    GGCCCGCGGTGATGCCTTTGAGGGTGGCCG
    CATCCATCTGGTCAGAAAAGACAATCTTTT
    TGTTGTCAAGCTTGGTGGCAAACGACCCGT
    AGAGGGCGTTGGACAGCAACTTGGCGATGG
    AGCGCAGGGTTTGGTTTTTGTCGCGATCGG
    CGCGCTCCTTGGCCGCGATGTTTAGCTGCA
    CGTATTCGCGCGCAACGCACCGCCATTCGG
    GAAAGACGGTGGTGCGCTCGTCGGGCACCA
    GGTGCACGCGCCAACCGCGGTTGTGCAGGG
    TGACAAGGTCAACGCTGGTGGCTACCTCTC
    CGCGTAGGCGCTCGTTGGTCCAGCAGAGGC
    GGCCGCCCTTGCGCGAGCAGAATGGCGGTA
    GGGGGTCTAGCTGCGTCTCGTCCGGGGGGT
    CTGCGTCCACGGTAAAGACCCCGGGCAGCA
    GGCGCGCGTCGAAGTAGTCTATCTTGCATC
    CTTGCAAGTCTAGCGCCTGCTGCCATGCGC
    GGGCGGCAAGCGCGCGCTCGTATGGGTTGA
    GTGGGGGACCCCATGGCATGGGGTGGGTGA
    GCGCGGAGGCGTACATGCCGCAAATGTCGT
    AAACGTAGAGGGGCTCTCTGAGTATTCCAA
    GATATGTAGGGTAGCATCTTCCACCGCGGA
    TGCTGGCGCGCACGTAATCGTATAGTTCGT
    GCGAGGGAGCGAGGAGGTCGGGACCGAGG
    TTGCTACGGGCGGGCTGCTCTGCTCGGAAG
    ACTATCTGCCTGAAGATGGCATGTGAGTTG
    GATGATATGGTTGGACGCTGGAAGACGTTG
    AAGCTGGCGTCTGTGAGACCTACCGCGTCA
    CGCACGAAGGAGGCGTAGGAGTCGCGCAG
    CTTGTTGACCAGCTCGGCGGTGACCTGCAC
    GTCTAGGGCGCAGTAGTCCAGGGTTTCCTT
    GATGATGTCATACTTATCCTGTCCCTTTTTT
    TTCCACAGCTCGCGGTTGAGGACAAACTCT
    TCGCGGTCTTTCCAGTACTCTTGGATCGGAA
    ACCCGTCGGCCTCCGAACGGTAAGAGCCTA
    GCATGTAGAACTGGTTGACGGCCTGGTAGG
    CGCAGCATCCCTTTTCTACGGGTAGCGCGT
    ATGCCTGCGCGGCCTTCCGGAGCGAGGTGT
    GGGTGAGCGCAAAGGTGTCCCTGACCATGA
    CTTTGAGGTACTGGTATTTGAAGTCAGTGTC
    GTCGCATCCGCCCTGCTCCCAGAGCAAAAA
    GTCCGTGCGCTTTTTGGAACGCGGATTTGG
    CAGGGCGAAGGTGACATCGTTGAAGAGTAT
    CTTTCCCGCGCGAGGCATAAAGTTGCGTGT
    GATGCGGAAGGGTCCCGGCACCTCGGAACG
    GTTGTTAATTACCTGGGCGGCGAGCACGAT
    CTCGTCAAAGCCGTTGATGTTGTGGCCCAC
    AATGTAAAGTTCCAAGAAGCGCGGGATGCC
    CTTGATGGAAGGCAATTTTTTAAGTTCCTCG
    TAGGTGAGCTCTTCAGGGGAGCTGAGCCCG
    TGCTCTGAAAGGGCCCAGTCTGCAAGATGA
    GGGTTGGAAGCGACGAATGAGCTCCACAG
    GTCACGGGCCATTAGCATTTGCAGGTGGTC
    GCGAAAGGTCCTAAACTGGCGACCTATGGC
    CATTTTTTCTGGGGTGATGCAGTAGAAGGT
    AAGCGGGTCTTGTTCCCAGCGGTCCCATCC
    AAGGTTCGCGGCTAGGTCTCGCGCGGCAGT
    CACTAGAGGCTCATCTCCGCCGAACTTCAT
    GACCAGCATGAAGGGCACGAGCTGCTTCCC
    AAAGGCCCCCATCCAAGTATAGGTCTCTAC
    ATCGTAGGTGACAAAGAGACGCTCGGTGCG
    AGGATGCGAGCCGATCGGGAAGAACTGGA
    TCTCCCGCCACCAATTGGAGGAGTGGCTAT
    TGATGTGGTGAAAGTAGAAGTCCCTGCGAC
    GGGCCGAACACTCGTGCTGGCTTTTGTAAA
    AACGTGCGCAGTACTGGCAGCGGTGCACGG
    GCTGTACATCCTGCACGAGGTTGACCTGAC
    GACCGCGCACAAGGAAGCAGAGTGGGAAT
    TTGAGCCCCTCGCCTGGCGGGTTTGGCTGG
    TGGTCTTCTACTTCGGCTGCTTGTCCTTGAC
    CGTCTGGCTGCTCGAGGGGAGTTACGGTGG
    ATCGGACCACCACGCCGCGCGAGCCCAAAG
    TCCAGATGTCCGCGCGCGGCGGTCGGAGCT
    TGATGACAACATCGCGCAGATGGGAGCTGT
    CCATGGTCTGGAGCTCCCGCGGCGTCAGGT
    CAGGCGGGAGCTCCTGCAGGTTTACCTCGC
    ATAGACGGGTCAGGGCGCGGGCTAGATCCA
    GGTGATACCTAATTTCCAGGGGCTGGTTGG
    TGGCGGCGTCGATGGCTTGCAAGAGGCCGC
    ATCCCCGCGGCGCGACTACGGTACCGCGCG
    GCGGGCGGTGGGCCGCGGGGGTGTCCTTGG
    ATGATGCATCTAAAAGCGGTGACGCGGGCG
    AGCCCCCGGAGGTAGGGGGGGCTCCGGAC
    CCGCCGGGAGAGGGGGCAGGGGCACGTCG
    GCGCCGCGCGCGGGCAGGAGCTGGTGCTGC
    GCGCGTAGGTTGCTGGCGAACGCGACGACG
    CGGCGGTTGATCTCCTGAATCTGGCGCCTCT
    GCGTGAAGACGACGGGCCCGGTGAGCTTGA
    ACCTGAAAGAGAGTTCGACAGAATCAATTT
    CGGTGTCGTTGACGGCGGCCTGGCGCAAAA
    TCTCCTGCACGTCTCCTGAGTTGTCTTGATA
    GGCGATCTCGGCCATGAACTGCTCGATCTC
    TTCCTCCTGGAGATCTCCGCGTCCGGCTCGC
    TCCACGGTGGCGGCGAGGTCGTTGGAAATG
    CGGGCCATGAGCTGCGAGAAGGCGTTGAG
    GCCTCCCTCGTTCCAGACGCGGCTGTAGAC
    CACGCCCCCTTCGGCATCGCGGGCGCGCAT
    GACCACCTGCGCGAGATTGAGCTCCACGTG
    CCGGGCGAAGACGGCGTAGTTTCGCAGGCG
    CTGAAAGAGGTAGTTGAGGGTGGTGGCGGT
    GTGTTCTGCCACGAAGAAGTACATAACCCA
    GCGTCGCAACGTGGATTCGTTGATATCCCC
    CAAGGCCTCAAGGCGCTCCATGGCCTCGTA
    GAAGTCCACGGCGAAGTTGAAAAACTGGG
    AGTTGCGCGCCGACACGGTTAACTCCTCCT
    CCAGAAGACGGATGAGCTCGGCGACAGTGT
    CGCGCACCTCGCGCTCAAAGGCTACAGGGG
    CCTCTTCTTCTTCTTCAATCTCCTCTTCCATA
    AGGGCCTCCCCTTCTTCTTCTTCTGGCGGCG
    GTGGGGGAGGGGGGACACGGCGGCGACGA
    CGGCGCACCGGGAGGCGGTCGACAAAGCG
    CTCGATCATCTCCCCGCGGCGACGGCGCAT
    GGTCTCGGTGACGGCGCGGCCGTTCTCGCG
    GGGGCGCAGTTGGAAGACGCCGCCCGTCAT
    GTCCCGGTTATGGGTTGGCGGGGGGCTGCC
    ATGCGGCAGGGATACGGCGCTAACGATGCA
    TCTCAACAATTGTTGTGTAGGTACTCCGCCG
    CCGAGGGACCTGAGCGAGTCCGCATCGACC
    GGATCGGAAAACCTCTCGAGAAAGGCGTCT
    AACCAGTCACAGTCGCAAGGTAGGCTGAGC
    ACCGTGGCGGGCGGCAGCGGGCGGCGGTC
    GGGGTTGTTTCTGGCGGAGGTGCTGCTGAT
    GATGTAATTAAAGTAGGCGGTCTTGAGACG
    GCGGATGGTCGACAGAAGCACCATGTCCTT
    GGGTCCGGCCTGCTGAATGCGCAGGCGGTC
    GGCCATGCCCCAGGCTTCGTTTTGACATCG
    GCGCAGGTCTTTGTAGTAGTCTTGCATGAG
    CCTTTCTACCGGCACTTCTTCTTCTCCTTCCT
    CTTGTCCTGCATCTCTTGCATCTATCGCTGC
    GGCGGCGGCGGAGTTTGGCCGTAGGTGGCG
    CCCTCTTCCTCCCATGCGTGTGACCCCGAAG
    CCCCTCATCGGCTGAAGCAGGGCTAGGTCG
    GCGACAACGCGCTCGGCTAATATGGCCTGC
    TGCACCTGCGTGAGGGTAGACTGGAAGTCA
    TCCATGTCCACAAAGCGGTGGTATGCGCCC
    GTGTTGATGGTGTAAGTGCAGTTGGCCATA
    ACGGACCAGTTAACGGTCTGGTGACCCGGC
    TGCGAGAGCTCGGTGTACCTGAGACGCGAG
    TAAGCCCTCGAGTCAAATACGTAGTCGTTG
    CAAGTCCGCACCAGGTACTGGTATCCCACC
    AAAAAGTGCGGCGGCGGCTGGCGGTAGAG
    GGGCCAGCGTAGGGTGGCCGGGGCTCCGG
    GGGCGAGATCTTCCAACATAAGGCGATGAT
    ATCCGTAGATGTACCTGGACATCCAGGTGA
    TGCCGGCGGCGGTGGTGGAGGCGCGCGGA
    AAGTCGCGGACGCGGTTCCAGATGTTGCGC
    AGCGGCAAAAAGTGCTCCATGGTCGGGACG
    CTCTGGCCGGTCAGGCGCGCGCAATCGTTG
    ACGCTCTAGCGTGCAAAAGGAGAGCCTGTA
    AGCGGGCACTCTTCCGTGGTCTGGTGGATA
    AATTCGCAAGGGTATCATGGCGGACGACCG
    GGGTTCGAGCCCCGTATCCGGCCGTCCGCC
    GTGATCCATGCGGTTACCGCCCGCGTGTCG
    AACCCAGGTGTGCGACGTCAGACAACGGG
    GGAGTGCTCCTTTTGGCTTCCTTCCAGGCGC
    GGCGGCTGCTGCGCTAGCTTTTTTGGCCACT
    GGCCGCGCGCAGCGTAAGCGGTTAGGCTGG
    AAAGCGAAAGCATTAAGTGGCTCGCTCCCT
    GTAGCCGGAGGGTTATTTTCCAAGGGTTGA
    GTCGCGGGACCCCCGGTTCGAGTCTCGGAC
    CGGCCGGACTGCGGCGAACGGGGGTTTGCC
    TCCCCGTCATGCAAGACCCCGCTTGCAAAT
    TCCTCCGGAAACAGGGACGAGCCCCTTTTT
    TGCTTTTCCCAGATGCATCCGGTGCTGCGGC
    AGATGCGCCCCCCTCCTCAGCAGCGGCAAG
    AGCAAGAGCAGCGGCAGACATGCAGGGCA
    CCCTCCCCTCCTCCTACCGCGTCAGGAGGG
    GCGACATCCGCGGTTGACGCGGCAGCAGAT
    GGTGATTACGAACCCCCGCGGCGCCGGGCC
    CGGCACTACCTGGACTTGGAGGAGGGCGAG
    GGCCTGGCGCGGCTAGGAGCGCCCTCTCCT
    GAGCGGCACCCAAGGGTGCAGCTGAAGCG
    TGATACGCGTGAGGCGTACGTGCCGCGGCA
    GAACCTGTTTCGCGACCGCGAGGGAGAGGA
    GCCCGAGGAGATGCGGGATCGAAAGTTCCA
    CGCAGGGCGCGAGCTGCGGCATGGCCTGAA
    TCGCGAGCGGTTGCTGCGCGAGGAGGACTT
    TGAGCCCGACGCGCGAACCGGGATTAGTCC
    CGCGCGCGCACACGTGGCGGCCGCCGACCT
    GGTAACCGCATACGAGCAGACGGTGAACC
    AGGAGATTAACTTTCAAAAAAGCTTTAACA
    ACCACGTGCGTACGCTTGTGGCGCGCGAGG
    AGGTGGCTATAGGACTGATGCATCTGTGGG
    ACTTTGTAAGCGCGCTGGAGCAAAACCCAA
    ATAGCAAGCCGCTCATGGCGCAGCTGTTCC
    TTATAGTGCAGCACAGCAGGGACAACGAG
    GCATTCAGGGATGCGCTGCTAAACATAGTA
    GAGCCCGAGGGCCGCTGGCTGCTCGATTTG
    ATAAACATCCTGCAGAGCATAGTGGTGCAG
    GAGCGCAGCTTGAGCCTGGCTGACAAGGTG
    GCCGCCATCAACTATTCCATGCTTAGCCTG
    GGCAAGTTTTACGCCCGCAAGATATACCAT
    ACCCCTTACGTTCCCATAGACAAGGAGGTA
    AAGATCGAGGGGTTCTACATGCGCATGGCG
    CTGAAGGTGCTTACCTTGAGCGACGACCTG
    GGCGTTTATCGCAACGAGCGCATCCACAAG
    GCCGTGAGCGTGAGCCGGCGGCGCGAGCTC
    AGCGACCGCGAGCTGATGCACAGCCTGCAA
    AGGGCCCTGGCTGGCACGGGCAGCGGCGAT
    AGAGAGGCCGAGTCCTACTTTGACGCGGGC
    GCTGACCTGCGCTGGGCCCCAAGCCGACGC
    GCCCTGGAGGCAGCTGGGGCCGGACCTGGG
    CTGGCGGTGGCACCCGCGCGCGCTGGCAAC
    GTCGGCGGCGTGGAGGAATATGACGAGGA
    CGATGAGTACGAGCCAGAGGACGGCGAGT
    ACTAAGCGGTGATGTTTCTGATCAGATGAT
    GCAAGACGCAACGGACCCGGCGGTGCGGG
    CGGCGCTGCAGAGCCAGCCGTCCGGCCTTA
    ACTCCACGGACGACTGGCGCCAGGTCATGG
    ACCGCATCATGTCGCTGACTGCGCGCAATC
    CTGACGCGTTCCGGCAGCAGCCGCAGGCCA
    ACCGGCTCTCCGCAATTCTGGAAGCGGTGG
    TCCCGGCGCGCGCAAACCCCACGCACGAGA
    AGGTGCTGGCGATCGTAAACGCGCTGGCCG
    AAAACAGGGCCATCCGGCCCGACGAGGCC
    GGCCTGGTCTACGACGCGCTGCTTCAGCGC
    GTGGCTCGTTACAACAGCGGCAACGTGCAG
    ACCAACCTGGACCGGCTGGTGGGGGATGTG
    CGCGAGGCCGTGGCGCAGCGTGAGCGCGC
    GCAGCAGCAGGGCAACCTGGGCTCCATGGT
    TGCACTAAACGCCTTCCTGAGTACACAGCC
    CGCCAACGTGCCGCGGGGACAGGAGGACT
    ACACCAACTTTGTGAGCGCACTGCGGCTAA
    TGGTGACTGAGACACCGCAAAGTGAGGTGT
    ACCAGTCTGGGCCAGACTATTTTTTCCAGA
    CCAGTAGACAAGGCCTGCAGACCGTAAACC
    TGAGCCAGGCTTTCAAAAACTTGCAGGGGC
    TGTGGGGGGTGCGGGCTCCCACAGGCGACC
    GCGCGACCGTGTCTAGCTTGCTGACGCCCA
    ACTCGCGCCTGTTGCTGCTGCTAATAGCGC
    CCTTCACGGACAGTGGCAGCGTGTCCCGGG
    ACACATACCTAGGTCACTTGCTGACACTGT
    ACCGCGAGGCCATAGGTCAGGCGCATGTGG
    ACGAGCATACTTTCCAGGAGATTACAAGTG
    TCAGCCGCGCGCTGGGGCAGGAGGACACG
    GGCAGCCTGGAGGCAACCCTAAACTACCTG
    CTGACCAACCGGCGGCAGAAGATCCCCTCG
    TTGCACAGTTTAAACAGCGAGGAGGAGCGC
    ATTTTGCGCTACGTGCAGCAGAGCGTGAGC
    CTTAACCTGATGCGCGACGGGGTAACGCCC
    AGCGTGGCGCTGGACATGACCGCGCGCAAC
    ATGGAACCGGGCATGTATGCCTCAAACCGG
    CCGTTTATCAACCGCCTAATGGACTACTTGC
    ATCGCGCGGCCGCCGTGAACCCCGAGTATT
    TCACCAATGCCATCTTGAACCCGCACTGGC
    TACCGCCCCCTGGTTTCTACACCGGGGGAT
    TCGAGGTGCCCGAGGGTAACGATGGATTCC
    TCTGGGACGACATAGACGACAGCGTGTTTT
    CCCCGCAACCGCAGACCCTGCTAGAGTTGC
    AACAGCGCGAGCAGGCAGAGGCGGCGCTG
    CGAAAGGAAAGCTTCCGCAGGCCAAGCAG
    CTTGTCCGATCTAGGCGCTGCGGCCCCGCG
    GTCAGATGCTAGTAGCCCATTTCCAAGCTT
    GATAGGGTCTCTTACCAGCACTCGCACCAC
    CCGCCCGCGCCTGCTGGGCGAGGAGGAGTA
    CCTAAACAACTCGCTGCTGCAGCCGCAGCG
    CGAAAAAAACCTGCCTCCGGCATTTCCCAA
    CAACGGGATAGAGAGCCTAGTGGACAAGA
    TGAGTAGATGGAAGACGTACGCGCAGGAG
    CACAGGGACGTGCCAGGCCCGCGCCCGCCC
    ACCCGTCGTCAAAGGCACGACCGTCAGCGG
    GGTCTGGTGTGGGAGGACGATGACTCGGCA
    GACGACAGCAGCGTCCTGGATTTGGGAGGG
    AGTGGCAACCCGTTTGCGCACCTTCGCCCC
    AGGCTGGGGAGAATGTTTTAAAAAAAAAA
    AAGCATGATGCAAAATAAAAAACTCACCA
    AGGCCATGGCACCGAGCGTTGGTTTTCTTG
    TATTCCCCTTAGTATGCGGCGCGCGGCGAT
    GTATGAGGAAGGTCCTCCTCCCTCCTACGA
    GAGTGTGGTGAGCGCGGCGCCAGTGGCGGC
    GGCGCTGGGTTCTCCCTTCGATGCTCCCCTG
    GACCCGCCGTTTGTGCCTCCGCGGTACCTG
    CGGCCTACCGGGGGGAGAAACAGCATCCGT
    TACTCTGAGTTGGCACCCCTATTCGACACC
    ACCCGTGTGTACCTGGTGGACAACAAGTCA
    ACGGATGTGGCATCCCTGAACTACCAGAAC
    GACCACAGCAACTTTCTGACCACGGTCATT
    CAAAACAATGACTACAGCCCGGGGGAGGC
    AAGCACACAGACCATCAATCTTGACGACCG
    GTCGCACTGGGGCGGCGACCTGAAAACCAT
    CCTGCATACCAACATGCCAAATGTGAACGA
    GTTCATGTTTACCAATAAGTTTAAGGCGCG
    GGTGATGGTGTCGCGCTTGCCTACTAAGGA
    CAATCAGGTGGAGCTGAAATACGAGTGGGT
    GGAGTTCACGCTGCCCGAGGGCAACTACTC
    CGAGACCATGACCATAGACCTTATGAACAA
    CGCGATCGTGGAGCACTACTTGAAAGTGGG
    CAGACAGAACGGGGTTCTGGAAAGCGACA
    TCGGGGTAAAGTTTGACACCCGCAACTTCA
    GACTGGGGTTTGACCCCGTCACTGGTCTTGT
    CATGCCTGGGGTATATACAAACGAAGCCTT
    CCATCCAGACATCATTTTGCTGCCAGGATG
    CGGGGTGGACTTCACCCACAGCCGCCTGAG
    CAACTTGTTGGGCATCCGCAAGCGGCAACC
    CTTCCAGGAGGGCTTTAGGATCACCTACGA
    TGATCTGGAGGGTGGTAACATTCCCGCACT
    GTTGGATGTGGACGCCTACCAGGCGAGCTT
    GAAAGATGACACCGAACAGGGCGGGGGTG
    GCGCAGGCGGCAGCAACAGCAGTGGCAGC
    GGCGCGGAAGAGAACTCCAACGCGGCAGC
    CGCGGCAATGCAGCCGGTGGAGGACATGA
    ACGATCATGCCATTCGCGGCGACACCTTTG
    CCACACGGGCTGAGGAGAAGCGCGCTGAG
    GCCGAAGCAGCGGCCGAAGCTGCCGCCCCC
    GCTGCGCAACCCGAGGTCGAGAAGCCTCAG
    AAGAAACCGGTGATCAAACCCCTGACAGA
    GGACAGCAAGAAACGCAGTTACAACCTAAT
    AAGCAATGACAGCACCTTCACCCAGTACCG
    CAGCTGGTACCTTGCATACAACTACGGCGA
    CCCTCAGACCGGAATCCGCTCATGGACCCT
    GCTTTGCACTCCTGACGTAACCTGCGGCTC
    GGAGCAGGTCTACTGGTCGTTGCCAGACAT
    GATGCAAGACCCCGTGACCTTCCGCTCCAC
    GCGCCAGATCAGCAACTTTCCGGTGGTGGG
    CGCCGAGCTGTTGCCCGTGCACTCCAAGAG
    CTTCTACAACGACCAGGCCGTCTACTCCCA
    ACTCATCCGCCAGTTTACCTCTCTGACCCAC
    GTGTTCAATCGCTTTCCCGAGAACCAGATTT
    TGGCGCGCCCGCCAGCCCCCACCATCACCA
    CCGTCAGTGAAAACGTTCCTGCTCTCACAG
    ATCACGGGACGCTACCGCTGCGCAACAGCA
    TCGGAGGAGTCCAGCGAGTGACCATTACTG
    ACGCCAGACGCCGCACCTGCCCCTACGTTT
    ACAAGGCCCTGGGCATAGTCTCGCCGCGCG
    TCCTATCGAGCCGCACTTTTTGAGCAAGCA
    TGTCCATCCTTATATCGCCCAGCAATAACA
    CAGGCTGGGGCCTGCGCTTCCCAAGCAAGA
    TGTTTGGCGGGGCCAAGAAGCGCTCCGACC
    AACACCCAGTGCGCGTGCGCGGGCACTACC
    GCGCGCCCTGGGGCGCGCACAAACGCGGCC
    GCACTGGGCGCACCACCGTCGATGACGCCA
    TCGACGCGGTGGTGGAGGAGGCGCGCAACT
    ACACGCCCACGCCGCCACCAGTGTCCACAG
    TGGACGCGGCCATTCAGACCGTGGTGCGCG
    GAGCCCGGCGCTATGCTAAAATGAAGAGAC
    GGCGGAGGCGCGTAGCACGTCGCCACCGCC
    GCCGACCCGGCACTGCCGCCCAACGCGCGG
    CGGCGGCCCTGCTTAACCGCGCACGTCGCA
    CCGGCCGACGGGCGGCCATGCGGGCCGCTC
    GAAGGCTGGCCGCGGGTATTGTCACTGTGC
    CCCCCAGGTCCAGGCGACGAGCGGCCGCCG
    CAGCAGCCGCGGCCATTAGTGCTATGACTC
    AGGGTCGCAGGGGCAACGTGTATTGGGTGC
    GCGACTCGGTTAGCGGCCTGCGCGTGCCCG
    TGCGCACCCGCCCCCCGCGCAACTAGATTG
    CAAGAAAAAACTACTTAGACTCGTACTGTT
    GTATGTATCCAGCGGCGGCGGCGCGCAACG
    AAGCTATGTCCAAGCGCAAAATCAAAGAA
    GAGATGCTCCAGGTCATCGCGCCGGAGATC
    TATGGCCCCCCGAAGAAGGAAGAGCAGGA
    TTACAAGCCCCGAAAGCTAAAGCGGGTCAA
    AAAGAAAAAGAAAGATGATGATGATGAAC
    TTGACGACGAGGTGGAACTGCTGCACGCTA
    CCGCGCCCAGGCGACGGGTACAGTGGAAA
    GGTCGACGCGTAAAACGTGTTTTGCGACCC
    GGCACCACCGTAGTCTTTACGCCCGGTGAG
    CGCTCCACCCGCACCTACAAGCGCGTGTAT
    GATGAGGTGTACGGCGACGAGGACCTGCTT
    GAGCAGGCCAACGAGCGCCTCGGGGAGTTT
    GCCTACGGAAAGCGGCATAAGGACATGCTG
    GCGTTGCCGCTGGACGAGGGCAACCCAACA
    CCTAGCCTAAAGCCCGTAACACTGCAGCAG
    GTGCTGCCCGCGCTTGCACCGTCCGAAGAA
    AAGCGCGGCCTAAAGCGCGAGTCTGGTGAC
    TTGGCACCCACCGTGCAGCTGATGGTACCC
    AAGCGCCAGCGACTGGAAGATGTCTTGGAA
    AAAATGACCGTGGAACCTGGGCTGGAGCCC
    GAGGTCCGCGTGCGGCCAATCAAGCAGGTG
    GCGCCGGGACTGGGCGTGCAGACCGTGGAC
    GTTCAGATACCCACTACCAGTAGCACCAGT
    ATTGCCACCGCCACAGAGGGCATGGAGACA
    CAAACGTCCCCGGTTGCCTCAGCGGTGGCG
    GATGCCGCGGTGCAGGCGGTCGCTGCGGCC
    GCGTCCAAGACCTCTACGGAGGTGCAAACG
    GACCCGTGGATGTTTCGCGTTTCAGCCCCCC
    GGCGCCCGCGCCGTTCGAGGAAGTACGGCG
    CCGCCAGCGCGCTACTGCCCGAATATGCCC
    TACATCCTTCCATTGCGCCTACCCCCGGCTA
    TCGTGGCTACACCTACCGCCCCAGAAGACG
    AGCAACTACCCGACGCCGAACCACCACTGG
    AACCCGCCGCCGCCGTCGCCGTCGCCAGCC
    CGTGCTGGCCCCGATTTCCGTGCGCAGGGT
    GGCTCGCGAAGGAGGCAGGACCCTGGTGCT
    GCCAACAGCGCGCTACCACCCCAGCATCGT
    TTAAAAGCCGGTCTTTGTGGTTCTTGCAGAT
    ATGGCCCTCACCTGCCGCCTCCGTTTCCCGG
    TGCCGGGATTCCGAGGAAGAATGCACCGTA
    GGAGGGGCATGGCCGGCCACGGCCTGACG
    GGCGGCATGCGTCGTGCGCACCACCGGCGG
    CGGCGCGCGTCGCACCGTCGCATGCGCGGC
    GGTATCCTGCCCCTCCTTATTCCACTGATCG
    CCGCGGCGATTGGCGCCGTGCCCGGAATTG
    CATCCGTGGCCTTGCAGGCGCAGAGACACT
    GATTAAAAACAAGTTGCATGTGGAAAAATC
    AAAATAAAAAGTCTGGACTCTCACGCTCGC
    TTGGTCCTGTAACTATTTTGTAGAATGGAA
    GACATCAACTTTGCGTCTCTGGCCCCGCGA
    CACGGCTCGCGCCCGTTCATGGGAAACTGG
    CAAGATATCGGCACCAGCAATATGAGCGGT
    GGCGCCTTCAGCTGGGGCTCGCTGTGGAGC
    GGCATTAAAAATTTCGGTTCCACCGTTAAG
    AACTATGGCAGCAAGGCCTGGAACAGCAG
    CACAGGCCAGATGCTGAGGGATAAGTTGAA
    AGAGCAAAATTTCCAACAAAAGGTGGTAG
    ATGGCCTGGCCTCTGGCATTAGCGGGGTGG
    TGGACCTGGCCAACCAGGCAGTGCAAAATA
    AGATTAACAGTAAGCTTGATCCCCGCCCTC
    CCGTAGAGGAGCCTCCACCGGCCGTGGAGA
    CAGTGTCTCCAGAGGGGCGTGGCGAAAAGC
    GTCCGCGCCCCGACAGGGAAGAAACTCTGG
    TGACGCAAATAGACGAGCCTCCCTCGTACG
    AGGAGGCACTAAAGCAAGGCCTGCCCACC
    ACCCGTCCCATCGCGCCCATGGCTACCGGA
    GTGCTGGGCCAGCACACACCCGTAACGCTG
    GACCTGCCTCCCCCCGCCGACACCCAGCAG
    AAACCTGTGCTGCCAGGCCCGACCGCCGTT
    GTTGTAACCCGTCCTAGCCGCGCGTCCCTG
    CGCCGCGCCGCCAGCGGTCCGCGATCGTTG
    CGGCCCGTAGCCAGTGGCAACTGGCAAAGC
    ACACTGAACAGCATCGTGGGTCTGGGGGTG
    CAATCCCTGAAGCGCCGACGATGCTTCTGA
    TAGCTAACGTGTCGTATGTGTGTCATGTATG
    CGTCCATGTCGCCGCCAGAGGAGCTGCTGA
    GCCGCCGCGCGCCCGCTTTCCAAGATGGCT
    ACCCCTTCGATGATGCCGCAGTGGTCTTAC
    ATGCACATCTCGGGCCAGGACGCCTCGGAG
    TACCTGAGCCCCGGGCTGGTGCAGTTTGCC
    CGCGCCACCGAGACGTACTTCAGCCTGAAT
    AACAAGTTTAGAAACCCCACGGTGGCGCCT
    ACGCACGACGTGACCACAGACCGGTCCCAG
    CGTTTGACGCTGCGGTTCATCCCTGTGGACC
    GTGAGGATACTGCGTACTCGTACAAGGCGC
    GGTTCACCCTAGCTGTGGGTGATAACCGTG
    TGCTGGACATGGCTTCCACGTACTTTGACAT
    CCGCGGCGTGCTGGACAGGGGCCCTACTTT
    TAAGCCCTACTCTGGCACTGCCTACAACGC
    CCTGGCTCCCAAGGGTGCCCCAAATCCTTG
    CGAATGGGATGAAGCTGCTACTGCTCTTGA
    AATAAACCTAGAAGAAGAGGACGATGACA
    ACGAAGACGAAGTAGACGAGCAAGCTGAG
    CAGCAAAAAACTCACGTATTTGGGCAGGCG
    CCTTATTCTGGTATAAATATTACAAAGGAG
    GGTATTCAAATAGGTGTCGAAGGTCAAACA
    CCTAAATATGCCGATAAAACATTTCAACCT
    GAACCTCAAATAGGAGAATCTCAGTGGTAC
    GAAACAGAAATTAATCATGCAGCTGGGAG
    AGTCCTAAAAAAGACTACCCCAATGAAACC
    ATGTTACGGTTCATATGCAAAACCCACAAA
    TGAAAATGGAGGGCAAGGCATTCTTGTAAA
    GCAACAAAATGGAAAGCTAGAAAGTCAAG
    TGGAAATGCAATTTTTCTCAACTACTGAGG
    CAGCCGCAGGCAATGGTGATAACTTGACTC
    CTAAAGTGGTATTGTACAGTGAAGATGTAG
    ATATAGAAACCCCAGACACTCATATTTCTT
    ACATGCCCACTATTAAGGAAGGTAACTCAC
    GAGAACTAATGGGCCAACAATCTATGCCCA
    ACAGGCCTAATTACATTGCTTTTAGGGACA
    ATTTTATTGGTCTAATGTATTACAACAGCAC
    GGGTAATATGGGTGTTCTGGCGGGCCAAGC
    ATCGCAGTTGAATGCTGTTGTAGATTTGCA
    AGACAGAAACACAGAGCTTTCATACCAGCT
    TTTGCTTGATTCCATTGGTGATAGAACCAG
    GTACTTTTCTATGTGGAATCAGGCTGTTGAC
    AGCTATGATCCAGATGTTAGAATTATTGAA
    AATCATGGAACTGAAGATGAACTTCCAAAT
    TACTGCTTTCCACTGGGAGGTGTGATTAAT
    ACAGAGACTCTTACCAAGGTAAAACCTAAA
    ACAGGTCAGGAAAATGGATGGGAAAAAGA
    TGCTACAGAATTTTCAGATAAAAATGAAAT
    AAGAGTTGGAAATAATTTTGCCATGGAAAT
    CAATCTAAATGCCAACCTGTGGAGAAATTT
    CCTGTACTCCAACATAGCGCTGTATTTGCCC
    GACAAGCTAAAGTACAGTCCTTCCAACGTA
    AAAATTTCTGATAACCCAAACACCTACGAC
    TACATGAACAAGCGAGTGGTGGCTCCCGGG
    CTAGTGGACTGCTACATTAACCTTGGAGCA
    CGCTGGTCCCTTGACTATATGGACAACGTC
    AACCCATTTAACCACCACCGCAATGCTGGC
    CTGCGCTACCGCTCAATGTTGCTGGGCAAT
    GGTCGCTATGTGCCCTTCCACATCCAGGTG
    CCTCAGAAGTTCTTTGCCATTAAAAACCTCC
    TTCTCCTGCCGGGCTCATACACCTACGAGT
    GGAACTTCAGGAAGGATGTTAACATGGTTC
    TGCAGAGCTCCCTAGGAAATGACCTAAGGG
    TTGACGGAGCCAGCATTAAGTTTGATAGCA
    TTTGCCTTTACGCCACCTTCTTCCCCATGGC
    CCACAACACCGCCTCCACGCTTGAGGCCAT
    GCTTAGAAACGACACCAACGACCAGTCCTT
    TAACGACTATCTCTCCGCCGCCAACATGCT
    CTACCCTATACCCGCCAACGCTACCAACGT
    GCCCATATCCATCCCCTCCCGCAACTGGGC
    GGCTTTCCGCGGCTGGGCCTTCACGCGCCTT
    AAGACTAAGGAAACCCCATCACTGGGCTCG
    GGCTACGACCCTTATTACACCTACTCTGGCT
    CTATACCCTACCTAGATGGAACCTTTTACCT
    CAACCACACCTTTAAGAAGGTGGCCATTAC
    CTTTGACTCTTCTGTCAGCTGGCCTGGCAAT
    GACCGCCTGCTTACCCCCAACGAGTTTGAA
    ATTAAGCGCTCAGTTGACGGGGAGGGTTAC
    AACGTTGCCCAGTGTAACATGACCAAAGAC
    TGGTTCCTGGTACAAATGCTAGCTAACTAT
    AACATTGGCTACCAGGGCTTCTATATCCCA
    GAGAGCTACAAGGACCGCATGTACTCCTTC
    TTTAGAAACTTCCAGCCCATGAGCCGTCAG
    GTGGTGGATGATACTAAATACAAGGACTAC
    CAACAGGTGGGCATCCTACACCAACACAAC
    AACTCTGGATTTGTTGGCTACCTTGCCCCCA
    CCATGCGCGAAGGACAGGCCTACCCTGCTA
    ACTTCCCCTATCCGCTTATAGGCAAGACCG
    CAGTTGACAGCATTACCCAGAAAAAGTTTC
    TTTGCGATCGCACCCTTTGGCGCATCCCATT
    CTCCAGTAACTTTATGTCCATGGGCGCACTC
    ACAGACCTGGGCCAAAACCTTCTCTACGCC
    AACTCCGCCCACGCGCTAGACATGACTTTT
    GAGGTGGATCCCATGGACGAGCCCACCCTT
    CTTTATGTTTTGTTTGAAGTCTTTGACGTGG
    TCCGTGTGCACCAGCCGCACCGCGGCGTCA
    TCGAAACCGTGTACCTGCGCACGCCCTTCT
    CGGCCGGCAACGCCACAACATAAAGAAGC
    AAGCAACATCAACAACAGCTGCCGCCATGG
    GCTCCAGTGAGCAGGAACTGAAAGCCATTG
    TCAAAGATCTTGGTTGTGGGCCATATTTTTT
    GGGCACCTATGACAAGCGCTTTCCAGGCTT
    TGTTTCTCCACACAAGCTCGCCTGCGCCATA
    GTCAATACGGCCGGTCGCGAGACTGGGGGC
    GTACACTGGATGGCCTTTGCCTGGAACCCG
    CACTCAAAAACATGCTACCTCTTTGAGCCC
    TTTGGCTTTTCTGACCAGCGACTCAAGCAG
    GTTTACCAGTTTGAGTACGAGTCACTCCTGC
    GCCGTAGCGCCATTGCTTCTTCCCCCGACCG
    CTGTATAACGCTGGAAAAGTCCACCCAAAG
    CGTACAGGGGCCCAACTCGGCCGCCTGTGG
    ACTATTCTGCTGCATGTTTCTCCACGCCTTT
    GCCAACTGGCCCCAAACTCCCATGGATCAC
    AACCCCACCATGAACCTTATTACCGGGGTA
    CCCAACTCCATGCTCAACAGTCCCCAGGTA
    CAGCCCACCCTGCGTCGCAACCAGGAACAG
    CTCTACAGCTTCCTGGAGCGCCACTCGCCCT
    ACTTCCGCAGCCACAGTGCGCAGATTAGGA
    GCGCCACTTCTTTTTGTCACTTGAAAAACAT
    GTAAAAATAATGTACTAGAGACACTTTCAA
    TAAAGGCAAATGCTTTTATTTGTACACTCTC
    GGGTGATTATTTACCCCCACCCTTGCCGTCT
    GCGCCGTTTAAAAATCAAAGGGGTTCTGCC
    GCGCATCGCTATGCGCCACTGGCAGGGACA
    CGTTGCGATACTGGTGTTTAGTGCTCCACTT
    AAACTCAGGCACAACCATCCGCGGCAGCTC
    GGTGAAGTTTTCACTCCACAGGCTGCGCAC
    CATCACCAACGCGTTTAGCAGGTCGGGCGC
    CGATATCTTGAAGTCGCAGTTGGGGCCTCC
    GCCCTGCGCGCGCGAGTTGCGATACACAGG
    GTTGCAGCACTGGAACACTATCAGCGCCGG
    GTGGTGCACGCTGGCCAGCACGCTCTTGTC
    GGAGATCAGATCCGCGTCCAGGTCCTCCGC
    GTTGCTCAGGGCGAACGGAGTCAACTTTGG
    TAGCTGCCTTCCCAAAAAGGGCGCGTGCCC
    AGGCTTTGAGTTGCACTCGCACCGTAGTGG
    CATCAAAAGGTGACCGTGCCCGGTCTGGGC
    GTTAGGATACAGCGCCTGCATAAAAGCCTT
    GATCTGCTTAAAAGCCACCTGAGCCTTTGC
    GCCTTCAGAGAAGAACATGCCGCAAGACTT
    GCCGGAAAACTGATTGGCCGGACAGGCCGC
    GTCGTGCACGCAGCACCTTGCGTCGGTGTT
    GGAGATCTGCACCACATTTCGGCCCCACCG
    GTTCTTCACGATCTTGGCCTTGCTAGACTGC
    TCCTTCAGCGCGCGCTGCCCGTTTTCGCTCG
    TCACATCCATTTCAATCACGTGCTCCTTATT
    TATCATAATGCTTCCGTGTAGACACTTAAG
    CTCGCCTTCGATCTCAGCGCAGCGGTGCAG
    CCACAACGCGCAGCCCGTGGGCTCGTGATG
    CTTGTAGGTCACCTCTGCAAACGACTGCAG
    GTACGCCTGCAGGAATCGCCCCATCATCGT
    CACAAAGGTCTTGTTGCTGGTGAAGGTCAG
    CTGCAACCCGCGGTGCTCCTCGTTCAGCCA
    GGTCTTGCATACGGCCGCCAGAGCTTCCAC
    TTGGTCAGGCAGTAGTTTGAAGTTCGCCTTT
    AGATCGTTATCCACGTGGTACTTGTCCATCA
    GCGCGCGCGCAGCCTCCATGCCCTTCTCCC
    ACGCAGACACGATCGGCACACTCAGCGGGT
    TCATCACCGTAATTTCACTTTCCGCTTCGCT
    GGGCTCTTCCTCTTCCTCTTGCGTCCGCATA
    CCACGCGCCACTGGGTCGTCTTCATTCAGC
    CGCCGCACTGTGCGCTTACCTCCTTTGCCAT
    GCTTGATTAGCACCGGTGGGTTGCTGAAAC
    CCACCATTTGTAGCGCCACATCTTCTCTTTC
    TTCCTCGCTGTCCACGATTACCTCTGGTGAT
    GGCGGGCGCTCGGGCTTGGGAGAAGGGCG
    CTTCTTTTTCTTCTTGGGCGCAATGGCCAAA
    TCCGCCGCCGAGGTCGATGGCCGCGGGCTG
    GGTGTGCGCGGCACCAGCGCGTCTTGTGAT
    GAGTCTTCCTCGTCCTCGGACTCGATACGCC
    GCCTCATCCGCTTTTTTGGGGGCGCCCGGG
    GAGGCGGCGGCGACGGGGACGGGGACGAC
    ACGTCCTCCATGGTTGGGGGACGTCGCGCC
    GCACCGCGTCCGCGCTCGGGGGTGGTTTCG
    CGCTGCTCCTCTTCCCGACTGGCCATTTCCT
    TCTCCTATAGGCAGAAAAAGATCATGGAGT
    CAGTCGAGAAGAAGGACAGCCTAACCGCC
    CCCTCTGAGTTCGCCACCACCGCCTCCACC
    GATGCCGCCAACGCGCCTACCACCTTCCCC
    GTCGAGGCACCCCCGCTTGAGGAGGAGGA
    AGTGATTATCGAGCAGGACCCAGGTTTTGT
    AAGCGAAGACGACGAGGACCGCTCAGTAC
    CAACAGAGGATAAAAAGCAAGACCAGGAC
    AACGCAGAGGCAAACGAGGAACAAGTCGG
    GCGGGGGGACGAAAGGCATGGCGACTACC
    TAGATGTGGGAGACGACGTGCTGTTGAAGC
    ATCTGCAGCGCCAGTGCGCCATTATCTGCG
    ACGCGTTGCAAGAGCGCAGCGATGTGCCCC
    TCGCCATAGCGGATGTCAGCCTTGCCTACG
    AACGCCACCTATTCTCACCGCGCGTACCCC
    CCAAACGCCAAGAAAACGGCACATGCGAG
    CCCAACCCGCGCCTCAACTTCTACCCCGTAT
    TTGCCGTGCCAGAGGTGCTTGCCACCTATC
    ACATCTTTTTCCAAAACTGCAAGATACCCCT
    ATCCTGCCGTGCCAACCGCAGCCGAGCGGA
    CAAGCAGCTGGCCTTGCGGCAGGGCGCTGT
    CATACCTGATATCGCCTCGCTCAACGAAGT
    GCCAAAAATCTTTGAGGGTCTTGGACGCGA
    CGAGAAGCGCGCGGCAAACGCTCTGCAAC
    AGGAAAACAGCGAAAATGAAAGTCACTCT
    GGAGTGTTGGTGGAACTCGAGGGTGACAAC
    GCGCGCCTAGCCGTACTAAAACGCAGCATC
    GAGGTCACCCACTTTGCCTACCCGGCACTT
    AACCTACCCCCCAAGGTCATGAGCACAGTC
    ATGAGTGAGCTGATCGTGCGCCGTGCGCAG
    CCCCTGGAGAGGGATGCAAATTTGCAAGAA
    CAAACAGAGGAGGGCCTACCCGCAGTTGGC
    GACGAGCAGCTAGCGCGCTGGCTTCAAACG
    CGCGAGCCTGCCGACTTGGAGGAGCGACGC
    AAACTAATGATGGCCGCAGTGCTCGTTACC
    GTGGAGCTTGAGTGCATGCAGCGGTTCTTT
    GCTGACCCGGAGATGCAGCGCAAGCTAGA
    GGAAACATTGCACTACACCTTTCGACAGGG
    CTACGTACGCCAGGCCTGCAAGATCTCCAA
    CGTGGAGCTCTGCAACCTGGTCTCCTACCTT
    GGAATTTTGCACGAAAACCGCCTTGGGCAA
    AACGTGCTTCATTCCACGCTCAAGGGCGAG
    GCGCGCCGCGACTACGTCCGCGACTGCGTT
    TACTTATTTCTATGCTACACCTGGCAGACGG
    CCATGGGCGTTTGGCAGCAGTGCTTGGAGG
    AGTGCAACCTCAAGGAGCTGCAGAAACTGC
    TAAAGCAAAACTTGAAGGACCTATGGACGG
    CCTTCAACGAGCGCTCCGTGGCCGCGCACC
    TGGCGGACATCATTTTCCCCGAACGCCTGC
    TTAAAACCCTGCAACAGGGTCTGCCAGACT
    TCACCAGTCAAAGCATGTTGCAGAACTTTA
    GGAACTTTATCCTAGAGCGCTCAGGAATCT
    TGCCCGCCACCTGCTGTGCACTTCCTAGCG
    ACTTTGTGCCCATTAAGTACCGCGAATGCC
    CTCCGCCGCTTTGGGGCCACTGCTACCTTCT
    GCAGCTAGCCAACTACCTTGCCTACCACTC
    TGACATAATGGAAGACGTGAGCGGTGACG
    GTCTACTGGAGTGTCACTGTCGCTGCAACC
    TATGCACCCCGCACCGCTCCCTGGTTTGCA
    ATTCGCAGCTGCTTAACGAAAGTCAAATTA
    TCGGTACCTTTGAGCTGCAGGGTCCCTCGC
    CTGACGAAAAGTCCGCGGCTCCGGGGTTGA
    AACTCACTCCGGGGCTGTGGACGTCGGCTT
    ACCTTCGCAAATTTGTACCTGAGGACTACC
    ACGCCCACGAGATTAGGTTCTACGAAGACC
    AATCCCGCCCGCCTAATGCGGAGCTTACCG
    CCTGCGTCATTACCCAGGGCCACATTCTTG
    GCCAATTGCAAGCCATCAACAAAGCCCGCC
    AAGAGTTTCTGCTACGAAAGGGACGGGGG
    GTTTACTTGGACCCCCAGTCCGGCGAGGAG
    CTCAACCCAATCCCCCCGCCGCCGCAGCCC
    TATCAGCAGCAGCCGCGGGCCCTTGCTTCC
    CAGGATGGCACCCAAAAAGAAGCTGCAGC
    TGCCGCCGCCACCCACGGACGAGGAGGAAT
    ACTGGGACAGTCAGGCAGAGGAGGTTTTGG
    ACGAGGAGGAGGAGGACATGATGGAAGAC
    TGGGAGAGCCTAGACGAGGAAGCTTCCGA
    GGTCGAAGAGGTGTCAGACGAAACACCGTC
    ACCCTCGGTCGCATTCCCCTCGCCGGCGCC
    CCAGAAATCGGCAACCGGTTCCAGCATGGC
    TACAACCTCCGCTCCTCAGGCGCCGCCGGC
    ACTGCCCGTTCGCCGACCCAACCGTAGATG
    GGACACCACTGGAACCAGGGCCGGTAAGTC
    CAAGCAGCCGCCGCCGTTAGCCCAAGAGCA
    ACAACAGCGCCAAGGCTACCGCTCATGGCG
    CGGGCACAAGAACGCCATAGTTGCTTGCTT
    GCAAGACTGTGGGGGCAACATCTCCTTCGC
    CCGCCGCTTTCTTCTCTACCATCACGGCGTG
    GCCTTCCCCCGTAACATCCTGCATTACTACC
    GTCATCTCTACAGCCCATACTGCACCGGCG
    GCAGCGGCAGCAACAGCAGCGGCCACACA
    GAAGCAAAGGCGACCGGATAGCAAGACTC
    TGACAAAGCCCAAGAAATCCACAGCGGCG
    GCAGCAGCAGGAGGAGGAGCGCTGCGTCT
    GGCGCCCAACGAACCCGTATCGACCCGCGA
    GCTTAGAAACAGGATTTTTCCCACTCTGTAT
    GCTATATTTCAACAGAGCAGGGGCCAAGAA
    CAAGAGCTGAAAATAAAAAACAGGTCTCTG
    CGATCCCTCACCCGCAGCTGCCTGTATCAC
    AAAAGCGAAGATCAGCTTCGGCGCACGCTG
    GAAGACGCGGAGGCTCTCTTCAGTAAATAC
    TGCGCGCTGACTCTTAAGGACTAGTTTCGC
    GCCCTTTCTCAAATTTAAGCGCGAAAACTA
    CGTCATCTCCAGCGGCCACACCCGGCGCCA
    GCACCTGTTGTCAGCGCCATTATGAGCAAG
    GAAATTCCCACGCCCTACATGTGGAGTTAC
    CAGCCACAAATGGGACTTGCGGCTGGAGCT
    GCCCAAGACTACTCAACCCGAATAAACTAC
    ATGAGCGCGGGACCCCACATGATATCCCGG
    GTCAACGGAATACGCGCCCACCGAAACCGA
    ATTCTCCTGGAACAGGCGGCTATTACCACC
    ACACCTCGTAATAACCTTAATCCCCGTAGTT
    GGCCCGCTGCCCTGGTGTACCAGGAAAGTC
    CCGCTCCCACCACTGTGGTACTTCCCAGAG
    ACGCCCAGGCCGAAGTTCAGATGACTAACT
    CAGGGGCGCAGCTTGCGGGCGGCTTTCGTC
    ACAGGGTGCGGTCGCCCGGGCAGGGTATAA
    CTCACCTGACAATCAGAGGGCGAGGTATTC
    AGCTCAACGACGAGTCGGTGAGCTCCTCGC
    TTGGTCTCCGTCCGGACGGGACATTTCAGA
    TCGGCGGCGCCGGCCGCTCTTCATTCACGC
    CTCGTCAGGCAATCCTAACTCTGCAGACCT
    CGTCCTCTGAGCCGCGCTCTGGAGGCATTG
    GAACTCTGCAATTTATTGAGGAGTTTGTGC
    CATCGGTCTACTTTAACCCCTTCTCGGGACC
    TCCCGGCCACTATCCGGATCAATTTATTCCT
    AACTTTGACGCGGTAAAGGACTCGGCGGAC
    GGCTACGACTGAATGTTAAGTGGAGAGGCA
    GAGCAACTGCGCCTGAAACACCTGGTCCAC
    TGTCGCCGCCACAAGTGCTTTGCCCGCGAC
    TCCGGTGAGTTTTGCTACTTTGAATTGCCCG
    AGGATCATATCGAGGGCCCGGCGCACGGCG
    TCCGGCTTACCGCCCAGGGAGAGCTTGCCC
    GTAGCCTGATTCGGGAGTTTACCCAGCGCC
    CCCTGCTAGTTGAGCGGGACAGGGGACCCT
    GTGTTCTCACTGTGATTTGCAACTGTCCTAA
    CCCTGGATTACATCAAGATCCTCTAGTTAAT
    GTCAGGTCGCCTAAGTCGATTAACTAGAGT
    ACCCGGGGATCTTATTCCCTTTAACTAATAA
    AAAAAAATAATAAAGCATCACTTACTTAAA
    ATCAGTTAGCAAATTTCTGTCCAGTTTATTC
    AGCAGCACCTCCTTGCCCTCCTCCCAGCTCT
    GGTATTGCAGCTTCCTCCTGGCTGCAAACTT
    TCTCCACAATCTAAATGGAATGTCAGTTTCC
    TCCTGTTCCTGTCCATCCGCACCCACTATCT
    TCATGTTGTTGCAGATGAAGCGCGCAAGAC
    CGTCTGAAGATACCTTCAACCCCGTGTATC
    CATATGACACGGAAACCGGTCCTCCAACTG
    TGCCTTTTCTTACTCCTCCCTTTGTATCCCCC
    AATGGGTTTCAAGAGAGTCCCCCTGGGGTA
    CTCTCTTTGCGCCTATCCGAACCTCTAGTTA
    CCTCCAATGGCATGCTTGCGCTCAAAATGG
    GCAACGGCCTCTCTCTGGACGAGGCCGGCA
    ACCTTACCTCCCAAAATGTAACCACTGTGA
    GCCCACCTCTCAAAAAAACCAAGTCAAACA
    TAAACCTGGAAATATCTGCACCCCTCACAG
    TTACCTCAGAAGCCCTAACTGTGGCTGCCG
    CCGCACCTCTAATGGTCGCGGGCAACACAC
    TCACCATGCAATCACAGGCCCCGCTAACCG
    TGCACGACTCCAAACTTAGCATTGCCACCC
    AAGGACCCCTCACAGTGTCAGAAGGAAAG
    CTAGCCCTGCAAACATCAGGCCCCCTCACC
    ACCACCGATAGCAGTACCCTTACTATCACT
    GCCTCACCCCCTCTAACTACTGCCACTGGTA
    GCTTGGGCATTGACTTGAAAGAGCCCATTT
    ATACACAAAATGGAAAACTAGGACTAAAG
    TACGGGGCTCCTTTGCATGTAACAGACGAC
    CTAAACACTTTGACCGTAGCAACTGGTCCA
    GGTGTGACTATTAATAATACTTCCTTGCAA
    ACTAAAGTTACTGGAGCCTTGGGTTTTGATT
    CACAAGGCAATATGCAACTTAATGTAGCAG
    GAGGACTAAGGATTGATTCTCAAAACAGAC
    GCCTTATACTTGATGTTAGTTATCCGTTTGA
    TGCTCAAAACCAACTAAATCTAAGACTAGG
    ACAGGGCCCTCTTTTTATAAACTCAGCCCA
    CAACTTGGATATTAACTACAACAAAGGCCT
    TTACTTGTTTACAGCTTCAAACAATTCCAAA
    AAGCTTGAGGTTAACCTAAGCACTGCCAAG
    GGGTTGATGTTTGACGCTACAGCCATAGCC
    ATTAATGCAGGAGATGGGCTTGAATTTGGT
    TCACCTAATGCACCAAACACAAATCCCCTC
    AAAACAAAAATTGGCCATGGCCTAGAATTT
    GATTCAAACAAGGCTATGGTTCCTAAACTA
    GGAACTGGCCTTAGTTTTGACAGCACAGGT
    GCCATTACAGTAGGAAACAAAAATAATGAT
    AAGCTAACTTTGTGGACCACACCAGCTCCA
    TCTCCTAACTGTAGACTAAATGCAGAGAAA
    GATGCTAAACTCACTTTGGTCTTAACAAAA
    TGTGGCAGTCAAATACTTGCTACAGTTTCA
    GTTTTGGCTGTTAAAGGCAGTTTGGCTCCA
    ATATCTGGAACAGTTCAAAGTGCTCATCTT
    ATTATAAGATTTGACGAAAATGGAGTGCTA
    CTAAACAATTCCTTCCTGGACCCAGAATAT
    TGGAACTTTAGAAATGGAGATCTTACTGAA
    GGCACAGCCTATACAAACGCTGTTGGATTT
    ATGCCTAACCTATCAGCTTATCCAAAATCTC
    ACGGTAAAACTGCCAAAAGTAACATTGTCA
    GTCAAGTTTACTTAAACGGAGACAAAACTA
    AACCTGTAACACTAACCATTACACTAAACG
    GTACACAGGAAACAGGAGACACAACTCCA
    AGTGCATACTCTATGTCATTTTCATGGGACT
    GGTCTGGCCACAACTACATTAATGAAATAT
    TTGCCACATCCTCTTACACTTTTTCATACAT
    TGCCCAAGAATAAAGAATCGTTTGTGTTAT
    GTTTCAACGTGTTTATTTTTCAATTGCAGAA
    AATTTCAAGTCATTTTTCATTCAGTAGTATA
    GCCCCACCACCACATAGCTTATACAGATCA
    CCGTACCTTAATCAAACTCACAGAACCCTA
    GTATTCAACCTGCCACCTCCCTCCCAACAC
    ACAGAGTACACAGTCCTTTCTCCCCGGCTG
    GCCTTAAAAAGCATCATATCATGGGTAACA
    GACATATTCTTAGGTGTTATATTCCACACGG
    TTTCCTGTCGAGCCAAACGCTCATCAGTGA
    TATTAATAAACTCCCCGGGCAGCTCACTTA
    AGTTCATGTCGCTGTCCAGCTGCTGAGCCA
    CAGGCTGCTGTCCAACTTGCGGTTGCTTAA
    CGGGCGGCGAAGGAGAAGTCCACGCCTAC
    ATGGGGGTAGAGTCATAATCGTGCATCAGG
    ATAGGGCGGTGGTGCTGCAGCAGCGCGCGA
    ATAAACTGCTGCCGCCGCCGCTCCGTCCTG
    CAGGAATACAACATGGCAGTGGTCTCCTCA
    GCGATGATTCGCACCGCCCGCAGCATAAGG
    CGCCTTGTCCTCCGGGCACAGCAGCGCACC
    CTGATCTCACTTAAATCAGCACAGTAACTG
    CAGCACAGCACCACAATATTGTTCAAAATC
    CCACAGTGCAAGGCGCTGTATCCAAAGCTC
    ATGGCGGGGACCACAGAACCCACGTGGCC
    ATCATACCACAAGCGCAGGTAGATTAAGTG
    GCGACCCCTCATAAACACGCTGGACATAAA
    CATTACCTCTTTTGGCATGTTGTAATTCACC
    ACCTCCCGGTACCATATAAACCTCTGATTA
    AACATGGCGCCATCCACCACCATCCTAAAC
    CAGCTGGCCAAAACCTGCCCGCCGGCTATA
    CACTGCAGGGAACCGGGACTGGAACAATG
    ACAGTGGAGAGCCCAGGACTCGTAACCATG
    GATCATCATGCTCGTCATGATATCAATGTTG
    GCACAACACAGGCACACGTGCATACACTTC
    CTCAGGATTACAAGCTCCTCCCGCGTTAGA
    ACCATATCCCAGGGAACAACCCATTCCTGA
    ATCAGCGTAAATCCCACACTGCAGGGAAGA
    CCTCGCACGTAACTCACGTTGTGCATTGTCA
    AAGTGTTACATTCGGGCAGCAGCGGATGAT
    CCTCCAGTATGGTAGCGCGGGTTTCTGTCTC
    AAAAGGAGGTAGACGATCCCTACTGTACGG
    AGTGCGCCGAGACAACCGAGATCGTGTTGG
    TCGTAGTGTCATGCCAAATGGAACGCCGGA
    CGTAGTCATATTTCCTGAAGCAAAACCAGG
    TGCGGGCGTGACAAACAGATCTGCGTCTCC
    GGTCTCGCCGCTTAGATCGCTCTGTGTAGTA
    GTTGTAGTATATCCACTCTCTCAAAGCATCC
    AGGCGCCCCCTGGCTTCGGGTTCTATGTAA
    ACTCCTTCATGCGCCGCTGCCCTGATAACAT
    CCACCACCGCAGAATAAGCCACACCCAGCC
    AACCTACACATTCGTTCTGCGAGTCACACA
    CGGGAGGAGCGGGAAGAGCTGGAAGAACC
    ATGTTTTTTTTTTTATTCCAAAAGATTATCC
    AAAACCTCAAAATGAAGATCTATTAAGTGA
    ACGCGCTCCCCTCCGGTGGCGTGGTCAAAC
    TCTACAGCCAAAGAACAGATAATGGCATTT
    GTAAGATGTTGCACAATGGCTTCCAAAAGG
    CAAACGGCCCTCACGTCCAAGTGGACGTAA
    AGGCTAAACCCTTCAGGGTGAATCTCCTCT
    ATAAACATTCCAGCACCTTCAACCATGCCC
    AAATAATTCTCATCTCGCCACCTTCTCAATA
    TATCTCTAAGCAAATCCCGAATATTAAGTC
    CGGCCATTGTAAAAATCTGCTCCAGAGCGC
    CCTCCACCTTCAGCCTCAAGCAGCGAATCA
    TGATTGCAAAAATTCAGGTTCCTCACAGAC
    CTGTATAAGATTCAAAAGCGGAACATTAAC
    AAAAATACCGCGATCCCGTAGGTCCCTTCG
    CAGGGCCAGCTGAACATAATCGTGCAGGTC
    TGCACGGACCAGCGCGGCCACTTCCCCGCC
    AGGAACCATGACAAAAGAACCCACACTGA
    TTATGACACGCATACTCGGAGCTATGCTAA
    CCAGCGTAGCCCCGATGTAAGCTTGTTGCA
    TGGGCGGCGATATAAAATGCAAGGTGCTGC
    TCAAAAAATCAGGCAAAGCCTCGCGCAAA
    AAAGAAAGCACATCGTAGTCATGCTCATGC
    AGATAAAGGCAGGTAAGCTCCGGAACCAC
    CACAGAAAAAGACACCATTTTTCTCTCAAA
    CATGTCTGCGGGTTTCTGCATAAACACAAA
    ATAAAATAACAAAAAAACATTTAAACATTA
    GAAGCCTGTCTTACAACAGGAAAAACAACC
    CTTATAAGCATAAGACGGACTACGGCCATG
    CCGGCGTGACCGTAAAAAAACTGGTCACCG
    TGATTAAAAAGCACCACCGACAGCTCCTCG
    GTCATGTCCGGAGTCATAATGTAAGACTCG
    GTAAACACATCAGGTTGATTCACATCGGTC
    AGTGCTAAAAAGCGACCGAAATAGCCCGG
    GGGAATACATACCCGCAGGCGTAGAGACA
    ACATTACAGCCCCCATAGGAGGTATAACAA
    AATTAATAGGAGAGAAAAACACATAAACA
    CCTGAAAAACCCTCCTGCCTAGGCAAAATA
    GCACCCTCCCGCTCCAGAACAACATACAGC
    GCTTCCACAGCGGCAGCCATAACAGTCAGC
    CTTACCAGTAAAAAAGAAAACCTATTAAAA
    AAACACCACTCGACACGGCACCAGCTCAAT
    CAGTCACAGTGTAAAAAAGGGCCAAGTGC
    AGAGCGAGTATATATAGGACTAAAAAATG
    ACGTAACGGTTAAAGTCCACAAAAAACACC
    CAGAAAACCGCACGCGAACCTACGCCCAG
    AAACGAAAGCCAAAAAACCCACAACTTCCT
    CAAATCGTCACTTCCGTTTTCCCACGTTACG
    TCACTTCCCATTTTAAGAAAACTACAATTCC
    CAACACATACAAGTTACTCCGCCCTAAAAC
    CTACGTCACCCGCCCCGTTCCCACGCCCCG
    CGCCACGTCACAAACTCCACCCCCTCATTA
    TCATATTGGCTTCAATCCAAAATAAGGTAT
    ATTATTGATGAT
    96 human IL-12 DNA GTCGACGCCACCATGTGTCACCAGCAGCT
    insert Sequence  CGTGATTAGCTGGTTCAGCCTGGTGTTTCTG
    (including GCTAGCCCTCTGGTGGCCATCTGGGAGCTG
    restriction AAGAAGGACGTGTACGTGGTGGAGCTCGAC
    sites and TGGTACCCTGACGCTCCCGGCGAGATGGTC
    Kozak GTGCTGACCTGCGACACCCCTGAGGAAGAT
    sequence) GGCATCACCTGGACCCTGGATCAAAGCTCC
    GAAGTGCTCGGCAGCGGCAAGACACTCACC
    ATCCAGGTGAAAGAGTTCGGAGACGCCGGC
    CAGTACACCTGCCACAAAGGCGGCGAGGTG
    CTGTCCCATTCCCTGCTGCTGCTGCACAAGA
    AAGAGGATGGCATCTGGTCCACCGACATCC
    TGAAGGACCAGAAGGAACCCAAGAACAAG
    ACCTTTCTGAGATGTGAGGCCAAGAACTAC
    AGCGGCAGGTTCACCTGCTGGTGGCTGACA
    ACAATCTCCACCGACCTGACCTTCAGCGTC
    AAGAGCAGCAGGGGCAGCAGCGACCCTCA
    AGGCGTGACATGTGGAGCCGCTACCCTGAG
    CGCTGAGAGAGTCAGGGGCGACAATAAGG
    AGTACGAGTACTCCGTGGAATGCCAGGAGG
    ACTCCGCCTGCCCTGCCGCCGAAGAGTCCC
    TCCCTATCGAAGTGATGGTTGATGCCGTGC
    ACAAGCTCAAGTATGAGAATTACACCAGCA
    GCTTTTTCATCAGGGACATCATCAAGCCCG
    ACCCCCCCAAAAACCTCCAGCTGAAACCCC
    TCAAGAATAGCAGGCAGGTGGAGGTCTCCT
    GGGAGTATCCTGACACCTGGAGCACCCCCC
    ACAGCTACTTCTCCCTGACCTTCTGTGTGCA
    GGTGCAGGGCAAGAGCAAAAGGGAGAAGA
    AGGATAGGGTCTTTACCGACAAGACCAGCG
    CCACAGTGATCTGCAGGAAGAACGCCAGCA
    TTTCCGTCAGGGCCCAGGACAGGTACTACA
    GCAGCAGCTGGTCCGAGTGGGCTAGCGTGC
    CTTGTTCCGGCGGCGGAGGATCTGGCGGAG
    GCGGAAGTGGCGGAGGGGGCTCTAGAAAC
    CTCCCCGTGGCCACACCCGACCCTGGCATG
    TTCCCCTGCCTCCACCACAGCCAGAACCTG
    CTGAGAGCCGTGAGCAATATGCTGCAGAAG
    GCCAGGCAAACCCTGGAGTTCTACCCCTGT
    ACCTCCGAGGAGATTGACCATGAGGACATC
    ACAAAGGACAAAACCAGCACCGTGGAGGC
    CTGTCTCCCCCTCGAACTGACCAAGAACGA
    GTCCTGCCTGAACTCCAGGGAGACATCCTT
    CATCACCAACGGCTCCTGCCTGGCCTCCAG
    AAAGACCAGCTTCATGATGGCCCTCTGCCT
    GAGCAGCATCTACGAGGACCTCAAGATGTA
    CCAGGTGGAGTTTAAAACAATGAACGCCAA
    GCTCCTCATGGACCCTAAGAGGCAGATTTT
    CCTCGACCAGAATATGCTGGCTGTCATTGA
    CGAGCTGATGCAGGCCCTCAATTTCAACTC
    CGAGACCGTCCCCCAGAAGTCCTCCCTGGA
    AGAGCCCGACTTTTACAAGACCAAGATCAA
    GCTCTGCATCCTGCTGCACGCCTTCAGAATT
    AGAGCCGTGACCATTGACAGGGTGATGAGC
    TACCTCAACGCCTCCTGATGACTCGAG
    97 human IL- GTCGACGCCACCACATCCGCGGCAACGCC
    insert TCCTTGGTGTCGTCCGCTTCCAATAACCCAG
    CTTGCGTCCTGC
    ACACTTGTGGCTTCCGTGCACACATTAACA
    ACTCATGGTTCTAGCTCCCAGTCGCCAAGC
    GTTGCCAAGGCGTTGAGAGATCATCTGGGA
    AGTCTTTTACCCAGAATTGCTTTGATTCAG
    GCCAGCTGGTTTTTCCTGCGGTGATTCGGA
    AATTCGCGAATTCCTCTGGTCCTCATCCAG
    GTGCGCGGGAAGCAGGTGCCCAGGAGAGA
    GGGGATAATGAAGATTCCATGCTGATGATC
    C
    CAAAGATTGAACCTGCAGACCAAGCGCAA
    AGTAGAAACTGAAAGTACACTGCTGGCGGA
    T
    CCTACGGAAGTTATGGAAAAGGCAAAGCG
    CAGAGCCACGCCGTAGTGTGTGCCGCCCCC
    C
    TTGGGATGGATGAAACTGCAGTCGCGGCGT
    GGGTAAGAGGAACCAGCTGCAGAGATCAC
    C
    CTGCCCAACACAGACTCGGCAACTCCGCGG
    AAGACCAGGGTCCTGGGAGTGACTATGGGC
    GGTGAGAGCTTGCTCCTGCTCCAGTTGCGG
    TCATCATGACTACGCCCGCCTCCCGCAGAC
    CATGTTCCATGTTTCTTTTAGGTATATCTTT
    GGACTTCCTCCCCTGATCCTTGTTCTGTT
    GCCAGTAGCATCATCTGATTGTGATATTGA
    AGGTAAAGATGGCAAACAATATGAGAGTG
    T
    TCTAATGGTCAGCATCGATCAATTATTGGA
    CAGCATGAAAGAAATTGGTAGCAATTGCCT
    GAATAATGAATTTAACTTTTTTAAAAGACA
    TATCTGTGATGCTAATAAGGAAGGTATGTT
    TTTATTCCGTGCTGCTCGCAAGTTGAGGCA
    ATTTCTTAAAATGAATAGCACTGGTGATTT
    TGATCTCCACTTATTAAAAGTTTCAGAAGG
    CACAACAATACTGTTGAACTGCACTGGCCA
    GGTTAAAGGAAGAAAACCAGCTGCCCTGG
    GTGAAGCCCAACCAACAAAGAGTTTGGAA
    GA
    AAATAAATCTTTAAAGGAACAGAAAAAACT
    GAATGACTTGTGTTTCCTAAAGAGACTATT
    ACAAGAGATAAAAACTTGTTGGAATAAAAT
    TTTGATGGGCACTAAAGAACACTGAAAAAT
    ATGGAGTGGCAATATAGAAACACGAACTTT
    AGCTGCATCCTCCAAGAATCTATCTGCTTA
    TGCAGTTTTTCAGAGTGGAATGCTTCCTAG
    AAGTTACTGAATGCACCATGGTCAAAACGG
    ATTAGGGCATTTGAGAAATGCATATTGTAT
    TACTAGAAGATGAATACAAACAATGGAAA
    C
    TGAATGCTCCAGTCAACAAACTATTTCTTAT
    ATATGTGAACATTTATCAATCAGTATAAT
    TCTGTACTGATTTTTGTAAGACAATCCATGT
    AAGGTATCAGTTGCAATAATACTTCTCAA
    ACCTGTTTAAATATTTCAAGACATTAAATCT
    ATGAAGTATATAATGGTTTCAAAGATTCA
    AAATTGACATTGCTTTACTGTCAAAATAATT
    TTATGGCTCACTATGAATCTATTATACTG
    TATTAAGAGTGAAAATTGTCTTCTTCTGTGC
    TGGAGATGTTTTAGAGTTAACAATGATAT
    ATGGATAATGCCGGTGAGAATAAGAGAGTC
    ATAAACCTTAAGTAAGCAACAGCATAACAA
    GGTCCAAGATACCTAAAAGAGATTTCAAGA
    GATTTAATTAATCATGAATGTGTAACACAG
    TGCCTTCAATAAATGGTATAGCAAATGTTTT
    GACATGAAAAAAGGACAATTTCAAAAAAA
    TAAAATAAAATAAAAATAAATTCACCTAGT
    CTAAGGATGCTAAACCTTAGTACTGAGTTA
    CATTGTCATTTATATAGATTATAACTTGTCT
    AAATAAGTTTGCAATTTGGGAGATATATT
    TTTAAGATAATAATATATGTTTACCTTTTAA
    TTAATGAAATATCTGTATTTAATTTTGAC
    ACTATATCTGTATATAAAATATTTTCATACA
    GCATTACAAATTGCTTACTTTGGAATACA
    TTTCTCCTTTGATAAAATAAATGAGCTATGT
    ATTAAAAAAAAAAAAAAA
    98 human CD70 NCBI GTCGACGCCACCCCAGAGAGGGGCAGGCT
    insert Reference GGTCCCCTGACAGGTTGAAGCAAGTAGACG
    Sequence: CCCAGGAGCCCCG
    NM_001252.4 GGAGGGGGCTGCAGTTTCCTTCCTTCCTTCT
    CGGCAGCGCTCCGCGCCCCCATCGCCCCT
    CCTGCGCTAGCGGAGGTGATCGCCGCGGCG
    ATGCCGGAGGAGGGTTCGGGCTGCTCGGTG
    CGGCGCAGGCCCTATGGGTGCGTCCTGCGG
    GCTGCTTTGGTCCCATTGGTCGCGGGCTTG
    GTGATCTGCCTCGTGGTGTGCATCCAGCGC
    TTCGCACAGGCTCAGCAGCAGCTGCCGCTC
    GAGTCACTTGGGTGGGACGTAGCTGAGCTG
    CAGCTGAATCACACAGGACCTCAGCAGGAC
    CCCAGGCTATACTGGCAGGGGGGCCCAGCA
    CTGGGCCGCTCCTTCCTGCATGGACCAGAG
    CTGGACAAGGGGCAGCTACGTATCCATCGT
    GATGGCATCTACATGGTACACATCCAGGTG
    ACGCTGGCCATCTGCTCCTCCACGACGGCC
    TCCAGGCACCACCCCACCACCCTGGCCGTG
    GGAATCTGCTCTCCCGCCTCCCGTAGCATC
    AGCCTGCTGCGTCTCAGCTTCCACCAAGGT
    TGTACCATTGCCTCCCAGCGCCTGACGCCC
    CTGGCCCGAGGGGACACACTCTGCACCAAC
    CTCACTGGGACACTTTTGCCTTCCCGAAAC
    ACTGATGAGACCTTCTTTGGAGTGCAGTGG
    GTGCGCCCCTGACCACTGCTGCTGATTAGG
    GTTTTTTAAATTTTATTTTATTTTATTTAA
    GTTCAAGAGAAAAAGTGTACACACAGGGG
    CCACCCGGGGTTGGGGTGGGAGTGTGGTGG
    G
    GGGTAGTGGTGGCAGGACAAGAGAAGGCA
    TTGAGCTTTTTCTTTCATTTTCCTATTAAAA
    AATACAAAAATCA
    99 NV1 Adenovirus GGCGGAAGTGTGATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCGCAA
    Enhancer GTCTATGTTGTAGTAAATTTGGGCGTAACC
    Region GAGTAAGATTTGGCCATTTTCGCGGGAAAA
    Mutant 1 CTGAATAAGAGGAAGTGAAATC
    100 NV2 Adenovirus ACAGGAAGTGACATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCGCAA
    Enhancer GTCTATGTTGTAGTAAATTTGGGCGTAACC
    Region GAGTAAGATTTGGCCATTTTCGCGGGAAAA
    Mutant 2 CTGAATAAGAGGAAGTGAAATCT
    101 NV3 Adenovirus ACAGGAAGTGACAATTTTCGCGCGGTTTTA
    E1a GGCGGATGTGGCGCAAGTCTATGTTGTAGT
    Enhancer AAATTTGGGCGTAACCGAGTAAGATTTGGC
    Region CATTTTCGCGGGAAAACTGAATAAGAGGAA
    Mutant 3 GTGAAATCT
    102 NV4 Adenovirus GGCGGAAGTGTGATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCAAAA
    Enhancer GTGACGTTTTTGGTGTGCGCCGGTGTACGG
    Region CGGAAGTGTGAATTTTCGCGCGGTTTTAGA
    Mutant 4 CGGATGTGGCAGTAAATTTGGGCGTAACCG
    AGTAAGATTTGGCCATTTTCGCGGGAAAAC
    TGAATAAGAGGAAGTGAAATCT
    103 NV5 Adenovirus GGCGGAAGTGTGATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCAAAA
    Enhancer GTGACGTTTTTGGTGTGCGCCGGTGTACGG
    Region CGGAAGTGTGAATTTTCGCGCGGTTTTAGA
    Mutant 5 CGGATGTGGCAGTAAATTTGGGCGTAACCG
    AGTAAGATTTGGCCATTTTCGCGGGAAAAC
    TGAATAAGAGGATGTGAAATCT
    104 NV6 Adenovirus GGCGGAAGTGTGATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCAAAA
    Enhancer GTGACGTTTTTGGTGTGCGCCGGTGTACGG
    Region CGGAAGTGTGAATTTTCGCGCGGTTTTAGA
    Mutant 6 CGGATGTGGCAGTAAATTTGGGCGTAACCG
    AGTAAGATTTGGCCATTTTCGCGGGAAAAC
    TGAATAGGCGGAAGTGTGATCT
    105  NV7 Adenovirus GGCGGAAGTGTGATGTTGCAAGTGTGGCGG
    E1a AACACATGTAAGCGACGGATGTGGCAAAA
    Enhancer GTGACGTTTTTGGTGTGCGCCGGTGTACAC
    Region AGGAAGTGACAATTTTCGCGCGGTTTTAGA
    Mutant 7 CGGATGTGGCAGTAAATTTGGGCGTAACCG
    AGTAAGATTTGGCCATTTTCGCGGGAAAAC
    TGAATAGGCGGAAGTGTGATCT

Claims (20)

What is claimed is:
1. A pharmaceutical composition comprising an effective amount of a recombinant adenoviral vector comprising:
(a) a first transgene insertion site located between the start site of adenoviral E1b-19K and the start site of adenoviral E1b-55K, the transgene insertion site comprising:
a first DNA sequence encoding a first polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, wherein the first DNA sequence is present in the first transgene insertion site and is operably linked to a first endogenous promoter, and
a second DNA sequence encoding a second polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, wherein the second DNA sequence is present in the second transgene insertion site and is operably linked to a second endogenous promoter,
wherein the first DNA sequence and the second DNA sequence are operably linked or the first polypeptide and the second polypeptide are operably linked, and
(b) a modified adenoviral E1a regulatory sequence, wherein at least one Pea3 binding site of the adenoviral E1a regulatory sequence is modified or deleted.
2. The pharmaceutical composition of claim 1, wherein the first DNA sequence and the second DNA sequence are operably linked by an IRES element.
3. The pharmaceutical composition of claim 1, wherein the first polypeptide and the second polypeptide are operably linked by a self-cleaving 2A peptide.
4. The pharmaceutical composition of claim 1, wherein a sequence between two Pea3 sites of the adenoviral E1a regulatory sequence is deleted.
5. The pharmaceutical composition of claim 1, wherein the first DNA sequence or the second DNA sequence encodes the chimeric human IL-12, wherein the chimeric human IL-12 comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide.
6. The pharmaceutical composition of claim 1, wherein the first DNA sequence or the second DNA sequence encodes human IL-2.
7. The pharmaceutical composition of claim 1, wherein the first DNA sequence or the second DNA sequence encodes human IL-7.
8. The pharmaceutical composition of claim 1, wherein the first polypeptide comprises human IL-7 and the second polypeptide comprises chimeric human IL-12.
9. A pharmaceutical composition comprising an effective amount of a recombinant adenoviral vector comprising:
(a) a first transgene insertion site located in the adenoviral E3 region, the transgene insertion site comprising a first DNA sequence encoding a first polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, wherein the first DNA sequence is present in the first transgene insertion site and is operably linked to a first endogenous promoter, and
(b) a modified adenoviral E1a regulatory sequence, wherein at least one Pea3 binding site of the adenoviral E1a regulatory sequence is modified or deleted.
10. The pharmaceutical composition of claim 9, further comprising in the transgene insertion site a second DNA sequence encoding a second polypeptide selected from the group consisting of: a chimeric human IL-12, a human IL-7, an anti-CTLA-4 antibody, an IL-10Rtrap, a human CD70, a human IL-2 polypeptide, a human CD40 ligand, and a human OX40 ligand, wherein the second DNA sequence is present in the second transgene insertion site and is operably linked to a second endogenous promoter.
11. The pharmaceutical composition of claim 9, wherein a sequence between two Pea3 sites of the adenoviral E1a regulatory sequence is deleted.
12. The pharmaceutical composition of claim 10, wherein the first DNA sequence or the second DNA sequence encodes the chimeric human IL-12, wherein the chimeric human IL-12 comprises a p40 polypeptide, a p35 polypeptide, and a linker polypeptide.
13. The pharmaceutical composition of claim 10, wherein the first DNA sequence or the second DNA sequence encodes human IL-2.
14. The pharmaceutical composition of claim 10, wherein the first DNA sequence or the second DNA sequence encodes human IL-7.
15. The pharmaceutical composition of claim 10, comprising a truncation in the E3 12.5K coding region, the E3 7.1K coding region, the E3 gp19K, the E3 10.5, or a combination thereof.
16. A method for treating a tumor in a human subject in need thereof, comprising administering to the human with a tumor a therapeutic amount of the pharmaceutical composition of claim 1 by systemic or intratumor administration.
17. The method of claim 16, further comprising treating the human subject with an anti-PD-1 antibody or an anti-PD-L1 antibody.
18. The method of claim 16, further comprising treating the human subject with:
(a) an agonist of a co-stimulatory signal selected from glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB (CD137), CD40, lymphotoxin alpha (LT alpha), LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes), CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA), or
(b) an antagonist of an inhibitory molecule selected from cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and T-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD 160, adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD 160.
19. A method for treating a tumor in a human subject in need thereof, comprising administering to the human with a tumor a therapeutic amount of the pharmaceutical composition of claim 9 by systemic or intratumor administration.
20. The method of claim 19, further comprising treating the human subject with:
(a) an agonist of a co-stimulatory signal selected from glucocorticoid-induced tumor necrosis factor receptor (GITR), Inducible T-cell co-stimulator (ICOS or CD278), OX40 (CD134), CD27, CD28, 4-IBB (CD137), CD40, lymphotoxin alpha (LT alpha), LIGHT (lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes), CD226, cytotoxic and regulatory T cell molecule (CRT AM), death receptor 3 (DR3), lymphotoxin-beta receptor (LTBR), transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA), or
(b) an antagonist of an inhibitory molecule selected from cytotoxic T lymphocyte-associated antigen 4 (CTLA-4 or CD52), programmed cell death protein 1 (PD1 or CD279), B and I-lymphocyte attenuator (BTLA), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), T-cell membrane protein 3 (TIM3), CD 160, adenosine A2a receptor (A2aR), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and CD 160.
US16/253,056 2016-12-30 2019-01-21 Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer Abandoned US20190142967A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/253,056 US20190142967A1 (en) 2016-12-30 2019-01-21 Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201662440670P 2016-12-30 2016-12-30
US201662440646P 2016-12-30 2016-12-30
US201762572206P 2017-10-13 2017-10-13
US15/834,690 US10232053B2 (en) 2016-12-30 2017-12-07 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US16/253,056 US20190142967A1 (en) 2016-12-30 2019-01-21 Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/834,690 Continuation US10232053B2 (en) 2016-12-30 2017-12-07 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer

Publications (1)

Publication Number Publication Date
US20190142967A1 true US20190142967A1 (en) 2019-05-16

Family

ID=62708668

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/834,690 Active US10232053B2 (en) 2016-12-30 2017-12-07 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US16/474,898 Abandoned US20200014798A1 (en) 2016-12-30 2018-01-02 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US16/253,056 Abandoned US20190142967A1 (en) 2016-12-30 2019-01-21 Immunomodulatory Oncolytic Adenoviral Vectors, and Methods of Production and Use Thereof for Treatment of Cancer
US17/385,265 Pending US20210393803A1 (en) 2016-12-30 2021-07-26 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/834,690 Active US10232053B2 (en) 2016-12-30 2017-12-07 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US16/474,898 Abandoned US20200014798A1 (en) 2016-12-30 2018-01-02 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/385,265 Pending US20210393803A1 (en) 2016-12-30 2021-07-26 Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer

Country Status (5)

Country Link
US (4) US10232053B2 (en)
EP (1) EP3562516A1 (en)
JP (1) JP2020503342A (en)
CA (1) CA3048998A1 (en)
WO (1) WO2018126282A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10676516B2 (en) 2017-05-24 2020-06-09 Pandion Therapeutics, Inc. Targeted immunotolerance
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
US10961310B2 (en) 2017-03-15 2021-03-30 Pandion Operations, Inc. Targeted immunotolerance
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11091526B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
WO2021249035A1 (en) * 2020-06-10 2021-12-16 广州恩宝生物医药科技有限公司 Replication-type human adenovirus and application thereof
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
WO2022203451A1 (en) * 2021-03-26 2022-09-29 (주) 와이디생명과학 Composition for preventing or treating cancer comprising dual expression vector for simultaneously expressing protein present in cell and protein secreted out of cell
US11739146B2 (en) 2019-05-20 2023-08-29 Pandion Operations, Inc. MAdCAM targeted immunotolerance
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US11981715B2 (en) 2020-02-21 2024-05-14 Pandion Operations, Inc. Tissue targeted immunotolerance with a CD39 effector

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3038526A1 (en) 2016-09-27 2018-04-05 Epicentrx, Inc. Immunomodulatory fusion proteins
US10232053B2 (en) * 2016-12-30 2019-03-19 Trieza Therapeutics, Inc. Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US20190352616A1 (en) * 2017-01-30 2019-11-21 Epicentrx, Inc. Multiple transgene recombinant adenovirus
EA202090838A1 (en) * 2017-09-27 2020-08-28 Эписентарикс, Инк. IMMUNOMODULATING FUSION PROTEINS
CA3088609C (en) * 2018-01-19 2024-02-13 Kolon Life Science, Inc. Recombinant vaccinia virus and pharmaceutical composition comprising same
JP7484717B2 (en) * 2018-09-26 2024-05-16 アステラス製薬株式会社 Cancer therapy using a combination of oncolytic vaccinia virus and immune checkpoint inhibitor, and pharmaceutical composition and combination drug for use therein
JP2022507606A (en) * 2018-11-16 2022-01-18 ネオイミューンテック, インコーポレイテッド How to Treat Tumors with a Combination of IL-7 Protein and Immune Checkpoint Inhibitors
US20220272953A1 (en) * 2019-07-29 2022-09-01 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Genetically modified non-human animal with human or chimeric il33
EP4019641A4 (en) * 2019-08-19 2022-12-21 Nanjing Novel Biotechnology Co., Ltd. Replicative oncolytic adenovirus for regulating lipid metabolism and use thereof
KR20220092523A (en) * 2019-10-11 2022-07-01 틸트 바이오세러퓨틱스 오이 Oncolytic viral vector encoding variant interleukin-2 (vIL-2) polypeptide
WO2021212083A2 (en) * 2020-04-17 2021-10-21 The Board Of Trustees Of The Leland Stanford Junior University Engineered il-12 and il-23 polypeptides and uses thereof
CN114350709A (en) * 2021-09-18 2022-04-15 梁亚龙 Modified oncolytic adenovirus vector

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6051427A (en) 1993-06-11 2000-04-18 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US6210939B1 (en) * 1993-10-25 2001-04-03 Canji, Inc. Recombinant adenoviral vector and methods of use
US5994104A (en) * 1996-11-08 1999-11-30 Royal Free Hospital School Of Medicine Interleukin-12 fusion protein
US6002068A (en) 1996-12-19 1999-12-14 Novartis Finance Corporation Methods for conferring insect resistance to a monocot using a perioxidase coding sequence
US6692736B2 (en) 2000-03-24 2004-02-17 Cell Genesys, Inc. Cell-specific adenovirus vectors comprising an internal ribosome entry site
US7364727B2 (en) * 2002-07-22 2008-04-29 Cell Genesys, Inc. Metastatic colon cancer specific promoter and uses thereof
WO2006124667A2 (en) 2005-05-12 2006-11-23 Zymogenetics, Inc. Compositions and methods for modulating immune responses
US20090258013A1 (en) 2008-04-09 2009-10-15 Genentech, Inc. Novel compositions and methods for the treatment of immune related diseases
PT2403951E (en) * 2009-03-02 2016-01-29 Univ California Tumor-selective adenovirus e1a and e1b mutants
KR101253276B1 (en) 2010-07-27 2013-04-10 윤채옥 Anti-tumor Compositions for Combined Gene Therapy
CA2850575C (en) * 2011-10-11 2020-12-15 Universitat Zurich Prorektorat Mnw Combination medicament comprising il-12 and an agent for blockade of t-cell inhibitory molecules for tumour therapy
WO2014170389A1 (en) 2013-04-18 2014-10-23 Tilt Biotherapeutics Oy Enhanced adoptive cell therapy
RS60826B1 (en) 2013-07-16 2020-10-30 Hoffmann La Roche Methods of treating cancer using pd-1 axis binding antagonists and tigit inhibitors
EP3083666A4 (en) * 2013-12-18 2017-09-20 Intrexon Corporation Single chain il-12 nucleic acids, polypeptids, and uses thereof
MA39818A (en) * 2014-03-30 2017-02-08 Benevir Biopharm Inc Exogenous tap inhibitor "armed" oncolytic viruses and therapeutic uses thereof
JP6920194B2 (en) 2014-08-19 2021-08-18 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Anti-TIGIT antibody
MA40662B1 (en) 2014-12-23 2020-12-31 Bristol Myers Squibb Co Antibodies against tigit
US10232053B2 (en) * 2016-12-30 2019-03-19 Trieza Therapeutics, Inc. Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Szymczak et al, Correction of multi-gene deficiency in vivo using a single ‘self-cleaving’ 2A peptide–based retroviral vector, Nature Biotechnology 22(5): 589-594, 2004 *
Tan et al, Coexpression of double or triple copies of the rabies virus glycoprotein gene using a ‘self-cleaving’ 2A peptide-based replication-defective human adenovirus serotype 5 vector, Biologicals 38: 586-593, 2010 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11077156B2 (en) 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US10961310B2 (en) 2017-03-15 2021-03-30 Pandion Operations, Inc. Targeted immunotolerance
US10676516B2 (en) 2017-05-24 2020-06-09 Pandion Therapeutics, Inc. Targeted immunotolerance
US11466068B2 (en) 2017-05-24 2022-10-11 Pandion Operations, Inc. Targeted immunotolerance
US11091526B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11091527B2 (en) 2017-12-06 2021-08-17 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11779632B2 (en) 2017-12-06 2023-10-10 Pandion Operation, Inc. IL-2 muteins and uses thereof
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11945852B2 (en) 2017-12-06 2024-04-02 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11965008B2 (en) 2017-12-06 2024-04-23 Pandion Operations, Inc. IL-2 muteins and uses thereof
US11739146B2 (en) 2019-05-20 2023-08-29 Pandion Operations, Inc. MAdCAM targeted immunotolerance
US11981715B2 (en) 2020-02-21 2024-05-14 Pandion Operations, Inc. Tissue targeted immunotolerance with a CD39 effector
WO2021249035A1 (en) * 2020-06-10 2021-12-16 广州恩宝生物医药科技有限公司 Replication-type human adenovirus and application thereof
WO2022203451A1 (en) * 2021-03-26 2022-09-29 (주) 와이디생명과학 Composition for preventing or treating cancer comprising dual expression vector for simultaneously expressing protein present in cell and protein secreted out of cell

Also Published As

Publication number Publication date
US20210393803A1 (en) 2021-12-23
WO2018126282A1 (en) 2018-07-05
EP3562516A1 (en) 2019-11-06
CA3048998A1 (en) 2018-07-05
US10232053B2 (en) 2019-03-19
US20180185515A1 (en) 2018-07-05
US20200014798A1 (en) 2020-01-09
JP2020503342A (en) 2020-01-30

Similar Documents

Publication Publication Date Title
US20210393803A1 (en) Immunomodulatory oncolytic adenoviral vectors, and methods of production and use thereof for treatment of cancer
US11938159B2 (en) Oncolytic adenoviruses armed with heterologous genes
US11155622B2 (en) Virus encoding an anti-TCR-complex antibody or fragment
EP3503918B1 (en) Adenovirus armed with bispecific t cell engager (bite)
JP2019500909A (en) Oncolytic virus strain
US11142580B2 (en) Oncolytic virus and method
WO2018083259A1 (en) Oncolytic adenovirus encoding transgenes
WO2018083258A1 (en) Oncolytic adenovirus encoding at least three transgenes
WO2018083257A1 (en) Oncolytic adenovirus encoding transgenes
CN109320602B (en) Siglec-9 targeted chimeric antigen receptor T cell and application thereof
US20220152134A1 (en) Oncolytic adenoviral vector expressing a member of the b7 family of costimulatory ligands and ada
MEDICHE Generation and in vitro characterization of cancer immunotherapeutics based on oncolytic viruses and immune checkpoint inhibitors
NZ791667A (en) Adenovirus armed with bispecific T-cell activator

Legal Events

Date Code Title Description
AS Assignment

Owner name: TRIEZA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HICKLIN, DANIEL;WILLS, KENNETH NELSON;SEIDEL-DUGAN, CYNTHIA;AND OTHERS;SIGNING DATES FROM 20171221 TO 20171230;REEL/FRAME:048076/0074

Owner name: TRIEZA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:STEINER, PHILIPP;REEL/FRAME:048076/0081

Effective date: 20180102

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: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: PRE-INTERVIEW COMMUNICATION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

AS Assignment

Owner name: EPICENTRX, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TRIEZA THERAPEUTICS, INC.;REEL/FRAME:066412/0279

Effective date: 20230517

STCB Information on status: application discontinuation

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