WO2024182707A1 - Interleukin-2 and interleukin-12 for cancer therapy - Google Patents
Interleukin-2 and interleukin-12 for cancer therapy Download PDFInfo
- Publication number
- WO2024182707A1 WO2024182707A1 PCT/US2024/018084 US2024018084W WO2024182707A1 WO 2024182707 A1 WO2024182707 A1 WO 2024182707A1 US 2024018084 W US2024018084 W US 2024018084W WO 2024182707 A1 WO2024182707 A1 WO 2024182707A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- recombinant
- genome
- cancer
- virus
- pharmaceutical composition
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/761—Adenovirus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/0008—Medicinal 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
- A61K48/0025—Medicinal 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 wherein the non-active part clearly interacts with the delivered nucleic acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal 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
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/5434—IL-12
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/55—IL-2
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/87—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10341—Use of virus, viral particle or viral elements as a vector
- C12N2710/10343—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/14011—Baculoviridae
- C12N2710/14041—Use of virus, viral particle or viral elements as a vector
- C12N2710/14043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vectore
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/16011—Herpesviridae
- C12N2710/16611—Simplexvirus, e.g. human herpesvirus 1, 2
- C12N2710/16641—Use of virus, viral particle or viral elements as a vector
- C12N2710/16643—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24041—Use of virus, viral particle or viral elements as a vector
- C12N2710/24043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/10041—Use of virus, viral particle or viral elements as a vector
- C12N2740/10043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/12011—Geminiviridae
- C12N2750/12041—Use of virus, viral particle or viral elements as a vector
- C12N2750/12043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2760/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
- C12N2760/00011—Details
- C12N2760/18011—Paramyxoviridae
- C12N2760/18111—Avulavirus, e.g. Newcastle disease virus
- C12N2760/18141—Use of virus, viral particle or viral elements as a vector
- C12N2760/18143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/32011—Picornaviridae
- C12N2770/32041—Use of virus, viral particle or viral elements as a vector
- C12N2770/32043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2770/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
- C12N2770/00011—Details
- C12N2770/36011—Togaviridae
- C12N2770/36111—Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
- C12N2770/36141—Use of virus, viral particle or viral elements as a vector
- C12N2770/36143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2795/00—Bacteriophages
- C12N2795/00011—Details
- C12N2795/00041—Use of virus, viral particle or viral elements as a vector
- C12N2795/00043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2800/00—Nucleic acids vectors
- C12N2800/22—Vectors comprising a coding region that has been codon optimised for expression in a respective host
Definitions
- the present disclosure relates, in part, to a first recombinant polynucleotide encoding an Interleukin (IL)-2 polypeptide and a second recombinant polynucleotide encoding an IL-12 polypeptide, recombinant nucleic acids and/or gene delivery vehicles comprising the same, pharmaceutical compositions and formulations thereof, and methods of their use (e.g., for treating cancer).
- IL Interleukin
- recombinant nucleic acids and/or gene delivery vehicles comprising the same, pharmaceutical compositions and formulations thereof, and methods of their use (e.g., for treating cancer).
- Cancer is among the leading causes of death worldwide. Despite significant advances in clinical care and treatment methods, more effective cancer treatment options are still needed to prolong survival and decrease cancer death rates.
- polynucleotides e.g., recombinant and/or isolated polynucleotides
- a first polynucleotide encoding an Interleukin (IL) 2 polypeptide
- a second polynucleotide encoding an IL-12 polypeptide
- recombinant nucleic acids and/or gene delivery vehicles e.g., a viral vector and/or a non-viral vector such as a lipid nanoparticle
- pharmaceutical compositions and/or formulations comprising the polynucleotides, recombinant nucleic acids, and/or gene delivery vehicles
- medicaments comprising the polynucleotides, recombinant nucleic acids, gene delivery vehicles, and/or pharmaceutical compositions or formulations, and/or methods of their use e.g., for treating cancer in a subject in need thereof.
- compositions or a pharmaceutical composition comprising a first recombinant polynucleotide encoding an IL-2 polypeptide and a second recombinant polynucleotide encoding an IL-12 polypeptide.
- the first polynucleotide and the second polynucleotide are contiguous.
- the first polynucleotide and the second polynucleotide are non-contiguous.
- the first polynucleotide and/or the second polynucleotide comprises deoxyribonucleic acid (DNA).
- the DNA is linear DNA or circular DNA.
- the first polynucleotide and/or the second polynucleotide comprises ribonucleic acid (RNA).
- the RNA is self-replicating. In some embodiments, the RNA is self-amplifying. In some embodiments, the RNA is not self-replicating. In some embodiments, the RNA is not self-amplifying.
- the RNA is a messenger RNA (mRNA) or a modified mRNA (mmRNA). In some embodiments, the mRNA and/or the mmRNA further comprises a 5' untranslated region (UTR), a 3' UTR, a polyadenylation (poly(A)) tail, and/or a 5' cap analog.
- the pharmaceutical composition comprises recombinant nucleic acids comprising any one or more of the recombinant polynucleotides described herein.
- the recombinant nucleic acid is a linear DNA, a circular DNA, an RNA, a mRNA, and/or a modified mRNA.
- the recombinant nucleic acid is a recombinant viral genome.
- the pharmaceutical composition comprises one or more gene delivery vehicles comprising the first polynucleotide and/or the second polynucleotide.
- the one or more gene delivery vehicles are one or more of a viral gene delivery vehicle and/or a non-viral gene delivery vehicle. In some embodiments, the one or more gene delivery vehicles are one or more of a viral vector and/or a non-viral vector.
- the one or more gene delivery vehicles are one or more viral vectors.
- the viral vector is pseudotyped.
- the viral vector comprises a recombinant viral genome.
- the recombinant viral genome comprises the first polynucleotide and/or the second polynucleotide.
- the recombinant viral genome is replication competent.
- the recombinant viral genome is replication defective.
- the recombinant viral genome is a recombinant oncolytic virus genome. In some embodiments, the recombinant viral genome is not a recombinant oncolytic virus genome.
- the recombinant viral genome is selected from a recombinant adenovirus genome, a recombinant retrovirus genome, a recombinant adeno-associated virus (AAV) genome, a recombinant herpes virus genome, a recombinant poxvirus genome, a recombinant bacteriophage genome, a recombinant alphavirus genome, a recombinant picornavirus genome, a recombinant iridovirus genome, a recombinant Newcastle disease virus genome, a recombinant baculovirus genome, a recombinant geminivirus genome, a recombinant caulimovirus genome, and any combinations and/or derivatives thereof.
- AAV recombinant adeno-associated virus
- the one or more gene delivery vehicles are one or more viral vectors.
- the viral vector is pseudotyped.
- the viral vector comprises the first polynucleotide and/or the second polynucleotide.
- the viral vector is replication competent.
- the viral vector is replication defective.
- the viral vector is an oncolytic virus. In some embodiments, the viral vector is not an oncolytic virus.
- the viral vector is selected from an adenovirus, a retrovirus, an AAV, a herpes virus, a poxvirus, a bacteriophage, an alphavirus, a picornavirus, a iridovirus, a Newcastle disease virus, a baculovirus, a geminivirus, a caulimovirus, and any combinations and/or derivatives thereof.
- the one or more gene delivery vehicles are one or more non-viral vectors.
- the non-viral vector is a chemical compound, a bacterium, a mammalian cell, or a physical delivery system.
- the chemical compound is selected from a polymer compound, a lipid compound, an inorganic compound, and any combinations and/or derivatives thereof.
- the polymer compound is a natural polymer, a synthetic polymer, a biopolymer, a biodegradable polymer, a cationic polymer, a protein polymer, a polysaccharide polymer, or any combinations and/or derivatives thereof.
- the lipid compound is selected from a nanoparticle, a lipid nanoparticle, a liposome, a cationic liposome, a solid lipid nanoparticle, a lipid emulsion, a lipidoid, a cytofectin, a lipid emulsion, a surfactant, a gemini surfactant, and any combinations and/or derivatives thereof.
- the inorganic compound is selected from a nanocarrier, a DNA nanclew, a gold nanoparticle, a carbon nanotube, a graphene, a quantum dot, an up-conversion nanoparticle, a silica nanoparticle, an iron oxide, a ferritin, and any combinations and/or derivatives thereof.
- the physical delivery system is selected from electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, hydroporation, and any combinations and/or derivatives thereof.
- aspects of the present disclosure relate to the use of any of the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, and/or pharmaceutical compositions described herein as a medicament.
- Other aspects of the present disclosure relate to the use of any of the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, and/or pharmaceutical compositions described herein in a therapy.
- aspects of the present disclosure relate to a method of expressing, enhancing, increasing, augmenting, and/or supplementing the levels of an IL-2 and/or IL-12 polypeptide in one or more cells of a subject comprising administering to the subject an effective amount of any of the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments described herein.
- the one or more cells are one or more cells of the respiratory tract, airway epithelial, and/or lung.
- the subject is a human.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered topically, transdermally, subcutaneously, epicutaneously, intradermally, orally, sublingually, buccally, rectally, vaginally, intravenously, intraarterially, intramuscularly, intraosseously, intracardially, intraperitoneally, transmucosally, intravitreally, subretinally, suprachoroidally, intracranially, intrathecally, intraventricularly, intraarticularly, peri- articularly, intratumorally, locally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered orally, intranasally, intratracheally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered intranasally or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a dry powder inhaler, a pressurized metered dose inhaler, a soft mist inhaler, a nebulizer, or an electrohydrodynamic aerosol device.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a nebulizer.
- the nebulizer is a vibrating mesh nebulizer.
- aspects of the present disclosure relate to a method of providing prophylactic, palliative, or therapeutic relief of one or more signs or symptoms of cancer in a subject in need thereof comprising administering to the subject an effective amount of any of the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments described herein.
- the cancer is selected from a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, skin cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- the cancer is small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, or squamous carcinoma of the lung.
- the cancer is non-small cell lung cancer.
- the cancer is osteosarcoma.
- the subject is a human.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered topically, transdermally, subcutaneously, epicutaneously, intradermally, orally, sublingually, buccally, rectally, vaginally, intravenously, intraarterially, intramuscularly, intraosseously, intracardially, intraperitoneally, transmucosally, intravitreally, subretinally, suprachoroidally, intracranially, intrathecally, intraventricularly, intraarticularly, peri-articularly, intratumorally, locally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered orally, intranasally, intratracheally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered orally, intranasally, intratracheally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered intranasally or via inhalation to the subject. In some embodiments, the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a dry powder inhaler, a pressurized metered dose inhaler, a soft mist inhaler, a nebulizer, or an electrohydrodynamic aerosol device.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a nebulizer.
- the nebulizer is a vibrating mesh nebulizer.
- cancers are selected from carcinoma, lymphoma, blastoma, sarcoma, a neuroendocrine tumor, mesothelioma, schwannoma, meningioma, adenocarcinoma, melanoma, leukemia, and lymphoid malignancy.
- the cancer is selected from a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, skin cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- the cancer is small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, or squamous carcinoma of the lung.
- the cancer is non-small cell lung cancer.
- the cancer is osteosarcoma.
- the subject is a human.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered topically, transdermally, subcutaneously, epicutaneously, intradermally, orally, sublingually, buccally, rectally, vaginally, intravenously, intraarterially, intramuscularly, intraosseously, intracardially, intraperitoneally, transmucosally, intravitreally, subretinally, suprachoroidally, intracranially, intrathecally, intraventricularly, intraarticularly, peri- articularly, intratumorally, locally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered orally, intranasally, intratracheally, or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered intranasally or via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered via inhalation to the subject.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a dry powder inhaler, a pressurized metered dose inhaler, a soft mist inhaler, a nebulizer, or an electrohydrodynamic aerosol device.
- the recombinant polynucleotides, recombinant nucleic acids and/or gene delivery vehicles comprising the recombinant polynucleotides, pharmaceutical compositions, and/or medicaments are administered using a nebulizer.
- the nebulizer is a vibrating mesh nebulizer.
- FIGS. 1A-1B depict secreted cytokine levels (IL-12, FIG. 1A; IL-2, FIG IB) by HEK293FT cells at 24-hours post infection with a modified herpes simplex virus encoding human IL-12 (FIG. 1A) and IL-2 (FIG. IB) transgene at a multiplicity of infection (MOI) of 1.
- IL-12 FIG. 1A
- IL-2 FIG IB
- MOI multiplicity of infection
- FIGS. 2A-2B depict in vitro bioactivity assay(s) of HSV-derived IL-12 (FIG. 2A) and IL-2 (FIG. 2B) compared to commercially available recombinant proteins as a function of IFNy release from splenocytes (FIG. 2A) or HEK-BlueTM IL2 reporter cells (FIG. 2B). Data indicative of cells assayed in triplicate, and data are presented as means ⁇ standard deviation (SD).
- SD standard deviation
- FIGS. 3A-3H show the in vivo pharmacokinetic evaluation of HSV-IL12 / HSV-IL2 following intradermal administration in healthy mice.
- FIG. 3A and FIG. 3C depict genome (circles) and transcript (square) levels in skin of C57BL/6 mice post-intradermal administration of HSV-IL12 / HSV-IL2 at the indicated time points.
- qPCR FIG. 3A circles and FIG. 3C circles
- qRT-PCT FIG. 3A squares and FIG. 3C squares
- FIG. 3E and FIG. 3F depict IL-12 (FIG. 3E) and IL-2 (FIG.
- FIG. 3G and FIG. 3H depict IL-12 (FIG. 3G) and IL-2 (FIG.
- FIGS. 4A-4/ show the in vivo evaluation of HSV-IL12 / HSV-IL2 following intratracheal administration in healthy mice.
- FIG. 4A depicts once weekly body weights in BALB/c female mice. Data are displayed as means ⁇ SEM.
- BALF bronchoalveolar lavage fluid
- FIGS. 4B-4C whole lungs
- FIGS. 4D-4E whole lungs
- 4H-4/ depict whole lungs that were taken at indicated time points to measure IL-12 and IL-2 concentrations. Cytokine concentrations were normalized to total protein.
- FIGS. 5A-5S depict HSV-IL12 / HSV-IL2 efficacy in an in vivo murine model of melanoma.
- FIG. 5A shows the study design. Euthanasia criteria were either tumor area > 150mm 2 or body weight loss > 20% of their pre-study body weight. Vectors were administered at ⁇ 10 8 total PFU. SC: subcutaneous.
- FIG. 5C shows the study design. Euthanasia criteria were either tumor area > 150mm 2 or body weight loss > 20% of their pre-study body weight.
- FIG. 5F shows the study design. Euthanasia criteria were either tumor area > 150mm 2 or body weight loss > 20% of their pre-study body weight. Vectors were administered at ⁇ 10 8 total PFU. SC: subcutaneous.
- For the rechallenge phase 5 naive age-matched C57BL/6 animals were inoculated with tumors to serve as positive controls for tumor growth.
- FIG. 5/ shows survival data displayed as individual animals.
- FIG. 5J-5S depict HSV-IL12 / HSV-IL2 treatment of a primary B16F10 melanoma resulted in an abscopal effect against a secondary B16F10 tumor.
- FIG. 5J shows a schematic of tumor inoculation and the study design. Euthanasia criteria were either tumor area > 150mm 2 or body weight loss > 20% of their pre-study body weight. Vectors were administered at ⁇ 10 8 total PFU.
- FIG. 5K primary/treated tumor
- FIG. 5L secondary tumor
- FIGS. 5M depicts survival data in the day 0 secondary tumor inoculation group. Data are displayed as individual animals.
- FIG. 5P depicts survival data in the day 4 secondary tumor inoculation group. Data are displayed as individual animals.
- FIGS. 6A-6H depict HSV-IL12 / HSV-IL2 efficacy in an in vivo murine model of osteosarcoma (e.g. osteosarcoma lung metastases).
- FIGS. 6A-6D show combinatorial HSV-IL12 / HSV-IL2 administered intratracheally enhanced tumor regression and survival in a K7M2 osteosarcoma lung metastasis model compared to control or single vector treatment.
- FIG. 6A depicts the study design. Euthanasia was performed if body weight loss was > 20% of their pre-study body weight. All vectors were administered at ⁇ 10 7 total PFU. IV: intravenous.
- FIGS. 6B-6C show body weight (FIG. 6B) and survival (FIG.
- FIG. 6C depicts representative H&E stained lung sections from animals that survived to day 100.
- FIGS. 6E-6H show intratracheal administration of HSV- IL12 / HSV-IL2 minimizes initial K7M2 lung tumor outgrowth and delays tumor recurrence without additional therapeutic intervention.
- FIG. 6E depicts a schematic of the study design. Euthanasia was performed if body weight loss was > 20% of their pre-study body weight. All vectors were administered at ⁇ 10 7 total PFU.
- the term “and/or” may include any and all combinations of one or more of the associated listed items.
- the term “a and/or b” may refer to “a alone”, “b alone”, “a or b”, or “a and b”;
- the term “a, b, and/or c” may refer to “a alone”, “b alone”, “c alone”, “a or b”, “a or c", “b or c”, “a, b, or c", “a and b”, “a and c", “b and c", or “a, b, and c”; etc.
- aspects and embodiments of the present disclosure include “comprising”, “consisting”, and “consisting essentially of” aspects and embodiments.
- polynucleotide As used herein, the terms “polynucleotide”, “nucleic acid sequence”, “nucleic acid”, and variations thereof shall be generic to polydeoxyribonucleotides (containing 2-deoxy-D-ribose), to polyribonucleotides (containing D-ribose), to any other type of polynucleotide that is an N-glycoside of a purine or pyrimidine base, and to other polymers containing non-nucleotidic backbones, provided that the polymers contain nucleobases in a configuration that allows for base pairing and base stacking, as found in DNA and RNA.
- these terms include known types of nucleic acid sequence modifications, for example, substitution of one or more of the naturally occurring nucleotides with an analog, and inter-nucleotide modifications.
- a nucleic acid is "operatively linked” or “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
- a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence, or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
- operatively linked or “operably linked” means that the DNA or RNA sequences being linked are contiguous.
- an "open reading frame” or “ORF” refers to a continuous stretch of nucleic acids, either DNA or RNA, that encode a protein or polypeptide.
- the nucleic acids comprise a translation start signal or initiation codon, such as ATG or AUG, and a termination codon.
- an "untranslated region” or “UTR” refers to untranslated nucleic acids at the 5' and/or 3' ends of an open reading frame.
- UTRs include one or more UTRs in a polynucleotide may affect post-transcriptional regulation, mRNA stability, translation of the polynucleotide, etc.
- polypeptide As used herein, the terms “polypeptide,” “protein,” and “peptide” are used interchangeably and may refer to a polymer of two or more amino acids.
- a "subject", “host”, or an “individual” refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, as well as animals used in research, such as mice, rats, hamsters, rabbits, and nonhuman primates, etc.
- the mammal is human.
- the terms “pharmaceutical formulation” or “pharmaceutical composition” refer to a preparation which is in such a form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition or formulation would be administered.
- “Pharmaceutically acceptable” excipients e.g., vehicles, additives
- an "effective amount” is at least the minimum amount required to affect a measurable improvement or prevention of one or more symptoms of a particular disorder.
- An “effective amount” may vary according to factors such as the disease state, age, sex, and weight of the patient.
- An effective amount is also one in which any toxic or detrimental effects of the treatment are outweighed by the therapeutically beneficial effects.
- beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
- beneficial or desired results include clinical results such as decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications used to treat symptoms of the disease, delaying the progression of the disease, and/or prolonging survival.
- An effective amount can be administered in one or more administrations.
- an effective amount of a recombinant polynucleotide, gene delivery vehicle comprising the recombinant polynucleotide, pharmaceutical composition, and/or medicament is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
- an effective amount of a recombinant polynucleotide, gene delivery vehicle comprising the recombinant polynucleotide, pharmaceutical composition, and/or medicament may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
- an "effective amount" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
- treatment refers to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease/disorder/defect progression, ameliorating, or palliating the disease/disorder/defect state, and remission or improved prognosis.
- the term "delaying progression of" a disease/disorder/defect refers to deferring, hindering, slowing, retarding, stabilizing, and/or postponing development of the disease/disorder/defect. This delay can be of varying lengths or time, depending on the history of the disease/disorder/defect and/or the individual being treated. As is evident to one of ordinary skill in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
- the present disclosure relates to a first polynucleotide (e.g., a first recombinant polynucleotide, a first isolated polynucleotide) encoding an IL-2 polypeptide and a second polynucleotide (e.g., a second recombinant polynucleotide, a second isolated polynucleotide) encoding an IL-12 polypeptide.
- a first polynucleotide e.g., a first recombinant polynucleotide, a first isolated polynucleotide
- a second polynucleotide e.g., a second recombinant polynucleotide, a second isolated polynucleotide
- Any suitable IL-2 and/or IL-12 polypeptide described herein or known in the art may be encoded by one or more polynucleotides of the present disclosure, including, for example
- a polynucleotide of the present disclosure comprises the wild-type coding sequence of any IL-2 and/or IL-12 gene described herein or known in the art (including any isoform or splice variant thereof), including, for example, an /..? gene (see e.g., NCBI Gene ID: 3558; SEQ ID NO: 5), an IL12A gene (see e.g., NCBI Gene ID: 3592; SEQ ID NO: 6), an IL12B gene (see e.g., NCBI Gene ID: 3593; SEQ ID NO: 7), etc.
- an /..? gene see e.g., NCBI Gene ID: 3558; SEQ ID NO: 5
- an IL12A gene see e.g., NCBI Gene ID: 3592; SEQ ID NO: 6
- an IL12B gene see e.g., NCBI Gene ID: 3593; SEQ ID NO: 7
- a polynucleotide of the present disclosure comprises a sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the sequence of any of the human IL-2 and/or IL-12 genes (and/or coding sequences thereof) described herein or known in the art.
- a polynucleotide of the present disclosure comprises a codon-optimized variant of the wild-type coding sequence of any IL-2 and/or IL-12 gene described herein or known in the art.
- use a of a codon-optimized variant of the coding sequence of a gene increases stability and/or yield of heterologous expression (RNA and/or protein) of the encoded polypeptide in a target cell, as compared to the stability and/or yield of heterologous expression of a corresponding, non-codon-optimized, wild-type sequence.
- a first polynucleotide of the present disclosure encodes an IL-2 polypeptide.
- the IL-2 polypeptide is a human IL-2 polypeptide (see e.g., UniProt accession number: P60568).
- the polynucleotide comprises the coding sequence of a wild-type IL2 gene (see e.g., NCBI Gene ID: 3558, SEQ ID NO: 5), or a codon-optimized variant thereof (see e.g., SEQ ID NO: 8).
- a polynucleotide encoding an IL-2 polypeptide is a polynucleotide that encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 1.
- a polynucleotide encoding an IL-2 polypeptide is a polynucleotide that encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 1.
- a first polynucleotide encoding an IL-2 polypeptide is a polynucleotide that encodes an N-terminal truncation, a C-terminal truncation, or a fragment of the amino acid sequence of SEQ ID NO: 1.
- N-terminal truncations, C-terminal truncations, or fragments may comprise at least 10, at least 12, at least 14, at least 16, at least 18, at least 20, at least 30, at least 40, at least 50, at least 75, at least 100, but fewer than 153, consecutive amino acids of SEQ ID NO: 1.
- a second polynucleotide of the present disclosure encodes an IL- 12 subunit a polypeptide.
- the IL-12 subunit a polypeptide is a human IL-12 subunit a polypeptide (see e.g., UniProt accession number: P29459).
- the polynucleotide comprises the coding sequence of a wild-type IL12A gene (see e.g., NCBI Gene ID: 3592, SEQ ID NO: 6), or a codon-optimized variant thereof (see e.g., SEQ ID NO: 9).
- a polynucleotide encoding an IL-12 subunit a polypeptide is a polynucleotide that encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 2.
- a polynucleotide encoding an IL-12 subunit a polypeptide is a polynucleotide that encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 2.
- a second polynucleotide encoding an IL-12 subunit a polypeptide is a polynucleotide that encodes an N-terminal truncation, a C-terminal truncation, or a fragment of the amino acid sequence of SEQ ID NO: 2.
- N-terminal truncations, C-terminal truncations, or fragments may comprise at least 10, at least 12, at least 14, at least 16, at least 18, at least 20, at least 30, at least 40, at least 50, at least 75, at least 100, at least 200, but fewer than 219, consecutive amino acids of SEQ ID NO: 2.
- a second polynucleotide of the present disclosure encodes an IL- 12 subunit p polypeptide.
- the IL-12 subunit polypeptide is a human IL-12 subunit p polypeptide (see e.g., UniProt accession number: P29460).
- the polynucleotide comprises the coding sequence of a wild-type IL12B gene (see e.g., NCBI Gene ID: 3593, SEQ ID NO: 7), or a codon-optimized variant thereof (see e.g., SEQ ID NO: 10).
- a recombinant polynucleotide encoding an IL-12 subunit p polypeptide is a polynucleotide that encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 3.
- a polynucleotide encoding an IL-12 subunit p polypeptide is a polynucleotide that encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 3.
- a second polynucleotide encoding an IL-12 subunit p polypeptide is a polynucleotide that encodes an N-terminal truncation, a C-terminal truncation, or a fragment of the amino acid sequence of SEQ ID NO: 3.
- N-terminal truncations, C-terminal truncations, or fragments may comprise at least 10, at least 12, at least 14, at least 16, at least 18, at least 20, at least 30, at least 40, at least 50, at least 75, at least 100, at least 200, at least 300, but fewer than 328, consecutive amino acids of SEQ ID NO: 3.
- a polynucleotide encoding an IL-2 polypeptide, a polynucleotide encoding an IL-12 subunit a polypeptide, and/or a polynucleotide encoding an IL-12 subunit p polypeptide is a single contiguous polynucleotide (e.g., a single contiguous DNA, a single contiguous RNA (such as a synthetic mRNA)).
- a polynucleotide encoding an IL-2 polypeptide, a polynucleotide encoding an IL-12 subunit a polypeptide, and/or a polynucleotide encoding an IL-12 subunit p polypeptide is a single contiguous polynucleotide contained within a single DNA molecule.
- a polynucleotide encoding an IL-2 polypeptide, a polynucleotide encoding an IL-12 subunit a polypeptide, and/or a polynucleotide encoding an IL-12 subunit p polypeptide is a single contiguous polynucleotide contained within a single RNA molecule (e.g., a synthetic RNA molecule).
- a polynucleotide encoding an IL-2 polypeptide, a polynucleotide encoding an IL-12 subunit a polypeptide, and/or a polynucleotide encoding an IL-12 subunit p polypeptide is a single contiguous polynucleotide contained within a single mRNA (e.g., a single synthetic mRNA or mmRNA).
- the single contiguous polynucleotide encodes an IL-2 polypeptide on a first open reading frame (ORF) and an IL-12 polypeptide on a second open reading frame (ORF).
- the two ORFs are separated by an internal ribosomal entry site (IRES).
- IRES internal ribosomal entry site
- the single contiguous polynucleotide encodes an IL-2 polypeptide on a first open reading frame (ORF), an IL-12 subunit a polypeptide on a second open reading frame (ORF), and an IL-12 subunit p polypeptide on a third open reading frame (ORF).
- the first, second, and third ORFs are each separated by an IRES.
- IRES's may include, but are not limited to, a virally-derived IRES (e.g. an IRES derived from a poliovirus, rhinovirus, encephalomyocarditis virus, foot-and-mouth disease virus, hepatitis C virus, classic swine fever virus, rous sarcoma virus, human immunodeficiency virus, cricket paralysis virus, Kaposi's sarcoma-associated herpesvirus, etc.) and a cellular mRNA-derived IRES (e.g.
- a virally-derived IRES e.g. an IRES derived from a poliovirus, rhinovirus, encephalomyocarditis virus, foot-and-mouth disease virus, hepatitis C virus, classic swine fever virus, rous sarcoma virus, human immunodeficiency virus, cricket paralysis virus, Kaposi's sarcoma-associated herpesvirus, etc.
- a virally-derived IRES e.
- an IRES derived from growth factor mRNAs such as fibroblast growth factor 2, platelet- derived growth factor B, and vascular endothelial growth factor
- an IRES derived from transcription factor mRNAs such as antennapedia, ultrapithoraxm, and NF-KB repressing factor
- an IRES derived from oncogene mRNAs such as c-myc, pim-1, and protein kinase p58PITSLRE, etc.
- a polynucleotide encoding any of the chimeric polypeptides described herein is encoded on a single ORF.
- a polynucleotide of the present disclosure encodes any one or more of an lnterleukin-2 (IL-2) peptide, an Interleukin-12 subunit alpha (IL-12a) peptide, an Interleukin-12 subunit beta (IL-12P) peptide, and/or any chimeric polypeptides thereof, in any combination.
- a chimeric polypeptide comprises a linker polypeptide.
- the linker polypeptide is a cleavable linker polypeptide. Any cleavable linker polypeptide known in the art may be used in the chimeric polypeptides of the present disclosure, including, for example, a T2A linker, a P2A linker, a E2A linker, and F2A linker, etc.
- the linker polypeptide is a T2A linker polypeptide.
- An exemplary nucleic acid sequence encoding a T2A linker polypeptide is provided as SEQ ID NO: 12.
- An exemplary amino acid sequence of a T2A linker polypeptide is provided as SEQ ID NO: 16.
- the linker polypeptide is a P2A linker polypeptide.
- An exemplary nucleic acid sequence encoding a P2A linker polypeptide is provided as SEQ ID NO: 13.
- An exemplary amino acid sequence of a P2A linker polypeptide is provided as SEQ ID NO: 17.
- the linker polypeptide is an E2A linker polypeptide.
- An exemplary nucleic acid sequence encoding an E2A linker polypeptide is provided as SEQ ID NO: 14.
- An exemplary amino acid sequence of an E2A linker polypeptide is provided as SEQ ID NO: 18.
- the linker polypeptide is an F2A linker polypeptide.
- an exemplary nucleic acid sequence encoding an F2A linker polypeptide is provided as SEQ ID NO: 15.
- An exemplary amino acid sequence of an F2A linker polypeptide is provided as SEQ ID NO: 19.
- the linker polypeptide comprises a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to an amino acid sequence selected from SEQ ID NOS: 16-19.
- the linker polypeptide comprises a sequence selected from SEQ ID NOS: 16-19.
- the linker polypeptide is a non-cleavable linker polypeptide.
- Any non-cleavable linker polypeptide known in the art may be used in the chimeric polypeptides of the present disclosure, including, for example, a GGGGSGGGGSGGGGS (SEQ ID NO: 20) linker, a GGSSRSSSSGGGGSGGGG (SEQ ID NO: 21) linker, a GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 22) linker, a CGGGSGGGGSGGGGS (SEQ ID NO: 23) linker, a SHGGHGGGGSGGGGS (SEQ ID NO: 24) linker, a MGGMSGGGGSGGGGS (SEQ ID NO: 25) linker, a YGGYSGGGGSGGGGS (SEQ ID NO: 26) linker, a WGGYSGGGGSGGGGS (SEQ ID NO: 27) linker, a SVSVGMKPSPRP (SEQ ID NO: 20
- the linker polypeptide comprises a sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to an amino acid sequence selected from SEQ ID NOS: 20-31.
- the linker polypeptide comprises a sequence selected from SEQ ID NOS: 20-31.
- a second polynucleotide of the present disclosure encodes an IL- 12 subunit a polypeptide and an IL-12 subunit p polypeptide.
- the IL-12 subunit a polypeptide is a human IL-12 subunit a polypeptide (see e.g., UniProt accession number: P29459).
- the polynucleotide comprises the coding sequence of a wild-type IL12A gene (see e.g., NCBI Gene ID: 3592, SEQ ID NO: 6), or a codon-optimized variant thereof (see e.g., SEQ ID NO: 9).
- a polynucleotide encoding an IL-12 subunit a polypeptide is a polynucleotide that encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 2.
- a polynucleotide encoding an IL-12 subunit a polypeptide is a polynucleotide that encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 2.
- the IL-12 subunit p polypeptide is a human IL-12 subunit polypeptide (see e.g., UniProt accession number: P29460).
- the polynucleotide comprises the coding sequence of a wild-type IL12B gene (see e.g., NCBI Gene ID: 3593, SEQ ID NO: 7), or a codon-optimized variant thereof (see e.g., SEQ ID NO: 10).
- a recombinant polynucleotide encoding an IL-12 subunit p polypeptide is a polynucleotide that encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 3.
- a polynucleotide encoding an IL-12 subunit p polypeptide is a polynucleotide that encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 3.
- the IL-as subunit a and polypeptides are separated by a linker polypeptide.
- a second polynucleotide of the present disclosure encodes a chimeric polypeptide comprising an IL-12 subunit p polypeptide and an IL-12 subunit a polypeptide.
- the chimeric polypeptide further comprises a linker polypeptide linking the IL- 12 subunit p polypeptide and the IL-12 subunit a polypeptide.
- the chimeric polypeptide comprises, from N-terminus to C-terminus, the IL-12 subunit a polypeptide-the linker polypeptide-the IL-12 subunit p polypeptide.
- the chimeric polypeptide comprises, from N-terminus to C-terminus, the IL-12 subunit p polypeptide-the linker polypeptide-the IL-12 subunit a polypeptide.
- An exemplary nucleic acid sequence encoding a chimeric polypeptide comprising a human IL12 subunit p polypeptide, a linker polypeptide, and a human IL12 subunit a polypeptide is provided as SEQ ID NO: 11.
- An exemplary amino acid sequence of a chimeric polypeptide comprising a human IL12 subunit p polypeptide, a linker polypeptide, and a human I L12 subunit a polypeptide is provided as SEQ ID NO: 4.
- a polynucleotide of the present disclosure encodes a polypeptide comprising an amino acid sequence having at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to the sequence of SEQ ID NO: 4.
- a polynucleotide of the present disclosure encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 4.
- a polynucleotide of the present disclosure does not encode an lnterleukin-4 (IL-4) polypeptide. In some embodiments, a polynucleotide of the present disclosure does not encode an Interleukin-10 (IL-10) polypeptide. In some embodiments, a polynucleotide of the present disclosure does not encode a Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) polypeptide.
- GM-CSF Granulocyte-Macrophage Colony-Stimulating Factor
- a polynucleotide of the present disclosure does not encode an lnterleukin-4 (IL-4) polypeptide, an Interleukin-10 (IL-10) polypeptide, and/or a Granulocyte- Macrophage Colony-Stimulating Factor (GM-CSF) polypeptide.
- IL-4 lnterleukin-4
- IL-10 Interleukin-10
- GM-CSF Granulocyte- Macrophage Colony-Stimulating Factor
- the first polynucleotide encoding an IL-2 polypeptide and/or the second polynucleotide encoding an IL-12 polypeptide encodes one or more additional polypeptides.
- the first polynucleotide encoding an IL-2 polypeptide and/or the second polynucleotide encoding an IL-12 polypeptide does not encode any additional polypeptides. In some embodiments, the first polynucleotide encoding an IL-2 polypeptide and/or the second polynucleotide encoding an IL-12 polypeptide encodes one or more additional cytokines and/or chemokines. In some embodiments, the first polynucleotide encoding an IL-2 polypeptide and/or the second polynucleotide encoding an IL-12 polypeptide does not encode any additional cytokines and/or chemokines.
- a polynucleotide of the present disclosure (e.g., a recombinant polynucleotide) encoding a polypeptide (e.g., an IL-2 and/or an IL-12 polypeptide) may further encode additional coding and non-coding sequences.
- a polypeptide e.g., an IL-2 and/or an IL-12 polypeptide
- additional coding and non-coding sequences may include, but are not limited to, sequences encoding additional polypeptide tags (e.g., encoded in-frame with the polypeptide in order to produce a fusion protein), introns (e.g., native, modified, or heterologous introns), 5' and/or 3' UTRs (e.g., native, modified, or heterologous 5' and/or 3' UTRs), and the like.
- additional polypeptide tags e.g., encoded in-frame with the polypeptide in order to produce a fusion protein
- introns e.g., native, modified, or heterologous introns
- 5' and/or 3' UTRs e.g., native, modified, or heterologous 5' and/or 3' UTRs
- suitable polypeptide tags may include, but are not limited, to any combination of purification tags, such as his-tags, flag-tags, maltose binding protein and glutathione-S-transferase tags, detection tags, such as tags that may be detected photometrically (e.g., green fluorescent protein, red fluorescent protein, etc.) and tags that have a detectable enzymatic activity (e.g., alkaline phosphatase, etc.), tags containing secretory sequences, signal sequences, leader sequences, and/or stabilizing sequences, protease cleavage sites (e.g., furin cleavage sites, TEV cleavage sites, Thrombin cleavage sites, etc.), and the like.
- purification tags such as his-tags, flag-tags, maltose binding protein and glutathione-S-transferase tags
- detection tags such as tags that may be detected photometrically (e.g., green fluorescent protein, red fluorescent protein, etc.) and tags
- the 5' and/or 3'UTRs increase the stability, localization, and/or translational efficiency of the polynucleotides. In some embodiments, the 5' and/or 3'UTRs improve the level and/or duration of protein expression. In some embodiments, the 5' and/or 3'UTRs include elements (e.g., one or more miRNA binding sites, etc.) that may block or reduce off-target expression (e.g., inhibiting expression in specific cell types (e.g., neuronal cells), at specific times in the cell cycle, at specific developmental stages, etc.). In some embodiments, the 5' and/or 3'UTRs include elements (e.g., one or more miRNA binding sites, etc.) that may enhance expression of the encoded polypeptide in specific cell types.
- elements e.g., one or more miRNA binding sites, etc.
- a polynucleotide of the present disclosure e.g., a recombinant polynucleotide
- a polypeptide e.g., an IL-2 and/or an IL-12 polypeptide
- a regulatory sequence may include enhancers, insulators, promoters, and other expression control elements (e.g., polyadenylation signals).
- enhancer(s) known in the art may be used, including, for example, enhancer sequences from mammalian genes (such as globin, elastase, albumin, a-fetoprotein, insulin and the like), enhancer sequences from a eukaryotic cell virus (such as SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, adenovirus enhancers, and the like), and any combinations thereof.
- mammalian genes such as globin, elastase, albumin, a-fetoprotein, insulin and the like
- enhancer sequences from a eukaryotic cell virus such as SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, adenovirus enhancers, and
- any suitable insulator(s) known in the art may be used, including, for example, HSV chromatin boundary (CTRL/CTCF-binding/insulator) elements CTRL1 and/or CTRL2, chicken hypersensitive site 4 insulator (cHS4), human HNRPA2B1— CBX3 ubiquitous chromatin opening element (UCOE), the scaffold/matrix attachment region (S/MAR) from the human interferon beta gene (IFNB1), and any combinations thereof.
- HSV chromatin boundary (CTRL/CTCF-binding/insulator) elements CTRL1 and/or CTRL2
- cHS4 chicken hypersensitive site 4 insulator
- UCOE ubiquitous chromatin opening element
- S/MAR scaffold/matrix attachment region from the human interferon beta gene
- any suitable promoter e.g., suitable for transcription in mammalian host cells
- suitable promoters including, for example, promoters obtained from the genomes of viruses (such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus, Simian Virus 40 (SV40), and the like), promoters from heterologous mammalian genes (such as the actin promoter (e.g., the p-actin promoter), a ubiquitin promoter (e.g., a ubiquitin C (UbC) promoter), a phosphoglycerate kinase (PGK) promoter, an immunoglobulin promoter, from heat-shock promoters, and the like), promoters from homologous mammalian genes, synthetic promoters (such as
- a polynucleotide of the present disclosure (e.g., a recombinant polynucleotide) is operably linked to one or more heterologous promoters.
- the one or more heterologous promoters are one or more of constitutive promoters, tissue-specific promoters, temporal promoters, spatial promoters, inducible promoters, and repressible promoters.
- the one or more heterologous promoters are one or more of the human cytomegalovirus (HCMV) immediate early promoter, the human elongation factor-1 (EFl) promoter, the human p-actin promoter, the human UbC promoter, the human PGK promoter, the synthetic CAG promoter, and any combinations thereof.
- HCMV human cytomegalovirus
- EFl human elongation factor-1
- p-actin promoter the human p-actin promoter
- the human UbC promoter the human PGK promoter
- synthetic CAG promoter and any combinations thereof.
- a polynucleotide of the present disclosure e.g., a recombinant polynucleotide
- a polypeptide e.g., an IL-2 and/or an IL-12 polypeptide
- a polypeptide expresses the polypeptide when the polynucleotide is delivered into one or more target cells of a subject (e.g., one or more cancer cells, one or more healthy (non-cancerous) cells, one or more cells of the respiratory tract, airway, lungs, etc. of the subject).
- expression of the polypeptide enhances, increases, augments, and/or supplements the levels, function, and/or activity of the polypeptide in one or more target cells of a subject (e.g., as compared to prior to expression of the polypeptide, as compared to levels of the endogenous polypeptide expressed in the cell, etc.).
- expression of the polypeptide provides prophylactic, palliative, or therapeutic relief of one or more signs or symptoms of cancer (e.g., solid tumor, hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, skin cancer, stomach cancer, thymic epithelial cancer, thyroid cancer, etc.) in a subject (e.g., as compared to prior to expression of the polypeptide).
- cancer e.g., solid tumor, hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, skin cancer
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Collagen alpha-1 (VII) chain polypeptide (C0L7).
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Lysyl hydroxylase 3 polypeptide (LH3).
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Keratin type I cytoskeletal 17 polypeptide (KRT17). In some embodiments, a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a transglutaminase (TGM) polypeptide (e.g., a human transglutaminase polypeptide such as a human TGM1 polypeptide and/or a human TGM5 polypeptide).
- TGM transglutaminase
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a cosmetic protein (e.g., collagen proteins, fibronectins, elastins, lumicans, vitronectins/vitronectin receptors, laminins, neuromodulators, fibrillins, additional dermal extracellular matrix proteins, etc.).
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) an antibody (e.g., a full-length antibody, an antibody fragment, etc.).
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Serine Protease Inhibitor Kazal-type (SPINK) polypeptide (e.g., a human SPINK polypeptide, such as a SPINK5 polypeptide).
- SPINK Serine Protease Inhibitor Kazal-type
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a filaggrin or filaggrin 2 polypeptide (e.g., a human filaggrin or filaggrin 2 polypeptide).
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) polypeptide (e.g., a human CFTR polypeptide).
- CFTR Cystic Fibrosis Transmembrane Conductance Regulator
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) an ichthyosis-associated polypeptide (e.g., an ATP-binding cassette sub-family A member 12 polypeptide, a l-acylglycerol-3-phosphate O-acyltransferase ABHD5 polypeptide, an Aldehyde dehydrogenase family 3 member A2 polypeptide, an Arachidonate 12-lipoxygenase 12R-type polypeptide, a Hydroperoxide isomerase ALOXE3 polypeptide, an AP-1 complex subunit sigma-lA polypeptide, an Arylsulfatase E polypeptide, a Caspase-14 polypeptide, a Corneodesmosin polypeptide, a Ceramide synthase 3 polypeptide, a Carbohydrate sulfotransferase 8 polypeptide
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Collagen alpha-1 (VII) chain polypeptide, a Lysyl hydroxylase 3 polypeptide, a Keratin type I cytoskeletal 17 polypeptide, and/or any chimeric polypeptides thereof.
- a polynucleotide of the present disclosure does not comprise the coding sequence of (e.g., a transgene encoding) a Collagen alpha-1 (VII) chain polypeptide, a Lysyl hydroxylase 3 polypeptide, a Keratin type I cytoskeletal 17 polypeptide, a transglutaminase (TGM) polypeptide, a filaggrin polypeptide, a cosmetic protein, an antibody, a SPINK polypeptide, a CFTR polypeptide, an ichthyosis-associated polypeptide, an Alpha-l-antitrypsin polypeptide, a Sodium-dependent phosphate transport protein 2B polypeptide, a Dynein heavy chain 5 axonemal polypeptide, a Dynein heavy chain 11 axonemal polypeptide, a Coiled-coil domain-containing protein 39 polypeptide, a Dynein intermediate chain 1 axonemal polypeptide, a Coiled-
- the present disclosure relates to synthetic RNAs containing one or more polynucleotides described herein.
- the synthetic RNAs are synthetic mRNAs containing one or more polynucleotides described herein.
- the synthetic RNAs comprise a 5' -cap structure. Examples of 5' -cap structures may include, but are not limited to, cap-0, cap-1, cap-2, and cap-3 structures, and any combinations and/or derivatives thereof.
- the synthetic RNAs comprise a 3'-poly(A) tail.
- the synthetic RNAs comprise one or more 5' and/or 3' UTRs flanking the one or more coding sequences contained within the synthetic RNAs.
- the 5' and/or 3' UTRs increase the stability, localization, and/or translational efficiency of the synthetic RNAs.
- the 5' and/or 3' UTRs are modified to increase the stability, localization, and/or translational efficiency of the synthetic RNAs.
- the 5' and/or 3' UTRs improve the level and/or duration of protein expression.
- the 5' and/or 3' UTRs are modified to improve the level and/or duration of protein expression.
- the 5' and/or 3'UTRs include elements (e.g., miRNA binding sites, etc.) that may limit off-target expression (e.g., inhibiting expression in specific cell types (e.g., neuronal cells), at specific times in the cell cycle, at specific developmental stages, etc.).
- the 5' UTRs comprise a Kozak sequence.
- the Kozak sequence is the same or substantially similar to the Kozak consensus sequence.
- modified ribonucleotides may include, but are not limited to, 2-thiouridine, 5-azauridine, pseudouridine, 4-thiouridine, 5-methyluridine, 5-aminouridine, 5- hydroxyuridine, 5-methyl-5-azauridine, 5-amino-5-azauridine, 5-hydroxy-5-azauridine, 5- methylpseudouridine, 5-aminopseudouridine, 5-hydroxypseudouridine, 4-thio-5-azauridine, 4- thiopseudouridine, 4-thio-5-methyluridine, 4-thio-5-aminouridine, 4-thio-5-hydroxyuridine, 4-thio-5- methyl-5-azauridine, 4-thio-5-amino-5-azauridine, 4-thio-5-hydroxy-5-azauridine, 4-thio-5-methylpseudouridine, 4-thio-5-aminopseudouridine, 4-thio-5-hydroxypseudouridine, 2-thiocytidine, 5-azacyt
- a polynucleotide encoding an IL-2 polypeptide and a polynucleotide encoding an IL-12 polypeptide are contained within two separate synthetic RNA molecules (e.g., two separate synthetic mRNAs).
- a polynucleotide encoding an IL-2 polypeptide, a polynucleotide encoding an IL-12 subunit a polypeptide, and/or a polynucleotide encoding an IL-12 subunit p polypeptide are contained within three separate synthetic RNA molecules (e.g., three separate synthetic mRNAs).
- recombinant nucleic acids comprising any one or more of the polynucleotides described herein (e.g., the first recombinant polynucleotide encoding an IL-2 polypeptide and/or the second recombinant polynucleotide encoding an IL-12 polypeptide).
- the recombinant nucleic acid is a linear DNA, a circular DNA, a closed ended DNA (ceDNA), an RNA (e.g., a synthetic RNA), an mRNA, and/or an mmRNA.
- the recombinant nucleic acid is a recombinant viral genome.
- a recombinant nucleic acid of the present disclosure is a linear DNA.
- the linear DNA is a linear expression template (LET).
- LET linear expression template
- a LET may comprise a promoter region, one or more recombinant polynucleotides described herein, and/or a transcriptional terminator.
- a LET may be produced in vitro via polymerase chain reaction (PCR) from , for example, plasmid DNA or genomic DNA.
- a recombinant nucleic acid of the present disclosure is a close ended DNA (ceDNA; e.g., as disclsoed in U.S. Patent Application Publication NO. 2021/0059953 and/or U.S. Patent Application Publication NO. 2020/0283794, the contents of which are hereby incorporated by reference in its entirety).
- a recombinant nucleic acid of the present disclosure is a circular DNA.
- the circular DNA is a plasmid (e.g., as disclosed in U.S. Patent Application Publication No. 2013/0195800 and U.S. Patent Nos. 5,693,622, 5,589,466, and 5,580,859, the contents of which are hereby incorporated by reference in its entirety), a cosmid, or a bacterial artificial chromosome (BAC).
- the circular DNA is a plasmid.
- a plasmid can be in the form of circular double-stranded DNA.
- the plasmid may contain autonomously replicating sequences, genome integrating sequences, and/or phage or nucleotide sequences.
- the plasmid may be linear, circular, or supercoiled, of a single- or double-stranded DNA or RNA.
- the plasmids described herein may be derived from any source in which a number of nucleotide sequences have been joined or recombined into a unique construction which is capable of introducing a promoter fragment and DNA sequence for a selected gene product along with appropriate 3' untranslated sequence into a cell.
- the circular DNA is a cosmid.
- a cosmid is a type of plasmid that contains a Lambda phage cos sequence.
- Cosmids may contain 37 to 52 kb of DNA.
- cosmids may replicate if they have an origin of replication (e.g. an SV40 in mammalian cells).
- cosmids may be packaged into phage capsids.
- the cosmid is a plasmid with a bacterial oriV, an antibiotic selection marker, and/or a cloning site.
- the cosmid may have one or more (e.g. two) cos sites derived from bacteriophage lambda.
- the circular DNA is a bacterial artificial chromosome (BAC).
- a BAC is a DNA construct, based on functional fertility plasmid (e.g. F-plasmid).
- the BAC may accommodate an insert size of 150-350 kbp.
- the BAC may comprise: an oriS and/or a repE'F for plasmid replication and/or regulation of copy number; parA and/or parB which may partition F plasmid DNA into daughter cells during division and may ensure stable maintenance of the BAC; a selection marker; and/or T7 and/or Sp6 for transcription of inserted genes.
- a recombinant nucleic acid of the present disclosure is an RNA, mRNA , and/or mmRNA (e.g., as disclosed in U.S. Patent Nos. 10,898,574, 10,702,600, and 10,933,127, the contents of which are hereby incorporated by reference in their entirety).
- the RNA, mRNA, and/or mmRNA may enhance the efficiency of protein production, intracellular retention of nucleic acids, and viability of contacted cells, as well as possess reduced immunogenicity.
- the RNA, mRNA, and/or mmRNA may have a length that is greater than about 30 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000 or up to and including 100,000 nucleotides).
- nucleotides in length e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500
- the RNA, mRNA, and/or mmRNA may contain one or more modified nucleosides (e.g., as described above) which may have useful properties including the lack of a substantial induction of the innate immune system of a cell into which the RNA, mRNA, and/or mmRNA is introduced.
- modified nucleosides e.g., as described above
- the modified nucleosides may not be uniformly modified along the entire length of the molecule.
- Different nucleotide modifications and/or backbone structures may exist at various positions in the nucleic acid.
- the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the function of the nucleic acid is not substantially decreased.
- a modification may also be a 5' or 3' terminal modification.
- the nucleic acids may contain at a minimum one and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 50% modified nucleotides, at least 80% modified nucleotides, or at least 90% modified nucleotides.
- the modified nucleosides may include modified nucleosides include pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio- pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1- carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5- taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1- taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-l-methyl- pseudouridine, 2-thio-l-methyl-pseudouridine, 1-methyl-l-deaza-pseudouridine, 2-thi
- modified nucleosides include 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5- hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2- thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-l-methyl-pseudoisocytidine, 4-thio-l-methyl-l-deaza-pseudoisocytidine, 1-methyl-l-deaza-pseudoisocytidine, zebularine, 5-aza- zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine,
- the modified nucleosides may include 2-aminopurine, 2, 6- diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2- aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6- methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio- N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6- threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N
- the modified nucleosides may include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7- deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2- dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2- methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
- RNA, mRNA, and/or mmRNA multiple distinct 5'-cap structures can be used to generate the 5'-cap of the RNA, mRNA, and/or mmRNA described herein.
- Many chemical cap analogs are used to co-transcriptionally cap a synthetic mRNA molecule.
- the Anti-Reverse Cap Analog (ARCA) cap contains a 5'-5'- triphosphate guanine-guanine linkage where one guanine contains an N7 methyl group as well as a 3'-O-methyl group.
- chemical cap analogs allow for the concomitant capping of an RNA molecule, up to 20% of transcripts remain uncapped and the synthetic cap analog is not identical to an endogenous 5'-cap structure of an authentic cellular mRNA. This may lead to reduced translational competency and reduced cellular stability.
- the RNA, mRNA, and/or mmRNA includes the poly-A tail.
- the poly-A tail is designed relative to the length of the overall modified RNA, mRNA, and/or mmRNA. This design may be based on the length of the coding region of the modified RNA, the length of a particular feature or region of the modified RNA (such as the mRNA), or based on the length of the ultimate product expressed from the modified RNA.
- the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% greater in length than the modified RNA or feature thereof.
- the poly-A tail may also be designed as a fraction of the modified RNA to which it belongs.
- a recombinant nucleic acid of the present disclosure is one or more recombinant viral genomes and/or one or more non-viral nucleic acids.
- the one or more recombinant viral genomes and/or one or more non-viral nucleic acids comprises a DNA vector or an RNA vector.
- vectors suitable to maintain, propagate, and/or express polynucleotides to produce one or more polypeptides in a subject may be used.
- suitable vectors may include, for example, plasmids, cosmids, episomes, transposons, and viral vectors (e.g., adenoviral vectors, adeno-associated viral vectors, vaccinia viral vectors, pox viral vectors, bacteriophage vector, alpha viral vector, picornaviral vector, iridoviral vector, Newcastle disease viral vector, baculoviral vector, geminiviral vector, caulimoviral vector, Sindbis-viral vectors, measles vectors, herpes viral vectors, lentiviral vectors, retroviral vectors, etc.).
- viral vectors e.g., adenoviral vectors, adeno-associated viral vectors, vaccinia viral vectors, pox viral vectors, bacteriophage vector, alpha viral vector, picornaviral vector, iridoviral vector, Newcastle disease viral vector, baculoviral vector, geminiviral vector, caulimoviral vector, Sindbis-vir
- one or more recombinant viral genomes and/or one or more non-viral nucleic acids is replication competent in a host cell. In some embodiments, the one or more recombinant viral genomes and/or one or more non-viral nucleic acids is replication incompetent in a host cell. In some embodiments, the one or more recombinant viral genomes and/or one or more non-viral nucleic acids can integrate into a host DNA. In some embodiments, the one or more recombinant viral genomes and/or one or more non-viral nucleic acids cannot integrate into a host DNA (e.g., is episomal).
- one or more of the recombinant nucleic acids described herein is a recombinant adenovirus genome.
- the recombinant adenovirus genome may be a recombinant genome from any member of the Adenoviridae family of viruses known in the art, including, for example, a recombinant Atadenovirus genome, a recombinant Aviadenovirus genome, a recombinant Ichtadenovirus genome, a recombinant Mastadenovirus genome, a recombinant Siadenovirus genome, a recombinant Testadenovirus genome, and any combinations or derivatives thereof.
- the recombinant adenovirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- an "inactivating mutation” may refer to any mutation that results in a gene or regulon product (RNA or protein) having reduced, undetectable, or eliminated quantity and/or function (e.g., as compared to a corresponding sequence lacking the inactivating mutation).
- inactivating mutations may include, but are not limited to, deletions, insertions, point mutations, and rearrangements in transcriptional control sequences (promoters, enhancers, insulators, etc.) and/or coding sequences of a given gene or regulon. Any suitable method of measuring the quantity of a gene or regulon product known in the art may be used, including, for example, qPCR, Northern blots, RNAseq, western blots, ELISAs, etc.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) adenovirus genes.
- the recombinant Atadenovirus genome may be a recombinant Bovine atadenovirus D genome, a recombinant Bovine atadenovirus E genome, a recombinant Deer atadenovirus A genome, a recombinant Duck atadenovirus A genome, a recombinant Lizard atadenovirus A genome, a recombinant Lizard atadenovirus B genome, a recombinant Ovine atadenovirus D genome, a recombinant Possum atadenovirus A genome, a recombinant Psittacine atadenovirus A genome, a recombinant Snake atadenovirus A genome, and any combinations or derivatives thereof.
- the recombinant Aviadenovirus genome may be a recombinant Duck aviadenovirus B genome, a recombinant Falcon aviadenovirus A genome, a recombinant Fowl aviadenovirus A genome, a recombinant Fowl aviadenovirus B genome, a recombinant Fowl aviadenovirus C genome, a recombinant Fowl aviadenovirus D genome, a recombinant Fowl aviadenovirus E genome, a recombinant Goose aviadenovirus A genome, a recombinant Pigeon aviadenovirus A genome, a recombinant Pigeon aviadenovirus B genome, a recombinant Psittacine aviadenovirus B genome, a recombinant Psittacine aviadenovirus C genome, a recombinant
- the recombinant Ichtadenovirus genome may be a recombinant Sturgeon ichtadenovirus A.
- the recombinant Mastadenovirus genome may be a recombinant Bat mastadenovirus A genome, a recombinant Bat mastadenovirus B genome, a recombinant Bat mastadenovirus C genome, a recombinant Bat mastadenovirus D genome, a recombinant Bat mastadenovirus E genome, a recombinant Bat mastadenovirus F genome, a recombinant Bat mastadenovirus G genome, a recombinant Bat mastadenovirus H genome, a recombinant Bat mastadenovirus I genome, a recombinant Bat mastadenovirus J genome, a recombinant Bovine mastadenovirus A genome, a recombinant Bovine mastadenovirus B genome, a recombinant Bovine mastadenovirus C genome, a recombinant Canine mastadenovirus A genome, a recombinant Bat mastadeno
- the recombinant Mastadenovirus genome may be the recombinant Human mastadenovirus A genome, the recombinant Human mastadenovirus B genome, the recombinant Human mastadenovirus C genome, the recombinant Human mastadenovirus D genome, the recombinant Human mastadenovirus E genome, the recombinant Human mastadenovirus F genome, or the recombinant Human mastadenovirus G genome, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus A genome may be a recombinant Human mastadenovirus A serotype 12, 18, 31, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus B genome may be a recombinant Human mastadenovirus B serotype 3, 7, 11, 14, 16, 21, 34, 35, 50, 55, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus C genome may be a recombinant Human mastadenovirus C serotype 1, 2, 5, 6, 57, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus D genome may be a recombinant Human mastadenovirus D serotype 8, 9, 10, 13, 15, 17, 19, 20, 22, 23, 24, 25, 26, 1 , 28, 29, 30, 32, 33, 36, 37, 38, 39, 42, 43, 44, 45, 46, 47, 48, 49, 51, 53, 54, 56, 58, 59, 60, 62, 63, 64, 65, 67, 69, 70, 71, 72, 73, 74, 75, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus E genome may be a recombinant Human mastadenovirus E serotype 4.
- the recombinant Human mastadenovirus F genome may be a recombinant Human mastadenovirus F serotype 40, 41, and any combinations or derivatives thereof.
- the recombinant Human mastadenovirus G genome may be a recombinant Human mastadenovirus G serotype 52.
- the recombinant Siadenovirus genome may be a recombinant Frog siadenovirus A genome, a recombinant Great tit siadenovirus A genome, a recombinant Rail siadenovirus A genome, a recombinant Psittacine siadenovirus D genome, a recombinant Psittacine siadenovirus E genome, a recombinant Raptor siadenovirus A genome, a recombinant Skua siadenovirus A genome, a recombinant Turkey siadenovirus A genome, and any combinations or derivatives thereof.
- the recombinant adenovirus genome may be the recombinant Testadenovirus genome.
- one or more of the recombinant nucleic acids described herein are a recombinant retrovirus genome.
- the recombinant retrovirus genome may be a recombinant genome from any member of the Retroviridae family of viruses known in the art, including, for example, a recombinant Alpharetrovirus genome, a recombinant Betaretrovirus genome, a recombinant Gammaretrovirus genome, a recombinant Deltaretrovirus genome, a recombinant Epsilonretrovirus genome, a recombinant Lentivirus genome, a recombinant Bovispumavirus genome, a recombinant Equispumavirus genome, a recombinant Felispumavirus genome, a recombinant Prosimiispumavirus genome, a recombinant Simiispumavirus genome, and any combinations or derivatives thereof.
- the recombinant retrovirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) retrovirus genes.
- the recombinant Alpharetrovirus genome may be a recombinant Avian carcinoma Mill Hill virus 2 genome, a recombinant Avian leukosis virus genome, a recombinant Avian myeloblastosis virus genome, a recombinant Avian myelocytomatosis virus 29 genome, a recombinant Avian sarcoma virus CT10 genome, a recombinant Fujinami sarcoma virus genome, a recombinant Rous sarcoma virus genome, a recombinant UR2 sarcoma virus genome, a recombinant Y73 sarcoma virus genome, and any combinations or derivatives thereof.
- the recombinant Betaretrovirus genome may be a recombinant Jaagsiekte sheep retrovirus genome, a recombinant Langur virus genome, a recombinant Mason- Pfizer monkey virus genome, a recombinant Mouse mammary tumor virus genome, a recombinant Squirrel monkey retrovirus genome, and any combinations or derivatives thereof.
- the recombinant Gammaretrovirus genome may be a recombinant Chick syncytial virus genome, a recombinant Feline leukemia virus genome, a recombinant Finkel- Biskis-Jinkins murine sarcoma virus genome, a recombinant Gardner-Arnstein feline sarcoma virus genome, a recombinant Gibbon ape leukemia virus genome, a recombinant Guinea pig type-C oncovirus genome, a recombinant Hardy-Zuckerman feline sarcoma virus genome, a recombinant Harvey murine sarcoma virus genome, a recombinant Kirsten murine sarcoma virus genome, a recombinant Koala retrovirus genome, a recombinant Moloney murine sarcoma virus genome, a recombinant Murine leukemia virus genome, a recombinant recombinant Mur
- the recombinant Deltaretrovirus genome may be a recombinant Bovine leukemia virus genome, a recombinant Primate T-lymphotropic virus 1 genome, a recombinant Primate T-lymphotropic virus 2 genome, a recombinant Primate T-lymphotropic virus 3 genome, and any combinations or derivatives thereof.
- the recombinant Epsilonretrovirus genome may be a recombinant Walleye dermal sarcoma virus genome, a recombinant Walleye epidermal hyperplasia virus 1 genome, a recombinant Walleye epidermal hyperplasia virus 2 genome, and any combinations or derivatives thereof.
- the recombinant Lentivirus genome may be a recombinant Bovine immunodeficiency virus genome, a recombinant Caprine arthritis encephalitis virus genome, a recombinant Equine infectious anemia virus genome, a recombinant Feline immunodeficiency virus genome, a recombinant Human immunodeficiency virus 1 genome, a recombinant Human immunodeficiency virus 2 genome, a recombinant Jembrana disease virus genome, a recombinant Puma lentivirus genome, a recombinant Simian immunodeficiency virus genome, a recombinant Visna-maedi virus genome, and any combinations or derivatives thereof.
- the recombinant retrovirus genome may be the recombinant
- the recombinant retrovirus genome may be the recombinant
- the recombinant retrovirus genome may be the recombinant
- the recombinant retrovirus genome may be the recombinant
- the recombinant Simiispumavirus genome may be a recombinant Simian foamy virus genome, a recombinant Human foamy virus genome, and any combinations or derivatives thereof.
- one or more recombinant nucleic acids described herein are a recombinant adeno-associated virus genome.
- the recombinant adeno-associated virus genome may be a recombinant genome from any member of the Dependoparvovirus family of viruses known in the art, including, for example, a recombinant Adeno-associated dependoparvovirus A genome, a recombinant Adeno-associated dependoparvovirus B genome, and any combinations or derivatives thereof.
- the recombinant adeno-associated virus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) adeno-associated virus genes.
- the recombinant adeno-associated virus (AAV) genome may be a recombinant AAV serotype 1 genome, a recombinant AAV serotype 2 genome, a recombinant AAV serotype 3 genome, a recombinant AAV serotype 4 genome, a recombinant AAV serotype 5 genome, a recombinant AAV serotype 6 genome, a recombinant AAV serotype 7 genome, a recombinant AAV serotype 8 genome, a recombinant AAV serotype 9 genome, a recombinant AAV serotype 10 genome, a recombinant AAV serotype 11 genome, and any combinations or derivatives thereof.
- AAV serotype 1 genome a recombinant AAV serotype 2 genome, a recombinant AAV serotype 3 genome, a recombinant AAV serotype 4 genome, a recombinant
- one or more of the recombinant nucleic acids described herein are a recombinant herpes simplex virus (HSV) amplicon.
- HSV herpes simplex virus
- Herpes virus amplicons including the structural features and methods of making the same, are generally known to one of ordinary skill in the art (see e.g., de Silva S. and Bowers W. "Herpes Virus Amplicon Vectors". Viruses 2009, 1, 594-629).
- the herpes simplex virus amplicon is an HSV-1 amplicon.
- the herpes simplex virus amplicon is an HSV-1 hybrid amplicon.
- HSV-1 hybrid amplicons may include, but are not limited to, HSV/AAV hybrid amplicons, HSV/EBV hybrid amplicons, HSV/EBV/RV hybrid amplicons, and/or HSV/Sleeping Beauty hybrid amplicons.
- the amplicon is an HSV/AAV hybrid amplicon.
- the amplicon is an S ⁇ //Sleeping Beauty hybrid amplicon.
- one or more of the recombinant nucleic acids described herein are a recombinant herpes virus genome.
- the recombinant herpes virus genome may be a recombinant genome from any member of the Herpesviridae family of DNA viruses known in the art, including, for example, a recombinant herpes simplex virus genome, a recombinant varicella zoster virus genome, a recombinant human cytomegalovirus genome, a recombinant herpesvirus 6A genome, a recombinant herpesvirus 6B genome, a recombinant herpesvirus 7 genome, a recombinant Epstein- Barr virus genome, a recombinant Kaposi's sarcoma-associated herpesvirus genome, and any combinations or derivatives thereof.
- the recombinant herpes virus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) herpes virus genes.
- the recombinant nucleic acid is a recombinant herpes simplex virus (HSV) genome.
- the recombinant herpes virus genome is a recombinant herpes simplex virus type 1 (HSV-1) genome, a recombinant herpes simplex virus type 2 (HSV-2) genome, or any derivatives thereof.
- the recombinant herpes simplex virus genome is a recombinant HSV-1 genome.
- the recombinant HSV-1 genome may be from any HSV-1 strain known in the art, including, for example, strains 17, Ty25, R62, S25, Ku86, S23, Rll, Tyl48, Ku47, H166 sy n, 1319-2005, F-13, M-12, 90237, F-17, KOS, 3083-2008, F12g, L2, CD38, H193, M-15, India 2011, 0116209, F-111, 66-207, 2762, 369-2007, 3355, MacIntyre, McKrae, 7862, 7- hse, HF10, 1394,2005, 270-2007, OD4, SC16, M-19, 4J1037, 5J1060, J1060, KOS79, 132-1988, 160- 1982, H166, 2158-2007, RE, 78326, F18g, Fll, 172-2010, H129, F, E4, CJ994, F14g, E03, E
- the recombinant HSV-1 genome is from the KOS strain. In some embodiments, the recombinant HSV-1 genome is not from the McKrae strain.
- one or more of the recombinant nucleic acids described herein are a recombinant poxvirus genome.
- the recombinant poxvirus genome may be a recombinant genome from any member of the Poxviridae family of viruses known in the art, including, for example, a recombinant smallpox virus genome, a recombinant vaccinia virus genome, a recombinant cowpox virus genome, a recombinant monkeypox virus genome, a recombinant orf virus genome, a recombinant pseudocowpox virus genome, a recombinant bovine papular stomatitis virus genome, a recombinant tanapox virus genome, a recombinant yaba monkey tumor virus genome, a recombinant molluscum contagiosum virus genome, and any combinations or derivatives thereof.
- the recombinant poxvirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) poxvirus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant bacteriophage genome.
- the recombinant bacteriophage genome may be, for example, a recombinant 186 phage genome, a recombinant Escherichia virus Lambda genome, a recombinant Pseudomonas virus phi6 genome, a recombinant Bacillus virus (029 genome, a recombinant Escherichia virus (0X174 genome, a recombinant Bacteriophage cfCbS genome, a recombinant Escherichia virus G4 genome, a recombinant Escherichia virus M13 genome, a recombinant Emesvirus zinderi genome, a recombinant N4 phage genome, a recombinant Escherichia virus Pl genome, a recombinant Escherichi
- the recombinant bacteriophage genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) bacteriophage genes.
- one or more of the recombinant nucleic acids described herein are a recombinant alphavirus genome.
- the recombinant alphavirus genome may be a recombinant genome from any member of the Alphavirus family of viruses known in the art, including, for example, a recombinant Aura virus genome, a recombinant Barmah Forest virus genome, a recombinant Bebaru virus genome, a recombinant Caaingua virus genome, a recombinant Cabassou virus genome, a recombinant Chikungunya virus genome, a recombinant Eastern equine encephalitis virus genome, a recombinant Eilat virus genome, a recombinant Everglades virus genome, a recombinant Fort Morgan virus genome, a recombinant Getah virus genome, a recombinant Highlands J virus genome, a recombinant Mad
- the recombinant alphavirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) alphavirus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant picornavirus genome.
- the recombinant piconavirus genome may be a recombinant genome from any member of the Picornaviridae family of viruses known in the art, including, for example, a recombinant Aalivirus genome, a recombinant Ailurivirus genome, a recombinant Ampivirus genome, a recombinant Anativirus genome, a recombinant Aphthovirus genome, a recombinant Aquamavirus genome, a recombinant Avihepatovirus genome, a recombinant Avisivirus genome, a recombinant Boosepivirus genome, a recombinant Bopivirus genome, a recombinant Caecilivirus genome, a recombinant Cardiovirus genome, a recombinant Cosavirus genome,
- the recombinant picornavirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) picornavirus genes.
- the recombinant Enterovirus genome may be a recombinant Coxsackievirus genome, a recombinant Poliovirus genome, a recombinant Echovirus genome, a recombinant Bovine enterovirus genome, and any combinations or derivatives thereof.
- the recombinant Senecavirus genome may be a recombinant Seneca Valley virus genome.
- the recombinant Cardiovirus genome may be a recombinant Theiler's Murine Encephalomyelitis genome, a recombinant Encephalomyocarditis virus genome, a recombinant Mengovirus genome, and any combinations or derivatives thereof.
- one or more of the recombinant nucleic acids described herein are a recombinant iridovirus genome.
- the recombinant iridovirus genome may be a recombinant genome from any member of the Iridovirus family of viruses known in the art, including, for example, a recombinant Invertebrate iridescent virus 6 (IIV-6) genome, a recombinant Invertebrate iridescent virus 31 (IIV-31) genome, and any combinations or derivatives thereof.
- the recombinant iridovirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) iridovirus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant Newcastle disease virus genome.
- the recombinant Newcastle disease virus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) Newcastle virus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant baculovirus genome.
- the recombinant baculovirus genome may be a recombinant genome from any member of the Baculoviridae family of viruses known in the art, including, for example, a recombinant alphabaculovirus genome, a recombinant betabaculovirus genome, a recombinant deltabaculovirus genome, a recombinant gammabaculovirus genome, and any combinations or derivatives thereof.
- the recombinant baculoviridae genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) baculovirus genes.
- the recombinant alphabaculovirus genome may be a recombinant Adoxophyes honmai nucleopolyhedrovirus genome, a recombinant Agrotis ipsilon multiple nucleopolyhedrovirus genome, a recombinant Agrotis segetum nucleopolyhedrovirus A genome, a recombinant Agrotis segetum nucleopolyhedrovirus B genome, a recombinant Antheraea pernyi nucleopolyhedrovirus genome, a recombinant Anticarsia gemmatalis multiple nucleopolyhedrovirus genome, a recombinant Autographa californica multiple nucleopolyhedrovirus genome, a recombinant Bombyx mori nucleopolyhedrovirus genome, a recombinant Buzura suppressaria nucleopolyhe
- the recombinant betabaculovirus genome may be a recombinant Adoxophyes orana granulovirus genome, a recombinant Agrotis segetum granulovirus genome, a recombinant Artogeia rapae granulovirus genome, a recombinant Choristoneura fumiferana granulovirus genome, a recombinant Clostera anachoreta granulovirus genome, a recombinant Clostera anastomosis granulovirus A genome, a recombinant Clostera anastomosis granulovirus B genome, a recombinant Cnaphalocrocis medinalis granulovirus genome, a recombinant Cryptophlebia leucotreta granulovirus genome, a recombinant Cydia pomonella granulovirus genome, a recombinant Cryptophle
- the recombinant deltabaculovirus genome may be a recombinant Culex nigripalpus nucleopolyhedrovirus genome.
- the recombinant gammabaculovirus genome may be a recombinant Neodiprion lecontei nucleopolyhedrovirus genome, a recombinant Neodiprion sertifer nucleopolyhedrovirus genome, and any combinations or derivatives thereof.
- one or more of the recombinant nucleic acids described herein are a recombinant geminivirus genome.
- the recombinant geminivirus genome may be a recombinant genome from any member of the Geminiviridae family of viruses known in the art, including, for example, a recombinant Becurtovirus genome, a recombinant Begomovirus genome, a recombinant Capulavirus genome, a recombinant Citlodavirus genome, a recombinant Curtovirus genome, a recombinant Eragrovirus genome, a recombinant Grablovirus genome, a recombinant Maldovirus genome, a recombinant Mastrevirus genome, a recombinant Mulcrilevirus genome, a recombinant Opunvirus genome, a recombinant Topilevirus genome, a recombinant Topocuvirus genome, a recombinant Becurto
- the recombinant geminivirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) geminivirus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant caulimovirus genome.
- the recombinant caulimovirus genome may be a recombinant genome from any member of the Caulimovirus family of viruses known in the art, including, for example, a recombinant Angelica bushy stunt virus genome, a recombinant Atractylodes mild mottle virus genome, a recombinant Carnation etched ring virus genome, a recombinant Cauliflower mosaic virus genome, a recombinant Dahlia mosaic virus genome, a recombinant Figwort mosaic virus genome, a recombinant Horseradish latent virus genome, a recombinant Lamium leaf distortion virus genome, a recombinant Mirabilis mosaic virus genome, a recombinant Soybean Putnam virus genome, a recombinant Strawberry vein banding virus genome, a recombinant Thistle mottle virus genome, and any combinations or derivatives thereof.
- a recombinant Angelica bushy stunt virus genome a
- the recombinant caulimovirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) caulimovirus genes.
- one or more of the recombinant nucleic acids described herein are a recombinant anellovirus genome.
- the recombinant anellovirus genome may be a recombinant genome from any member of the anellovirus family of viruses known in the art, including, for example, a recombinant Aleptorquevirus genome, a recombinant Alphatorquevirus genome, a recombinant Betatorquevirus genome, a recombinant Chitorquevirus genome, a recombinant Dalettorquevirus genome, a recombinant Deltatorquevirus genome, a recombinant Epsilontorquevirus genome, a recombinant Etatorquevirus genome, a recombinant Gammatorquevirus genome, a recombinant Gimeltorquevirus genome, a recombinant Gyrovirus genome, a recombinant Hetorquevirus genome, a recombinant recombin
- the recombinant anellovirus genome comprises one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) inactivating mutations.
- the one or more inactivating mutations are in one or more (e.g., one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, etc.) anellovirus genes.
- viral vectors or viral gene delivery vehicles comprising any of the polynucleotides and/or recombinant nucleic acids described herein.
- the virus is capable of infecting one or more target cells of a subject (e.g., a human).
- the virus is suitable for delivering the polynucleotides and/or recombinant nucleic acids into one or more target cells of a subject (e.g., a human).
- the one or more target cells are human cells.
- the one or more target cells are one or more cancerous cells.
- the one or more target cells are one or more non-cancerous (e.g., healthy) cells.
- the one or more target cells are one or more airway epithelial cells.
- the one or more target cells are one or more cells of the respiratory tract (e.g., airway epithelial cells (such as goblet cells, ciliated cells, Clara cells, neuroendocrine cells, basal cells, intermediate or parabasal cells, Serous cells, brush cells, oncocytes, non-ciliated columnar cells, and/or metaplastic cells); alveolar cells (such as type 1 pneumocytes, type 2 pneumocytes, and/or cuboidal non-ciliated cells); salivary gland cells in bronchi (such as Serous cells, mucous cells, and/or ductal cells); etc.).
- the one or more target cells are one or more cells of the lung.
- any suitable virus known in the art may be used, including, for example, adenovirus, adeno-associated virus, retrovirus, lentivirus, sendai virus, papillomavirus, herpes virus (e.g., a herpes simplex virus), vaccinia virus (e.g., a poxvirus), a bacteriophage, an alphavirus, a picornavirus, an iridovirus, a Newcastle virus, a baculovirus, a geminivirus, a caulimovirus, anellovirus, and/or any hybrid or derivative viruses thereof.
- the virus is attenuated.
- the virus is replication competent.
- the virus is replication defective. In some embodiments, the virus is not oncolytic. In some embodiments, the virus has been modified to alter its tissue tropism relative to the tissue tropism of a corresponding unmodified, wildtype virus. In some embodiments, the virus has reduced cytotoxicity (e.g., in a target cell) as compared to a corresponding wild-type virus.
- the viral gene delivery vehicle is an adenovirus.
- the adenovirus may be from any member of the Adenoviridae family of viruses known in the art, including, for example, a recombinant Atadenovirus, a recombinant Aviadenovirus, a recombinant Ichtadenovirus, a recombinant Mastadenovirus, a recombinant Siadenovirus, a recombinant Testadenovirus, and any combinations or derivatives thereof.
- the adenovirus is replication competent.
- the adenovirus is replication defective or replication incompetent.
- the adenovirus is not oncolytic.
- the viral gene delivery vehicle is a retrovirus.
- the retrovirus may be from any member of the Retroviridae family of viruses known in the art, including, for example, a recombinant Alpharetrovirus, a recombinant Betaretrovirus, a recombinant Gammaretrovirus, a recombinant Deltaretrovirus, a recombinant Epsilonretrovirus, a recombinant Lentivirus, a recombinant Bovispumavirus, a recombinant Equispumavirus, a recombinant Felispumavirus, a recombinant Prosimiispumavirus, a recombinant Simiispumavirus, and any combinations or derivatives thereof.
- the retrovirus is replication competent.
- the retrovirus is replication defective or replication incompetent.
- the retrovirus is not oncolytic.
- the viral gene delivery vehicle is an adeno-associated virus (AAV).
- AAV may be from any member of the Dependoparvovirus family of viruses known in the art, including, for example, a recombinant Adeno-associated dependoparvovirus A, a recombinant Adeno- associated dependoparvovirus B, and any combinations or derivatives thereof.
- the AAV is replication competent.
- the AAV is replication defective or replication incompetent.
- the AAV is not oncolytic.
- the AAV may be from any serotype known in the art, including, for example, serotype 1, serotype 2, serotype 3, serotype 4, serotype 5, serotype 6, serotype 7, serotype 8, AAV serotype 9, serotype 10, serotype 11, and any combinations or derivatives thereof.
- the viral gene delivery vehicle is a member of the Herpesviridae family of DNA viruses, including, for example, a herpes simplex virus, a varicella zoster virus, a human cytomegalovirus, a herpesvirus 6A, a herpesvirus 6B, a herpesvirus 7, an Epstein-Barr virus, and a Kaposi's sarcoma-associated herpesvirus, and any combinations or derivatives thereof.
- the Herpesviridae virus is replication competent.
- the Herpesviridae virus is replication defective or replication incompetent.
- the Herpesviridae virus is not oncolytic.
- the herpesvirus is a herpes simplex virus.
- the herpes simplex virus is an HSV-1, an HSV-2, or any combinations or derivatives thereof.
- the herpes simplex virus is an HSV-1 virus.
- the viral gene delivery vehicle is a poxvirus.
- the poxvirus may be from any member of the Poxviridae family of viruses known in the art, including, for example, a recombinant smallpox virus, a recombinant vaccinia virus, a recombinant cowpox virus, a recombinant monkeypox virus, a recombinant orf virus, a recombinant pseudocowpox virus, a recombinant bovine papular stomatitis virus, a recombinant tanapox virus, a recombinant yaba monkey tumor virus, a recombinant molluscum contagiosum virus, and any combinations or derivatives thereof.
- the poxvirus is replication competent. In some embodiments, the poxvirus is replication defective or replication incompetent. In some embodiments, the poxvirus is not oncolytic. [0146] In some embodiments, the viral gene delivery vehicle is a bacteriophage.
- the bacteriophage may be any bacteriophage known in the art, including, for example, a recombinant 186 phage, a recombinant Escherichia virus Lambda, a recombinant Pseudomonas virus phi6, a recombinant Bacillus virus (029, a recombinant Escherichia virus (0X174, a recombinant Bacteriophage tfiCbS, a recombinant Escherichia virus G4, a recombinant Escherichia virus M13, a recombinant Emesvirus zinderi, a recombinant N4 phage, a recombinant Escherichia virus Pl, a recombinant Escherichia virus P2, a recombinant Enterobacteria phage P4, a recombinant R17 phage, a recombinant Enterobacteria
- the viral gene delivery vehicle is an alphavirus.
- the alphavirus may be from any member of the Alphavirus family of viruses known in the art, including, for example, a recombinant Aura virus, a recombinant Barmah Forest virus, a recombinant Bebaru virus, a recombinant Caaingua virus, a recombinant Cabassou virus, a recombinant Chikungunya virus, a recombinant Eastern equine encephalitis virus, a recombinant Eilat virus, a recombinant Everglades virus, a recombinant Fort Morgan virus, a recombinant Getah virus, a recombinant Highlands J virus, a recombinant Madariaga virus, a recombinant Mayaro virus, a recombinant Middelburg virus, a recombinant Mosso das Pedras virus,
- the viral gene delivery vehicle is a picornavirus.
- the picornavirus may be from any member of the Picornaviridae family of viruses known in the art, including, for example, a recombinant Aalivirus, a recombinant Ailurivirus, a recombinant Ampivirus, a recombinant Anativirus, a recombinant Aphthovirus, a recombinant Aquamavirus, a recombinant Avihepatovirus, a recombinant Avisivirus, a recombinant Boosepivirus, a recombinant Bopivirus, a recombinant Caecilivirus, a recombinant Cardiovirus, a recombinant Cosavirus, a recombinant Crahelivirus, a recombinant Crohivirus, a recombinant Danipivirus, a recombinant Dicip
- the viral gene delivery vehicle is an iridovirus.
- the iridovirus may be from any member of the Iridovirus family of viruses known in the art, including, for example, a recombinant Invertebrate iridescent virus 6 (IIV-6), a recombinant Invertebrate iridescent virus 31 (II V- 31), and any combinations or derivatives thereof.
- the iridovirus is replication competent.
- the iridovirus is replication defective or replication incompetent.
- the iridovirus is not oncolytic.
- the viral gene delivery vehicle is a Newcastle disease virus.
- the Newcastle disease virus is replication competent.
- the Newcastle disease virus is replication defective or replication incompetent.
- the Newcastle disease virus is not oncolytic.
- the viral gene delivery vehicle is a baculovirus.
- the baculovirus may be from any member of the Baculoviridae family of viruses known in the art, including, for example, a recombinant alphabaculovirus, a recombinant betabaculovirus, a recombinant deltabaculovirus, a recombinant gammabaculovirus, and any combinations or derivatives thereof.
- the baculovirus is replication competent.
- the baculovirus is replication defective or replication incompetent.
- the baculovirus is not oncolytic.
- the viral gene delivery vehicle is a gemininvirus.
- the geminivirus may be from any member of the Geminiviridae family of viruses known in the art, including, for example, a recombinant Becurtovirus, a recombinant Begomovirus, a recombinant Capulavirus, a recombinant Citlodavirus, a recombinant Curtovirus, a recombinant Eragrovirus, a recombinant Grablovirus, a recombinant Maldovirus, a recombinant Mastrevirus, a recombinant Mulcrilevirus, a recombinant Opunvirus, a recombinant Topilevirus, a recombinant Topocuvirus, a recombinant Turncurtovirus, and any combinations or derivatives thereof.
- the gemininvirus is replication competent.
- the gemininvirus is replication defective or
- the viral gene delivery vehicle is a caulimovirus.
- the caulimovirus may be from any member of the Caulimovirus family of viruses known in the art, including, for example, a recombinant Angelica bushy stunt virus, a recombinant Atractylodes mild mottle virus, a recombinant Carnation etched ring virus, a recombinant Cauliflower mosaic virus, a recombinant Dahlia mosaic virus, a recombinant Figwort mosaic virus, a recombinant Horseradish latent virus, a recombinant Lamium leaf distortion virus, a recombinant Mirabilis mosaic virus, a recombinant Soybean Putnam virus, a recombinant Strawberry vein banding virus, a recombinant Thistle mottle virus, and any combinations or derivatives thereof.
- the caulimovirus is replication competent.
- the caulimovirus is replication competent.
- the viral gene delivery vehicle is an anellovirus.
- the anellovirus may be from any member of the Anelloviridae family of viruses known in the art, including, for example, a recombinant Aleptorquevirus , a recombinant Alphatorquevirus, a recombinant Betatorquevirus, a recombinant Chitorquevirus, a recombinant Dalettorquevirus, a recombinant Deltatorquevirus, a recombinant Epsilontorquevirus, a recombinant Etatorquevirus, a recombinant Gammatorquevirus, a recombinant Gimeltorquevirus, a recombinant Gyrovirus, a recombinant Hetorquevirus, a recombinant lotatorquevirus, a recombinant Kappatorquevirus, a recombinant Lambdatorquevirus, a recombinant Mutor
- the anellovirus is replication competent. In some embodiments, the anellovirus is replication defective or replication incompetent. In some embodiments, the anellovirus is not oncolytic.
- recombinant viruses which may or may not be pseudotyped, that produce IL-2 and IL-12 polypeptides for the treatment of cancer including solid tumors (e.g., advanced solid tumors) and hematologic malignancies.
- the therapeutic polypeptides produced by the recombinant viruses described herein mediate or enhance an anti-tumor effect, such as by effector-cell mediated lysis of tumor cells.
- non-viral vectors or non-viral gene delivery vehicles comprising any of the polynucleotides and/or recombinant nucleic acids described herein.
- the non-viral vector or non-viral gene delivery vehicle is capable of delivering the polynucleotides and/or recombinant nucleic acids to one or more target cells of a subject (e.g., a human).
- the non-viral vector or non-viral gene delivery vehicle is suitable for delivering the polynucleotides and/or recombinant nucleic acids into one or more target cells of a subject (e.g., a human).
- the one or more target cells are human cells. In some embodiments, the one or more target cells are one or more cancerous cells. In some embodiments, the one or more target cells are one or more non-cancerous (e.g., healthy) cells. In some embodiments, the one or more target cells are one or more airway epithelial cells.
- the one or more target cells are one or more cells of the respiratory tract (e.g., airway epithelial cells (such as goblet cells, ciliated cells, Clara cells, neuroendocrine cells, basal cells, intermediate or parabasal cells, Serous cells, brush cells, oncocytes, non-ciliated columnar cells, and/or metaplastic cells); alveolar cells (such as type 1 pneumocytes, type 2 pneumocytes, and/or cuboidal non-ciliated cells); salivary gland cells in bronchi (such as Serous cells, mucous cells, and/or ductal cells); etc.).
- the one or more target cells are one or more cells of the lung.
- Any non-viral vector or non-viral gene delivery vehicle known in the art may be used, including, for example, a chemical compound, a bacterium, a mammalian cell, or a physical delivery system and/or any hybrid or derivative thereof.
- Methods of producing a non-viral vector or non-viral gene delivery vehicle comprising polynucleotides and/or recombinant nucleic acids are well known to one of ordinary skill in the art.
- the non-viral vector or non-viral gene delivery vehicle is a chemical compound.
- the chemical compound may be a polymer compound, a lipid compound, an inorganic compound, and any combinations or derivatives thereof.
- the polymer compound may be a natural polymer, a synthetic polymer, a biopolymer, a biodegradable polymer, a cationic polymer, a protein polymer, a polysaccharide polymer, and any combinations or derivatives thereof.
- the polymer compound may be poly-L-lactide (PLA), chitosan, pectin, polyethylenimine (PEI), poly-L-lysine (PLL), poly 2-N-dimethylaminoethyl methacrylate (PDMAEMA), polyaminoamine, polyaminoesters, polyamidoamine, poly-vinylimidazole (PVI), Poly- beta-amino ester (PBAE), gelatin, albumin, chitosan, beta-cyclodextrin, dextran, hyaluronic acid, polyester, alpha-Pyrrolidinohexiophenone (PHP), Poly-2-acrylamidoglycolic acid (PAGA), Poly-delta- valerolactone (PVL), polyphenyl ethers (PPE), Poly-beta-amino-ester (PBAE), poly-amine-co-ester (PACE), poly-lactic-co-
- the lipid compound may be a nanoparticle (e.g., as disclosed in U.S. Patent Application Publication No. 2010/0015232, the contents of which are hereby incorporated by reference in its entirety), a lipid nanoparticle (LNP; e.g., as disclosed in U.S. Patent Nos. 10,898,574, 10,702,600, and 10,933,127, the contents of which are hereby incorporated by reference in its entirety), a cell targeted lipid nanoparticle (ctLNP; e.g., as disclosed in U.S. Patent Application Publication Nos.
- LNP lipid nanoparticle
- ctLNP cell targeted lipid nanoparticle
- a liposome a cationic liposome, a solid lipid nanoparticle, a lipid emulsion, a lipidoid, a cytofectin, a lipid emulsion, a surfactant, a gemini surfactant, and any combinations or derivatives thereof.
- the inorganic compound may be a nanocarrier, a DNA nanclew, a gold nanoparticle, a carbon nanotube, a graphene, a quantum dot, an up-conversion nanoparticle, a silica nanoparticle, an iron oxide, a ferritin, and any combinations or derivatives thereof.
- the non-viral vector or non-viral gene delivery vehicle is a physical delivery system.
- the physical delivery system may be electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, hydroporation, and any combinations or derivatives thereof.
- compositions or formulations comprising any of the polynucleotides, recombinant nucleic acids, and/or gene delivery vehicles comprising the polynucleotide(s) and/or recombinant nucleic acids, and a pharmaceutically acceptable excipient or carrier.
- compositions and formulations can be prepared by mixing the active ingredient(s) (such as a polynucleotide, a recombinant nucleic acid comprising the polynucleotide, a gene delivery vehicle comprising the polynucleotide and/or recombinant nucleic acid) having the desired degree of purity with one or more pharmaceutically acceptable carriers or excipients.
- active ingredient(s) such as a polynucleotide, a recombinant nucleic acid comprising the polynucleotide, a gene delivery vehicle comprising the polynucleotide and/or recombinant nucleic acid
- Pharmaceutically acceptable carriers or excipients are generally nontoxic to recipients at the dosages and concentrations employed, and may include, but are not limited to: buffers (such as phosphate, citrate, acetate, and other organic acids); antioxidants (such as ascorbic acid and methionine); preservatives (such as octadecyldimethylbenzyl ammonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol); amino acids (such as glycine, glutamine, asparagine, histidine, arginine, or lysine); low molecular weight (less than about 10 residues) polypeptides; proteins (such as serum albumin, gelatin, or immunoglobulins); polyols (such as glycerol, e.
- glycerol hydrophilic polymers (such as polyvinylpyrrolidone); monosaccharides, disaccharides, and other carbohydrates (including glucose, mannose, or dextrins); chelating agents (such as EDTA); sugars (such as sucrose, mannitol, trehalose, or sorbitol); salt-forming counter-ions (such as sodium); metal complexes (such as Zn-protein complexes); and/or non-ionic surfactants (such as polyethylene glycol (PEG)).
- hydrophilic polymers such as polyvinylpyrrolidone
- monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins
- chelating agents such as EDTA
- sugars such as sucrose, mannitol, trehalose, or sorbitol
- salt-forming counter-ions such as sodium
- metal complexes such as Zn-protein complexes
- non-ionic surfactants
- the pharmaceutical composition or formulation comprises one or more lipid (e.g., cationic lipid) carriers.
- the pharmaceutical composition or formulation comprises one or more nanoparticle carriers.
- a variety of molecules e.g., proteins, peptides, recombinant nucleic acids, etc.
- a molecule "encapsulated" in a nanoparticle may refer to a molecule that is contained within the nanoparticle or attached to and/or associated with the surface of the nanoparticle, or any combination thereof.
- Nanoparticles for use in the compositions or formulations described herein may be any type of biocompatible nanoparticle known in the art, including, for example, nanoparticles comprising poly(lactic acid), poly(glycolic acid), PLGA, PLA, PGA, and any combinations thereof (see e.g., Vauthier et al. Adv Drug Del Rev. (2003) 55: 519- 48; US 2007/0148074; US 2007/0092575; US 2006/0246139; US 5,753,234; US 7,081,483; and US 2008/0260851, all of which are incorporated by reference herein in their entirety).
- the pharmaceutically acceptable carrier or excipient may be adapted for or suitable for any administration route known in the art, including, for example, intratumoral, intravenous, intramuscular, subcutaneous, cutaneous, oral, intranasal, intratracheal, sublingual, buccal, topical, transdermal, intradermal, intraperitoneal, intraorbital, intravitreal, subretinal, transmucosal, intraarticular, by implantation, by inhalation, intrathecal, intraventricular, intracranial, and/or intranasal administration.
- the pharmaceutically acceptable carrier or excipient is adapted for or suitable for oral, intranasal, intratracheal, and/or inhaled administration.
- the pharmaceutically acceptable carrier or excipient is adapted for or suitable for intranasal and/or inhaled administration. In some embodiments, the pharmaceutically acceptable carrier or excipient is adapted for or suitable for inhaled administration. In some embodiments, the pharmaceutically acceptable carrier or excipient is adapted for or suitable for intratumoral administration.
- the pharmaceutical composition or formulation is adapted for or suitable for any administration route known in the art, including, for example, intratumoral, intravenous, intramuscular, subcutaneous, cutaneous, oral, intranasal, intratracheal, sublingual, buccal, topical, transdermal, intradermal, intraperitoneal, intraorbital, intravitreal, subretinal, transmucosal, intraarticular, by implantation, by inhalation, intrathecal, intraventricular, intracranial, or intranasal administration.
- the pharmaceutical composition or formulation is adapted for or suitable for oral, intranasal, intratracheal, and/or inhaled administration.
- the pharmaceutical composition or formulation is adapted for or suitable for intranasal and/or inhaled administration. In some embodiments, the pharmaceutical composition or formulation is adapted for or suitable for inhaled administration. In some embodiments, the pharmaceutical composition or formulation is adapted for or suitable for intratumoral administration.
- the pharmaceutical composition or formulation further comprises one or more additional components.
- additional components may include, but are not limited to, binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc.); lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.); disintegrants (e.g., starch, sodium starch glycolate, etc.); wetting agents (e.g., sodium lauryl sulphate, etc.); salt solutions; alcohols; polyethylene glycols
- the pharmaceutical composition or formulation comprises a methylcellulose gel (e.g., hydroxypropyl methylcellulose, carboxy methylcellulose, etc.).
- the pharmaceutical composition or formulation comprises a phosphate buffer.
- the pharmaceutical composition or formulation comprises glycerol (e.g., at about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, etc.).
- the pharmaceutical composition or formulation comprises a phosphate buffer and glycerol.
- compositions and formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used to deliver one or more polynucleotides encoding IL-2 and IL-12 polypeptides into one or more cells of a subject (e.g., one or more cells of the respiratory tract of the subject). In some embodiments, the subject suffers from cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of carcinoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of lymphoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of blastoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of sarcoma. In some embodiments, any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of a neuroendocrine tumor.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of mesothelioma. In some embodiments, any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of schwannoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of meningioma. In some embodiments, any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of adenocarcinoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of melanoma. In some embodiments, any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of leukemia.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of lymphoid malignancy.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used to deliver one or more polynucleotides encoding an IL-2 and IL-12 polypeptide into one or more cells of a subject (e.g., one or more cells of the respiratory tract of the subject). In some embodiments, the subject suffers from cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of a solid tumor.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of a hematologic cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of bladder cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of brain cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of breast cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of colon cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of gastric cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of glioma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of head cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of leukemia.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of liver cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of lung cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of lymphoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of myeloma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of neck cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of ovarian cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of melanoma. In some embodiments, any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of pancreatic cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of renal cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of salivary cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of skin cancer (e.g., melanoma, basal cell carcinoma, squamous cell carcinoma, actinic keratosis, an atypical mole, and/or Merkel cell carcinoma).
- skin cancer e.g., melanoma, basal cell carcinoma, squamous cell carcinoma, actinic keratosis, an atypical mole, and/or Merkel cell carcinoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of stomach cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of thymic epithelial cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of thyroid cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of osteosarcoma.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of leptomeningeal cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used to deliver one or more polynucleotides encoding an IL-2 and IL-12 polypeptide into one or more cells of a subject (e.g., one or more cells of the respiratory tract of the subject).
- the subject suffers from lung cancer.
- Lung cancers are often divided into the broad categories of smallcell lung cancer (SCLC), also called oat cell cancer, and non-small-cell lung cancer (NSCLC).
- SCLC smallcell lung cancer
- NSCLC non-small-cell lung cancer
- NSCLC is further divided into three major types, squamous, cell carcinoma (SCC), adenocarcinoma and large cell carcinomas.
- SCC cell carcinoma
- adenocarcinoma large cell carcinomas.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of small-cell lung cancer.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of adenocarcinoma of the lung.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of squamous carcinoma of the lung.
- any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations described herein may be used in the treatment of non-small cell lung cancer.
- Certain aspects of the present disclosure relate to a method of delivering one or more polynucleotides to one or more cells of a subject (e.g., one or more cancerous cells, one or more non- cancerous (e.g., healthy) cells, one or more cells of the respiratory tract, such as airway epithelial cells (goblet cells, ciliated cells, Clara cells, neuroendocrine cells, basal cells, intermediate or parabasal cells, Serous cells, brush cells, oncocytes, non-ciliated columnar cells, and/or metaplastic cells); alveolar cells (type 1 pneumocytes, type 2 pneumocytes, and/or cuboidal non-ciliated cells); salivary gland cells in bronchi (Serous cells, mucous cells, and/or ductal cells); etc.) comprising administering to the subject any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleot
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered topically, transdermally, subcutaneously, epicutaneously, intradermally, orally, sublingually, buccally, rectally, vaginally, intravenously, intraarterially, intramuscularly, intraosseously, intracardially, intraperitoneally, transmucosally, intravitreally, subretinally, suprachoroidally, intracranially, intrathecally, intraventricularly, intraarticularly, peri-articularly, intratumorally, locally, or via inhalation to the subject.
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered orally, intranasally, intratracheally, or via inhalation to the subject.
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered intranasally or via inhalation to the subject.
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered via inhalation to the subject.
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered using a dry powder inhaler, a pressurized metered dose inhaler, a soft mist inhaler, a nebulizer, or an electrohydrodynamic aerosol device.
- the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, and/or pharmaceutical compositions or formulations are administered using a nebulizer.
- the nebulizer is a vibrating mesh nebulizer.
- the subject is a human.
- the subject suffers from a cancer.
- the cancer is selected from acute myeloid leukemia (LAML or AML), acute lymphoblastic leukemia (ALL), adrenocortical carcinoma (ACC), bladder urothelial cancer (BLCA), brain stem glioma, brain lower grade glioma (LGG), brain tumor, breast cancer (BRCA), bronchial tumors, Burkitt lymphoma, cancer of unknown primary site, carcinoid tumor, carcinoma of unknown primary site, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumors, cervical squamous cell carcinoma, endocervical adenocarcinoma (CESC) cancer, childhood cancers, cholangiocarcinoma (CHOL), chordoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative
- LAML or AML acute myeloid leukemia
- the cancer is a virus- associated cancer.
- the cancer is a human papilloma virus (HPV)-associated cancer (e.g., an HPV-associated cancer of the back of the throat, cervix, anus, vulva, penis, and/or vagina).
- HPV human papilloma virus
- the cancer is not skin cancer (e.g., melanoma, basal cell carcinoma, squamous cell carcinoma, actinic keratosis, an atypical mole, and/or Merkel cell carcinoma).
- the cancer is not melanoma.
- the subject suffers from one or more of carcinoma, lymphoma, blastoma, sarcoma, a neuroendocrine tumor, mesothelioma, schwannoma, meningioma, adenocarcinoma, melanoma, leukemia, and lymphoid malignancy.
- the subject suffers from one or more of a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, skin cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- the subject suffers from one or more of small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, or squamous carcinoma of the lung.
- the subject suffers from osteosarcoma.
- aspects of the present disclosure relate to a method of providing prophylactic, palliative, or therapeutic relief to one or more signs or symptoms of cancer in a subject in need thereof comprising administering to the subject an effective amount of any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, medicaments, and/or pharmaceutical compositions or formulations described herein.
- the subject is a human.
- the subject suffers from a cancer.
- the cancer is selected from acute myeloid leukemia (LAML or AML), acute lymphoblastic leukemia (ALL), adrenocortical carcinoma (ACC), bladder urothelial cancer (BLCA), brain stem glioma, brain lower grade glioma (LGG), brain tumor, breast cancer (BRCA), bronchial tumors, Burkitt lymphoma, cancer of unknown primary site, carcinoid tumor, carcinoma of unknown primary site, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumors, cervical squamous cell carcinoma, endocervical adenocarcinoma (CESC) cancer, childhood cancers, cholangiocarcinoma (CHOL), chordoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon (adenocarcinoma) cancer (COAD), colorectal cancer, craniopharyngiom
- the cancer is a virus-associated cancer.
- the cancer is a human papilloma virus (HPV)-associated cancer (e.g., an HPV-associated cancer of the back of the throat, cervix, anus, vulva, penis, and/or vagina).
- HPV human papilloma virus
- the cancer is not skin cancer (e.g., melanoma, basal cell carcinoma, squamous cell carcinoma, actinic keratosis, an atypical mole, and/or Merkel cell carcinoma).
- the cancer is not melanoma.
- the subject suffers from one or more of carcinoma, lymphoma, blastoma, sarcoma, a neuroendocrine tumor, mesothelioma, schwannoma, meningioma, adenocarcinoma, melanoma, leukemia, and lymphoid malignancy.
- the subject suffers from one or more of a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- the subject suffers from one or more of small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, or squamous carcinoma of the lung.
- the subject suffers from osteosarcoma.
- prokaryotic cells comprising any of the polynucleotides and/or recombinant nucleic acids described herein.
- Any suitable host cell known in the art may be used, including, for example: prokaryotic cells including eubacteria, such as Gram-negative or Gram-positive organisms, for example Enterobacteriaceae such as Escherichia (e.g., E. coli), Enterobacter, Erminia, Klebsiella, Proteus, Salmonella (e.g., S. typhimurium), Serratia (e.g., S. marcescans), and Shigella, as well as Bacilli such as B.
- eubacteria such as Gram-negative or Gram-positive organisms
- Enterobacteriaceae such as Escherichia (e.g., E. coli)
- Enterobacter Erminia
- Klebsiella Proteus
- Salmonella e.g., S. typhimurium
- subtilis and B. licheniformis include subtilis and B. licheniformis; fungal cells (e.g., S. cerevisiae); insect cells (e.g., S2 cells, etc.); and mammalian cells, including monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651), human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture), baby hamster kidney cells (BHK, ATCC CCL 10), mouse Sertoli cells (TM4), monkey kidney cells (CV1 ATCC CCL 70), African green monkey kidney cells (VERO-76, ATCC CRL-1587), human cervical carcinoma cells (HELA, ATCC CCL 2), canine kidney cells (MDCK, ATCC CCL 34), buffalo rat liver cells (BRL 3A, ATCC CRL 1442), human lung cells (W138, ATCC CCL 75), human liver cells (Hep G2, HB 8065), mouse mammary tumor (MMT 060562, ATCC CCL51), TRI cells
- the host cell is a human cell (e.g., a T cell, NK cell, etc.) or non-human primate cell.
- the host cells are cells from a cell line. Examples of suitable host cells or cell lines may include, but are not limited to, 293, HeLa, SH-Sy5y, Hep G2, CACO-2, A549, L929, 3T3, K562, CHO-K1, MDCK, HUVEC, Vero, N20, COS-7, PSN1, VcaP, CHO cells, and the like.
- the present disclosure relate to an article of manufacture or a kit comprising any of the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, medicaments, and/or pharmaceutical compositions or formulations described herein.
- the article of manufacture or kit comprises a package insert comprising instructions for administering the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, medicaments, and/or pharmaceutical compositions or formulations.
- Suitable containers for the polynucleotides, recombinant nucleic acids comprising the polynucleotides, gene delivery vehicles comprising the polynucleotides and/or recombinant nucleic acids, medicaments, and/or pharmaceutical compositions or formulations may include, for example, bottles, vials, bags, tubes, and syringes.
- the container may be formed from a variety of materials such as glass, plastic (such as polyvinyl chloride or polyolefin), or metal alloy (such as stainless steel or hastelloy).
- the container comprises a label on, or associated with the container, wherein the label indicates directions for use.
- the article of manufacture or kit may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, a package insert, and the like.
- Embodiment 1 a composition comprising a first recombinant polynucleotide encoding an Interleukin (IL)-2 polypeptide and a second recombinant polynucleotide encoding an IL-12 polypeptide.
- IL Interleukin
- Embodiment 2 a pharmaceutical composition comprising the first recombinant polynucleotide and the second recombinant polynucleotide of embodiment 1.
- Embodiment 3 the pharmaceutical composition of embodiment 2, wherein the pharmaceutical composition comprises a recombinant nucleic acid comprising the first polynucleotide and a recombinant nucleic acid comprising the second polynucleotide.
- Embodiment 4 the pharmaceutical composition of any of embodiments 2 or 3, wherein the recombinant nucleic acid comprising the first polynucleotide and the recombinant nucleic acid comprising the second polynucleotide are the same recombinant nucleic acid.
- Embodiment 5 the pharmaceutical composition of any of embodiments 2-4, wherein the recombinant nucleic acid comprising the first polynucleotide and the recombinant nucleic acid comprising the second polynucleotide are different recombinant nucleic acids.
- Embodiment 6 the pharmaceutical composition of any of embodiments 2-5, wherein the pharmaceutical composition comprises one or more gene delivery vehicles comprising the first polynucleotide and the second polynucleotide.
- Embodiment 7 the pharmaceutical composition of any of embodiments 2-6, wherein the first polynucleotide and the second polynucleotide are contiguous.
- Embodiment 8 the pharmaceutical composition of any of embodiments 2-7, wherein the first polynucleotide and the second polynucleotide are non-contiguous.
- Embodiment 9 the pharmaceutical composition of any of embodiments 2-8, wherein the first polynucleotide and/or the second polynucleotide comprises a deoxyribonucleic acid (DNA).
- DNA deoxyribonucleic acid
- Embodiment 10 the pharmaceutical composition of any of embodiments 2-9, wherein the first polynucleotide or the second polynucleotide comprises a deoxyribonucleic acid (DNA).
- DNA deoxyribonucleic acid
- Embodiment 11 the pharmaceutical composition of any of embodiments 2-10, wherein the first polynucleotide and the second polynucleotide comprises a deoxyribonucleic acid (DNA).
- DNA deoxyribonucleic acid
- Embodiment 12 the pharmaceutical composition of any of embodiments 2-11, wherein the first polynucleotide comprises a deoxyribonucleic acid (DNA).
- DNA deoxyribonucleic acid
- Embodiment 13 the pharmaceutical composition of any of embodiments 2-12, wherein the second polynucleotide comprises a deoxyribonucleic acid (DNA).
- DNA deoxyribonucleic acid
- Embodiment 14 the pharmaceutical composition of any of embodiments 2-13, wherein the DNA is a linear DNA or a circular DNA.
- Embodiment 15 the pharmaceutical composition of any of embodiments 2-14, wherein the DNA is a linear DNA.
- Embodiment 16 the pharmaceutical composition of any of embodiments 2-15, wherein the DNA is a circular DNA.
- Embodiment 17 the pharmaceutical composition of any of embodiments 2-16, wherein the first polynucleotide and/or the second polynucleotide comprises a ribonucleic acid (RNA), a messenger RNA (mRNA), or a modified mRNA (mmRNA).
- RNA ribonucleic acid
- mRNA messenger RNA
- mmRNA modified mRNA
- Embodiment 18 the pharmaceutical composition of any of embodiments 2-17, wherein the first polynucleotide comprises an RNA.
- Embodiment 19 the pharmaceutical composition of any of embodiments 2-18, wherein the first polynucleotide comprises an mRNA.
- Embodiment 20 the pharmaceutical composition of any of embodiments 2-19, wherein the first polynucleotide comprises an mmRNA.
- Embodiment 21 the pharmaceutical composition of any of embodiments 2-20, wherein the second polynucleotide comprises an RNA.
- Embodiment 22 the pharmaceutical composition of any of embodiments 2-21, wherein the second polynucleotide comprises an mRNA.
- Embodiment 23 the pharmaceutical composition of any of embodiments 2-22, wherein the second polynucleotide comprises an mmRNA.
- Embodiment 24 the pharmaceutical composition of any of embodiments 2-23, wherein the first polynucleotide and the second polynucleotide comprise an RNA.
- Embodiment 25 the pharmaceutical composition of any of embodiments 2-24, wherein the first polynucleotide and the second polynucleotide comprise an mRNA.
- Embodiment 26 the pharmaceutical composition of any of embodiments 2-25, wherein the first polynucleotide and the second polynucleotide comprise an mmRNA.
- Embodiment 27 the pharmaceutical composition of any of embodiments 2-26, wherein the mRNA and/or the mmRNA further comprises a 5' untranslated region (UTR), a 3' UTR, a polyadenylation (poly(A)) tail, and/or a 5' cap analog.
- UTR 5' untranslated region
- poly(A) polyadenylation
- Embodiment 28 the pharmaceutical composition of any of embodiments 2-27, wherein the mRNA further comprises a 5' untranslated region (UTR), a 3' UTR, a polyadenylation (poly(A)) tail, and a 5' cap analog.
- UTR 5' untranslated region
- 3' UTR 3' UTR
- poly(A) polyadenylation
- 5' cap analog 5' cap analog
- Embodiment 29 the pharmaceutical composition of any of embodiments 2-28, wherein the mmRNA further comprises a 5' untranslated region (UTR), a 3' UTR, a polyadenylation (poly(A)) tail, and a 5' cap analog.
- UTR 5' untranslated region
- 3' UTR 3' UTR
- poly(A) polyadenylation
- 5' cap analog 5' cap analog
- Embodiment 30 the pharmaceutical composition of any of embodiments 2-29, wherein the 5' cap analog is 7mG(5')ppp(5')NlmpNp.
- Embodiment 31 the pharmaceutical composition of any of embodiments 2-30, wherein the mRNA and/or the mmRNA further comprises a 1-methylpseudouridine modification and/or a 1- ethylpseudouridine modification.
- Embodiment 32 the pharmaceutical composition of any of embodiments 2-31, wherein the mRNA further comprises a 1-methylpseudouridine modification.
- Embodiment 33 the pharmaceutical composition of any of embodiments 2-32, wherein the mRNA further comprises a 1-ethylpseudouridine modification.
- Embodiment 34 the pharmaceutical composition of any of embodiments 1-33, wherein the mmRNA further comprises a 1-methylpseudouridine modification.
- Embodiment 35 the pharmaceutical composition of any of embodiments 2-34, wherein the mmRNA further comprises a 1-ethylpseudouridine modification.
- Embodiment 36 the pharmaceutical composition of any of embodiments 2-35, wherein the mRNA further comprises a 1-methylpseudouridine modification and a 1-ethylpseudouridine modification.
- Embodiment 37 the pharmaceutical composition of any of embodiments 2-36, wherein the mmRNA further comprises a 1-methylpseudouridine modification and a 1-ethylpseudouridine modification.
- Embodiment 38 the pharmaceutical composition of any of embodiments 2-37, wherein the first polynucleotide and/or the second polynucleotide are codon-optimized.
- Embodiment 39 the pharmaceutical composition of any of embodiments 2-38, wherein the first polynucleotide is codon-optimized.
- Embodiment 40 the pharmaceutical composition of any of embodiments 2-29, wherein the second polynucleotide is codon-optimized.
- Embodiment 41 the pharmaceutical composition of any of embodiments 2-40, wherein the first polynucleotide and the second polynucleotide are codon-optimized.
- Embodiment 42 the pharmaceutical composition of any of embodiments 2-41, wherein the first polynucleotide and/or the second polynucleotide are codon-optimized for human codon usage.
- Embodiment 43 the pharmaceutical composition of any of embodiments 2-42, wherein the first polynucleotide is codon-optimized for human codon usage.
- Embodiment 44 the pharmaceutical composition of any of embodiments 2-43, wherein the second polynucleotide is codon-optimized for human codon usage.
- Embodiment 45 the pharmaceutical composition of any of embodiments 2-44, wherein the first polynucleotide and the second polynucleotide are codon-optimized for human codon usage.
- Embodiment 46 the pharmaceutical composition of any of embodiments 2-45, wherein the IL-2 polypeptide is a human IL-2 polypeptide.
- Embodiment 47 the pharmaceutical composition of any of embodiments 2-46, wherein the IL-2 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ. ID NO: 1.
- Embodiment 48 the pharmaceutical composition of any of embodiments 2-47, wherein the IL-12 polypeptide is a human IL-12 polypeptide.
- Embodiment 49 the pharmaceutical composition of any of embodiments 2-48, wherein the IL-12 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID Nos: 2-4.
- Embodiment 50 the pharmaceutical composition of any of embodiments 2-49, wherein the IL-12 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 2.
- Embodiment 51 the pharmaceutical composition of any of embodiments 2-50, wherein the IL-12 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 3.
- Embodiment 52 the pharmaceutical composition of any of embodiments 2-51, wherein the IL-12 polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 4.
- Embodiment 53 the pharmaceutical composition of any of embodiments 2-52, wherein the one or more gene delivery vehicles are one or more of a viral gene delivery vehicle and/or a non- viral gene delivery vehicle.
- Embodiment 54 the pharmaceutical composition of any of embodiments 2-53, wherein the one or more gene delivery vehicles is one or more viral gene delivery vehicle.
- Embodiment 55 the pharmaceutical composition of any of embodiments 2-54, wherein the one or more gene delivery vehicles is one or more of a non-viral gene delivery vehicle.
- Embodiment 56 the pharmaceutical composition of any of embodiments 2-55, wherein the one or more gene delivery vehicles are one or more of a viral gene delivery vehicle and a non-viral gene delivery vehicle.
- Embodiment 57 the pharmaceutical composition of any of embodiments 2-56, wherein the viral gene delivery vehicle comprises a recombinant viral genome.
- Embodiment 58 the pharmaceutical composition of any of embodiments 2-57, wherein the recombinant viral genome comprises the first polynucleotide and/or the second polynucleotide.
- Embodiment 59 the pharmaceutical composition of any of embodiments 2-58, wherein the recombinant viral genome comprises the first polynucleotide.
- Embodiment 60 the pharmaceutical composition of any of embodiments 2-59, wherein the recombinant viral genome comprises the second polynucleotide.
- Embodiment 61 the pharmaceutical composition of any of embodiments 2-60, wherein the recombinant viral genome comprises the first polynucleotide and the second polynucleotide.
- Embodiment 62 the pharmaceutical composition of any of embodiments 2-61, wherein the recombinant viral genome is replication competent or replication defective.
- Embodiment 63 the pharmaceutical composition of any of embodiments 2-62, wherein the recombinant viral genome is replication competent.
- Embodiment 64 the pharmaceutical composition of any of embodiments 2-63, wherein the recombinant viral genome is replication defective.
- Embodiment 65 the pharmaceutical composition of any of embodiments 2-64, wherein the recombinant viral genome is a recombinant oncolytic virus genome, or wherein the recombinant viral genome is not a recombinant oncolytic virus genome.
- Embodiment 66 the pharmaceutical composition of any of embodiments 2-65, wherein the recombinant viral genome is a recombinant oncolytic virus genome.
- Embodiment 67 the pharmaceutical composition of any of embodiments 2-66, wherein the recombinant viral genome is not a recombinant oncolytic virus genome.
- Embodiment 68 the pharmaceutical composition of any of embodiments 2-67, wherein the recombinant viral genome is selected from the group consisting of a recombinant adenovirus genome, a recombinant retrovirus genome, a recombinant adeno-associated virus genome, a recombinant herpes virus genome, a recombinant poxvirus genome, a recombinant bacteriophage genome, a recombinant alphavirus genome, a recombinant picornavirus genome, a recombinant iridovirus genome, a recombinant Newcastle disease virus genome, a recombinant baculovirus genome, a recombinant geminivirus genome, a recombinant caulimovirus genome, and a combination thereof.
- the recombinant viral genome is selected from the group consisting of a recombinant adenovirus genome, a recombinant retrovirus genome
- Embodiment 69 the pharmaceutical composition of any of embodiments 2-68, wherein the recombinant viral genome is a recombinant adenovirus genome.
- Embodiment 70 the pharmaceutical composition of any of embodiments 2-69, wherein the recombinant viral genome is a recombinant retrovirus genome.
- Embodiment 71 the pharmaceutical composition of any of embodiments 2-70, wherein the recombinant viral genome is a recombinant adeno-associated virus genome.
- Embodiment 72 the pharmaceutical composition of any of embodiments 2-71, wherein the recombinant viral genome is a recombinant herpes virus genome.
- Embodiment 73 the pharmaceutical composition of any of embodiments 2-72, wherein the recombinant viral genome is a recombinant poxvirus genome.
- Embodiment 74 the pharmaceutical composition of any of embodiments 2-73, wherein the recombinant viral genome is a recombinant bacteriophage genome.
- Embodiment 75 the pharmaceutical composition of any of embodiments 2-74, wherein the recombinant viral genome is a recombinant alphavirus genome.
- Embodiment 76 the pharmaceutical composition of any of embodiments 2-75, wherein the recombinant viral genome is a recombinant picornavirus genome.
- Embodiment 77 the pharmaceutical composition of any of embodiments 2-76, wherein the recombinant viral genome is a recombinant iridovirus genome.
- Embodiment 78 the pharmaceutical composition of any of embodiments 2-77, wherein the recombinant viral genome is a recombinant Newcastle disease virus genome.
- Embodiment 79 the pharmaceutical composition of any of embodiments 2-78, wherein the recombinant viral genome is a recombinant baculovirus genome.
- Embodiment 80 the pharmaceutical composition of any of embodiments 2-79, wherein the recombinant viral genome is a recombinant geminivirus genome.
- Embodiment 81 the pharmaceutical composition of any of embodiments 2-80, wherein the recombinant viral genome is a recombinant caulimovirus genome.
- Embodiment 82 the pharmaceutical composition of any of embodiments 2-81, wherein the recombinant adenovirus genome is selected from the group consisting of a recombinant Atadenovirus genome, a recombinant Aviadenovirus genome, a recombinant Ichtadenovirus genome, a recombinant Mastadenovirus genome, a recombinant Siadenovirus genome, a recombinant Testadenovirus genome, and any derivatives thereof.
- Embodiment 83 the pharmaceutical composition of any of embodiments 2-82, wherein the recombinant retrovirus genome is selected from the group consisting of a recombinant Alpharetrovirus genome, a recombinant Betaretrovirus genome, a recombinant Gammaretrovirus genome, a recombinant Deltaretrovirus genome, a recombinant Epsilonretrovirus genome, a recombinant Lentivirus genome, a recombinant Bovispumavirus genome, a recombinant Equispumavirus genome, a recombinant Felispumavirus genome, a recombinant Prosimiispumavirus genome, a recombinant Simiispumavirus genome, and any derivatives thereof.
- the recombinant retrovirus genome is selected from the group consisting of a recombinant Alpharetrovirus genome, a recombinant Betaretrovirus genome, a recombinant Gam
- Embodiment 84 the pharmaceutical composition of any of embodiments 2-83, wherein the recombinant adeno-associated virus genome is selected from the group consisting of a recombinant Adeno-associated dependoparvovirus A genome, a recombinant Adeno-associated dependoparvovirus B genome, and any derivates thereof.
- Embodiment 85 the pharmaceutical composition of any of embodiments 2-84, wherein the recombinant adeno-associated virus (AAV) genome is a recombinant AAV serotype 1 genome, a recombinant AAV serotype 2 genome, a recombinant AAV serotype 3 genome, a recombinant AAV serotype 4 genome, a recombinant AAV serotype 5 genome, a recombinant AAV serotype 6 genome, a recombinant AAV serotype 7 genome, a recombinant AAV serotype 8 genome, a recombinant AAV serotype 9 genome, a recombinant AAV serotype 10 genome, a recombinant AAV serotype 11 genome, or a combination thereof.
- AAV adeno-associated virus
- Embodiment 86 the pharmaceutical composition of any of embodiments 2-85, wherein the recombinant herpes virus genome is selected from the group consisting of a recombinant herpes simplex virus genome, a recombinant varicella zoster virus genome, a recombinant human cytomegalovirus genome, a recombinant herpesvirus 6A genome, a recombinant herpesvirus 6B genome, a recombinant herpesvirus 7 genome, a recombinant Epstein-Barr virus genome, a recombinant Kaposi's sarcoma-associated herpesvirus genome, and any derivatives thereof.
- the recombinant herpes virus genome is selected from the group consisting of a recombinant herpes simplex virus genome, a recombinant varicella zoster virus genome, a recombinant human cytomegalovirus genome, a recombinant herpes
- Embodiment 87 the pharmaceutical composition of any of embodiments 2-86, wherein the recombinant herpes simplex virus genome is a recombinant herpes simplex virus type 1 (HSV-1) genome, a recombinant herpes simplex virus type 2 (HSV-2) genome, or any derivatives thereof.
- HSV-1 herpes simplex virus type 1
- HSV-2 herpes simplex virus type 2
- Embodiment 88 the pharmaceutical composition of any of embodiments 2-87, wherein the recombinant poxvirus genome is selected from the group consisting of a recombinant smallpox virus genome, a recombinant vaccinia virus genome, a recombinant cowpox virus genome, a recombinant monkeypox virus genome, a recombinant orf virus genome, a recombinant pseudocowpox virus genome, a recombinant bovine papular stomatitis virus genome, a recombinant tanapox virus genome, a recombinant yaba monkey tumor virus genome, a recombinant molluscum contagiosum virus genome, and any derivatives thereof.
- the recombinant poxvirus genome is selected from the group consisting of a recombinant smallpox virus genome, a recombinant vaccinia virus genome, a recombinant cowpo
- Embodiment 89 the pharmaceutical composition of any of embodiments 2-88, wherein the recombinant bacteriophage genome is selected from the group consisting of a recombinant 186 phage genome, a recombinant Escherichia virus Lambda genome, a recombinant Pseudomonas virus phi6 genome, a recombinant Bacillus virus (029 genome, a recombinant Escherichia virus (0X174 genome, a recombinant Bacteriophage c[>Cb5 genome, a recombinant Escherichia virus G4 genome, a recombinant Escherichia virus M13 genome, a recombinant Emesvirus zinderi genome, a recombinant N4 phage genome, a recombinant Escherichia virus Pl genome, a recombinant Escherichia virus P2 genome, a recombinant Enter
- Embodiment 90 the pharmaceutical composition of any of embodiments 2-89, wherein the recombinant alphavirus genome is selected from the group consisting of a recombinant Aura virus genome, a recombinant Barmah Forest virus genome, a recombinant Bebaru virus genome, a recombinant Caaingua virus genome, a recombinant Cabassou virus genome, a recombinant Chikungunya virus genome, a recombinant Eastern equine encephalitis virus genome, a recombinant Eilat virus genome, a recombinant Everglades virus genome, a recombinant Fort Morgan virus genome, a recombinant Getah virus genome, a recombinant Highlands J virus genome, a recombinant Madariaga virus genome, a recombinant Mayaro virus genome, a recombinant Middelburg virus genome, a recombin
- Embodiment 91 the pharmaceutical composition of any of embodiments 2-90, wherein the recombinant picornavirus genome is selected from the group consisting of a recombinant Aalivirus genome, a recombinant Ailurivirus genome, a recombinant Ampivirus genome, a recombinant Anativirus genome, a recombinant Aphthovirus genome, a recombinant Aquamavirus genome, a recombinant Avihepatovirus genome, a recombinant Avisivirus genome, a recombinant Boosepivirus genome, a recombinant Bopivirus genome, a recombinant Caecilivirus genome, a recombinant Cardiovirus genome, a recombinant Cosavirus genome, a recombinant Crahelivirus genome, a recombinant Crohivirus genome, a recombinant Danipivirus genome, a
- Embodiment 92 the pharmaceutical composition of any of embodiments 2-91, wherein the recombinant iridovirus genome is a recombinant Invertebrate iridescent virus 6 (IIV-6) genome or a recombinant Invertebrate iridescent virus 31 (IIV-31) genome.
- the recombinant iridovirus genome is a recombinant Invertebrate iridescent virus 6 (IIV-6) genome or a recombinant Invertebrate iridescent virus 31 (IIV-31) genome.
- Embodiment 93 the pharmaceutical composition of any of embodiments 2-92, wherein the recombinant baculovirus genome is selected from the group consisting of a recombinant alphabaculovirus genome, a recombinant betabaculovirus genome, a recombinant deltabaculovirus genome, a recombinant gammabaculovirus genome, and any derivatives thereof.
- Embodiment 94 the pharmaceutical composition of any of embodiments 2-93, wherein the recombinant geminivirus genome is selected from the group consisting of a recombinant Becurtovirus genome, a recombinant Begomovirus genome, a recombinant Capulavirus genome, a recombinant Citlodavirus genome, a recombinant Curtovirus genome, a recombinant Eragrovirus genome, a recombinant Grablovirus genome, a recombinant Maldovirus genome, a recombinant Mastrevirus genome, a recombinant Mulcrilevirus genome, a recombinant Opunvirus genome, a recombinant Topilevirus genome, a recombinant Topocuvirus genome, a recombinant Turncurtovirus genome, and any derivatives thereof.
- the recombinant geminivirus genome is selected from the group consisting of a recombinant Becurto
- Embodiment 95 the pharmaceutical composition of any of embodiments 2-94, wherein the recombinant caulimovirus genome is selected from the group consisting of a recombinant Angelica bushy stunt virus genome, a recombinant Atractylodes mild mottle virus genome, a recombinant Carnation etched ring virus genome, a recombinant Cauliflower mosaic virus genome, a recombinant Dahlia mosaic virus genome, a recombinant Figwort mosaic virus genome, a recombinant Horseradish latent virus genome, a recombinant Lamium leaf distortion virus genome, a recombinant Mirabilis mosaic virus genome, a recombinant Soybean Putnam virus genome, a recombinant Strawberry vein banding virus genome, a recombinant Thistle mottle virus genome, and any derivatives thereof.
- Embodiment 96 the pharmaceutical composition of any of embodiments 2-95, wherein the recombinant
- Embodiment 97 the pharmaceutical composition of any of embodiments 2-96, wherein the non-viral gene delivery vehicle is a chemical compound, a bacterium, a mammalian cell, or a physical delivery system.
- the non-viral gene delivery vehicle is a chemical compound, a bacterium, a mammalian cell, or a physical delivery system.
- Embodiment 98 the pharmaceutical composition of any of embodiments 2-97, wherein the non-viral gene delivery vehicle is a chemical compound.
- Embodiment 99 the pharmaceutical composition of any of embodiments 2-98, wherein the non-viral gene delivery vehicle is a bacterium.
- Embodiment 100 the pharmaceutical composition of any of embodiments 2-99, wherein the non-viral gene delivery vehicle is a mammalian cell.
- Embodiment 101 the pharmaceutical composition of any of embodiments 2-100, wherein the non-viral gene delivery vehicle is a physical delivery system.
- Embodiment 102 the pharmaceutical composition of any of embodiments 2-101, wherein the chemical compound is selected from the groups consisting of a polymer compound, a lipid compound, an inorganic compound, and a combination thereof.
- Embodiment 103 the pharmaceutical composition of any of embodiments 2-102, wherein the chemical compound is a polymer compound.
- Embodiment 104 the pharmaceutical composition of any of embodiments 2-103, wherein the chemical compound is a lipid compound.
- Embodiment 105 the pharmaceutical composition of any of embodiments 2-104, wherein the chemical compound is an inorganic compound.
- Embodiment 106 the pharmaceutical composition of any of embodiments 2-105, wherein the polymer compound is a natural polymer, a synthetic polymer, a biopolymer, a biodegradable polymer, a cationic polymer, a protein polymer, a polysaccharide polymer, or any combination thereof.
- the polymer compound is a natural polymer, a synthetic polymer, a biopolymer, a biodegradable polymer, a cationic polymer, a protein polymer, a polysaccharide polymer, or any combination thereof.
- Embodiment 107 the pharmaceutical composition of any of embodiments 2-106, wherein the polymer compound is selected from the group consisting of poly-L-lactide (PLA), chitosan, pectin, polyethylenimine (PEI), poly-L-lysine (PLL), poly 2-N-dimethylaminoethyl methacrylate (PDMAEMA), polyaminoamine, polyaminoesters, polyamidoamine, poly-vinylimidazole (PVI), Poly- beta-amino ester (PBAE), gelatin, albumin, chitosan, beta-cyclodextrin, dextran, hyaluronic acid, polyester, alpha-Pyrrolidinohexiophenone (PHP), Poly-2-acrylamidoglycolic acid (PAGA), Poly-delta- valerolactone (PVL), polyphenyl ethers (PPE), Poly-beta-amino-ester (PLA),
- Embodiment 108 the pharmaceutical composition of any of embodiments 2-107, wherein the lipid compound is selected from the group consisting of a nanoparticle, a lipid nanoparticle, a liposome, a cationic liposome, a solid lipid nanoparticle, a lipid emulsion, a lipidoid, a cytofectin, a lipid emulsion, a surfactant, a gemini surfactant, and any derivatives thereof.
- the lipid compound is selected from the group consisting of a nanoparticle, a lipid nanoparticle, a liposome, a cationic liposome, a solid lipid nanoparticle, a lipid emulsion, a lipidoid, a cytofectin, a lipid emulsion, a surfactant, a gemini surfactant, and any derivatives thereof.
- Embodiment 109 the pharmaceutical composition of any of embodiments 2-108, wherein the inorganic compound is selected from the group consisting of a nanocarrier, a DNA nanclew, a gold nanoparticle, a carbon nanotube, a graphene, a quantum dot, an up-conversion nanoparticle, a silica nanoparticle, an iron oxide, a ferritin, and any derivatives thereof.
- the inorganic compound is selected from the group consisting of a nanocarrier, a DNA nanclew, a gold nanoparticle, a carbon nanotube, a graphene, a quantum dot, an up-conversion nanoparticle, a silica nanoparticle, an iron oxide, a ferritin, and any derivatives thereof.
- Embodiment 110 the pharmaceutical composition of any of embodiments 2-109, wherein the physical delivery system is selected from the group consisting of electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, hydroporation, and any combination thereof.
- the physical delivery system is selected from the group consisting of electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, hydroporation, and any combination thereof.
- Embodiment 111 use of the pharmaceutical composition of any of embodiments 2-110 in the manufacture of a medicament for treating cancer.
- Embodiment 112 use of embodiment 111, wherein the cancer is selected from the group consisting of acute myeloid leukemia, acute lymphoblastic leukemia, adrenocortical carcinoma, bladder urothelial cancer, brain stem glioma, brain lower grade glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, cancer of unknown primary site, carcinoid tumor, carcinoma of unknown primary site, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumors, cervical squamous cell carcinoma, endocervical adenocarcinoma cancer, childhood cancers, cholangiocarcinoma, chordoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, endocrine pancrea
- Embodiment 113 a method of expressing, enhancing, increasing, augmenting, and/or supplementing the levels of an IL-2 polypeptide and/or an IL-12 polypeptide in one or more cells of a subject, the method comprising administering to the subject an effective amount of the pharmaceutical composition of any of embodiment 2-112.
- Embodiment 114 the method of embodiment 113, wherein the one or more cells are one or more cells of the respiratory tract, airway epithelial, and/or lung.
- Embodiment 115 the method of any of embodiments 113 or 114, wherein the one or more cells is one or more cells of the respiratory tract.
- Embodiment 116 the method of any of embodiments 113-115, wherein the one or more cells is one or more of the airway epithelial.
- Embodiment 117 the method of any of embodiments 113-116, wherein the one or more cells is one or more of the lung.
- Embodiment 118 a method of providing prophylactic, palliative, or therapeutic relief of one or more signs or symptoms of cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of the pharmaceutical composition of any of embodiments 2-112.
- Embodiment 119 a method of treating cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of the pharmaceutical composition of any of embodiment 2-112.
- Embodiment 120 the method of any of embodiments 118 or 119, wherein the cancer is selected from the group consisting of acute myeloid leukemia, acute lymphoblastic leukemia, adrenocortical carcinoma, bladder urothelial cancer, brain stem glioma, brain lower grade glioma, brain tumor, breast cancer, bronchial tumors, Burkitt lymphoma, cancer of unknown primary site, carcinoid tumor, carcinoma of unknown primary site, central nervous system atypical teratoid/rhabdoid tumor, central nervous system embryonal tumors, cervical squamous cell carcinoma, endocervical adenocarcinoma cancer, childhood cancers, cholangiocarcinoma, chordoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloproliferative disorders, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma,
- Embodiment 121 the method of any of embodiments 118-120, wherein the cancer is selected from the group consisting of a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- the cancer is selected from the group consisting of a solid tumor, a hematologic cancer, bladder cancer, brain cancer, breast cancer, colon cancer, gastric cancer, glioma, head cancer, leukemia, liver cancer, lung cancer, lymphoma, myeloma, neck cancer, ovarian cancer, melanoma, pancreatic cancer, renal cancer, salivary cancer, stomach cancer, thymic epithelial cancer, and thyroid cancer.
- Embodiment 122 the method of any of embodiments 118-121, wherein the cancer is small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, or squamous carcinoma of the lung.
- Embodiment 123 the method of any of embodiments 118-122, wherein the cancer is non- small cell lung cancer.
- Embodiment 124 the method of any of embodiments 113-123, wherein the subject is a human.
- Embodiment 125 the method of any of embodiments 113-124, wherein the pharmaceutical composition is administered topically, transdermally, subcutaneously, epicutaneously, intradermally, orally, sublingually, buccally, rectally, vaginally, intravenously, intraarterially, intramuscularly, intraosseously, intracardially, intraperitoneally, transmucosally, intravitreally, subretinally, suprachoroidally, intracranially, intrathecally, intraventricularly, intraarticularly, peri-articularly, intratumorally, locally, or via inhalation to the subject.
- Embodiment 126 the method of any of embodiments 113-125, wherein the pharmaceutical composition is administered orally, intranasally, intratracheally, or via inhalation to the subject.
- Embodiment 127 the method of any of embodiments 113-126, wherein the pharmaceutical composition is administered intratumorally to the subject.
- Embodiment 128 the method of any of embodiments 113-127, wherein the pharmaceutical composition is administered intratracheally to the subject.
- the specification is considered to be sufficient to enable one skilled in the art to practice the present disclosure. Various modifications of the present disclosure in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims.
- Example 1 construction of a modified herpes simplex virus encoding human IL-12 and IL-2
- HSV-1 herpes simplex virus type 1
- HSV-1 was engineered to incorporate human IL12 and IL2 expression cassettes containing a heterologous promoter and polyA sequence. Viral plaques putatively containing the human IL-12 and IL-2 cassettes were picked and screened by infection in a complementing cell line to test for human IL-12 and IL-2 protein expression via western blot analysis (data not shown). High expressing clones termed HSV-1 L12/I L2 were subsequently selected for additional in vitro analysis.
- HEK 293FT cells Human embryonic kidney (HEK) 293FT cells were mock infected with vehicle control or were infected with HSV-IL12/IL2 at a multiplicity of infection (MOI) of 1 in serum-free cell culture medium. 24- or 48-hours post-infection, cell pellets were harvested, lysed in RIPA buffer containing protease inhibitors, and protein content was quantified via a BCA assay. 30-40pg of each sample was loaded and run on a 4-20% acrylamide gel, and expression of the HSV-encoded human protein was assessed via western blot analysis (data not shown). Recombinant human IL-12 or IL-2 was loaded on the gel as a positive control. While no human IL-12 or IL-2 was detected in the uninfected control cells, robust expression of human IL-12 (data not shown) and IL-2 (data not shown) was observed after infection with HSV-1 L12/I L2.
- MOI multiplicity of infection
- HEK293FT cell culture supernatants were also harvested and tested for the presence of the human protein by ELISA.
- human IL-12 over 3,000 ng/mL; FIG. 1A
- IL-2 over 400 ng/mL; FIG. IB
- FIG. IB human IL-12 and IL-2 were detected in the supernatants of HEK293FT cells infected with HSV-1 L12/I L2, at MOIs of 1, suggesting that the full-length human protein was being properly processed/secreted after expression from the recombinant vector.
- Example 2 in vitro murine IL-12 and IL-2 bioactivity assay
- HSV-1 herpes simplex virus type 1
- the objective of this study was, in part, to determine if the murine IL-12 protein and IL-2 proteins made from recombinant HSV-1 vectors were as bioactive as commercially available recombinant IL-12 and IL-2 protein.
- HEK293FT cells were infected with HSV-IL12 or HSV-IL2 at an MOI of 1 in serum-free cell culture medium for 24-hours. Cell culture supernatants were harvested and IL- 12 and IL-2 concentrations were determined by ELISA. As indicated in FIGS.
- murine splenocyte or HEK-BlueTM-IL2 cell culture supernatants were harvested and assayed by I FNy ELISA (FIG. 2A) or for secreted embryonic alkaline phosphatase (SEAP) activity (FIG. 2B).
- FIG. 2A the addition of aCD3/aCD28 coated beads resulted in the release of I FNy from murine splenocytes.
- the HSV-IL12 dependent release of IFNy was comparable to that induced by recombinant IL-12.
- FIG. 2B the HSV-IL2 dependent SEAP activity was comparable to that of recombinant IL-2 in HEK-BlueTM IL-2 reporter cells.
- Example 3 intradermal administration and in vivo evaluation of HSV-IL12 / HSV-IL2 in healthy mice [0319] As the above studies confirmed IL-12 and IL-2 transgene bioactivity, the next step was to test HSV-IL12 / HSV-IL2 in vivo. The present study evaluated IL-12 and/or IL-2 expression, following a single dose (8x10 s PFU) of HSV-IL12 / HSV-IL2 administered intradermally to C57BL/6 animals. IL-12 and IL-2 expression was assessed by nucleic acid and protein analysis in skin tissue at the indicated time points following vector administration. All procedures conducted were in compliance with applicable animal welfare acts and were approved by the local Institutional Animal Care and Use Committee (IACUC).
- IACUC Institutional Animal Care and Use Committee
- IL12 FIG. 3A
- IL2 FIG. 3C
- IL-12 FIG. 3B
- IL-2 FIG. 3D
- Peak cytokine protein levels were observed at 8-hours post-injection, and these protein levels remained detectable through day 7 post-HSV-IL12 / HSV-IL2 intradermal (ID) administration.
- Serum samples and skin biopsies were taken at five minutes or 24-hours post-treatment for analysis by ELISA.
- FIGS. 3E-3H there was minimal systemic cytokine exposure (FIGS. 3E-3F) following ID administration of HSV-IL12 / HSV-IL2, coupled with robust expression of vector-derived IL-12 and IL-2 observed in skin biopsies, compared to intravenous (IV) recombinant protein therapy (FIGS. 3G-3H).
- IV intravenous
- FIGS. 3G-3H intravenous recombinant protein therapy
- Example 4 intratracheal administration and in vivo evaluation of HSV-IL12 / HSV-IL2 in healthy mice [0322] The objective of this study was, in part, to evaluate transgene expression in the lungs following intratracheal (IT) administration of HSV-IL12 / HSV-IL2.
- BALB/c animals were administered a single dose of HSV-IL12 / HSV-IL2 intratracheally, at one of two doses (high or mid dose; ⁇ 10 8 or ⁇ 10 7 total PFU, respectively) on days 0 and 7 of the study. Animals were sacrificed on day 8, 24-hours following their last dose. Body weights were taken once weekly to assess toxicity, and IL-12 and IL-2 protein expression was assessed in lung tissue and bronchoalveolar lavage fluid (BALF) by ELISA. Results from this study indicate no significant differences in bodyweights between groups (FIG. 4A).
- IL-12 and IL-2 protein levels in the BALF and lung tissue were significantly higher in animals treated with HSV-IL12 / HSV-IL2 compared to vehicle control (FIGS. 4B-4D).
- HSV-IL12 / HSV-IL2 administered intratracheally is well-tolerated and results in detectable and dose-dependent IL-12 and IL-2 expression in the lungs.
- mice were treated with HSV-IL12 / HSV-IL2 ( ⁇ 10 7 total PFU) intratracheally or with IL-12 and IL-2 recombinant proteins (125 ng and 600 ng, respectively) intravenously at murine equivalent doses to human IL-12 and IL-2 that demonstrated clinical efficacy and toxicity in humans.
- Untreated animals served as negative controls. Serum samples and lungs were taken at five minutes or 24-hours post-treatment for analysis by ELISA (FIGS.
- FIGS. 4F-4/ there was minimal systemic cytokine exposure (FIGS. 4F-4G) following administration of HSV-IL12 / HSV-IL2 compared to IV recombinant protein treatment, in addition to more robust expression of IL-12 and IL-2 in lung homogenates (FIGS. 4H-4/).
- FIGS. 4F-4G systemic cytokine exposure
- FIGS. 4H-4/ lung homogenates
- Example 5 HSV-IL12 / HSV-IL2 efficacy in an in vivo murine model of melanoma
- the objective of this study was, in part, to evaluate the efficacy of intradermal administration of HSV-IL12 / HSV-IL2 therapy at inhibiting B16F10 melanoma tumor growth in the skin.
- the B16F10 model of melanoma following subcutaneous (SC) administration is well established.
- animals were dosed ( ⁇ 10 8 PFU) via intratumoral injection with the indicated vector at 7-, 14-, and 21-days following SC administration of B16F10 cells.
- FIG. 5B the greatest survival benefit was observed in animals treated with the combination HSV-IL12 / HSV-IL2 compared to groups treated with single vectors or vehicle control. Further, and as shown in FIGS.
- this survival benefit was potentiated when mice received a maintenance dose (FIG. 5C) of HSV-IL12 / HSV-IL2 at day 42 post SC administration of B16F10 cells.
- tumor area (FIG. 5D) was significantly reduced in the HSV-IL12 / HSV-IL2 treatment group compared to vehicle control.
- HSV-IL12 / HSV-IL2 Animals treated with HSV-IL12 / HSV-IL2 displayed a reduction in primary tumor outgrowth, independent of secondary tumor administration (FIGS. 5K, 5N, and 5Q). Notably, an abscopal effect was observed with HSV-IL12 / HSV-IL2 treatment in groups that received secondary tumors either on day 4 (FIG. 50) or day 10 (FIG. 5R) post-study initiation; no effect was demonstrated in day 0 secondary tumor recipients (FIG. 5L). Overall, a survival benefit in all groups was observed in mice receiving HSV-IL12 / HSV-IL2 treatment compared to vehicle control (FIGS. 5M, 5P, and 5S). Taken together, these data suggest HSV-IL12 / HSV-IL2 treatment of primary melanoma tumors results in an abscopal effect against a secondary melanoma tumor.
- Example 6 HSV-IL12 / HSV-IL2 efficacy in an in vivo murine model of osteosarcoma
- Example 7 a recombinant nucleic acid as a linear DNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as a linear DNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as a linear DNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as a linear DNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 8 a recombinant nucleic acid as a circular DNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as a circular DNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as a circular DNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as a circular DNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 9 a recombinant nucleic acid as a close ended DNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as a close ended DNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as a close ended DNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL- 2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as a close ended DNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 10 a recombinant nucleic acid as an RNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as an RNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as an RNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as an RNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 11 a recombinant nucleic acid as an mRNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as an mRNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as an mRNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as an mRNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 12 a recombinant nucleic acid as an mmRNA encoding IL-12 and IL-2
- a first recombinant nucleic acid is engineered as an mmRNA to include a recombinant polynucleotide encoding a human IL-12.
- a second recombinant nucleic acid is engineered as an mmRNA to include a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acids are tested for their in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as their efficacy as a cancer therapeutic. These results are not available at the time of filing.
- a recombinant nucleic acid is engineered as an mmRNA to include a recombinant polynucleotide encoding a human IL-12 and a recombinant polynucleotide encoding a human IL-2.
- the recombinant nucleic acid is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 13 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant adenovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant adenovirus genome, optionally in an adenovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 14 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant retrovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant retrovirus genome, optionally in a retrovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 15 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant adeno-associated virus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant adeno-associated virus genome, optionally in an adeno-associated virus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 16 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant poxvirus virus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant poxvirus genome, optionally in a poxvirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 17 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant bacteriophage genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7-
- Example 18 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant alphavirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant alphavirus genome, optionally in an alphavirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 19 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant picornavirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant picornavirus genome, optionally in a picornavirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 20 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant iridovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant iridovirus genome, optionally in an iridovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 21 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant Newcastle disease virus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant Newcastle disease virus genome, optionally in a Newcastle disease virus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 22 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant baculovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant baculovirus genome, optionally in a baculovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 23 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant geminivirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant geminivirus genome, optionally in a geminivirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 24 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant caulimovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant caulimovirus genome, optionally in a caulimovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 25 a recombinant nucleic acid as described in any one or more of examples 7-12 in a recombinant anellovirus genome
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in a recombinant anellovirus genome, optionally in an anellovirus gene delivery vehicle, and is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 26 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more chemical compounds, bacteria, mammalian cells, and/or physical delivery system
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more chemical compounds, bacteria, mammalian cells, and/or physical delivery systems as a non-viral gene delivery vehicle.
- the one or more chemical compounds, bacteria, mammalian cells, and/or physical delivery systems are tested for its in vitro and/or in vivo expression of IL-12 and/or IL- 2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 27 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more polymer compounds, lipid compounds, and/or inorganic compounds
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more polymer compounds, lipid compounds, and/or inorganic compounds as a non-viral gene delivery vehicle.
- the one or more polymer compounds, lipid compounds, and/or inorganic compounds are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 28 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more natural polymers, synthetic polymers, biopolymers, biodegradable polymers, cationic polymers, protein polymers, and/or polysaccharide polymers [0359]
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more natural polymers, synthetic polymers, biopolymers, biodegradable polymers, cationic polymers, protein polymers, and/or polysaccharide polymers as a non-viral gene delivery vehicle.
- the one or more natural polymers, synthetic polymers, biopolymers, biodegradable polymers, cationic polymers, protein polymers, and/or polysaccharide polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 29 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more natural polymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more natural polymers as a non-viral gene delivery vehicle.
- the one or more natural polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 30 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more synthetic polymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more synthetic polymers as a non-viral gene delivery vehicle.
- the one or more synthetic polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 31 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more biopolymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more biopolymers as a non-viral gene delivery vehicle.
- the one or more biopolymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 32 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more biodegradable polymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more biodegradable polymers as a non-viral gene delivery vehicle.
- the one or more biodegradable polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 33 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more cationic polymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more cationic polymers as a non-viral gene delivery vehicle.
- the one or more cationic polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 34 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more protein polymers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more protein polymers as a non-viral gene delivery vehicle.
- the one or more protein polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 35 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more polysaccharide polymer
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more polysaccharide polymers as a non-viral gene delivery vehicle.
- the one or more polysaccharide polymers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 36 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more nanoparticles, lipid nanoparticles, liposomes, cationic liposome, solid lipid nanoparticle, lipid emulsions, lipidoids, cytofectins, surfactants, and/or gemini surfactants
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more nanoparticles, lipid nanoparticles, liposomes, cationic liposome, solid lipid nanoparticle, lipid emulsions, lipidoids, cytofectins, surfactants, and/or gemini surfactants as a non- viral gene delivery vehicle.
- the one or more nanoparticles, lipid nanoparticles, liposomes, cationic liposome, solid lipid nanoparticle, lipid emulsions, lipidoids, cytofectins, surfactants, and/or gemini surfactants are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 37 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more nanoparticles as a non-viral gene delivery vehicle.
- the one or more nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 38 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more lipid nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more lipid nanoparticles as a non-viral gene delivery vehicle.
- the one or more lipid nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 39 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more liposomes
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more liposomes as a non-viral gene delivery vehicle.
- the one or more liposomes are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 40 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more cationic liposomes
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more cationic liposomes as a non-viral gene delivery vehicle.
- the one or more cationic liposomes are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 41 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more solid lipid nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more solid lipid nanoparticles as a non-viral gene delivery vehicle.
- the one or more solid lipid nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 42 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more lipid emulsions
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more lipid emulsions as a non-viral gene delivery vehicle.
- the one or more lipid emulsions are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 43 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more lipidoids
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more lipidoids as a non-viral gene delivery vehicle.
- the one or more lipidoids are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 44 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more cytofectins
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more cytofectins as a non-viral gene delivery vehicle.
- the one or more cytofectins are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 45 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more surfactants
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more surfactants as a non-viral gene delivery vehicle.
- the one or more surfactants are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 46 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more gemini surfactants
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more gemini surfactants as a non-viral gene delivery vehicle.
- the one or more gemini surfactants are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 47 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more nanocarriers, DNA nanclews, gold nanoparticles, carbon nanotubes, graphenes, quantum dots, up-conversion nanoparticles, silica nanoparticles, iron oxides, and/or ferritins
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more nanocarriers, DNA nanclews, gold nanoparticles, carbon nanotubes, graphenes, quantum dots, up-conversion nanoparticles, silica nanoparticles, iron oxides, and/or ferritins as a non- viral gene delivery vehicle.
- the one or more nanocarriers, DNA nanclews, gold nanoparticles, carbon nanotubes, graphenes, quantum dots, up-conversion nanoparticles, silica nanoparticles, iron oxides, and/or ferritins are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 48 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more nanocarriers
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more nanocarriers as a non-viral gene delivery vehicle.
- the one or more nanocarriers are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 49 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more DNA nanclews
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more DNA nanclews as a non-viral gene delivery vehicle.
- the one or more DNA nanclews are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 50 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more gold nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more gold nanoparticles as a non-viral gene delivery vehicle.
- the one or more gold nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 51 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more carbon nanotubes
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more carbon nanotubes as a non-viral gene delivery vehicle.
- the one or more carbon nanotubes are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 52 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more graphenes
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more graphenes as a non-viral gene delivery vehicle.
- the one or more graphenes are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 53 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more quantum dots
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more quantum dots as a non-viral gene delivery vehicle.
- the one or more quantum dots are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 54 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more up-conversion nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more up-conversion nanoparticles as a non-viral gene delivery vehicle.
- the one or more up-conversion nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 55 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more silica nanoparticles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more silica nanoparticles as a non-viral gene delivery vehicle.
- the one or more silica nanoparticles are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 56 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more iron oxides
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more iron oxides as a non-viral gene delivery vehicle.
- the one or more iron oxides are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 57 a recombinant nucleic acid as described in any one or more of examples 7-12 in one or more ferritins
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 in one or more ferritins as a non-viral gene delivery vehicle.
- the one or more ferritins are tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 58 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by one or more of electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, and/or hydroporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by one or more of electroporation, gene gun, jet gun, ultrasound, nucleofection, hydrodynamic gene delivery, needle injection, microinjection, ballistic DNA injection, sonoporation, photoporation, laser pulse, magnetofection, magnetoporation, magnetic particles, and/or hydroporation as a non-viral gene delivery vehicle.
- Example 59 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by electroporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by electroporation as a non-viral gene delivery vehicle.
- the delivery by electroporation is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 60 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by gene gun
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by gene gun as a non-viral gene delivery vehicle.
- the delivery by gene gun is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 61 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by jet gun
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by jet gun as a non-viral gene delivery vehicle.
- the delivery by jet gun is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 62 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by ultrasound
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by ultrasound as a non-viral gene delivery vehicle.
- the delivery by ultrasound is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 63 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by nucleofection
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by nucleofection as a non-viral gene delivery vehicle.
- the delivery by nucleofection is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 64 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by hydrodynamic gene delivery
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by hydrodynamic gene delivery as a non-viral gene delivery vehicle.
- the delivery by hydrodynamic gene delivery is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 65 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by needle injection
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by needle injection as a non-viral gene delivery vehicle.
- the delivery by needle injection is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 66 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by microinjection
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by microinjection as a non-viral gene delivery vehicle.
- the delivery by microinjection is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 67 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by ballistic DNA injection
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by ballistic DNA injection as a non-viral gene delivery vehicle.
- the delivery by ballistic DNA injection is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 68 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by sonoporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by sonoporation as a non-viral gene delivery vehicle.
- the delivery by sonoporation is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 69 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by photoporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by photoporation as a non-viral gene delivery vehicle.
- the delivery by photoporation is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 70 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by laser pulse [0401]
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12delivered by laser pulse as a non-viral gene delivery vehicle. The delivery by laser pulse is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic.
- Example 71 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by magnetofection
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by magnetofection as a non-viral gene delivery vehicle.
- the delivery by magnetofection is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 72 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by magnetoporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by magnetoporation as a non-viral gene delivery vehicle.
- the delivery by magnetoporation is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 73 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by magnetic particles
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by magnetic particles as a non-viral gene delivery vehicle.
- the delivery by magnetic particles is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
- Example 74 a recombinant nucleic acid as described in any one or more of examples 7-12 delivered by hydroporation
- a recombinant nucleic acid is engineered as described in any one or more of examples 7- 12 delivered by hydroporation as a non-viral gene delivery vehicle.
- the delivery by hydroporation is tested for its in vitro and/or in vivo expression of IL-12 and/or IL-2, as well as its efficacy as a cancer therapeutic. These results are not available at the time of filing.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Molecular Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Biophysics (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Toxicology (AREA)
- Mycology (AREA)
- Immunology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU2024229184A AU2024229184A1 (en) | 2023-03-02 | 2024-03-01 | Interleukin-2 and interleukin-12 for cancer therapy |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363449533P | 2023-03-02 | 2023-03-02 | |
| US63/449,533 | 2023-03-02 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2024182707A1 true WO2024182707A1 (en) | 2024-09-06 |
| WO2024182707A9 WO2024182707A9 (en) | 2024-12-05 |
Family
ID=90719353
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/018084 Pending WO2024182707A1 (en) | 2023-03-02 | 2024-03-01 | Interleukin-2 and interleukin-12 for cancer therapy |
Country Status (2)
| Country | Link |
|---|---|
| AU (1) | AU2024229184A1 (en) |
| WO (1) | WO2024182707A1 (en) |
Citations (17)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5580859A (en) | 1989-03-21 | 1996-12-03 | Vical Incorporated | Delivery of exogenous DNA sequences in a mammal |
| US5693622A (en) | 1989-03-21 | 1997-12-02 | Vical Incorporated | Expression of exogenous polynucleotide sequences cardiac muscle of a mammal |
| US5753234A (en) | 1995-03-16 | 1998-05-19 | Lg Chemical Ltd. | Single-shot vaccine formulation |
| US7081483B2 (en) | 2001-07-03 | 2006-07-25 | The University Of Chicago | Polyfunctional dispersants for controlling viscosity of phyllosilicates |
| US20060246139A1 (en) | 2005-04-28 | 2006-11-02 | Nipro Corporation | Bioabsorbable pharmaceutical formulation |
| US20070092575A1 (en) | 2005-05-10 | 2007-04-26 | Naomi Balaban | Compositions for administering RNAIII-inhibiting peptides |
| US20070148074A1 (en) | 2003-01-16 | 2007-06-28 | Mostafa Sadoqi | Nanoparticle based stabilization of ir fluorescent dyes |
| US20080260851A1 (en) | 2004-05-13 | 2008-10-23 | The Trustees Of Columbia University In The City Of New York | Polymeric Nanoparticles and Nanogels for Extraction and Release of Compounds |
| US20100015232A1 (en) | 2006-07-07 | 2010-01-21 | Aarhus Universitet | Nanoparticles for nucleic acid delivery |
| US20130195800A1 (en) | 2010-03-23 | 2013-08-01 | Intrexon Corporation | Vectors Conditionally Expressing Therapeutic Proteins, Host Cells Comprising the Vectors, and Uses Thereof |
| US10702600B1 (en) | 2015-10-22 | 2020-07-07 | Modernatx, Inc. | Betacoronavirus mRNA vaccine |
| US20200283794A1 (en) | 2017-09-08 | 2020-09-10 | Generation Bio Co. | Modified closed-ended dna (cedna) |
| US10898574B2 (en) | 2011-03-31 | 2021-01-26 | Modernatx, Inc. | Delivery and formulation of engineered nucleic acids |
| US20210059953A1 (en) | 2017-09-08 | 2021-03-04 | Generation Bio Co. | Lipid nanoparticle formulations of non-viral, capsid-free dna vectors |
| US20210108228A1 (en) | 2015-05-26 | 2021-04-15 | Ramot At Tel-Aviv University Ltd. | Targeted lipid particles for systemic delivery of nucleic acid molecules to leukocytes |
| WO2022109503A1 (en) * | 2020-11-23 | 2022-05-27 | Devacell, Inc. | Recombinant oncolytic viruses, surface-engineered delivery systems and related methods |
| WO2022212896A1 (en) * | 2021-04-02 | 2022-10-06 | Krystal Biotech, Inc. | Viral vectors for cancer therapy |
-
2024
- 2024-03-01 WO PCT/US2024/018084 patent/WO2024182707A1/en active Pending
- 2024-03-01 AU AU2024229184A patent/AU2024229184A1/en active Pending
Patent Citations (19)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5589466A (en) | 1989-03-21 | 1996-12-31 | Vical Incorporated | Induction of a protective immune response in a mammal by injecting a DNA sequence |
| US5693622A (en) | 1989-03-21 | 1997-12-02 | Vical Incorporated | Expression of exogenous polynucleotide sequences cardiac muscle of a mammal |
| US5580859A (en) | 1989-03-21 | 1996-12-03 | Vical Incorporated | Delivery of exogenous DNA sequences in a mammal |
| US5753234A (en) | 1995-03-16 | 1998-05-19 | Lg Chemical Ltd. | Single-shot vaccine formulation |
| US7081483B2 (en) | 2001-07-03 | 2006-07-25 | The University Of Chicago | Polyfunctional dispersants for controlling viscosity of phyllosilicates |
| US20070148074A1 (en) | 2003-01-16 | 2007-06-28 | Mostafa Sadoqi | Nanoparticle based stabilization of ir fluorescent dyes |
| US20080260851A1 (en) | 2004-05-13 | 2008-10-23 | The Trustees Of Columbia University In The City Of New York | Polymeric Nanoparticles and Nanogels for Extraction and Release of Compounds |
| US20060246139A1 (en) | 2005-04-28 | 2006-11-02 | Nipro Corporation | Bioabsorbable pharmaceutical formulation |
| US20070092575A1 (en) | 2005-05-10 | 2007-04-26 | Naomi Balaban | Compositions for administering RNAIII-inhibiting peptides |
| US20100015232A1 (en) | 2006-07-07 | 2010-01-21 | Aarhus Universitet | Nanoparticles for nucleic acid delivery |
| US20130195800A1 (en) | 2010-03-23 | 2013-08-01 | Intrexon Corporation | Vectors Conditionally Expressing Therapeutic Proteins, Host Cells Comprising the Vectors, and Uses Thereof |
| US10898574B2 (en) | 2011-03-31 | 2021-01-26 | Modernatx, Inc. | Delivery and formulation of engineered nucleic acids |
| US20210108228A1 (en) | 2015-05-26 | 2021-04-15 | Ramot At Tel-Aviv University Ltd. | Targeted lipid particles for systemic delivery of nucleic acid molecules to leukocytes |
| US10702600B1 (en) | 2015-10-22 | 2020-07-07 | Modernatx, Inc. | Betacoronavirus mRNA vaccine |
| US10933127B2 (en) | 2015-10-22 | 2021-03-02 | Modernatx, Inc. | Betacoronavirus mRNA vaccine |
| US20200283794A1 (en) | 2017-09-08 | 2020-09-10 | Generation Bio Co. | Modified closed-ended dna (cedna) |
| US20210059953A1 (en) | 2017-09-08 | 2021-03-04 | Generation Bio Co. | Lipid nanoparticle formulations of non-viral, capsid-free dna vectors |
| WO2022109503A1 (en) * | 2020-11-23 | 2022-05-27 | Devacell, Inc. | Recombinant oncolytic viruses, surface-engineered delivery systems and related methods |
| WO2022212896A1 (en) * | 2021-04-02 | 2022-10-06 | Krystal Biotech, Inc. | Viral vectors for cancer therapy |
Non-Patent Citations (19)
| Title |
|---|
| "Cell and Tissue Culture: Laboratory Procedures", August 1993, J. WILEY AND SONS |
| "Gene Transfer Vectors for Mammalian Cells", 1987 |
| "Methods in Enzymology", 2003, ACADEMIC PRESS, INC., article "Current Protocols in Molecular Biology" |
| "NCBI", Database accession no. 3.593 |
| "Oligonucleotide Synthesis", 1984 |
| "PCR 2: A Practical Approach", 1988 |
| "PCR: The Polymerase Chain Reaction", 1994 |
| "Short Protocols in Molecular Biology", 1999, WILEY AND SONS |
| "UniProt", Database accession no. P29460 |
| AKIO HIRAKI: "Interleukin-12 augments cytolytic activity of peripheral blood mononuclear cells against autologous lung cancer cells in combination with IL-2", LUNG CANCER., vol. 35, no. 3, 1 March 2002 (2002-03-01), NL, pages 329 - 333, XP093166057, ISSN: 0169-5002, DOI: 10.1016/S0169-5002(01)00424-X * |
| ANONYMOUS: "Study Details | Combination Therapy of Interleukin-12 and Interleukin-2 to Treat Advanced Cancer | ClinicalTrials.gov", 17 December 2019 (2019-12-17), XP093165922, Retrieved from the Internet <URL:https://clinicaltrials.gov/study/NCT00005655?cond=cancer&term=il-2%20and%20il-12&rank=1> * |
| BOWEN ET AL., J VIROL., vol. 93, no. 8, 3 April 2019 (2019-04-03) |
| DE SILVA S.BOWERS W.: "Herpes Virus Amplicon Vectors", VIRUSES, vol. 1, 2009, pages 594 - 629 |
| FATH ET AL., PLOS ONE, vol. 6, no. 3, 3 March 2011 (2011-03-03), pages e17596 |
| J.P. MATHERP.E. ROBERTS: "Introduction to Cell and Tissue Culture", 1998, ACADEMIC PRESS |
| KAUFMAN H L ET AL: "Insertion of interleukin-2 (IL-2) and interleukin-12 (IL-12) genes into vaccinia virus results in effective anti-tumor responses without toxicity", VACCINE, ELSEVIER, AMSTERDAM, NL, vol. 20, no. 13-14, 15 March 2002 (2002-03-15), pages 1862 - 1869, XP004366025, ISSN: 0264-410X, DOI: 10.1016/S0264-410X(02)00032-4 * |
| SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS |
| VAUTHIER ET AL., ADV DRUG DEL REV., vol. 55, 2003, pages 519 - 48 |
| WIGGINTON JON M ET AL: "IL-12/IL-2 combination cytokine therapy for solid tumours: translation from bench to bedside", EXPERT OPINION ON BIOLOGICAL THERAPY, INFORMA HEALTHCARE, vol. 2, no. 5, 1 June 2002 (2002-06-01), pages 513 - 524, XP009152161, ISSN: 1471-2598 * |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2024182707A9 (en) | 2024-12-05 |
| AU2024229184A1 (en) | 2025-08-28 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7480105B2 (en) | Compositions and methods for treating skin wounds, disorders, and diseases | |
| US20210221851A1 (en) | Vsv/ndv hybrid viruses for oncolytic therapy of cancer | |
| JP3989541B2 (en) | Method for producing viral vector by intermolecular homologous recombination | |
| KR102814622B1 (en) | Methods and compositions for increasing protein expression and/or treating hepatotoxicity disorders | |
| JP2014516536A5 (en) | ||
| CN110741081B (en) | Tumor-selective TATA box and CAAT box mutants | |
| JP2023515692A (en) | Compositions and methods for the treatment of metabolic liver injury | |
| JP2024513826A (en) | Viral vectors for cancer treatment | |
| JP2022546545A (en) | Compositions and methods for the treatment of congenital ichthyosis | |
| WO2024182707A1 (en) | Interleukin-2 and interleukin-12 for cancer therapy | |
| US20210317174A1 (en) | Compositions and methods for delivering cftr polypeptides | |
| JP7560449B2 (en) | Compositions and methods for treating skin disorders | |
| JP7336791B2 (en) | Coxsackievirus group B for treating tumors | |
| CN1934253A (en) | Subgroup B adenoviral vectors for treating disease | |
| JP7773468B2 (en) | Compositions and methods for gene delivery to the airways and/or lungs | |
| JP7250031B2 (en) | Novel viral vectors and methods for producing and using the same | |
| CN117568338A (en) | An optimized polyA sequence and its application | |
| CN111100847A (en) | Telomere gene therapy product with higher safety for human health to reverse aging | |
| EP3997106B1 (en) | Oncolytic non-human adenoviruses and uses thereof | |
| CN113549605B (en) | Compositions and vector systems for inhibiting SARS-CoV-2 | |
| CN113573741B (en) | Modified adenovirus and medicine containing the same | |
| CN1869242A (en) | Recombination adenovirus and its application | |
| Brimble | Characterisation of adeno-associated virus DNA contaminants and development of a P5 promoter replacement, high purity adeno-associated virus production system | |
| WO2024221102A1 (en) | Gm2 activator vectors and methods for use thereof |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24717386 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: AU2024229184 Country of ref document: AU |
|
| ENP | Entry into the national phase |
Ref document number: 2024229184 Country of ref document: AU Date of ref document: 20240301 Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024717386 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2024717386 Country of ref document: EP Effective date: 20251002 |
|
| ENP | Entry into the national phase |
Ref document number: 2024717386 Country of ref document: EP Effective date: 20251002 |
|
| ENP | Entry into the national phase |
Ref document number: 2024717386 Country of ref document: EP Effective date: 20251002 |