[go: up one dir, main page]

WO2025106374A1 - Procédé de production de vaa - Google Patents

Procédé de production de vaa Download PDF

Info

Publication number
WO2025106374A1
WO2025106374A1 PCT/US2024/055351 US2024055351W WO2025106374A1 WO 2025106374 A1 WO2025106374 A1 WO 2025106374A1 US 2024055351 W US2024055351 W US 2024055351W WO 2025106374 A1 WO2025106374 A1 WO 2025106374A1
Authority
WO
WIPO (PCT)
Prior art keywords
aav
cell culture
cells
nucleic acid
antifoam agent
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
Application number
PCT/US2024/055351
Other languages
English (en)
Inventor
Dah-ve BELL
Daniel Hurwit
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Juno Therapeutics Inc
Original Assignee
Juno Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Juno Therapeutics Inc filed Critical Juno Therapeutics Inc
Publication of WO2025106374A1 publication Critical patent/WO2025106374A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/755Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0091Purification or manufacturing processes for gene therapy compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14151Methods of production or purification of viral material

Definitions

  • compositions and methods related to producing an adeno-associated virus such as methods comprising culturing a packaging cell culture in the presence of an antifoam agent prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Adeno-associated viruses such as recombinant AAVs
  • AAVs are typically produced in permissive (packaging) host cell cultures.
  • Cell damage and loss due to foam formation in culture systems can limit cell culture-based bioproduction efficiencies, including AAV production.
  • increasing the cell density of a culture leads to increased concentrations of cellular proteins and increased oxygen demand, which can each lead to excessive foam formation in the culture.
  • Foam formation can damage and destroy AAV-producing cells (AAV packaging cells) and can interfere with bioreactor components (e.g., reducing pump efficiencies (due to cavitation, for example), reducing capacities of pumps and storage tanks, promoting unwanted bacterial growth, causing drainage problems, and leading to culture downtime as a result of necessary cleanings).
  • bioreactor components e.g., reducing pump efficiencies (due to cavitation, for example), reducing capacities of pumps and storage tanks, promoting unwanted bacterial growth, causing drainage problems, and leading to culture downtime as a result of necessary cleanings.
  • AAVs such as recombinant AAVs
  • gene therapy and vaccine approaches have been described.
  • AAVs including solutions for reducing or preventing foam formation in these methods.
  • the present disclosure provides improved, scalable methods of producing AAVs, such as for therapeutic use in a subject, including improved solutions for preventing and/or reducing foam formation in AAV packaging cell cultures.
  • AAVs such as for therapeutic use in a subject
  • improved solutions for preventing and/or reducing foam formation in AAV packaging cell cultures are provided.
  • Embodiment 1 is a method of producing adeno-associated virus (AAV), comprising
  • Embodiment 2 is a method of producing an AAV, comprising culturing packaging cells comprising at least one nucleic acid encoding an AAV under conditions suitable for production of the AAV, wherein the packaging cells are cultured in the presence of an antifoam agent prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 3 is the method of embodiment 1 or embodiment 2, further comprising harvesting the AAV from the cell culture.
  • Embodiment 5 is the method of any one of the preceding embodiments, wherein the antifoam agent is an inorganic antifoam agent.
  • Embodiment 6 is the method of any one of the preceding embodiments, wherein the antifoam agent comprises silicone.
  • Embodiment 8 is the method of any one of the preceding embodiments, wherein the antifoam agent comprises 10-20%, 20-30%, 30-40%, 40-50%, 10-40%, 10-30%, 20-50%, 30- 50%, 20-40%, 25-35%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% simethicone.
  • Embodiment 9 is the method of any one of embodiments 1-5, wherein the antifoam agent comprises a mineral oil.
  • Embodiment 10 is the method of any one of the preceding embodiments, wherein the cell culture comprises 15-100, 15-80, 15-60, 20-80, 20-70, 20-60, 30-60, 24-45, 55-75, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 parts per million of the antifoam agent prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 11 is the method of any one of the preceding embodiments, wherein the antifoam agent is present in the cell culture between 20 minutes and 120 hours prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 12 is the method of any one of the preceding embodiments, wherein the antifoam agent is present in the cell culture at least 20 minutes, at least 30 minutes, at least 40 minutes, at least 50 minutes, at least one hour, at least 1.5 hours, at least 2 hours, at least 2.5 hours, at least 5 hours, at least 10 hours, at least 15 hours, at least 20 hours, at least 24 hours, at least 48 hours, at least 72 hours, at least 96 hours, or at least 120 hours prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 13 is the method of any one of the preceding embodiments, wherein the AAV is or is derived from an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV 10, AAV11, AAV 12, or Myo AAV.
  • Embodiment 14 is the method of any one of the preceding embodiments, wherein the AAV is a chimeric AAV.
  • Embodiment 15 is the method of any one of the preceding embodiments, wherein, after transfecting the cell culture with the at least one nucleic acid encoding the AAV, the AAV is harvested from the cell culture every 1-6 days, every 1-5 days, every 1-4 days, every 1-3 days, every 2-6 days, every 2-5 days, or every 2-4 days.
  • Embodiment 16 is the method of any one of the preceding embodiments, wherein the AAV is harvested from the cell culture 1-6 days, 1-5 days, 1-4 days, 1-3 days, 2-6 days, 2-5 days, or 2-4 days after transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 23 is the method of embodiment 22, wherein the one or more helper genes comprise at least one polynucleotide encoding an adenovirus packaging helper gene, at least one polynucleotide encoding an AAV replication protein sufficient for packaging, and/or at least one polynucleotide encoding an AAV capsid protein sufficient for packaging.
  • Embodiment 24 is the method of embodiment 23, wherein the packaging cells are stably transformed with the at least one polynucleotide encoding an adenovirus packaging helper gene, the at least one polynucleotide encoding an AAV replication protein sufficient for packaging, and/or the at least one polynucleotide encoding an AAV capsid protein sufficient for packaging.
  • Embodiment 25 is the method of any one of the preceding embodiments, wherein the packaging cells comprise:
  • Embodiment 26 is the method of any one of the preceding embodiments, wherein the AAV comprises a nucleic acid molecule encoding at least one protein or RNA of interest.
  • Embodiment 27 is the method of embodiment 26, wherein the nucleic acid molecule encodes at least one protein or RNA of therapeutic interest.
  • Embodiment 28 is the method of embodiment 26 or embodiment 27, wherein the nucleic acid molecule encodes a chimeric antigen receptor.
  • Embodiment 29 is the method of any one of embodiments 22-28, wherein the one or more helper genes is expressed under a constitutive promoter, an activatable promoter, or an inducible promoter.
  • Embodiment 30 is the method of any one of embodiments 22 or 24-29, wherein the one or more helper viruses comprises an adenovirus, a baculovirus, or a herpes simplex virus.
  • Embodiment 31 is the method of embodiment 30, wherein the adenovirus is a wild-type adenovirus.
  • Embodiment 32 is the method of any one of the preceding embodiments, wherein a titer of the AAV at harvest is higher than that of a control.
  • Embodiment 33 is the method of embodiment 32, wherein the control comprises the same AAV harvested from a cell culture not comprising the antifoam agent.
  • Embodiment 34 is the method of embodiment 32 or embodiment 33, wherein the control comprises the same AAV harvested from a control cell culture to which the antifoam agent was only added after transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 35 is the method of any one of embodiments 32-34, wherein the control comprises the same AAV harvested from a control cell culture wherein the antifoam agent is not present in the control cell culture prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV, and wherein 5-10 parts per million of the antifoam agent is added to the control cell culture at least once after transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 36 is the method of embodiment 32, wherein the control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added in an amount at least 1.5-fold, at least 1.75-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold less than an amount of the antifoam agent added to the cell culture, prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 37 is the method of embodiment 32, wherein the control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added at least 1 hour, at least 2 hours, at least 5 hours, at least 10 hours, at least 15 hours, at least 20 hours, or at least 24 hours later than the antifoam agent was added to the cell culture, and prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • Embodiment 38 is the method of any one of embodiments 32-37, wherein the titer of the AAV at harvest is at least 1.25-fold, at least 1.5-fold, at least 1.75-fold, or at least 2-fold greater than that of the control.
  • Embodiment 39 is the method of any one of embodiments 32-38, wherein the titer of the AAV at harvest is at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50% greater, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% greater than that of the control.
  • Embodiment 40 is the method of any one of the preceding embodiments, wherein the cell culture comprises a HEK cell culture medium.
  • Embodiment 41 is the method of any one of the preceding embodiments, wherein the cell culture comprises glutamine and/or a shear protectant.
  • Embodiment 42 is the method of any one of the preceding embodiments, wherein the cell culture is serum-free.
  • Embodiment 43 is the method of any one of the preceding embodiments, wherein the cell culture is a suspension culture.
  • Embodiment 44 is the method of any one of the preceding embodiments, wherein the cell culture is in a bioreactor.
  • Embodiment 45 is the method of any one of the preceding embodiments, wherein the cell culture is a batch-fed cell culture.
  • Embodiment 46 is the method of any one of embodiments 1-44, wherein the cell culture is a continuous cell culture.
  • Embodiment 47 is the method of any one of embodiments 1-44 or 46, wherein the cell culture is a perfusion cell culture.
  • Embodiment 48 is the method of any one of the preceding embodiments, wherein the packaging cells are mammalian cells.
  • Embodiment 49 is the method of any one of the preceding embodiments, wherein the mammalian cells are Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) cells, Madin-Darby canine kidney (MDCK) cells, or Vero cells.
  • the mammalian cells are Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) cells, Madin-Darby canine kidney (MDCK) cells, or Vero cells.
  • Embodiment 50 is the method of embodiment 48 or embodiment 49, wherein the mammalian cells are Chinese hamster ovary (CHO) cells.
  • the mammalian cells are Chinese hamster ovary (CHO) cells.
  • Embodiment 51 is the method of embodiment 48 or embodiment 49, wherein the mammalian cells are human cells.
  • Embodiment 52 is the method of embodiment 51, wherein the human cells are HEK293 cells.
  • Embodiment 53 is the method of any one of embodiments 1-47, wherein the packaging cells are insect cells.
  • Embodiment 54 is the method of any one of the preceding embodiments, wherein the method does not comprise adding an antifoam agent to the cell culture after transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • FIG. 1 illustrates the use of an antifoam agent in an AAV6 suspension culture process to increase AAV vector production.
  • An antifoam agent (30 ppm or 60 ppm) was added to HEK293 cell cultures prior to transfecting the cell cultures with at least one nucleic acid encoding the AAV. Control cultures did not comprise an antifoam agent.
  • FIG.2 illustrates the use of an antifoam agent in an AAV5 suspension culture process to increase AAV vector production.
  • An antifoam agent (5 ppm, 30 ppm, or 60 ppm) was added to HEK293 cell cultures post-inoculation (DO), at the time of plasmid transfection on day 3 (D3), or 24 hours post-transfection on day 4 (D4). Control cultures did not comprise an antifoam agent.
  • adeno-associated virus refers to an adeno-associated virus vector, including any AAV serotype or variant, including but not limited to an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrhlO (see, e.g, SEQ ID NO: 81 of US 9,790,472), AAVrh74 (see, e.g, SEQ ID NO: 1 of US 2015/0111955), AAV9, AAV9P also known as AAVMYO (see, e.g., Weinmann etal., Nature Communications, 2020, 11 :5432), AAV11, AAV 12, and Myo- AAV (as described, for example, in Tabebordbar etal., 2021, Cell, 184: 1-20 (e.g., MyoAAV 1A, 2 A, 3 A, 4 A, 4C, or 4E)), and chimeras thereof (
  • AAV can also refer to any known AAV (vector) system.
  • the AAV vector is a single-stranded AAV (ssAAV).
  • the AAV vector is a double-stranded AAV (dsAAV).
  • AAVs are small ( ⁇ 25 nm), singlestranded DNA, non-enveloped viruses with an icosahedral capsid.
  • AAV can refer to naturally occurring or engineered AAV serotypes and recombinant AAVs (rAAVs) and variants that can differ in the composition and structure of their capsid protein, and can have varying tropism, z. ?., ability to transduce different cell types. When combined with active promoters, this tropism defines the site of gene expression, e.g., in a host.
  • recombinant AAV refers to an AAV with a capsid having packaged therein a heterologous nucleic acid molecule comprising an expression cassette for a desired product, such as a gene product.
  • a desired product such as a gene product.
  • Such an expression cassette may contain an AAV 5’ and/or 3’ inverted terminal repeat sequence flanking a nucleic acid of interest, such as a gene sequence, in which the nucleic acid of interest is operably linked to expression control sequences.
  • An expression cassette is thus useful for effecting the expression of the desired product (e.g., protein or RNA) in an intended target cell.
  • Expression cassettes of use herein are known and available in the art or can be readily constructed from components that are available in the art.
  • heterologous nucleic acid as used herein is a nucleic acid derived from a genotypically distinct entity from that of the rest of the entity to which it is being compared.
  • a nucleic acid introduced by genetic engineering techniques into a plasmid or vector derived from a different species is a heterologous nucleic acid.
  • a promoter removed from its native coding sequence and operatively linked to a coding sequence with which it is not naturally found linked is a heterologous promoter.
  • heterologous nucleic acid as used herein includes coding as well as non-coding nucleotide sequences.
  • antifoam agent refers to an agent that can be added to a cell culture to prevent or counter foam generation, e.g., in the culture medium.
  • an antifoam agent can reduce or prevent formation of foam in a cell culture.
  • an antifoam agent may also be useful for reducing foam already present in a cell culture. Such agents may also be referred to as a “defoam agent” or “defoaming agent.”
  • Antifoam agents of use herein include synthetic (i.e., inorganic) agents, such as silicone-based antifoams (e.g., siloxane polymers).
  • an antifoam agent comprises simethicone.
  • isolated refers to separation of a biological component (such as an AAV) from some or all other components of a mixture (such as cell culture media, whole cells, cellular materials, and/or cell lysate).
  • a biological component such as an AAV
  • a “packaging cell” refers to a cell wherein an AAV may be “packaged.” “Packaging” refers to a series of intracellular events that result in the assembly of the capsid proteins and encapsidation of the vector genome to form an AAV particle, thereby “packaging” the vector (such as a vector comprising a nucleic acid molecule of interest) in a delivery vehicle (the AAV particle).
  • the terms “cell line” or “cell culture” refer to a packaging cell line or packaging cell culture.
  • the term “packaging cell” as used herein can also mean a “producer cell” as known in the art and as described herein.
  • a producer cell may comprise (e.g., be transfected with) the AAV genome to be produced.
  • a producer cell may be generated by stably integrating AAV genes (such as along with an ITR-flanked therapeutic gene of interest) into a cell line (such as CHO, HEK293, or SF9 cells).
  • AAV production may then be triggered by the addition of a helper virus to provide functional genes for AAV replication.
  • a cell has been “transfected,” e.g., with at least one nucleic acid encoding an AAV, when such nucleic acids have been introduced inside the cell.
  • a cell may be transfected, e.g., with one or more (such as one, two, three, or four) recombinant plasmids or other nucleic acids through any process known in the art, including but not limited to electroporation, calcium phosphate precipitation, or contacting with a polynucleotide-liposome complex.
  • transfection encompasses any means of introducing one or more (such as one, two, three, or four) nucleic acids inside a cell, such as, but not limited to, transduction or infection with a DNA or RNA virus or viral vector.
  • a transfected nucleic acid may be introduced into a chromosome or mini -chromosome in the cell.
  • foam formation can damage and destroy AAV-producing cells (AAV packaging cells) and can interfere with bioreactor components (e.g., reducing pump efficiencies (due to cavitation, for example), reducing capacities of pumps and storage tanks, promoting unwanted bacterial growth, causing drainage problems, and leading to culture downtime as a result of necessary cleanings), and thus can limit AAV production efficiencies.
  • AAV packaging cells AAV packaging cells
  • bioreactor components e.g., reducing pump efficiencies (due to cavitation, for example), reducing capacities of pumps and storage tanks, promoting unwanted bacterial growth, causing drainage problems, and leading to culture downtime as a result of necessary cleanings
  • higher density packaging cell cultures may have increased concentrations of cellular proteins and substantially increased oxygen demands, each of which can lead to increased foam formation.
  • Current commercial methods for producing AAVs may employ mechanical mechanisms and/or chemical additives in attempting to reduce or prevent foam formation in AAV packaging cell culture systems.
  • Antifoam agents are typically added to packaging cell culture systems following inoculation of the cells into the culture media, e.g., when foaming is observed in the culture media.
  • foam production in AAV packaging cell cultures remains a limiting factor in the efficient production of AAVs, such as recombinant AAVs for use in therapeutic applications.
  • the present disclosure provides improved methods for AAV production using packaging cell cultures.
  • the method comprises culturing packaging cells in the presence of at least one antifoam agent and transfecting the packaging cell culture with at least one nucleic acid encoding an AAV.
  • the at least one nucleic acid encoding an AAV encodes, e.g., AAV rep and/or cap genes, a nucleotide sequence of interest (e.g., a nucleic acid encoding a protein or RNA of interest, or a noncoding sequence of interest), one or more helper functions, an additional element (such as one or more elements that can enhance viral second-strand DNA synthesis, one or more aspects of viral assembly and production, and/or AAV-mediated transgene expression (See, e.g., Ma W et al., Hum Gene Ther. 2011, 22(5):633-40; Khan N et al. Cancer Med. 2020, 9(9):3188-3201), or any combination thereof.
  • AAV rep and/or cap genes e.g., AAV rep and/or cap genes, a nucleotide sequence of interest (e.g., a nucleic acid encoding a protein or RNA of interest, or a noncoding sequence of interest), one or more helper functions
  • the at least one nucleic acid encoding an AAV encodes AAV rep and/or cap genes. In some embodiments, the at least one nucleic acid encoding an AAV encodes a nucleotide sequence of interest (e.g., a nucleic acid encoding a protein or RNA of interest, or a noncoding sequence of interest). In some embodiments, the at least one nucleic acid encoding an AAV encodes one or more helper functions. In some embodiments, the at least one nucleic acid encoding an AAV encodes an additional element, such as one or more elements that can enhance viral second-strand DNA synthesis, one or more aspects of viral assembly and production, and/or AAV-mediated transgene expression.
  • a nucleotide sequence of interest e.g., a nucleic acid encoding a protein or RNA of interest, or a noncoding sequence of interest.
  • the at least one nucleic acid encoding an AAV encodes one or more helper functions.
  • the at least one nucleic acid encoding an AAV comprises a final component for production of the AAV in the packaging cell.
  • a packaging cell has been transfected and/or infected and/or co-infected with other components needed to produce the AAV, and transfection of the at least one nucleic acid encoding the AAV triggers production of the AAV.
  • the packaging cells are cultured in the presence of the at least one antifoam agent prior to transfecting the cell culture with the AAV.
  • the packaging cells are inoculated into cell culture media comprising the at least one antifoam agent.
  • the cell culture media comprises at least one antifoam agent prior to inoculation of the AAV packaging cells into the cell culture media.
  • the method comprises culturing packaging cells in the presence of an AAV under conditions suitable for production of the AAV, wherein the packaging cells are cultured in the presence of at least one antifoam agent prior to transfecting the cell culture with at least one nucleic acid encoding the AAV.
  • the at least one antifoam agent is a non-ionic antifoam agent, such as an inorganic antifoam agent.
  • the at least one antifoam agent comprises simethicone.
  • the disclosed methods do not comprise adding the at least one antifoam agent to the cell culture after transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • Some embodiments of the present disclosure further comprise harvesting the AAV from the cell culture.
  • a titer of an AAV at harvest is higher than that of a control, such as a control comprising the same AAV harvested from a cell culture not comprising the antifoam agent, e.g., a control comprising the same AAV harvested from a cell culture that did not comprise the antifoam agent prior to transfection of the cells with at least one nucleic acid encoding an AAV.
  • the titer of the AAV at harvest is at least 1.25-fold, at least 1.5-fold, at least 1.75-fold, or at least 2-fold greater than that of the control.
  • the titer of the AAV at harvest is at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50% greater, at least 55%, at least 60%, at least 65%, at least 70%, or at least 75% greater than that of the control.
  • the control comprises the same AAV harvested from a control cell culture to which the antifoam agent was only added after transfecting the control cell culture with at least one nucleic acid encoding the AAV.
  • the control comprises the same AAV harvested from a control cell culture wherein the antifoam agent is not present in the control cell culture prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV, and wherein the antifoam agent is added to the control cell culture at least once (such as once, twice, three times, for times, five times, or more) after transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • the control comprises the same AAV harvested from a control cell culture wherein the antifoam agent is not present in the control cell culture prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV, and wherein 5-100 parts per million, such as 5-90, 5-80, 5-70, 5-60, 5-50-, 5-40, 5-30, 5-20, or 5-10 parts per million, of the antifoam agent is added to the control cell culture at least once (such as once, twice, three times, for times, five times, or more) after transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • 5-100 parts per million such as 5-90, 5-80, 5-70, 5-60, 5-50-, 5-40, 5-30, 5-20, or 5-10 parts per million
  • control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added in an amount less than an amount of the antifoam agent added to the non-control cell culture, prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added in an amount at least 1.5-fold, at least 1.75-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold less than an amount of the antifoam agent added to the cell culture, prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • control comprises the same AAV harvested from a control cell culture to which the antifoam agent was added at least 1 hour, at least 2 hours, at least 5 hours, at least 10 hours, at least 15 hours, at least 20 hours, or at least 24 hours later than the antifoam agent was added to the cell culture, and prior to transfecting the control cell culture with the at least one nucleic acid encoding the AAV.
  • Antifoam agents are added to a cell culture to prevent or reduce foam generation, e.g., in the culture medium.
  • these agents may have surface active properties and/or may be insoluble in the culture medium.
  • antifoam agents are typically added prior to foam formation in order to reduce or prevent the foam formation, but alternatively or in addition may be added after foam formation to reduce foam already present (e.g., in culture media).
  • Antifoam agents can also be applied directly to cell culture system components, e.g., to filling nozzles, rims of processing vats, screens, etc.
  • antifoam agents can act by reducing the surface tension of a solution or emulsion, thereby inhibiting or modifying the formation of a foam.
  • Antifoam agents of include synthetic agents, such as those known in the art, including silicone-based antifoam agents (e.g., siloxane polymers), and antifoam agents comprising polyethylene glycol and polypropylene glycol copolymers.
  • an antifoam agent is silicone-based and has a molecular weight range of 3,000 to 17,000 Daltons.
  • an antifoam agent is silicone- based and comprises particles ranging in size from 5 to 50 microns. Such particles may be removable from a cell culture, in some embodiments, such as by filtration.
  • Antifoam agents may be formulated for use in a cell culture system, and thus may be in the form of a fluid, suspension, emulsion (such as an oil-based emulsion or a water-based emulsion), powder, and/or any other form suitable for such use.
  • antifoam agents of use herein may comprise additional components, such as one or more of an emulsifier (such as a non-ionic emulsifier) or diluent (such as propylene glycol, water, or vegetable oil).
  • an emulsifier such as a non-ionic emulsifier
  • diluent such as propylene glycol, water, or vegetable oil
  • a non-ionic emulsifier comprises an olyoxyethylene fatty acid derivative of the sorbitan esters (for example, Tween), a polyoxyethylene fatty alcohol ether, a sorbitan fatty acid ester, a polyoxyethylene alkyl ether (a macrogol), a polyoxyethylene sorbitan fatty acid ester, a polyoxyethylene polyoxypropylene block copolymer (poloxamer), a polyethylene glycol 400 monostearate, a lanolin alcohol, an ethoxylated lanolin, a castor oil ethoxylate, a tri-styryl phenol ethoxylate, and/or an alcohol alkoxylate.
  • sorbitan esters for example, Tween
  • a polyoxyethylene fatty alcohol ether for example, a sorbitan fatty acid ester, a polyoxyethylene alkyl ether (a macrogol), a polyoxyethylene sorbitan fatty acid ester, a poly
  • the antifoam agent does not include an emulsifier and/or does not include a diluent.
  • Exemplary commercial antifoam agent formulations of use herein include, but are not limited to, Antifoam A Concentrate (A5633 and A6582; Sigma Aldrich), Antifoam B Emulsion (A5757; Sigma Aldrich), Antifoam C Emulsion (A8011 ; Sigma Aldrich), Antifoam Y-30 Emulsion (A5758 and A6457; Sigma Aldrich), and Antifoam SE-15 (A8582; Sigma Aldrich).
  • an antifoam agent is a non-ionic antifoam agent, such as an inorganic antifoam agent.
  • the antifoam agent comprises a mineral oil.
  • the antifoam agent comprises 10- 20%, 20-30%, 30-40%, 40-50%, 10-40%, 10-30%, 20-50%, 30-50%, 20-40%, 25-35%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% mineral oil.
  • the antifoam agent comprises silicone.
  • the antifoam agent comprises simethicone.
  • Simethicone (C2HeOSi)n (SiO2)m] is a mixture of fully methylated linear siloxane polymers comprising repeating units of the formula [- (CH3)2SiO]n, stabilized with trimethylsiloxy end-blocking units of the formula [(CHj ⁇ SiO-], and silicon dioxide. Simethicone can reduce and/or prevent foaming by reducing the surface tension of formed gas bubbles in a cell culture medium or cell culture system.
  • the antifoam agent comprises 10-20%, 20-30%, 30-40%, 40-50%, 10-40%, 10- 30%, 20-50%, 30-50%, 20-40%, 25-35%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% simethicone.
  • an antifoam agent is present in an AAV packaging cell culture prior to transfecting the cell culture with at least one nucleic acid encoding an AAV.
  • an AAV packaging cell culture comprises 15-100, 15-80, 15-60, 20-80, 20- 70, 20-60, 30-60, 24-45, 55-75, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 parts per million of the antifoam agent prior to transfecting the cell culture with at least one nucleic acid encoding the AAV.
  • an antifoam agent is present in the AAV packaging cell culture at least 10 minutes, at least 20 minutes, at least 30 minutes, at least 40 minutes, or at least 50 minutes prior to transfecting the cell culture with at least one nucleic acid encoding the AAV.
  • the antifoam agent is present in the AAV packaging cell culture at least 1 hour, such as at least 1.25, at least 1.5, at least 1.75, at least 2, at least 2.5, at least 3, at least 3.5, at least 4, at least 4.5, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 36, at least 48, at least 60, at least 72, at least 84, at least 96, at least 108, or at least 120 hours prior to transfecting the cell culture with at least one nucleic acid encoding the AAV.
  • the antifoam agent is added to the cell culture at least 20 minutes prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • the antifoam agent is present in the AAV packaging cell culture between 20 minutes and 120 hours prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • the antifoam agent is present in the cell culture 20 minutes - 110 hours, 20 minutes - 100 hours, 20 minutes - 90 hours, 20 minutes - 80 hours, 20 minutes - 70 hours, 20 minutes - 60 hours, 20 minutes - 50 hours, 20 minutes - 40 hours, 20 minutes - 30 hours, 20 minutes - 20 hours, 20 minutes - 15 hours, 20 minutes - 10 hours, 20 minutes - 9 hours, 20 minutes - 8 hours, 20 minutes - 7 hours, 20 minutes - 6 hours, 20 minutes - 5 hours, 20 minutes - 4 hours, 20 minutes - 3 hours, 20 minutes - 2 hours, 20 minutes - 1.5 hours, 20 minutes - 1 hour, 20-50 minutes, 20-40 minutes, 20-35 minutes, 20-30 minutes, or 20-25 minutes prior to transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • the antifoam agent is added to the cell culture 20 minutes - 120 hours prior to transfecting the cell culture with
  • Embodiments of the disclosed methods comprise culturing packaging cells in the presence of at least one antifoam agent and transfecting the cell culture with at least one nucleic acid encoding an AAV.
  • the packaging cells are cultured in the presence of the at least one antifoam agent prior to inoculating the cell culture with the AAV.
  • the packaging cells are inoculated into cell culture media comprising the at least one antifoam agent.
  • the cell culture media comprises at least one antifoam agent prior to inoculation of the AAV packaging cells into the cell culture media.
  • the method comprises culturing packaging cells in the presence of an AAV under conditions suitable for production of the AAV, wherein the packaging cells are cultured in the presence of at least one antifoam agent prior to transfecting the cell culture with at least one nucleic acid encoding the AAV.
  • the disclosed methods do not comprise adding an antifoam agent to the cell culture after transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • a packaging cell useful in the present disclosure can be a eukaryotic cell, a fungal cell, an insect cell, a prokaryotic cell (c.g, bacterial or archaeal cell), or a cell from a multicellular organism (e.g, a cell line) cultured as a unicellular entity, and include the progeny of the original cell if such cell has been transformed by the nucleic acid.
  • the cell is a eukaryotic cell, such as a 293T cell (such as a HEK293T cell).
  • Exemplary mammalian cells include without limitation, Chinese hamster ovary (CHO) cells, CHO-K1 cells, CHO-derived cells, human embryonic kidney (HEK) cells (such as HEK293 cells, which express functional adenoviral El, and HEK-derived cells such as HEK293T or HEK293F), Madin-Darby canine kidney (MDCK) cells, Vero cells, EB66 cells, chicken embryo cells, RK cells, RAF cells, PK15 cells, MRC-5 cells, A549 cells, WEHI cells, 3T3 cells, 10T1/2 cells, BHK cells, COS 1 cells, COS 7 cells, BSC 1 cells, BSC 40 cells, BMT 10 cells, WI38 cells, HeLa cells, Saos cells, C2C12 cells, L cells, HT1080 cells, HepG2 cells, NS-1 cells, and primary fibroblast, hepatocyte, and myoblast cells derived from mammals including human, monkey, mouse, rat, rabbit, and
  • the cells are suspension-adapted cells.
  • the selection of the mammalian species providing the cells is not a limitation of this invention; nor is the type of mammalian cell, i.e., fibroblast, hepatocyte, tumor cell, etc.
  • the cell is a fungal cell, such as a yeast cell, such as a yeast cell of species Saccharomyces (such as Saccharomyces cerevisiae).
  • the cell is an insect cell e.g., for use in baculovirus-based AAV production systems), such as an SF-9 cell.
  • a “recombinant packaging cell” (also referred to as a “genetically modified packaging cell”) is a packaging cell into which has been introduced a heterologous nucleic acid, e.g., an expression vector.
  • a bacterial packaging cell is a genetically modified bacterial packaging cell by virtue of introduction of an exogenous nucleic acid (e g., a plasmid or recombinant expression vector) into a suitable bacterial packaging cell
  • a eukaryotic packaging cell is a genetically modified eukaryotic packaging cell (e.g., a mammalian cell), by virtue of introduction of an exogenous nucleic acid into a suitable eukaryotic packaging cell.
  • the packaging cells are Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) cells (such as HEK293 cells), Madin-Darby canine kidney (MDCK) cells, or Vero cells.
  • CHO Chinese hamster ovary
  • HEK human embryonic kidney
  • MDCK Madin-Darby canine kidney
  • Vero cells Humidity
  • the packaging cells are CHO cells.
  • the packaging cells are HEK293 cells.
  • the disclosed methods comprise culturing an AAV packaging cell in media that allows production of the AAV.
  • suitable media known in the art may be used for AAV production including, without limitation, media produced by Hyclone Laboratories and JRH including Modified Eagle Medium (MEM), Dulbecco's Modified Eagle Medium (DMEM), and custom formulations such as those described in U.S. Pat. No. 6,566,118, and Sf-900 II SFM media as described in U.S. Pat. No. 6,723,551.
  • AAV production culture media may be supplemented with serum or serum-derived recombinant proteins, e.g., at a level of 0.5%-20% (v/v or w/v).
  • AAV vectors may be produced in serum-free conditions which may also be referred to as media with no animal-derived products.
  • Commercial or custom media designed to support production of AAVs may also be supplemented with one or more cell culture components know in the art, including without limitation, glucose, vitamins, amino acids, and/or growth factors, in order to increase the titer or yield of AAV in production cultures.
  • the cell culture comprises a culture medium suitable for culturing HEK293 cells, such as a chemically defined HEK293 cell culture medium.
  • the cell culture comprises glutamine and/or a shear protectant.
  • the cell culture is serum-free.
  • Exemplary commercially available cell culture media include, but are not limited to, BalanCD media (e.g., BalanCD HEK293 medium or BalanCD CHO medium; Fujifilm and Irvine Scientific), Viral Production Media (e.g., Gibco LV-MAXTM Production Medium), and F 17 medium (Invitrogen).
  • Methods of culturing an AAV packaging cell to produce an AAV are known in the art, and exemplary methods are discussed herein.
  • the disclosed methods comprise culturing an AAV packaging cell in media that allows production of the AAV.
  • Cell culture procedures for both large and small-scale production of AAVs are encompassed by the present disclosure.
  • AAV production cultures can be grown under a variety of conditions (over a wide temperature range, for varying lengths of time, and the like) suitable to the particular host cell being utilized.
  • AAV production cultures include attachment-dependent cultures that can be cultured in suitable attachment-dependent vessels such as, for example, roller bottles, hollow fiber filters, microcarriers, and packed-bed or fluidized-bed bioreactors.
  • AAV vector production cultures may also include suspension-adapted host cells such as HEK293, HeLa, and SF-9 cells that can be cultured in a variety of attachment-independent vessels including, for example, spinner flasks, stirred tank bioreactors, batch bioreactors, fed batch bioreactors, continuous culture bioreactors (e.g., perfusion bioreactors), and disposable systems such as the Wave bag system.
  • cells are initially ‘bulked up’ in tissue culture flasks or bioreactors and subsequently grown in multi-layered culture vessels or larger bioreactors (e.g., greater than 50 L).
  • media into which the cells are inoculated generally comprises an antifoam agent prior to the inoculation.
  • Suitable conditions for culturing cells are known, and further, numerous suspension culture systems are known in the art for production of AAV (such as rAAV) particles (See, e.g., U.S. Pat. Nos. 6,995,006 and 9,783,826; U.S. Pat. Appl. Pub. No. 20120122155; Tissue Culture, Academic Press, Kruse and Paterson, editors (1973); and R. I. Freshney, Culture of animal cells: A manual of basic technique, fourth edition, Wiley -Liss Inc., 2000, ISBN 0-471-34889-9, each of which is incorporated herein by reference in its entirety).
  • AAV such as rAAV
  • a cell culture disclosed herein is a suspension culture.
  • a cell culture is a suspension culture comprising HEK293 cells.
  • a cell culture is a suspension culture comprising HEK293 cells adapted for growth in suspension culture.
  • a suspension cell culture comprises a serum-free medium, an animal-component free medium, and/or a chemically defined medium.
  • suspension-adapted cells are cultured in a shaker flask, a spinner flask, a cellbag, or a bioreactor.
  • a cell culture comprises cells attached to a substrate (e.g., microcarriers) that are themselves in suspension in a medium.
  • the cells are HEK293 cells.
  • a cell culture is an adherent culture.
  • a cell culture disclosed herein is an adherent culture comprising HEK293 cells.
  • an adherent cell culture comprises a serum-free medium, an animal-component free medium, and/or or a chemically defined medium.
  • a cell culture of the disclosed methods comprises a high-density cell culture.
  • the cell culture has a total cell density of at least about 2xl0 6 vc/mL, at least about 2xl0 7 vc/mL, or at least about 2xl0 8 vc/mL.
  • the cell culture has a total cell density of between IxlO 6 and 2xl0 7 vc/mL, such as between IxlO 6 and IxlO 7 vc/mL, IxlO 6 and 75xl0 6 vc/mL, IxlO 6 and 50xl0 6 vc/mL, or IxlO 6 and 25xl0 6 vc/mL. In some embodiments, more than about 50% of the cells of the cell culture are viable cells.
  • the cells are HeLa cells, HEK293 cells, HEK293 derived cells (e.g., HEK293T cells, HEK293F cells), CHO cells, Vero cells, or SF-9 cells.
  • the cells are HEK293 cells.
  • the cells are HEK293 cells adapted for growth in suspension culture.
  • culturing is performed by incubating a mammalian packaging cell line (e.g., HEK293 or CHO cells) under humidified conditions.
  • the humidified conditions comprise incubating the transfected cells at about 37°C and about 5% CO2.
  • large volumes of cell culture can be present (e.g., during the commercial manufacturing processes).
  • the methods disclosed herein are suitable for the processing of a large volume of cell culture comprising AAV (such as rAAV) particles.
  • the cell culture is a suspension culture, a batch fed culture, a continuous cell culture, or a perfusion cell culture.
  • the methods disclosed herein are scalable, and thus can be carried out in any desired volume of culture medium, e.g., from 10 ml (e.g., in shaker flasks) to 0.5 L, 1 L, 5 L, 10 L, 20 L, 30 L, 40 L, 50 L, 60 L, 70 L, 80 L, 90 L, 100 L, 500 L, 1,000 L, 5,000 L, 10,000 L, or more (e.g. in bioreactors such as wave bioreactor systems and stirred tanks).
  • the cell culture is cultured (such as in a bioreactor) in a volume of at least 0.01 L, at least 0.1 L, at least 0.2 L, at least 0.3 L, at least 0.4 L, at least 0.5 L, at least 0.6 L, at least 0.7 L, at least 0.8 L, at least 0.9 L, at least 1 L, at least 1.25 L, at least 1.5 L, at least 1.75 L, at least 2 L, at least 3 L, at least 4 L, at least 5 L, at least 6 L, at least 7 L, at least 8 L, at least 9 L, at least 10 L, at least 15 L, at least 20 L, at least 25 L, at least 50 L, at least 75 L, at least 100 L, at least 200 L, at least 300 L, at least 400 L, at least 500 L, or at least 1,000 L.
  • the cell culture is cultured (such as in a bioreactor) in a volume of 0.1-1,000 L, such as 1-1,000 L, 20-1,000 L, 50-1,000 L, 75-1,000 L, 100-1,000 L, 500-1,000 L, 1-900 L, 1-800 L, 1-700 L, 1- 600 L, 1-500 L, 1-400 L, 1-300 L, 1-200 L, 0.1-90 L, 0.1-80 L, 0.1-70 L, 0.1-60 L, 0.1-50 L, 0.1- 40 L, 0.1-30 L, 0.1-25 L, 0.1-20 L, 0.1-15 L, 0.1-10 L, 0.1-9 L, 0.1-8 L, 0.1-7 L, 0.1-6 L, 0.1-5 L, 0.1-4 L, 0.1-3 L, 0.1-2 L, 0.1-1 L, 0.1-0.75 L, 0.1-0.5 L, 0.1-0.25 L, 90-100 L, 80-100 L, 70- 100 L, 60-100 L, 50-100 L, 40-100 L, 30-100 L, 25-100 L, 20-100
  • the cell culture is in a bioreactor.
  • the cell culture is cultured at a pH of between about 6 and about 8, such as at a pH between about 6.1 and 7.9, 6.2 and 7.8, 6.3 and 7.7, 6.4 and 7.6, 6.5 and 7.5, 6.6 and 7.4, 6.7 and 7.4, 6.8 and 7.4, 6.8 and 7.3, or 6.8 and 7.2.
  • the cell culture is cultured with a dissolved oxygen (DO) setpoint of about 20-60%, such as about 25-60%, about 30-60%, about 35-55%, about 40-55%, about 45-55%, about 20-55%, about 20-50%, about 20-45%, about 20-35%, or about 20-25%.
  • the cell culture is cultured at an agitation rate that imparts a power input per volume (P/V) of about 15-30 W/m 3 , such as about 15-25, 15-20, 20-30, or 25- 35 W/m 3 .
  • P/V power input per volume
  • a packaging cell (such as a packaging cell cultured in the presence of an antifoam agent) is transfected with at least one nucleic acid (such as a plasmid) encoding the AAV.
  • the preparation of a packaging cell suitable for use in the disclosed methods involves techniques such as assembly of selected DNA sequences. This assembly may be accomplished utilizing conventional techniques. Such techniques include cDNA and genomic cloning, which are well known and are described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., including polymerase chain reaction, synthetic methods, and any other suitable methods which provide the desired nucleotide sequence.
  • a cell (such as an AAV packaging cell) has been transfected by an exogenous nucleic acid, e.g., a recombinant expression vector, when such a nucleic acid (e.g., DNA) has been introduced inside the cell.
  • an exogenous nucleic acid e.g., DNA
  • the presence of an exogenous nucleic acid can result in permanent or transient genetic change.
  • the transfected exogenous nucleic acid may or may not be integrated (covalently linked) into the genome of the cell.
  • a cell wherein an exogenous nucleic acid has been integrated into the genome of the cell is “stably transformed.”
  • helper virus On its own, AAV does not possess the ability to efficiently replicate its genetic material, and thus typically requires the presence of a helper virus.
  • Three helper viruses are commonly used in AAV production: adenovirus (Ad), herpes simplex virus (HSV), and baculovirus (Bac).
  • Ad adenovirus
  • HSV herpes simplex virus
  • Bac baculovirus
  • the packaging cells are infected with one or more helper viruses, such as an adenovirus (such as a wild-type adenovirus), a baculovirus, or a herpes simplex virus.
  • Ad and HSV helper AAV production methods use mammalian cell lines while the Bac system uses insect cells.
  • an AAV production system useful in the disclosed methods may be helper free.
  • the one or more helper functions, the AAV Rep protein, the AAV capsid protein, and/or the nucleic acid molecule of interest is expressed under an activatable or inducible promoter.
  • the nucleic acid molecule of interest encodes a chimeric antigen receptor (CAR) or T cell receptor (TCR), such as are known in the art and/or described elsewhere herein.
  • Genetic material can be introduced into cells using any of a variety of means.
  • such techniques include, for example, transfection with viral, bacteria, or yeast- derived plasmids.
  • Transfection methods have been described in the art and include, for example, calcium phosphate co-precipitation, direct micro-injection into cultured cells, electroporation, liposome mediated gene transfer, lipid-mediated transfection or nucleic acid delivery using high- velocity microprojectiles.
  • Other suitable transfection media include strontium phosphate, poly cationic polymers, e.g., SuperfectTM (Qiagen), liposomes, and cationic polymers such as polyethylenimine (PEI).
  • the term transfection encompasses any means of introducing a nucleic acid inside a cell, including, but not limited to, transduction or infection with a DNA or RNA virus or viral vector.
  • any of these techniques can be used to introduce one or more exogenous nucleic acids, such as vector constructs, into suitable host cells.
  • the exogenous nucleic acid traverses the host cell plasma membrane in order to be exposed to the cell's transcription and replication machinery.
  • the resulting cell can be transiently transfected with the exogenous nucleic acid molecule, i.e., the exogenous DNA will not be integrated into the genome of a transfected cell, but rather can exist episomally.
  • the resulting cell can be stably transfected, i.e., the nucleic acid molecule becomes covalently linked with the host cell genome or is maintained and replicated as an episomal unit that can be passed on to progeny cells (e.g., is capable of extra- chromosomal replication at a sufficient rate).
  • transfecting packaging cells with an AAV comprises dual transfection, triple transfection, quad transfection, induction of a stable cell line, or co-infection with one or more helper viruses.
  • the packaging cells are HEK293 cells or derivatives thereof.
  • HEK, CHO, or SF9 cells are used as stable cell lines from which AAV is induced. Stable cell lines may be engineered by introducing either the AAV rep and cap genes (packaging cell lines) and/or the AAV genome to be produced (producer cells).
  • An AAV can be produced from packaging cell lines upon transfection of the AAV construct and coinfection with a helper virus (such as an Ad virus) or upon infection with a recombinant hybrid helper (such as an Ad/ AAV).
  • a helper virus such as an Ad virus
  • Ad/ AAV recombinant hybrid helper
  • co-infection with one or more helper viruses may comprise co-infection with a BAcv, Ad5, or HSV helper virus in SF9 cells, HEK293 cells, or BHK cells, respectively.
  • Such transfection methods are known in the art, such as described in Clement and Grieger, Mol Ther Methods Clin Dev. 2016 Mar 16;3 : 16002, which is incorporated by reference herein in its entirety.
  • “dual transfection” is performed using two plasmid vectors, typically wherein the first plasmid vector comprises a heterologous nucleotide sequence (such as a nucleic acid molecule of interest) flanked by ITRs and the second plasmid vector comprises the AAV rep and AAV cap gene sequences and adenovirus helper functions sequences, e.g., the Ad5 genes (VA RNAs, E2A, and E4OEF6).
  • HEK293 cells constitutively express Ela/b, the fourth Ad function for AAV replication.
  • the second plasmid vector comprises from 5' to 3' an AAV rep coding region, an AAV cap coding region and a nucleotide sequence comprising a promoter region, e.g., an AAV p5 promoter region.
  • “triple transfection” (also known as “3-plasmid transfection”) is performed using three plasmid vectors, such that the packaging cells are triple-transfected with at least one vector encoding a heterologous nucleic acid (such as a nucleic acid molecule of interest), at least one vector encoding AAV rep and cap genes, and at least one vector encoding adenoviral accessory functions, such as those described above for dual transfection.
  • dual and triple transfections are performed using calcium phosphate (CaPO4) plasmid precipitation in HEK293 cells.
  • quadruple-transfected with at least one vector encoding a heterologous nucleic acid (such as a nucleic acid molecule of interest), at least one vector encoding AAV rep and cap genes, at least one vector encoding adenoviral accessory functions, and at least one vector encoding an additional element, such as one or more elements that can enhance viral second-strand DNA synthesis, one or more aspects of viral assembly and production, and/or AAV-mediated transgene expression.
  • a heterologous nucleic acid such as a nucleic acid molecule of interest
  • the packaging cells comprise one or more nucleic acids encoding one or more helper genes, and/or the packaging cells are infected with one or more helper viruses.
  • the one or more helper genes comprise at least one nucleic acid encoding an adenovirus packaging helper gene, at least one nucleic acid encoding an AAV replication protein sufficient for packaging, and/or at least one nucleic acid encoding an AAV capsid protein sufficient for packaging.
  • the packaging cells are stably transformed with the at least one nucleic acid encoding an adenovirus packaging helper gene, the at least one nucleic acid encoding an AAV replication protein sufficient for packaging, and/or the at least one nucleic acid encoding an AAV capsid protein sufficient for packaging.
  • the packaging cells comprise (a) a nucleic acid encoding E2A, a nucleic acid encoding E4, and a nucleic acid encoding VA genes; (b) a nucleic acid encoding an AAV replication protein; (c) a nucleic acid encoding an AAV capsid protein; and/or (d) a nucleic acid encoding a gene of interest, optionally comprising an inverted terminal repeat.
  • the AAV (such as an AAV5, AAV6, or AAV8) comprises an AAV capsid protein and a nucleic acid molecule that comprises an AAV 5’ inverted terminal repeat (ITR), a nucleic acid molecule of interest to be packaged into the AAV capsid, and a 3’ ITR.
  • ITR inverted terminal repeat
  • the packaging cell can thus comprise (i) the at least one nucleic acid of interest to be packaged into the AAV capsid, (ii) a nucleic acid molecule encoding the AAV capsid protein under control of one or more sequences that direct its expression in the packaging cell, (iii) a nucleic acid molecule encoding an AAV Rep protein that expresses the AAV Rep protein in the cell to permit packaging of the nucleic acid of interest into the AAV capsid, and/or (iv) one or more helper functions required for packaging the nucleic acid molecule of interest into the AAV capsid.
  • the nucleic acid molecule of interest encodes a chimeric antigen receptor (CAR) or T cell receptor (TCR)).
  • the one or more helper genes is expressed under, for example, a constitutive promoter, an activatable promoter, or an inducible promoter.
  • An exemplary promoter useful in the present disclosure can be a constitutively active promoter (i.e., a promoter that is constitutively in an active/“ON” state), an activatable promoter, an inducible promoter (i.e., a promoter whose state, active/“ON” or inactive/ “OFF”, is controlled by an external stimulus, e.g., the presence of a particular temperature, compound, or protein), a spatially restricted promoter (i.e., transcriptional control element, enhancer, etc., e.g., a tissue specific promoter, a cell type specific promoter, etc.), or a temporally restricted promoter (i.e., the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process).
  • a constitutively active promoter i
  • Suitable promoters can be derived from viruses (viral promoters), or they can be derived from any organism, including prokaryotic or eukaryotic organisms. Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • viruses viral promoters
  • Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • Exemplary promoters include, but are not limited to the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a Rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6) (Miyagishi etal., Nature Biotechnology. 20: 497-500; 2002), an enhanced U6 promoter (e. ., Xia et al., Nucleic Acids Res. Sep 1(31): 17; 2003), a human Hl promoter (Hl), and the like.
  • LTR mouse mammary tumor virus long terminal repeat
  • Ad MLP adenovirus major late promoter
  • HSV herpes simplex virus
  • CMVIE cytomegalovirus
  • RSV
  • nucleic acid(s) introduced to the cell may be introduced simultaneously with or operably linked to one or more detectable or selectable markers as is known in the art.
  • a drug resistance gene can be used as a selectable marker.
  • Drug resistant cells can then be picked and grown, and then tested for expression of a desired sequence, such as a packaging gene product, or a product of the heterologous nucleic acid, as appropriate.
  • Testing for acquisition, localization, and/or maintenance of an introduced nucleic acid can be performed by means known in the art, for example, using DNA hybridization-based techniques (such as Southern blotting and other procedures as known in the art), Northern analysis of RNA extracted from the genetically altered cells, or by indirect immunofluorescence for the corresponding gene product.
  • DNA hybridization-based techniques such as Southern blotting and other procedures as known in the art
  • Northern analysis of RNA extracted from the genetically altered cells or by indirect immunofluorescence for the corresponding gene product.
  • an AAV (such as an AAV5, AAV6, or AAV8) is harvested from AAV packaging cells and/or from the cell culture media.
  • the AAV after transfecting the cell culture with at least one nucleic acid encoding the AAV, the AAV is harvested from the cell culture every 1-6 days, every 1-5 days, every 1-4 days, every 1-3 days, every 2-6 days, every 2-5 days, or every 2-4 days.
  • the AAV is harvested from the cell culture 1-6 days, 1-5 days, 1-4 days, 1-3 days, 2-6 days, 2-5 days, or 2-4 days after transfecting the cell culture with the at least one nucleic acid encoding the AAV.
  • the AAV harvesting may comprise cell disruption (such as cell lysis) or may substantially not comprise cell disruption (such as in embodiments wherein harvesting the AAV from the cell culture comprises harvesting the AAV from cell culture media (e.g., from a supernatant of the cell culture)).
  • AAVs of the invention may be harvested from AAV production cultures by lysis of the packaging cells of the production culture or by harvest of the media (“spent” media) from the production culture, provided the cells are cultured under conditions known in the art to cause release of AAV particles into the media from intact cells (such as described more fully in U.S. Pat. No. 6,566,118).
  • Suitable methods of lysing cells are also known in the art and include for example multiple freeze/thaw cycles, sonication, microfluidization, and treatment with chemicals, such as detergents and/or proteases.
  • harvesting the AAV from the cell culture comprises isolating the AAV from the cell culture.
  • Methods of isolating an AAV from a cell culture can include, but are not limited to, chemical lysis, mechanical lysis, depth filtration, microfiltration, ultrafiltration, alternating tangential flow filtration, tangential flow depth filtration, batch chromatography, acoustic wave separation, and/or centrifugation.
  • isolating the AAV from the cell culture further comprises concentrating the AAV using a method selected from chromatography, column-based chromatography, membrane-based chromatography, filtration, and/or precipitation.
  • an AAV of the disclosed methods can be an AAV packaging cell culture harvest without further processing, or can be treated, e.g., prior to loading on a column using one or more of a clarifying treatment (such as filtration and/or centrifugation), one or more nucleases and/or proteases (to digest contaminating nucleic acids and/or proteins), an additionally chromatography step (such as affinity chromatography), a concentrating step, and the like.
  • a clarifying treatment such as filtration and/or centrifugation
  • nucleases and/or proteases to digest contaminating nucleic acids and/or proteins
  • an additionally chromatography step such as affinity chromatography
  • a concentrating step and the like.
  • AAV particles produced according to a method disclosed herein can be isolated using methods known in the art.
  • methods of isolating AAV particles produced according to a method disclosed herein comprises downstream processing such as, for example, harvest of a cell culture, clarification of the harvested cell culture (e.g., by centrifugation or depth filtration), tangential flow filtration, affinity chromatography, anion exchange chromatography, cation exchange chromatography, size exclusion chromatography, hydrophobic interaction chromatography, hydroxylapatite chromatography, sterile filtration, or any combination thereof.
  • downstream processing includes at least 2, at least 3, at least 4, at least 5, or at least 6 of: harvest of a cell culture, clarification of the harvested cell culture (e.g., by centrifugation or depth filtration), tangential flow filtration, affinity chromatography, anion exchange chromatography, cation exchange chromatography, size exclusion chromatography, hydrophobic interaction chromatography, hydroxylapatite chromatography, and sterile filtration.
  • an AAV of the present disclosure may contain one or more of the following: packaging cell materials (such as packaging cell proteins and/or DNA); plasmid DNA; helper virus; helper virus proteins; helper virus DNA; and media components including, for example, serum proteins, amino acids, transferrins, and other low molecular weight proteins.
  • packaging cell materials such as packaging cell proteins and/or DNA
  • plasmid DNA such as packaging cell proteins and/or DNA
  • helper virus such as packaging cell proteins and/or DNA
  • helper virus proteins helper virus proteins
  • helper virus DNA such as plasmid DNA
  • media components including, for example, serum proteins, amino acids, transferrins, and other low molecular weight proteins.
  • the production culture harvest is clarified by filtration through a series of depth filters including, for example, a grade DOHC Millipore Millistak+HC Pod Filter, a grade A1HC Millipore Millistak+HC Pod Filter, and/or a 0.2 pm Filter Opticap XL10 Millipore Express SHC Hydrophilic Membrane filter. Clarification can also be achieved by a variety of other standard techniques known in the art, such as, centrifugation or filtration through any suitable filter (such as any suitable cellulose acetate filter of 0.2 pm or greater pore size) known in the art. Still other suitable depth filters, e.g., in the range of about 0.045 pm to about 0.2 pm, or other filtration techniques may be used.
  • the AAV starting preparation can be treated with a nuclease, or a combination of nucleases, to digest any contaminating high molecular weight nucleic acid present in the production culture.
  • Suitable nucleases include but are not limited to a DNAse, e.g., Benzonase® digestion performed under standard conditions known in the art. For example, a final concentration of 1 unit/mL to 2.5 units/mL of Benzonase® is used at a temperature ranging from ambient temperature to 37°C for a period of 30 minutes to several hours, or about 2 hours. In another example, a turbonuclease is used.
  • nucleases may be selected to degrade single stranded DNA and/or double-stranded DNA, and RNA. Such steps may contain a single nuclease, or mixtures of nucleases directed to different targets, and may be endonucleases or exonucleases.
  • AAVs produced using a disclosed method may be isolated or purified prior to loading onto a column chromatography resin (e.g., an anion exchange column or a cation exchange column) using one or more of the following steps: tangential flow filtration for concentrating the AAV particles, heat inactivation of helper virus, AAV capture by hydrophobic interaction chromatography, affinity capture chromatography to remove production system contaminants, buffer exchange by size exclusion chromatography (SEC), and/or nanofiltration. These steps may be used alone, in various combinations, or in different orders. In some embodiments, the method comprises all the steps in the order.
  • a column chromatography resin e.g., an anion exchange column or a cation exchange column
  • a nuclease (e.g., Benzonase®)-treated mixture is concentrated via tangential flow filtration.
  • Large scale concentration of viruses using tangential flow filtration ultrafiltration has been described by R. Paul et al., Human Gene Therapy, 4:609-615 (1993).
  • Tangential flow filtration concentration of the AAV enables a technically manageable volume of the preparation to be subjected to the methods of the present disclosure and allows for more reasonable sizing of a solid support (such as beads, such as in a column).
  • the AAV is concentrated between at least two-fold or at least ten-fold.
  • the AAV is concentrated between at least ten-fold and at least twenty -fold, such as at least 10-fold, at least 11 -fold, at least 12-fold, at least 13 -fold, at least 14-fold, at least 15-fold, at least 16-fold, at least 17-fold, at least 18-fold, at least 19-fold, or at least 20-fold. In some embodiments, the AAV is concentrated between at least twenty-fold and at least fifty-fold.
  • tangential flow filtration can also be used at any step in the disclosed methods where it is desirable to exchange buffers before performing the next step in the method.
  • AAVs produced using a disclosed method have been separated from contaminants (e.g., packaging cell, viral, and other nucleic acid or proteinaceous materials which are present in the production culture or are by-products thereof) present from the production system.
  • contaminants e.g., packaging cell, viral, and other nucleic acid or proteinaceous materials which are present in the production culture or are by-products thereof
  • the AAVs separated from contaminants contain less than about 10% contamination from non- AAV viral and cellular proteinaceous and nucleic acid materials, or less than about 5% contaminants, or less than about 1% contaminating viral and cellular proteinaceous and nucleic acid materials.
  • the AAV e.g., in some examples, prior to loading onto a column, such as a cation or anion exchange column, for a further separation step
  • the AAV is about 95% to about 99% free of contaminants.
  • affinity capture chromatography may be used to separate AAVs from production system contaminants. This affinity capture can be performed, e.g., using an antibody-capture affinity resin.
  • a solid support is a cross-linked poly(styrene-divinylbenzene) having an average particle size of about 50 pm and having an AAV-specific antibody.
  • POROSTM high performance affinity resin commercially available from Thermo Fisher Scientific. The resin contains ligands created by a proprietary technology based on camelid- derived single-domain antibody fragments coupled to the resin via carbonyldiimidazole (CDI).
  • the ligand is a 13-kDa single-domain fragment that comprises the 3 CDRs that form the antigen binding domain and is efficiently produced by the yeast Saccharomyces cerevisiae in a production process free of animal components.
  • suitable affinity resins may be selected or designed which contain an AAV-specific antibody (such as an AAV5, AAV6, or AAV8 specific antibody), or other immunoglobulin construct which is an AAV-specific ligand.
  • Such solid supports may be any suitable polymeric matrix material, e.g., agarose, sepharose, sephadex, amongst others.
  • an AAV (such as an AAV that has undergone one or more processing steps as described herein) is diluted, such as in a buffer, e.g., prior to loading onto a chromatography column for further purification.
  • An AAV packaging cell culture of the present disclosure may yield a mixture of AAV full capsids, AAV empty capsids, and/or AAV partially empty capsids.
  • an AAV is produced using an AAV packaging cell culture, wherein the packaging cells are cultured in the presence of an antifoam agent prior to transfecting the cell culture with at least one nucleic acid encoding the AAV, such as at least one nucleic acid as described herein.
  • Adeno-associated virus AAV
  • AAV a member of the Parvovirus family, is a small, non-enveloped virus.
  • AAV particles comprise an AAV capsid composed of 60 capsid protein subunits that are each made up of VP1, VP2, and VP3 proteins. The VP1, VP2, and VP3 proteins are present in a predicted ratio of about 1 :1 :10 and have icosahedral symmetry.
  • the AAV capsid encloses a small, single-stranded DNA (ssDNA) genome of about 4.8 kilobases (kb).
  • the ssDNA AAV genome includes two open reading frames, Rep and Cap, flanked by two 145-base inverted terminal repeats (ITRs). These ITRs base pair to allow for synthesis of the complementary DNA strand.
  • Rep and Cap are translated to produce multiple distinct proteins (Rep78, Rep68, Rep52, Rep40, which play important roles in the AAV life cycle; and the VP1, VP2, and VP3 capsid proteins).
  • the AAV is a recombinant or engineered AAV.
  • the AAV is a pseudotyped AAV.
  • Use of a recombinant or engineered AAV enables insertion, deletion, or substitution of target DNA sequences into the genomes of mammalian cells.
  • AAV comprises a protein capsid surrounding and protecting a single-stranded DNA genome of approximately 4.8 kilobases (kb). Naso et al., BioDrugs. 2017; 31(4): 317-334.
  • AAV belongs to the parvovirus family and is dependent on co-infection with other viruses, mainly adenoviruses, in order to replicate.
  • the viral capsid is comprised of approximately 60 proteins arranged into an icosahedral structure with the capsid proteins in a molar ratio of 1 : 1 : 10 (VP1 :VP2:VP3).
  • the aap gene encodes the assembly-activating protein (AAP) in an alternate reading frame overlapping the cap gene. This nuclear protein is thought to provide a scaffolding function for capsid assembly, but may be nonessential in certain AAV serotypes.
  • Recombinant AAV which lacks viral DNA
  • rAAV is a protein-based nanoparticle engineered to traverse the cell membrane, where it can ultimately traffic and deliver a DNA cargo (comprised within the viral capsid) into the nucleus of a cell.
  • rAAV Recombinant AAV
  • ITR-flanked transgenes encoded within rAAV can form circular concatemers that persist as episomes in the nucleus of a transduced cell. Because recombinant episomal DNA does not integrate into host genomes, it will eventually be diluted over time as the cell undergoes repeated rounds of replication.
  • the AAV that is produced using the disclosed methods is, or is derived from, an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrhlO, AAVrh74, AAV9, AAV9P, AAV10, AAV11, AAV12, or Myo-AAV, or novel chimeras thereof.
  • the AAV is AAV5, AAV6, or AAV8. Methods disclosed herein can be used in the production of rAAV particles comprising a capsid protein from any AAV capsid serotype.
  • the rAAV particles comprise a capsid protein from an AAV capsid serotype selected from AAV1, AAV2, rAAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV14, AAV15 and AAV16, AAV.rh8, AAV.rhlO, AAV.rh20, AAV.rh39, AAV.Rh74, AAV.RHM4-1, AAV.hu37, AAV.Anc80, AAV.Anc80L65, AAV.7m8, AAV.PHP.B, AAV2.5, AAV2tYF, AAV3B, AAV.LK03, AAV.HSC1, AAV.HSC2, AAV.HSC3, AAV.HSC4, AAV.HSC5, AAV.HSC6, AAV.HSC7, AAV.HSC8, AAV.HSC9, AAV.HSC10
  • the rAAV particles comprise a capsid protein that is a derivative, modification, or pseudotype of AAV1, AAV2, rAAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV14, AAV15 and AAV16, AAV.rh8, AAV.rhlO, AAV.rh20, AAV.rh39, AAV.Rh74, AAV.RHM4-1, AAV.hu37, AAV.Anc80, AAV.Anc80L65, AAV.7m8, AAV.PHP.B, AAV2.5, AAV2tYF, AAV3B, AAV.LK03, AAV.HSC1, AAV.HSC2, AAV.HSC3, AAV.HSC4, AAV.HSC5, AAV.HSC6, AAV.HSC7, AAV.HSC8, AAV.HSC9, AAV.HSC10
  • the AAV particles comprise a capsid protein from an AAV capsid serotype selected from AAV 5, AAV6, or AAV8. In some embodiments, the AAV particles have an AAV capsid serotype of AAV5. In some embodiments, the AAV particles have an AAV capsid serotype of AAV6. In some embodiments, the AAV particles have an AAV capsid serotype of AAV8.
  • AAV6 has been shown to have enhanced capsid-associated tropism in tissues, such as the lungs, cardiac muscle, and skeletal muscle (Halbert et al., J Virol. 2001, 75(14):6615-24; Rengo et al., Circulation. 2009, 119( 1): 89-98; Bortolanza et al. Mol Ther. 2011, 19(11):2055-64). Like other AAVs, AAV6 can transduce non-dividing cells (Halbert et al., J Virol. 2001, 75(14):6615- 24). AAV5 exhibits enhanced capsid-associated tropism in the central nervous system, lung, photoreceptor cells, and retinal pigment epithelium.
  • AAV8 exhibits enhanced capsid- associated tropism in the central nervous system, photoreceptor cells, and retinal pigment epithelium, as well as the heart, liver, and skeletal muscle.
  • AAV5 and AAV8 vectors are also currently being explored for gene therapy applications in hemophilia B and hemophilia A, respectively. See, e.g., Issa et al., Cells. 2023, 12(5), 785.
  • the AAV (such as the AAV5, AAV6, or AAV8) is less than 5 kb from ITR to ITR in size, inclusive of both ITRs. In particular embodiments, the AAV (such as the AAV5, AAV6, or AAV8) is less than 4.9 kb from ITR to ITR in size, inclusive of both ITRs. In further embodiments, the AAV (such as the AAV5, AAV6, or AAV8) is less than 4.85 kb from ITR to ITR in size, inclusive of both ITRs. In further embodiments, the AAV (such as the AAV5, AAV6, or AAV8) is less than 4.8 kb from ITR to ITR in size, inclusive of both ITRs.
  • the AAV (such as the AAV5, AAV6, or AAV8) is less than 4.75 kb from ITR to ITR in size, inclusive of both ITRs. In further embodiments, the AAV (such as the AAV5, AAV6, or AAV8) is less than 4.7 kb from ITR to ITR in size, inclusive of both ITRs.
  • the AAV (such as the AAV5, AAV6, or AAV8) is 3.9-5 kb, 4-5 kb, 4.2-5 kb, 4.4-5 kb, 4.6-5 kb, 4.7-5 kb, 3.9-4.9 kb, 4.2-4.9 kb, 4.4-4.9 kb, 4.7-4.9 kb, 3.9-4.85 kb, 4.2- 4.85 kb, 4.4-4.85 kb, 4.6-4.85 kb, 4.7-4.85 kb, 4.7-4.9 kb, 3.9-4.8 kb, 4.2-4.8 kb, 4.4-4.8 kb or 4.6-4.8 kb from ITR to ITR in size, inclusive of both ITRs.
  • the AAV (such as the AAV5, AAV6, or AAV8) is 4.4-4.85 kb from ITR to ITR in size, inclusive of both ITRs.
  • the AAV (such as the AAV5, AAV6, or AAV8) comprises a nucleic acid molecule (e.g., enclosed within the fully assembled AAV capsid) that encodes at least one protein or RNA of interest, such as at least one protein or RNA of therapeutic interest.
  • the nucleic acid of interest to be packaged into the AAV capsid may encode a chimeric antigen receptor (CAR) or T cell receptor (TCR)).
  • the CAR comprises an antigen-binding domain and an intracellular signaling region comprising an intracellular signaling domain.
  • the antigen-binding domain is or comprises an antibody or an antibody fragment thereof, which optionally is a single chain fragment.
  • the fragment comprises an scFv.
  • the intracellular signaling domain can comprise a primary signaling domain, a signaling domain that is capable of inducing a primary activation signal in a T cell, a signaling domain of a T cell receptor (TCR) component, and/or a signaling domain comprising an immunoreceptor tyrosine-based activation motif.
  • the intracellular signaling domain is or comprises an intracellular signaling domain of a CD3 chain, optionally a CD3-zeta chain, or a signaling portion thereof.
  • the CAR can comprise a transmembrane domain disposed between the extracellular domain and the intracellular signaling region, which can further comprise a costimulatory signaling domain.
  • the costimulatory signaling domain can comprise an intracellular signaling domain of a T cell costimulatory molecule or a signaling portion thereof, such as an intracellular signaling domain of a CD28, a 4-1BB or an ICOS or a signaling portion thereof.
  • the costimulatory signaling domain is between the transmembrane domain and the intracellular signaling domain.
  • the TCR comprises an alpha chain containing a variable alpha (Va) region and a beta chain containing a variable beta (VP) region, wherein the TCR is capable of binding to or recognizing a peptide epitope in the context of an MHC molecule, such as HLA-A2.
  • a nucleic acid molecule of interest can be operably linked to one or more regulatory elements, such as a promoter, such as a tissue-specific promoter.
  • a promoter such as a tissue-specific promoter.
  • the term “operably linked” means that the nucleic acid molecule of interest is linked to regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence.
  • AAVs produced using the disclosed methods can include any AAV comprising a tissue-specific promoter in facilitating administration of gene therapy, which can include any known gene editing system in the art.
  • a promoter as described herein can also be “cell specific,” meaning that the particular promoter selected for the AAV can direct expression of the selected transgene/nucleotide sequence of interest in a particular cell or cell type.
  • An exemplary promoter useful in the present disclosure can be a constitutively active promoter (i.e., a promoter that is constitutively in an active/“ON” state), an activatable promoter, an inducible promoter (i.e., a promoter whose state, active/“ON” or inactive/ “OFF”, is controlled by an external stimulus, e.g., the presence of a particular temperature, compound, or protein), a spatially restricted promoter (i.e., transcriptional control element, enhancer, etc., e.g., a tissue specific promoter, a cell type specific promoter, etc ), or a temporally restricted promoter (i.e., the promoter is in the “ON” state or “OFF” state during specific stages of embryonic development or during specific stages of a biological process).
  • a constitutively active promoter i.e., a promoter that is constitutively in an active/“ON” state
  • an activatable promoter i.e., an induc
  • Suitable promoters can be derived from viruses (viral promoters), or they can be derived from any organism, including prokaryotic or eukaryotic organisms. Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • viruses viral promoters
  • Suitable promoters can be used to drive expression by any RNA polymerase (e.g., pol I, pol II, pol III).
  • Exemplary promoters include, but are not limited to the SV40 early promoter, mouse mammary tumor virus long terminal repeat (LTR) promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a Rous sarcoma virus (RSV) promoter, a human U6 small nuclear promoter (U6) (Miyagishi et al., Nature Biotechnology. 20: 497-500; 2002), an enhanced U6 promoter (e. ., Xia el al., Nucleic Acids Res. Sep 1(31): 17; 2003), a human Hl promoter (Hl), and the like.
  • LTR mouse mammary tumor virus long terminal repeat
  • Ad MLP adenovirus major late promoter
  • HSV herpes simplex virus
  • CMVIE cytomegalovirus
  • Nucleic acids of interest useful herein can also include other regulatory elements, i.e., transcriptional and translational control sequences, such as enhancers, polyadenylation signals, terminators, protein degradation signals, and the like, that provide for and/or regulate transcription of a non-coding sequence (e.g., guide RNA) or a coding sequence (e.g., site- directed modifying polypeptide, or Cas9 polypeptide) and/or regulate translation of an encoded polypeptide.
  • a non-coding sequence e.g., guide RNA
  • a coding sequence e.g., site- directed modifying polypeptide, or Cas9 polypeptide
  • Exemplary regulatory sequences are known in the art and are described, for example, in Goeddel, Gene Expression Technology: Methods in Enzymology. Vol. 185, Academic Press, San Diego, CA (1990).
  • Example 1 Use of Antifoam in an AAV6 suspension culture process to increase vector production
  • FIG. 1 The workflow described in this example is illustrated in FIG. 1.
  • This Example shows the use of an antifoam agent in an AAV6 suspension culture process to increase AAV vector production.
  • HEK293 cells were inoculated into an Eppendorf 1 L bioreactor containing a chemically defined HEK293 cell culture medium comprising 30 ppm, 60 ppm, or 0 ppm (control cultures) Ex-Cell® Antifoam (Sigma Aldrich) formulated from a 30% simethicone emulsion from Dow Coming (Q7-2587). 72 hours after inoculation, cell cultures were transfected (using a triple transient transfection method) with at least one nucleic acid encoding the AAV.
  • Ex-Cell® Antifoam Sigma Aldrich
  • a triple transfection three separate plasmids encoding (1) a heterologous nucleotide sequence (encoding a gene of interest) flanked by ITRs, (2) the AAV rep and AAV cap gene sequences, and (3) adenovirus helper function sequences (the Ad5 genes: VA RNAs, E2A, and E4OEF6), respectively, were mixed and incubated with a cationic polymer to form complexes, and were then introduced to the HEK293 cells in the bioreactor.
  • HEK293 cells constitutively express Ela/b, the fourth adenovirus function for AAV replication.
  • the HEK293 packaging cell cultures were lysed, and the lysate subsequently clarification using depth filtration.
  • Additional AAV6 concentration and purification methods included tangential flow filtration, affinity chromatography (Poros CaptureSelect AAVX), anion exchange chromatography (CIMmultus QA monolith), nanofiltration (Planova 35N, Asahi Kasei), additional tangential flow filtration, and 0.2um sterile-grade filtration.
  • Chroros CaptureSelect AAVX affinity chromatography
  • CCMmultus QA monolith anion exchange chromatography
  • nanofiltration Plantova 35N, Asahi Kasei
  • additional tangential flow filtration additional tangential flow filtration
  • 0.2um sterile-grade filtration Column chromatography was performed on an AKTA Avantl50 FPLC (Cytiva, Massachusetts, USA).
  • Viral genome titers were calculated using a qPCR method. Capsid titers were calculated by an ELISA assay. Percent full was determined as viral genome titer divided by capsid titer. [0143] As shown in FIG. 1, study 1 yielded an average titer increase of 40% and 27% for the 30 ppm and 60 ppm antifoam conditions, respectively, compared to the control.
  • the average viral genome titer (vg/mL) was 2.47xlO u vg/mL, 2.25xlO u vg/mL, and 1.67xlO n vg/mL for the 30 ppm, 60 ppm, and control (0 ppm) antifoam conditions, respectively.
  • Study 2 yielded a titer increase of 48% for the 30 ppm antifoam condition (3.27xlO n vg/mL) compared to the control (2.00xl0 n vg/mL).
  • the average viral genome titer (vg/mL) was 3.19xl0 n vg/mL, 3.45xlO n vg/mL, and 2.29xlO n vg/mL for the 30 ppm, 60 ppm, and control (0 ppm) antifoam conditions, respectively.
  • Example 2 Use of Antifoam in a HEK293 cell suspension culture process producing AAV5 to increase viral vector production
  • This Example shows the use of an antifoam agent in a suspension culture process producing AAV5 to increase AAV vector production at development scales.
  • HEK293 VPC 2.0 cells (ThermoFisher Scientific) were inoculated into a 250 mL Ambr bioreactor (Ambr250) or a 125 mL shaker flask (SF125) containing a HEK293 cell culture medium containing 5 ppm, 30 ppm, 60 ppm, or 0 ppm (control cultures) of Ex-Cell® Antifoam (Sigma Aldrich) formulated from a 30% simethicone emulsion from Dow Corning (Q7-2587).
  • Antifoam was added postinoculation (DO), at the time of plasmid transfection on day 3 (D3), or 24 hours post-transfection on day 4 (D4).
  • DO postinoculation
  • D3 time of plasmid transfection on day 3
  • D4 24 hours post-transfection on day 4
  • Cell cultures were transfected (using a triple transient transfection method) with at least one plasmid encoding the AAV.
  • HEK293 cells constitutively express Ela/b, the fourth adenovirus function for AAV replication.
  • vg Viral genome
  • Capsid titers were calculated using an ELISA assay.
  • antifoam addition increased viral genome (vg) titer at the SF125 and Ambr250 scale in all but the 30 ppm D4 SF125 condition. The highest observed titer was 3.3xl0 n vg/mL with 1.9 xlO 12 capsid/mL in SF125.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Manufacturing & Machinery (AREA)
  • Plant Pathology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des procédés de production d'un VAA. Dans certains modes de réalisation, le procédé comprend la culture de cellules d'encapsulation en présence d'un agent anti-mousse et la transfection de la culture cellulaire avec au moins un acide nucléique codant pour un VAA. Dans des modes de réalisation particuliers, le procédé comprend la culture de cellules d'encapsulation comprenant au moins un acide nucléique codant pour un VAA dans des conditions appropriées pour la production du VAA, les cellules d'encapsulation étant cultivées en présence d'un agent antimousse avant la transfection de la culture cellulaire avec le ou les acides nucléiques codant pour le VAA.
PCT/US2024/055351 2023-11-13 2024-11-11 Procédé de production de vaa Pending WO2025106374A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363598260P 2023-11-13 2023-11-13
US63/598,260 2023-11-13

Publications (1)

Publication Number Publication Date
WO2025106374A1 true WO2025106374A1 (fr) 2025-05-22

Family

ID=93655970

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/055351 Pending WO2025106374A1 (fr) 2023-11-13 2024-11-11 Procédé de production de vaa

Country Status (1)

Country Link
WO (1) WO2025106374A1 (fr)

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
WO2003042397A2 (fr) 2001-11-13 2003-05-22 The Trustees Of The University Of Pennsylvania Methode de detection et/ou d'identification de sequences de virus associes aux adenovirus (aav) et d'isolation de nouvelles sequences ainsi identifiees
WO2003052051A2 (fr) 2001-12-17 2003-06-26 The Trustees Of The University Of Pennsylvania Sequences du serotype 8 du virus associe a l'adenovirus (aav), vecteurs les contenant et utilisations correspondantes
US6723551B2 (en) 2001-11-09 2004-04-20 The United States Of America As Represented By The Department Of Health And Human Services Production of adeno-associated virus in insect cells
WO2005033321A2 (fr) 2003-09-30 2005-04-14 The Trustees Of The University Of Pennsylvania Variantes des virus associes aux adenovirus (aav), sequences, vecteurs les contenant, et leur utilisation
US6995006B2 (en) 1997-09-05 2006-02-07 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
WO2006068888A1 (fr) 2004-12-22 2006-06-29 Raytheon Company Systeme et technique d'etalonnage de reseaux de radars
WO2006110689A2 (fr) 2005-04-07 2006-10-19 The Trustees Of The University Of Pennsylvania Procede d'augmentation de la fonction d'un vecteur aav
WO2009104964A1 (fr) 2008-02-19 2009-08-27 Amsterdam Molecular Therapeutics B.V. Optimisation de l'expression de protéines rep et cap parvovirales dans des cellules d'insectes
WO2010127097A1 (fr) 2009-04-30 2010-11-04 The Trustees Of The University Of Pennsylvania Compositions pour cibler des cellules des voies respiratoires conductrices comprenant des constructions de virus adéno-associé
US20120122155A9 (en) 2009-05-12 2012-05-17 Transgene S.A. Immortalized avian cell lines and use thereof
US20130224836A1 (en) 2010-10-27 2013-08-29 Jichi Medical University Adeno-Associated Virus Virion for Gene Transfer to Nervous System Cells
US8628966B2 (en) 2010-04-30 2014-01-14 City Of Hope CD34-derived recombinant adeno-associated vectors for stem cell transduction and systemic therapeutic gene transfer
US8734809B2 (en) 2009-05-28 2014-05-27 University Of Massachusetts AAV's and uses thereof
US8927514B2 (en) 2010-04-30 2015-01-06 City Of Hope Recombinant adeno-associated vectors for targeted treatment
US20150111955A1 (en) 2012-02-17 2015-04-23 The Children's Hospital Of Philadelphia Aav vector compositions and methods for gene transfer to cells, organs and tissues
US20150126588A1 (en) 2012-05-09 2015-05-07 Oregon Health & Science University Adeno associated virus plasmids and vectors
US9169299B2 (en) 2011-08-24 2015-10-27 The Board Of Trustees Of The Leleand Stanford Junior University AAV capsid proteins for nucleic acid transfer
US9193956B2 (en) 2011-04-22 2015-11-24 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
WO2015191508A1 (fr) 2014-06-09 2015-12-17 Voyager Therapeutics, Inc. Capsides chimériques
US20150374803A1 (en) 2013-03-13 2015-12-31 The Children's Hospital Of Philadelphia Adeno-associated virus vectors and methods of use thereof
US20160215024A1 (en) 2013-10-11 2016-07-28 Massachusetts Eye & Ear Infirmary Methods of Predicting Ancestral Virus Sequences and Uses Thereof
US9409953B2 (en) 2011-02-10 2016-08-09 The University Of North Carolina At Chapel Hill Viral vectors with modified transduction profiles and methods of making and using the same
US20170067908A1 (en) 2014-04-25 2017-03-09 Oregon Health & Science University Methods of viral neutralizing antibody epitope mapping
US9783826B2 (en) 2008-01-29 2017-10-10 Applied Genetic Technologies Corporation Recombinant virus production using mammalian cells in suspension
WO2020033842A1 (fr) * 2018-08-10 2020-02-13 Regenxbio Inc. Procédé évolutif pour la production d'aav recombinants
WO2022032153A1 (fr) * 2020-08-06 2022-02-10 Voyager Therapeutics, Inc. Milieu de culture cellulaire destiné à être utilisé dans la production de produits de thérapie génique dans des bioréacteurs

Patent Citations (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6995006B2 (en) 1997-09-05 2006-02-07 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6723551B2 (en) 2001-11-09 2004-04-20 The United States Of America As Represented By The Department Of Health And Human Services Production of adeno-associated virus in insect cells
US8524446B2 (en) 2001-11-13 2013-09-03 The Trustees Of The University Of Pennsylvania Method for detecting adeno-associated virus
WO2003042397A2 (fr) 2001-11-13 2003-05-22 The Trustees Of The University Of Pennsylvania Methode de detection et/ou d'identification de sequences de virus associes aux adenovirus (aav) et d'isolation de nouvelles sequences ainsi identifiees
US9790472B2 (en) 2001-11-13 2017-10-17 The Trustees Of The University Of Pennsylvania Method of detecting and/or identifying adeno-associated virus (AAV) sequences and isolating novel sequences identified thereby
WO2003052051A2 (fr) 2001-12-17 2003-06-26 The Trustees Of The University Of Pennsylvania Sequences du serotype 8 du virus associe a l'adenovirus (aav), vecteurs les contenant et utilisations correspondantes
US7282199B2 (en) 2001-12-17 2007-10-16 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) serotype 8 sequences, vectors containing same, and uses therefor
WO2005033321A2 (fr) 2003-09-30 2005-04-14 The Trustees Of The University Of Pennsylvania Variantes des virus associes aux adenovirus (aav), sequences, vecteurs les contenant, et leur utilisation
US7906111B2 (en) 2003-09-30 2011-03-15 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clades, sequences, vectors containing same, and uses therefor
WO2006068888A1 (fr) 2004-12-22 2006-06-29 Raytheon Company Systeme et technique d'etalonnage de reseaux de radars
WO2006110689A2 (fr) 2005-04-07 2006-10-19 The Trustees Of The University Of Pennsylvania Procede d'augmentation de la fonction d'un vecteur aav
US8999678B2 (en) 2005-04-07 2015-04-07 The Trustees Of The University Of Pennsylvania Method of increasing the function of an AAV vector
US9783826B2 (en) 2008-01-29 2017-10-10 Applied Genetic Technologies Corporation Recombinant virus production using mammalian cells in suspension
WO2009104964A1 (fr) 2008-02-19 2009-08-27 Amsterdam Molecular Therapeutics B.V. Optimisation de l'expression de protéines rep et cap parvovirales dans des cellules d'insectes
WO2010127097A1 (fr) 2009-04-30 2010-11-04 The Trustees Of The University Of Pennsylvania Compositions pour cibler des cellules des voies respiratoires conductrices comprenant des constructions de virus adéno-associé
US20120122155A9 (en) 2009-05-12 2012-05-17 Transgene S.A. Immortalized avian cell lines and use thereof
US9284357B2 (en) 2009-05-28 2016-03-15 University Of Massachusetts AAV's and uses thereof
US8734809B2 (en) 2009-05-28 2014-05-27 University Of Massachusetts AAV's and uses thereof
US8927514B2 (en) 2010-04-30 2015-01-06 City Of Hope Recombinant adeno-associated vectors for targeted treatment
US8628966B2 (en) 2010-04-30 2014-01-14 City Of Hope CD34-derived recombinant adeno-associated vectors for stem cell transduction and systemic therapeutic gene transfer
US20130224836A1 (en) 2010-10-27 2013-08-29 Jichi Medical University Adeno-Associated Virus Virion for Gene Transfer to Nervous System Cells
US9409953B2 (en) 2011-02-10 2016-08-09 The University Of North Carolina At Chapel Hill Viral vectors with modified transduction profiles and methods of making and using the same
US9587282B2 (en) 2011-04-22 2017-03-07 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
US9193956B2 (en) 2011-04-22 2015-11-24 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
US9458517B2 (en) 2011-04-22 2016-10-04 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
US9169299B2 (en) 2011-08-24 2015-10-27 The Board Of Trustees Of The Leleand Stanford Junior University AAV capsid proteins for nucleic acid transfer
US20150111955A1 (en) 2012-02-17 2015-04-23 The Children's Hospital Of Philadelphia Aav vector compositions and methods for gene transfer to cells, organs and tissues
US20150126588A1 (en) 2012-05-09 2015-05-07 Oregon Health & Science University Adeno associated virus plasmids and vectors
US20150374803A1 (en) 2013-03-13 2015-12-31 The Children's Hospital Of Philadelphia Adeno-associated virus vectors and methods of use thereof
US20160215024A1 (en) 2013-10-11 2016-07-28 Massachusetts Eye & Ear Infirmary Methods of Predicting Ancestral Virus Sequences and Uses Thereof
US20170051257A1 (en) 2013-10-11 2017-02-23 Massachusetts Eye And Ear Infirmary Methods of predicting ancestral virus sequences and uses thereof
US20170067908A1 (en) 2014-04-25 2017-03-09 Oregon Health & Science University Methods of viral neutralizing antibody epitope mapping
WO2015191508A1 (fr) 2014-06-09 2015-12-17 Voyager Therapeutics, Inc. Capsides chimériques
WO2020033842A1 (fr) * 2018-08-10 2020-02-13 Regenxbio Inc. Procédé évolutif pour la production d'aav recombinants
WO2022032153A1 (fr) * 2020-08-06 2022-02-10 Voyager Therapeutics, Inc. Milieu de culture cellulaire destiné à être utilisé dans la production de produits de thérapie génique dans des bioréacteurs

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Tissue Culture", 1973, ACADEMIC PRESS
ALMEIDA MAURO ET AL: "Poloxamers, poloxamines and polymeric micelles: Definition, structure and therapeutic applications in cancer", JOURNAL OF POLYMER RESEARCH, SPRINGER NETHERLANDS, DORDRECHT, vol. 25, no. 1, 30 December 2017 (2017-12-30), pages 1 - 14, XP036399648, ISSN: 1022-9760, [retrieved on 20171230], DOI: 10.1007/S10965-017-1426-X *
BORTOLANZA ET AL., MOL THER, vol. 19, no. 11, 2011, pages 2055 - 64
GOEDDEL: "Gene Expression Technology: Methods in Enzymology", vol. 185, 1990, ACADEMIC PRESS
HALBERT ET AL., J VIROL, vol. 75, no. 14, 2001, pages 6615 - 24
ISSA ET AL., CELLS, vol. 12, no. 5, 2023, pages 785
KHAN N ET AL., CANCER MED, vol. 9, no. 9, 2020, pages 3188 - 3201
MA W ET AL., HUM GENE THER, vol. 22, no. 5, 2011, pages 633 - 40
MA W ET AL., HUM GENE THER., vol. 22, no. 5, 2011, pages 633 - 40
MCCARTY ET AL., GENE THER., vol. 8, 2001, pages 1248 - 54
MEIERRIEKS FREDERIK ET AL: "A robust and efficient alluvial filtration method for the clarification of adeno-associated viruses from crude cell lysates", JOURNAL OF BIOTECHNOLOGY, vol. 367, 1 April 2023 (2023-04-01), Amsterdam NL, pages 31 - 41, XP093246381, ISSN: 0168-1656, Retrieved from the Internet <URL:https://pdf.sciencedirectassets.com/271109/1-s2.0-S0168165623X00066/1-s2.0-S0168165623000627/main.pdf?hash=3663d4cb61ed8296f35d7324473a162b4c93f72c7920e0a7167c8f9d2ddb7897&host=68042c943591013ac2b2430a89b270f6af2c76d8dfd086a07176afe7c76c2c61&pii=S0168165623000627&tid=spdf-c0e103d9-908b-4b24-ac18-54d> DOI: 10.1016/j.jbiotec.2023.03.010 *
MIYAGISHI ET AL., NATURE BIOTECHNOLOGY, vol. 20, 2002, pages 497 - 500
NASO ET AL., BIODRUGS, vol. 31, no. 4, 2017, pages 317 - 334
R. I. FRESHNEY: "Culture of animal cells: A manual of basic technique", 2000, WILEY-LISS INC
R. PAUL ET AL., HUMAN GENE THERAPY, vol. 4, 1993, pages 609 - 615
RENGO ET AL., CIRCULATION, vol. 119, no. 1, 2009, pages 89 - 98
RITACCO FRANK V ET AL: "Cell culture media for recombinant protein expression in Chinese hamster ovary (CHO) cells: History, key components, and optimization strategies", BIOTECHNOLOGY PROGRESS, WILEY-BLACKWELL PUBLISHING, INC, US, vol. 34, no. 6, 1 November 2018 (2018-11-01), pages 1407 - 1426, XP002799102, ISSN: 1520-6033, DOI: 10.1002/BTPR.2706 *
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", vol. 3, 16 March 2016, COLD SPRING HARBOR PRESS, pages: 16002
SARAH J ROUTLEDGE: "BEYOND DE-FOAMING: THE EFFECTS OF ANTIFOAMS ON BIOPROCESS PRODUCTIVITY", COMPUTATIONAL AND STRUCTURAL BIOTECHNOLOGY JOURNAL, vol. 3, no. 4, 1 October 2012 (2012-10-01), pages 1 - 7, XP055218287, ISSN: 2001-0370, DOI: 10.5936/csbj.201210014 *
TABEBORDBAR ET AL., CELL, vol. 184, 2021, pages 1 - 20
VINEY, J VIROL, vol. 95, no. 7, 2021, pages 02023 - 20
WEINMANN ET AL., NATURE COMMUNICATIONS, vol. 11, 2020, pages 5432
XIA ET AL., NUCLEIC ACIDS RES., vol. 1, no. 31, September 2003 (2003-09-01), pages 17
ZHANG S ET AL: "Foaming and media surfactant effects on the cultivation of animal cells in stirred and sparged bioreactors", JOURNAL OF BIOTECHNOLOGY, ELSEVIER, AMSTERDAM NL, vol. 25, no. 3, 1 September 1992 (1992-09-01), pages 289 - 306, XP024346500, ISSN: 0168-1656, [retrieved on 19920901], DOI: 10.1016/0168-1656(92)90162-3 *

Similar Documents

Publication Publication Date Title
US9441206B2 (en) Cell line for production of adeno-associated virus
ES3017247T3 (en) Method for the production of aav
WO2009154452A1 (fr) Capside parvovirale avec région de répétition gly-ala incorporée
WO2021198508A1 (fr) Vecteurs bifonctionnels doubles pour la production d&#39;aav
WO2023060113A1 (fr) Compositions et procédés de production d&#39;aav recombinants
KR20230011935A (ko) 아데노 연관 바이러스 입자 또는 아데노바이러스를 정제하기 위한 방법 및 조성물
US20240279682A1 (en) AAV Manufacturing Methods
WO2025106374A1 (fr) Procédé de production de vaa
WO2024211780A1 (fr) Compositions et procédés pour la production d&#39;aav recombiné
WO2024013239A1 (fr) Procédé de production de particules de virus adéno-associé recombinant
RU2850334C1 (ru) Способ получения aav
AU2020247133B2 (en) Methods for the manufacture of recombinant viral vectors
WO2025171001A1 (fr) Compositions et procédés pour la production améliorée de vecteurs viraux aav
AU2022360254A1 (en) Compositions and methods for recombinant aav production
WO2025090962A1 (fr) Compositions et procédés pour la production d&#39;aav recombiné
WO2024243563A1 (fr) Procédé de purification d&#39;aav
CN120835797A (zh) 用于产生重组aav颗粒制备物的方法
CN118974274A (zh) 用于确定aav基因组的方法
CN115851837A (zh) 一种提高杆状病毒系统生产腺相关病毒的方法及应用

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: 24813306

Country of ref document: EP

Kind code of ref document: A1